1
|
Idris AL, Fan X, Li W, Pei H, Muhammad MH, Guan X, Huang T. Galactose-1-phosphate uridylyltransferase GalT promotes biofilm formation and enhances UV-B resistance of Bacillus thuringiensis. World J Microbiol Biotechnol 2024; 40:383. [PMID: 39551829 DOI: 10.1007/s11274-024-04195-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/06/2024] [Indexed: 11/19/2024]
Abstract
Ultraviolet radiation (UV) is a major abiotic stress resulting in relative short duration of Bacillus thuringiensis (Bt) biopesticides in the field, which is expected to be solved by formation of Bt biofilm with higher UV resistance. Therefore, one of the important prerequisite works is to clarify the functions of biofilm-associated genes on biofilm formation and UV resistance of Bt. In this study, comparative genomics and bioinformatic analysis indicated that BTXL6_19475 gene involved in biofilm formation of Bt XL6 was likely to encode a galactose-1-phosphate uridylyltransferase (GalT, E.C. 2.7.7.12). Heterologous expression of the BTXL6_19475 gene in Escherichia coli and detection of its GalT enzyme activity in vitro proved that the gene did encode GalT. Comparing the wild type Bt strain XL6 with galT gene knockout mutant Bt XL6ΔgalT and its complementary strain Bt XL6ΔgalT::19,475, GalT promoted the biofilm formation and enhanced the UV-B resistance of Bt XL6 likely by increasing its D-ribose production and reducing its alanine aryldamidase activity. GalT did not affect the growth and the cell motility of Bt XL6. A regulation map had been proposed to elucidate how GalT promoted biofilm formation and enhanced UV-B resistance of Bt XL6 by the cross-talk between Leloir pathway, Embden-Meyerhof glycolysis pathway and pentose phosphate pathway. Our finding provides a theoretical basis for the efficient use of biofilm genes to improve the UV resistance of Bt biofilms and thus extend field duration of Bt formulations based on biofilm engineering.
Collapse
Affiliation(s)
- Aisha Lawan Idris
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops & Key Laboratory of Biopesticide and Chemical Biology of Ministry of Education & Biopesticide Research Center, College of Life Sciences & College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Xiao Fan
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops & Key Laboratory of Biopesticide and Chemical Biology of Ministry of Education & Biopesticide Research Center, College of Life Sciences & College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Wen Li
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops & Key Laboratory of Biopesticide and Chemical Biology of Ministry of Education & Biopesticide Research Center, College of Life Sciences & College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Hankun Pei
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops & Key Laboratory of Biopesticide and Chemical Biology of Ministry of Education & Biopesticide Research Center, College of Life Sciences & College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Musa Hassan Muhammad
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops & Key Laboratory of Biopesticide and Chemical Biology of Ministry of Education & Biopesticide Research Center, College of Life Sciences & College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Xiong Guan
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops & Key Laboratory of Biopesticide and Chemical Biology of Ministry of Education & Biopesticide Research Center, College of Life Sciences & College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Tianpei Huang
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops & Key Laboratory of Biopesticide and Chemical Biology of Ministry of Education & Biopesticide Research Center, College of Life Sciences & College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China.
| |
Collapse
|
2
|
Li K. Heterologous expression of a novel galactose-1-phosphate uridylyltransferase from Thermodesulfatator indicus and its application for bioproduction of Gal-β-1,4-GlcNAc-X. Protein Expr Purif 2024; 222:106538. [PMID: 38950762 DOI: 10.1016/j.pep.2024.106538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/28/2024] [Accepted: 06/28/2024] [Indexed: 07/03/2024]
Abstract
Nucleotide sugars (UDP-Sugars) are essential for the production of polysaccharides and glycoconjugates utilized in medicines, cosmetics, and food industries. The enzyme Galactose-1-phosphate uridylyltransferase (GalU; EC 2.7.7.12) is responsible for the synthesis of UDP-galactose from α-d-galactose-1-phosphate (Gal-1P) and UTP. A novel bacterial GalU (TiGalU) encoded from a thermophilic bacterium, Thermodesulfatator indicus, was successfully purified using the Ni-NTA column after being expressed in Escherichia coli. The optimal pH for recombinant TiGalU was determined to be 5.5. The optimum temperature of the enzyme was 45 °C. The activity of TiGalU was not dependent on Mg2+ and was strongly inhibited by SDS. When coupled with galactose kinase (GALK1) and β-1,4-galactosyltransferase 1 (B4GALT1), the enzyme enabled the one-pot synthesis of Gal-β-1,4-GlcNAc-X by utilizing galactose and UTP as substrates. This study reported the in vitro biosynthesis of Gal-β-1,4-GlcNAc-X for the first time, providing an environmentally friendly way to biosynthesis glycosides and other polysaccharides.
Collapse
Affiliation(s)
- Kaiqi Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| |
Collapse
|
3
|
Panis B, Vos EN, Barić I, Bosch AM, Brouwers MCGJ, Burlina A, Cassiman D, Coman DJ, Couce ML, Das AM, Demirbas D, Empain A, Gautschi M, Grafakou O, Grunewald S, Kingma SDK, Knerr I, Leão-Teles E, Möslinger D, Murphy E, Õunap K, Pané A, Paci S, Parini R, Rivera IA, Scholl-Bürgi S, Schwartz IVD, Sdogou T, Shakerdi LA, Skouma A, Stepien KM, Treacy EP, Waisbren S, Berry GT, Rubio-Gozalbo ME. Brain function in classic galactosemia, a galactosemia network (GalNet) members review. Front Genet 2024; 15:1355962. [PMID: 38425716 PMCID: PMC10902464 DOI: 10.3389/fgene.2024.1355962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 01/24/2024] [Indexed: 03/02/2024] Open
Abstract
Classic galactosemia (CG, OMIM #230400, ORPHA: 79,239) is a hereditary disorder of galactose metabolism that, despite treatment with galactose restriction, affects brain function in 85% of the patients. Problems with cognitive function, neuropsychological/social emotional difficulties, neurological symptoms, and abnormalities in neuroimaging and electrophysiological assessments are frequently reported in this group of patients, with an enormous individual variability. In this review, we describe the role of impaired galactose metabolism on brain dysfunction based on state of the art knowledge. Several proposed disease mechanisms are discussed, as well as the time of damage and potential treatment options. Furthermore, we combine data from longitudinal, cross-sectional and retrospective studies with the observations of specialist teams treating this disease to depict the brain disease course over time. Based on current data and insights, the majority of patients do not exhibit cognitive decline. A subset of patients, often with early onset cerebral and cerebellar volume loss, can nevertheless experience neurological worsening. While a large number of patients with CG suffer from anxiety and depression, the increased complaints about memory loss, anxiety and depression at an older age are likely multifactorial in origin.
Collapse
Affiliation(s)
- Bianca Panis
- Department of Pediatrics, MosaKids Children’s Hospital, Maastricht University Medical Centre, Maastricht, Netherlands
- European Reference Network for Hereditary Metabolic Disorders (MetabERN) Member, Padova, Italy
- United for Metabolic Diseases (UMD), Amsterdam, Netherlands
| | - E. Naomi Vos
- Department of Pediatrics, MosaKids Children’s Hospital, Maastricht University Medical Centre, Maastricht, Netherlands
- European Reference Network for Hereditary Metabolic Disorders (MetabERN) Member, Padova, Italy
- United for Metabolic Diseases (UMD), Amsterdam, Netherlands
- Department of Clinical Genetics, Maastricht University Medical Centre, Maastricht, Netherlands
- GROW School for Oncology and Reproduction, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Ivo Barić
- Department of Pediatrics, University Hospital Center Zagreb, Croatia, and School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Annet M. Bosch
- European Reference Network for Hereditary Metabolic Disorders (MetabERN) Member, Padova, Italy
- United for Metabolic Diseases (UMD), Amsterdam, Netherlands
- Department of Pediatrics, Division of Metabolic Diseases, Emma Children’s Hospital, Amsterdam University Medical Center, Amsterdam Gastroenterology Endocrinology Metabolism, Inborn Errors of Metabolism, Amsterdam, Netherlands
| | - Martijn C. G. J. Brouwers
- European Reference Network for Hereditary Metabolic Disorders (MetabERN) Member, Padova, Italy
- Department of Internal Medicine, Division of Endocrinology and Metabolic Disease, Maastricht University Medical Centre, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands
| | - Alberto Burlina
- European Reference Network for Hereditary Metabolic Disorders (MetabERN) Member, Padova, Italy
- Division of Inherited Metabolic Diseases, Reference Centre Expanded Newborn Screening, University Hospital Padova, Padova, Italy
| | - David Cassiman
- Laboratory of Hepatology, Department of Chronic Diseases, Metabolism and Ageing, Faculty of Medicine, KU Leuven, Leuven, Belgium
| | - David J. Coman
- Queensland Children’s Hospital, Children’s Health Queensland, Brisbane, QLD, Australia
| | - María L. Couce
- European Reference Network for Hereditary Metabolic Disorders (MetabERN) Member, Padova, Italy
- Department of Pediatrics, Diagnosis and Treatment Unit of Congenital Metabolic Diseases, University Clinical Hospital of Santiago de Compostela, IDIS-Health Research Institute of Santiago de Compostela, CIBERER, RICORS Instituto Salud Carlos III, Santiago de Compostela, Spain
| | - Anibh M. Das
- European Reference Network for Hereditary Metabolic Disorders (MetabERN) Member, Padova, Italy
- Department of Paediatrics, Pediatric Metabolic Medicine, Hannover Medical School, Hannover, Germany
| | - Didem Demirbas
- Division of Genetics and Genomics, Boston Children’s Hospital, Harvard Medical School, Manton Center for Orphan Disease Research, Boston, MA, United States
| | - Aurélie Empain
- European Reference Network for Hereditary Metabolic Disorders (MetabERN) Member, Padova, Italy
- Department of Paediatrics, Metabolic and Nutrition Unit, Division of Endocrinology, Diabetes and Metabolism, University Hospital for Children Queen Fabiola, Bruxelles, Belgium
| | - Matthias Gautschi
- Department of Paediatrics, Institute of Clinical Chemistry, Inselspital, Bern University Hospital, Swiss Reference Centre for Inborn Errors of Metabolism, Site Bern, Division of Pediatric Endocrinology, Diabetes and Metabolism, University of Bern, Bern, Switzerland
| | - Olga Grafakou
- European Reference Network for Hereditary Metabolic Disorders (MetabERN) Member, Padova, Italy
- IEM Clinic, Arch Makarios III Hospital, Nicosia, Cyprus
| | - Stephanie Grunewald
- Metabolic Unit Great Ormond Street Hospital and Institute for Child Health, University College London, London, United Kingdom
| | - Sandra D. K. Kingma
- European Reference Network for Hereditary Metabolic Disorders (MetabERN) Member, Padova, Italy
- Centre for Metabolic Diseases, University Hospital Antwerp, University of Antwerp, Antwerp, Belgium
| | - Ina Knerr
- National Centre for Inherited Metabolic Disorders, Children’s Health Ireland at Temple Street, University College Dublin, Dublin, Ireland
| | - Elisa Leão-Teles
- European Reference Network for Hereditary Metabolic Disorders (MetabERN) Member, Padova, Italy
- Reference Centre of Inherited Metabolic Diseases, Centro Hospitalar Universitário São João, Porto, Portugal
| | - Dorothea Möslinger
- European Reference Network for Hereditary Metabolic Disorders (MetabERN) Member, Padova, Italy
- Department of Paediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Elaine Murphy
- Charles Dent Metabolic Unit, National Hospital for Neurology and Neurosurgery (NHNN), London, United Kingdom
| | - Katrin Õunap
- European Reference Network for Hereditary Metabolic Disorders (MetabERN) Member, Padova, Italy
- Genetics and Personalized Medicine Clinic, Faculty of Medicine, Tartu University Hospital, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Adriana Pané
- European Reference Network for Hereditary Metabolic Disorders (MetabERN) Member, Padova, Italy
- Endocrinology and Nutrition Department, Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Sabrina Paci
- European Reference Network for Hereditary Metabolic Disorders (MetabERN) Member, Padova, Italy
- Inborn Errors of Metabolism, Clinical Department of Pediatrics, San Paolo Hospital - ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Rossella Parini
- European Reference Network for Hereditary Metabolic Disorders (MetabERN) Member, Padova, Italy
- Rare Diseases Unit, Department of Internal Medicine, San Gerardo Hospital IRCCS, Monza, Italy
| | - Isabel A. Rivera
- iMed.ULisboa–Instituto de Investigação do Medicamento, Faculdade de Farmácia, Universidade de Lisboa, Lisboa, Portugal
| | - Sabine Scholl-Bürgi
- Department of Child and Adolescent Health, Division of Pediatrics I-Inherited Metabolic Disorders, Medical University Innsbruck, Innsbruck, Austria
| | - Ida V. D. Schwartz
- Medical Genetics Service, Hospital de Clinicas de Porto Alegre, Porto Alegre, Brazil
| | - Triantafyllia Sdogou
- European Reference Network for Hereditary Metabolic Disorders (MetabERN) Member, Padova, Italy
- Newborn Screening Department, Institute of Child Health, Athens, Greece
| | - Loai A. Shakerdi
- Adult Metabolics/Genetics, National Centre for Inherited Metabolic Disorders, The Mater Misericordiae University Hospital, Dublin, Ireland
| | - Anastasia Skouma
- European Reference Network for Hereditary Metabolic Disorders (MetabERN) Member, Padova, Italy
- Newborn Screening Department, Institute of Child Health, Athens, Greece
| | - Karolina M. Stepien
- Salford Royal Organisation, Northern Care Alliance NHS Foundation Trust, Salford, United Kingdom
| | - Eileen P. Treacy
- School of Medicine, Trinity College Dublin, National Rare Diseases Office, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Susan Waisbren
- Division of Genetics and Genomics, Boston Children’s Hospital, Harvard Medical School, Manton Center for Orphan Disease Research, Boston, MA, United States
| | - Gerard T. Berry
- Division of Genetics and Genomics, Boston Children’s Hospital, Harvard Medical School, Manton Center for Orphan Disease Research, Boston, MA, United States
| | - M. Estela Rubio-Gozalbo
- Department of Pediatrics, MosaKids Children’s Hospital, Maastricht University Medical Centre, Maastricht, Netherlands
- European Reference Network for Hereditary Metabolic Disorders (MetabERN) Member, Padova, Italy
- United for Metabolic Diseases (UMD), Amsterdam, Netherlands
- Department of Clinical Genetics, Maastricht University Medical Centre, Maastricht, Netherlands
- GROW School for Oncology and Reproduction, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
4
|
Forte G, Buonadonna AL, Pantaleo A, Fasano C, Capodiferro D, Grossi V, Sanese P, Cariola F, De Marco K, Lepore Signorile M, Manghisi A, Guglielmi AF, Simonetti S, Laforgia N, Disciglio V, Simone C. Classic Galactosemia: Clinical and Computational Characterization of a Novel GALT Missense Variant (p.A303D) and a Literature Review. Int J Mol Sci 2023; 24:17388. [PMID: 38139222 PMCID: PMC10744227 DOI: 10.3390/ijms242417388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/30/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
Classic galactosemia is an autosomal recessive inherited liver disorder of carbohydrate metabolism caused by deficient activity of galactose-1-phosphate uridylyltransferase (GALT). While a galactose-restricted diet is lifesaving, most patients still develop long-term complications. In this study, we report on a two-week-old female patient who is a compound heterozygote for a known pathogenic variant (p.K285N) and a novel missense variant (p.A303D) in the GALT gene. Segregation analysis showed that the patient inherited the p.K285N pathogenic variant from her father and the p.A303D variant from her mother. A bioinformatics analysis to predict the impact of the p.A303D missense variant on the structure and stability of the GALT protein revealed that it may be pathogenic. Based on this finding, we performed a literature review of all GALT missense variants identified in homozygous and compound heterozygous galactosemia patients carrying the p.K285N pathogenic variant to explore their molecular effects on the clinical phenotype of the disease. Our analysis revealed that these missense variants are responsible for a wide range of molecular defects. This study expands the clinical and mutational spectrum in classic galactosemia and reinforces the importance of understanding the molecular consequences of genetic variants to incorporate genetic analysis into clinical care.
Collapse
Affiliation(s)
- Giovanna Forte
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (A.L.B.); (A.P.); (C.F.); (V.G.); (P.S.); (F.C.); (K.D.M.); (M.L.S.); (A.M.); (A.F.G.)
| | - Antonia Lucia Buonadonna
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (A.L.B.); (A.P.); (C.F.); (V.G.); (P.S.); (F.C.); (K.D.M.); (M.L.S.); (A.M.); (A.F.G.)
| | - Antonino Pantaleo
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (A.L.B.); (A.P.); (C.F.); (V.G.); (P.S.); (F.C.); (K.D.M.); (M.L.S.); (A.M.); (A.F.G.)
| | - Candida Fasano
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (A.L.B.); (A.P.); (C.F.); (V.G.); (P.S.); (F.C.); (K.D.M.); (M.L.S.); (A.M.); (A.F.G.)
| | - Donatella Capodiferro
- Section of Neonatology and Neonatal Intensive Care Unit, Department of Interdisciplinary Medicine, “Aldo Moro” University of Bari, 70121 Bari, Italy; (D.C.); (N.L.)
| | - Valentina Grossi
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (A.L.B.); (A.P.); (C.F.); (V.G.); (P.S.); (F.C.); (K.D.M.); (M.L.S.); (A.M.); (A.F.G.)
| | - Paola Sanese
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (A.L.B.); (A.P.); (C.F.); (V.G.); (P.S.); (F.C.); (K.D.M.); (M.L.S.); (A.M.); (A.F.G.)
| | - Filomena Cariola
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (A.L.B.); (A.P.); (C.F.); (V.G.); (P.S.); (F.C.); (K.D.M.); (M.L.S.); (A.M.); (A.F.G.)
| | - Katia De Marco
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (A.L.B.); (A.P.); (C.F.); (V.G.); (P.S.); (F.C.); (K.D.M.); (M.L.S.); (A.M.); (A.F.G.)
| | - Martina Lepore Signorile
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (A.L.B.); (A.P.); (C.F.); (V.G.); (P.S.); (F.C.); (K.D.M.); (M.L.S.); (A.M.); (A.F.G.)
| | - Andrea Manghisi
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (A.L.B.); (A.P.); (C.F.); (V.G.); (P.S.); (F.C.); (K.D.M.); (M.L.S.); (A.M.); (A.F.G.)
| | - Anna Filomena Guglielmi
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (A.L.B.); (A.P.); (C.F.); (V.G.); (P.S.); (F.C.); (K.D.M.); (M.L.S.); (A.M.); (A.F.G.)
| | - Simonetta Simonetti
- Clinical Pathology and Neonatal Screening, Azienda Ospedaliera Universitaria Policlinico-Giovanni XXIII, 70124 Bari, Italy;
| | - Nicola Laforgia
- Section of Neonatology and Neonatal Intensive Care Unit, Department of Interdisciplinary Medicine, “Aldo Moro” University of Bari, 70121 Bari, Italy; (D.C.); (N.L.)
| | - Vittoria Disciglio
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (A.L.B.); (A.P.); (C.F.); (V.G.); (P.S.); (F.C.); (K.D.M.); (M.L.S.); (A.M.); (A.F.G.)
| | - Cristiano Simone
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (A.L.B.); (A.P.); (C.F.); (V.G.); (P.S.); (F.C.); (K.D.M.); (M.L.S.); (A.M.); (A.F.G.)
- Medical Genetics, Department of Precision and Regenerative Medicine and Jonic Area (DiMePRe-J), University of Bari Aldo Moro, 70124 Bari, Italy
| |
Collapse
|
5
|
Delwing-de Lima D, Sasso S, Delwing-Dal Magro D, Pereira NR, Rodrigues AF, Schmitz F, Manoel Pereira E, Schramm do Nascimento MA, Wyse ATS. In vitro galactose impairs energy metabolism in the brain of young rats: protective role of antioxidants. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2023; 42:967-985. [PMID: 37317977 DOI: 10.1080/15257770.2023.2222776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 06/04/2023] [Indexed: 06/16/2023]
Abstract
We, herein, investigated the in vitro effects of galactose on the activity of pyruvate kinase, succinate dehydrogenase (SDH), complex II and IV (cytochrome c oxidase) of the respiratory chain and Na+K+-ATPase in the cerebral cortex, cerebellum and hippocampus of 30-day-old rats. We also determined the influence of the antioxidants, trolox, ascorbic acid and glutathione, on the effects elicited by galactose. Galactose was added to the assay at concentrations of 0.1, 3.0, 5.0 and 10.0 mM. Control experiments were performed without galactose. Galactose, at 3.0, 5.0 and 10.0 mM, decreased pyruvate kinase activity in the cerebral cortex and at 10.0 mM in the hippocampus. Galactose, at 10.0 mM, reduced SDH and complex II activities in the cerebellum and hippocampus, and reduced cytochrome c oxidase activity in the hippocampus. Additionally, decreased Na+K+-ATPase activity in the cerebral cortex and hippocampus; conversely, galactose, at 3.0 and 5.0 mM, increased this enzyme's activity in the cerebellum. Data show that galactose disrupts energy metabolism and trolox, ascorbic acid and glutathione addition prevented the majority of alterations in the parameters analyzed, suggesting the use of antioxidants as an adjuvant therapy in Classic galactosemia.
Collapse
Affiliation(s)
- Daniela Delwing-de Lima
- Department of Medicine, University of Joinville Region, Joinville, Santa Catarina, Brazil
- Post-Graduation Program in Health and Environment, University of Joinville Region, Joinville, Santa Catarina, Brazil
| | - Simone Sasso
- Post-Graduation Program in Health and Environment, University of Joinville Region, Joinville, Santa Catarina, Brazil
| | - Débora Delwing-Dal Magro
- Department of Natural Sciences, Center of Exact and Natural Sciences, Regional University of Blumenau, Blumenau, Santa Catarina, Brazil
| | - Nariana Regina Pereira
- Department of Pharmacy, University of Joinville Region, Joinville, Santa Catarina, Brazil
| | - André Felipe Rodrigues
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Max Delbrück Center (MDC), Berlin, Germany
| | - Felipe Schmitz
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Eduardo Manoel Pereira
- Department of Pharmacy, University of Joinville Region, Joinville, Santa Catarina, Brazil
| | | | - Angela T S Wyse
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
6
|
Thirugnanasambantham P, Bashian E, Zaleski R, Gopalan V. Demonstrating the utility of sugar-phosphate phosphatases in coupled enzyme assays: galactose-1-phosphate uridylyltransferase as proof-of-concept. Glycobiology 2023; 33:95-98. [PMID: 36585843 PMCID: PMC9990984 DOI: 10.1093/glycob/cwac085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/05/2022] [Accepted: 12/17/2022] [Indexed: 01/01/2023] Open
Abstract
During our biochemical characterization of select bacterial phosphatases belonging to the haloacid dehalogenase superfamily of hydrolases, we discovered a strong bias of Salmonella YidA for glucose-1-phosphate (Glc-1-P) over galactose-1-phosphate (Gal-1-P). We sought to exploit this ability of YidA to discriminate these two sugar-phosphate epimers in a simple coupled assay that could be a substitute for current cumbersome alternatives. To this end, we focused on Gal-1-P uridylyltransferase (GalT) that is defective in individuals with classical galactosemia, an inborn disorder. GalT catalyzes the conversion of Gal-1-P and UDP-glucose to Glc-1-P and UDP-galactose. When recombinant YidA was coupled to GalT, the final orthophosphate product (generated from selective hydrolysis of Glc-1-P by YidA) could be easily measured using the inexpensive malachite green reagent. When this new YidA-based colorimetric assay was benchmarked using a recombinant Duarte GalT variant, it yielded kcat/Km values that are ~2.5-fold higher than the standard coupled assay that employs phosphoglucomutase and glucose-6-phosphate dehydrogenase. Although the simpler design of our new GalT coupled assay might find appeal in diagnostics, a testable expectation, we spotlight the GalT example to showcase the untapped potential of sugar-phosphate phosphatases with distinctive substrate-recognition properties for measuring the activity of various metabolic enzymes (e.g. trehalose-6-phosphate synthase, N-acetyl-glucosamine-6-phosphate deacetylase, phosphofructokinase).
Collapse
Affiliation(s)
| | - Eleanor Bashian
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA
| | - Rosemary Zaleski
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Venkat Gopalan
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
7
|
Rodrigues KF, Yong WTL, Bhuiyan MSA, Siddiquee S, Shah MD, Venmathi Maran BA. Current Understanding on the Genetic Basis of Key Metabolic Disorders: A Review. BIOLOGY 2022; 11:biology11091308. [PMID: 36138787 PMCID: PMC9495729 DOI: 10.3390/biology11091308] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/27/2022] [Accepted: 08/29/2022] [Indexed: 12/02/2022]
Abstract
Simple Summary Metabolic disorders (MD) are a challenge to healthcare systems; the emergence of the modern socio-economic system has led to a profound change in lifestyles in terms of dietary habits, exercise regimens, and behavior, all of which complement the genetic factors associated with MD. Diabetes Mellitus and Familial hypercholesterolemia are two of the 14 most widely researched MD, as they pose the greatest challenge to the public healthcare system and have an impact on productivity and the economy. Research findings have led to the development of new therapeutic molecules for the mitigation of MD as well as the invention of experimental strategies, which target the genes themselves via gene editing and RNA interference. Although these approaches may herald the emergence of a new toolbox to treat MD, the current therapeutic approaches still heavily depend on substrate reduction, dietary restrictions based on genetic factors, exercise, and the maintenance of good mental health. The development of orphan drugs for the less common MD such as Krabbe, Farber, Fabry, and Gaucher diseases, remains in its infancy, owing to the lack of investment in research and development, and this has driven the development of personalized therapeutics based on gene silencing and related technologies. Abstract Advances in data acquisition via high resolution genomic, transcriptomic, proteomic and metabolomic platforms have driven the discovery of the underlying factors associated with metabolic disorders (MD) and led to interventions that target the underlying genetic causes as well as lifestyle changes and dietary regulation. The review focuses on fourteen of the most widely studied inherited MD, which are familial hypercholesterolemia, Gaucher disease, Hunter syndrome, Krabbe disease, Maple syrup urine disease, Metachromatic leukodystrophy, Mitochondrial encephalopathy lactic acidosis stroke-like episodes (MELAS), Niemann-Pick disease, Phenylketonuria (PKU), Porphyria, Tay-Sachs disease, Wilson’s disease, Familial hypertriglyceridemia (F-HTG) and Galactosemia based on genome wide association studies, epigenetic factors, transcript regulation, post-translational genetic modifications and biomarker discovery through metabolomic studies. We will delve into the current approaches being undertaken to analyze metadata using bioinformatic approaches and the emerging interventions using genome editing platforms as applied to animal models.
Collapse
Affiliation(s)
- Kenneth Francis Rodrigues
- Biotechnology Research Institute, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia
- Correspondence: (K.F.R.); (B.A.V.M.); Tel.: +60-16-2096905 (B.A.V.M.)
| | - Wilson Thau Lym Yong
- Biotechnology Research Institute, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia
| | | | | | - Muhammad Dawood Shah
- Borneo Marine Research Institute, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia
| | - Balu Alagar Venmathi Maran
- Borneo Marine Research Institute, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia
- Correspondence: (K.F.R.); (B.A.V.M.); Tel.: +60-16-2096905 (B.A.V.M.)
| |
Collapse
|
8
|
Succoio M, Sacchettini R, Rossi A, Parenti G, Ruoppolo M. Galactosemia: Biochemistry, Molecular Genetics, Newborn Screening, and Treatment. Biomolecules 2022; 12:biom12070968. [PMID: 35883524 PMCID: PMC9313126 DOI: 10.3390/biom12070968] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/02/2022] [Accepted: 07/08/2022] [Indexed: 12/16/2022] Open
Abstract
Galactosemia is an inborn disorder of carbohydrate metabolism characterized by the inability to metabolize galactose, a sugar contained in milk (the main source of nourishment for infants), and convert it into glucose, the sugar used by the body as the primary source of energy. Galactosemia is an autosomal recessive genetic disease that can be diagnosed at birth, even in the absence of symptoms, with newborn screening by assessing the level of galactose and the GALT enzyme activity, as GALT defect constitutes the most frequent cause of galactosemia. Currently, galactosemia cannot be cured, but only treated by means of a diet with a reduced content of galactose and lactose. Although the diet is able to reverse the neonatal clinical picture, it does not prevent the development of long-term complications. This review provides an overview of galactose metabolism, molecular genetics, newborn screening and therapy of galactosemia. Novel treatments for galactosemia currently being investigated in (pre)clinical studies and potentially able to prevent long-term complications are also presented.
Collapse
Affiliation(s)
- Mariangela Succoio
- CEINGE-Biotecnologie Avanzate s.c.ar.l., 80145 Naples, Italy; (M.S.); (R.S.)
| | - Rosa Sacchettini
- CEINGE-Biotecnologie Avanzate s.c.ar.l., 80145 Naples, Italy; (M.S.); (R.S.)
| | - Alessandro Rossi
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (A.R.); (G.P.)
| | - Giancarlo Parenti
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (A.R.); (G.P.)
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy
| | - Margherita Ruoppolo
- CEINGE-Biotecnologie Avanzate s.c.ar.l., 80145 Naples, Italy; (M.S.); (R.S.)
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
- Correspondence:
| |
Collapse
|
9
|
Delnoy B, Haskovic M, Vanoevelen J, Steinbusch LKM, Vos EN, Knoops K, Zimmermann LJI, Noga M, Lefeber DJ, Martini PGV, Coelho AI, Rubio‐Gozalbo ME. Novel mRNA therapy restores GALT protein and enzyme activity in a zebrafish model of classic galactosemia. J Inherit Metab Dis 2022; 45:748-758. [PMID: 35527402 PMCID: PMC9541528 DOI: 10.1002/jimd.12512] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 11/24/2022]
Abstract
Messenger RNA (mRNA) has emerged as a novel therapeutic approach for inborn errors of metabolism. Classic galactosemia (CG) is an inborn error of galactose metabolism caused by a severe deficiency of galactose-1-phosphate:uridylyltransferase (GALT) activity leading to neonatal illness and chronic impairments affecting the brain and female gonads. In this proof of concept study, we used our zebrafish model for CG to evaluate the potential of human GALT mRNA (hGALT mRNA) packaged in two different lipid nanoparticles to restore GALT expression and activity at early stages of development. Both one cell-stage and intravenous single-dose injections resulted in hGALT protein expression and enzyme activity in the CG zebrafish (galt knockout) at 5 days post fertilization (dpf). Moreover, the levels of galactose-1-phosphate (Gal-1-P) and galactonate, metabolites that accumulate because of the deficiency, showed a decreasing trend. LNP-packaged mRNA was effectively translated and processed in the CG zebrafish without signs of toxicity. This study shows that mRNA therapy restores GALT protein and enzyme activity in the CG zebrafish model, and that the zebrafish is a suitable system to test this approach. Further studies are warranted to assess whether repeated injections safely mitigate the chronic impairments of this disease.
Collapse
Affiliation(s)
- Britt Delnoy
- Department of PediatricsMaastricht University Medical Center+Maastrichtthe Netherlands
- GROW, Maastricht UniversityMaastrichtthe Netherlands
| | - Minela Haskovic
- Department of PediatricsMaastricht University Medical Center+Maastrichtthe Netherlands
- GROW, Maastricht UniversityMaastrichtthe Netherlands
| | - Jo Vanoevelen
- GROW, Maastricht UniversityMaastrichtthe Netherlands
- Department of Clinical GeneticsMaastricht University Medical Center+Maastrichtthe Netherlands
| | - Laura K. M. Steinbusch
- Department of Clinical GeneticsMaastricht University Medical Center+Maastrichtthe Netherlands
| | - Esther Naomi Vos
- Department of PediatricsMaastricht University Medical Center+Maastrichtthe Netherlands
| | - Kèvin Knoops
- Microscopy CORE LaboratoryMaastricht UniversityMaastrichtthe Netherlands
| | - Luc J. I. Zimmermann
- Department of PediatricsMaastricht University Medical Center+Maastrichtthe Netherlands
- GROW, Maastricht UniversityMaastrichtthe Netherlands
| | - Marek Noga
- Translational Metabolic LaboratoryRadboud University Medical CenterNijmegenthe Netherlands
| | - Dirk J. Lefeber
- Translational Metabolic LaboratoryRadboud University Medical CenterNijmegenthe Netherlands
- Department of NeurologyDonders Institute for Brain, Cognition and Behavior, Radboud University Medical CenterNijmegenthe Netherlands
| | | | - Ana I. Coelho
- Department of PediatricsMaastricht University Medical Center+Maastrichtthe Netherlands
| | - Maria Estela Rubio‐Gozalbo
- Department of PediatricsMaastricht University Medical Center+Maastrichtthe Netherlands
- GROW, Maastricht UniversityMaastrichtthe Netherlands
- Department of Clinical GeneticsMaastricht University Medical Center+Maastrichtthe Netherlands
| |
Collapse
|
10
|
Brophy ML, Stansfield JC, Ahn Y, Cheng SH, Murphy JE, Bell RD. AAV-mediated expression of galactose-1-phosphate uridyltransferase corrects defects of galactose metabolism in classic galactosemia patient fibroblasts. J Inherit Metab Dis 2022; 45:481-492. [PMID: 34918784 DOI: 10.1002/jimd.12468] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 11/22/2021] [Accepted: 12/08/2021] [Indexed: 11/10/2022]
Abstract
Classic galactosemia (CG) is a rare disorder of autosomal recessive inheritance. It is caused predominantly by point mutations as well as deletions in the gene encoding the enzyme galactose-1-phosphate uridyltransferase (GALT). The majority of the more than 350 mutations identified in the GALT gene cause a significant reduction in GALT enzyme activity resulting in the toxic buildup of galactose metabolites that in turn is associated with cellular stress and injury. Consequently, developing a therapeutic strategy that reverses both the oxidative and ER stress in CG cells may be helpful in combating this disease. Recombinant adeno-associated virus (AAV)-mediated gene therapy to restore GALT activity offers the potential to address the unmet medical needs of galactosemia patients. Here, utilizing fibroblasts derived from CG patients we demonstrated that AAV-mediated augmentation of GALT protein and activity resulted in the prevention of ER and oxidative stress. We also demonstrate that these CG patient fibroblasts exhibit reduced CD109 and TGFβRII protein levels and that these effectors of cellular homeostasis could be restored following AAV-mediated expression of GALT. Finally, we show initial in vivo proof-of-concept restoration of galactose metabolism in a GALT knockout mouse model following treatment with AAV-GALT.
Collapse
Affiliation(s)
- Megan L Brophy
- Rare Disease Research Unit, Pfizer, Inc., Cambridge, Massachusetts, USA
| | - John C Stansfield
- Early Clinical Development, Pfizer, Inc., Cambridge, Massachusetts, USA
| | - Youngwook Ahn
- Target Sciences, Pfizer, Inc., Cambridge, Massachusetts, USA
| | - Seng H Cheng
- Rare Disease Research Unit, Pfizer, Inc., Cambridge, Massachusetts, USA
| | - John E Murphy
- Rare Disease Research Unit, Pfizer, Inc., Cambridge, Massachusetts, USA
| | - Robert D Bell
- Rare Disease Research Unit, Pfizer, Inc., Cambridge, Massachusetts, USA
| |
Collapse
|
11
|
Verdino A, D’Urso G, Tammone C, Scafuri B, Catapano L, Marabotti A. Simulation of the Interactions of Arginine with Wild-Type GALT Enzyme and the Classic Galactosemia-Related Mutant p.Q188R by a Computational Approach. Molecules 2021; 26:6061. [PMID: 34641605 PMCID: PMC8513022 DOI: 10.3390/molecules26196061] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 09/30/2021] [Accepted: 10/05/2021] [Indexed: 11/18/2022] Open
Abstract
Classic galactosemia is an inborn error of metabolism associated with mutations that impair the activity and the stability of galactose-1-phosphate uridylyltransferase (GALT), catalyzing the third step in galactose metabolism. To date, no treatments (including dietary galactose deprivation) are able to prevent or alleviate the long-term complications affecting galactosemic patients. Evidence that arginine is able to improve the activity of the human enzyme expressed in a prokaryotic model of classic galactosemia has induced researchers to suppose that this amino acid could act as a pharmacochaperone, but no effects were detected in four galactosemic patients treated with this amino acid. Given that no molecular characterizations of the possible effects of arginine on GALT have been performed, and given that the samples of patients treated with arginine are extremely limited for drawing definitive conclusions at the clinical level, we performed computational simulations in order to predict the interactions (if any) between this amino acid and the enzyme. Our results do not support the possibility that arginine could function as a pharmacochaperone for GALT, but information obtained by this study could be useful for identifying, in the future, possible pharmacochaperones for this enzyme.
Collapse
Affiliation(s)
- Anna Verdino
- Department of Chemistry and Biology “A. Zambelli”, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, SA, Italy; (A.V.); (G.D.); (C.T.); (B.S.); (L.C.)
| | - Gaetano D’Urso
- Department of Chemistry and Biology “A. Zambelli”, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, SA, Italy; (A.V.); (G.D.); (C.T.); (B.S.); (L.C.)
| | - Carmen Tammone
- Department of Chemistry and Biology “A. Zambelli”, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, SA, Italy; (A.V.); (G.D.); (C.T.); (B.S.); (L.C.)
| | - Bernardina Scafuri
- Department of Chemistry and Biology “A. Zambelli”, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, SA, Italy; (A.V.); (G.D.); (C.T.); (B.S.); (L.C.)
- Interuniversity Center “ELFID—European Laboratory for Food Induced Diseases”, University of Salerno, 84084 Fisciano, SA, Italy
| | - Lucrezia Catapano
- Department of Chemistry and Biology “A. Zambelli”, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, SA, Italy; (A.V.); (G.D.); (C.T.); (B.S.); (L.C.)
| | - Anna Marabotti
- Department of Chemistry and Biology “A. Zambelli”, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, SA, Italy; (A.V.); (G.D.); (C.T.); (B.S.); (L.C.)
- Interuniversity Center “ELFID—European Laboratory for Food Induced Diseases”, University of Salerno, 84084 Fisciano, SA, Italy
| |
Collapse
|
12
|
Ghorbani MJ, Moravej H, Amirhakimi A, Geramizadeh B, Kalani M, Fallahi F. A novel missense mutation (c.516A>T; p.Glu172Asp) in the GALT gene as a cause of classic galactosemia: A case report. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
13
|
Banford S, McCorvie TJ, Pey AL, Timson DJ. Galactosemia: Towards Pharmacological Chaperones. J Pers Med 2021; 11:jpm11020106. [PMID: 33562227 PMCID: PMC7914515 DOI: 10.3390/jpm11020106] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 02/02/2021] [Accepted: 02/04/2021] [Indexed: 02/07/2023] Open
Abstract
Galactosemia is a rare inherited metabolic disease resulting from mutations in the four genes which encode enzymes involved in the metabolism of galactose. The current therapy, the removal of galactose from the diet, is inadequate. Consequently, many patients suffer lifelong physical and cognitive disability. The phenotype varies from almost asymptomatic to life-threatening disability. The fundamental biochemical cause of the disease is a decrease in enzymatic activity due to failure of the affected protein to fold and/or function correctly. Many novel therapies have been proposed for the treatment of galactosemia. Often, these are designed to treat the symptoms and not the fundamental cause. Pharmacological chaperones (PC) (small molecules which correct the folding of misfolded proteins) represent an exciting potential therapy for galactosemia. In theory, they would restore enzyme function, thus preventing downstream pathological consequences. In practice, no PCs have been identified for potential application in galactosemia. Here, we review the biochemical basis of the disease, identify opportunities for the application of PCs and describe how these might be discovered. We will conclude by considering some of the clinical issues which will affect the future use of PCs in the treatment of galactosemia.
Collapse
Affiliation(s)
- Samantha Banford
- South Eastern Health and Social Care Trust, Downpatrick BT30 6RL, UK;
| | - Thomas J. McCorvie
- Structural Genomics Consortium, University of Oxford, Oxford OX3 7DQ, UK;
| | - Angel L. Pey
- Departamento de Química Física, Unidad de Excelencia de Química aplicada a Biomedicina y Medioambiente e Instituto de Biotecnología, Facultad de Ciencias, Universidad de Granada, 18071 Granada, Spain;
| | - David J. Timson
- School of Pharmacy and Biomolecular Sciences, The University of Brighton, Brighton BN2 4GJ, UK
- Correspondence:
| |
Collapse
|
14
|
Delnoy B, Coelho AI, Rubio-Gozalbo ME. Current and Future Treatments for Classic Galactosemia. J Pers Med 2021; 11:jpm11020075. [PMID: 33525536 PMCID: PMC7911353 DOI: 10.3390/jpm11020075] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/23/2021] [Accepted: 01/24/2021] [Indexed: 02/07/2023] Open
Abstract
Type I (classic) galactosemia, galactose 1-phosphate uridylyltransferase (GALT)-deficiency is a hereditary disorder of galactose metabolism. The current therapeutic standard of care, a galactose-restricted diet, is effective in treating neonatal complications but is inadequate in preventing burdensome complications. The development of several animal models of classic galactosemia that (partly) mimic the biochemical and clinical phenotypes and the resolution of the crystal structure of GALT have provided important insights; however, precise pathophysiology remains to be elucidated. Novel therapeutic approaches currently being explored focus on several of the pathogenic factors that have been described, aiming to (i) restore GALT activity, (ii) influence the cascade of events and (iii) address the clinical picture. This review attempts to provide an overview on the latest advancements in therapy approaches.
Collapse
Affiliation(s)
- Britt Delnoy
- Department of Pediatrics, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands; (B.D.); (A.I.C.)
- Department of Clinical Genetics, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
- GROW-School for Oncology and Developmental Biology, Maastricht University, 6229 HX Maastricht, The Netherlands
| | - Ana I. Coelho
- Department of Pediatrics, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands; (B.D.); (A.I.C.)
| | - Maria Estela Rubio-Gozalbo
- Department of Pediatrics, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands; (B.D.); (A.I.C.)
- Department of Clinical Genetics, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
- GROW-School for Oncology and Developmental Biology, Maastricht University, 6229 HX Maastricht, The Netherlands
- Correspondence: ; Tel.: +31-43-3872920
| |
Collapse
|
15
|
Pacheco-García JL, Cano-Muñoz M, Sánchez-Ramos I, Salido E, Pey AL. Naturally-Occurring Rare Mutations Cause Mild to Catastrophic Effects in the Multifunctional and Cancer-Associated NQO1 Protein. J Pers Med 2020; 10:E207. [PMID: 33153185 PMCID: PMC7711955 DOI: 10.3390/jpm10040207] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 10/27/2020] [Accepted: 11/02/2020] [Indexed: 12/13/2022] Open
Abstract
The functional and pathological implications of the enormous genetic diversity of the human genome are mostly unknown, primarily due to our unability to predict pathogenicity in a high-throughput manner. In this work, we characterized the phenotypic consequences of eight naturally-occurring missense variants on the multifunctional and disease-associated NQO1 protein using biophysical and structural analyses on several protein traits. Mutations found in both exome-sequencing initiatives and in cancer cell lines cause mild to catastrophic effects on NQO1 stability and function. Importantly, some mutations perturb functional features located structurally far from the mutated site. These effects are well rationalized by considering the nature of the mutation, its location in protein structure and the local stability of its environment. Using a set of 22 experimentally characterized mutations in NQO1, we generated experimental scores for pathogenicity that correlate reasonably well with bioinformatic scores derived from a set of commonly used algorithms, although the latter fail to semiquantitatively predict the phenotypic alterations caused by a significant fraction of mutations individually. These results provide insight into the propagation of mutational effects on multifunctional proteins, the implementation of in silico approaches for establishing genotype-phenotype correlations and the molecular determinants underlying loss-of-function in genetic diseases.
Collapse
Affiliation(s)
- Juan Luis Pacheco-García
- Departamento de Química Física, Facultad de Ciencias, Universidad de Granada, 18071 Granada, Spain; (J.L.P.-G.); (M.C.-M.); (I.S.-R.)
| | - Mario Cano-Muñoz
- Departamento de Química Física, Facultad de Ciencias, Universidad de Granada, 18071 Granada, Spain; (J.L.P.-G.); (M.C.-M.); (I.S.-R.)
| | - Isabel Sánchez-Ramos
- Departamento de Química Física, Facultad de Ciencias, Universidad de Granada, 18071 Granada, Spain; (J.L.P.-G.); (M.C.-M.); (I.S.-R.)
| | - Eduardo Salido
- Centre for Biomedical Research on Rare Diseases (CIBERER), Hospital Universitario de Canarias, 38320 Tenerife, Spain;
| | - Angel L. Pey
- Departamento de Química Física y Unidad de Excelencia de Química Aplicada a Biomedicina y Medioambiente (UEQ), Facultad de Ciencias, Universidad de Granada, 18071 Granada, Spain
| |
Collapse
|
16
|
Haskovic M, Coelho AI, Bierau J, Vanoevelen JM, Steinbusch LKM, Zimmermann LJI, Villamor‐Martinez E, Berry GT, Rubio‐Gozalbo ME. Pathophysiology and targets for treatment in hereditary galactosemia: A systematic review of animal and cellular models. J Inherit Metab Dis 2020; 43:392-408. [PMID: 31808946 PMCID: PMC7317974 DOI: 10.1002/jimd.12202] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 11/25/2019] [Accepted: 12/03/2019] [Indexed: 12/18/2022]
Abstract
Since the first description of galactosemia in 1908 and despite decades of research, the pathophysiology is complex and not yet fully elucidated. Galactosemia is an inborn error of carbohydrate metabolism caused by deficient activity of any of the galactose metabolising enzymes. The current standard of care, a galactose-restricted diet, fails to prevent long-term complications. Studies in cellular and animal models in the past decades have led to an enormous progress and advancement of knowledge. Summarising current evidence in the pathophysiology underlying hereditary galactosemia may contribute to the identification of treatment targets for alternative therapies that may successfully prevent long-term complications. A systematic review of cellular and animal studies reporting on disease complications (clinical signs and/or biochemical findings) and/or treatment targets in hereditary galactosemia was performed. PubMed/MEDLINE, EMBASE, and Web of Science were searched, 46 original articles were included. Results revealed that Gal-1-P is not the sole pathophysiological agent responsible for the phenotype observed in galactosemia. Other currently described contributing factors include accumulation of galactose metabolites, uridine diphosphate (UDP)-hexose alterations and subsequent impaired glycosylation, endoplasmic reticulum (ER) stress, altered signalling pathways, and oxidative stress. galactokinase (GALK) inhibitors, UDP-glucose pyrophosphorylase (UGP) up-regulation, uridine supplementation, ER stress reducers, antioxidants and pharmacological chaperones have been studied, showing rescue of biochemical and/or clinical symptoms in galactosemia. Promising co-adjuvant therapies include antioxidant therapy and UGP up-regulation. This systematic review provides an overview of the scattered information resulting from animal and cellular studies performed in the past decades, summarising the complex pathophysiological mechanisms underlying hereditary galactosemia and providing insights on potential treatment targets.
Collapse
Affiliation(s)
- Minela Haskovic
- Department of PediatricsMaastricht University Medical Center+MaastrichtThe Netherlands
- Department of Clinical GeneticsMaastricht University Medical Center+MaastrichtThe Netherlands
- GROW‐School for Oncology and Developmental Biology, Maastricht UniversityMaastrichtThe Netherlands
| | - Ana I. Coelho
- Department of PediatricsMaastricht University Medical Center+MaastrichtThe Netherlands
- Department of Clinical GeneticsMaastricht University Medical Center+MaastrichtThe Netherlands
- GROW‐School for Oncology and Developmental Biology, Maastricht UniversityMaastrichtThe Netherlands
| | - Jörgen Bierau
- Department of Clinical GeneticsMaastricht University Medical Center+MaastrichtThe Netherlands
| | - Jo M. Vanoevelen
- Department of Clinical GeneticsMaastricht University Medical Center+MaastrichtThe Netherlands
- GROW‐School for Oncology and Developmental Biology, Maastricht UniversityMaastrichtThe Netherlands
| | - Laura K. M. Steinbusch
- Department of Clinical GeneticsMaastricht University Medical Center+MaastrichtThe Netherlands
| | - Luc J. I. Zimmermann
- Department of PediatricsMaastricht University Medical Center+MaastrichtThe Netherlands
- GROW‐School for Oncology and Developmental Biology, Maastricht UniversityMaastrichtThe Netherlands
| | - Eduardo Villamor‐Martinez
- Department of PediatricsMaastricht University Medical Center+MaastrichtThe Netherlands
- GROW‐School for Oncology and Developmental Biology, Maastricht UniversityMaastrichtThe Netherlands
| | - Gerard T. Berry
- The Manton Center for Orphan Disease Research, Division of Genetics and GenomicsBoston Children's Hospital, Harvard Medical SchoolBostonMassachusetts
| | - M. Estela Rubio‐Gozalbo
- Department of PediatricsMaastricht University Medical Center+MaastrichtThe Netherlands
- Department of Clinical GeneticsMaastricht University Medical Center+MaastrichtThe Netherlands
- GROW‐School for Oncology and Developmental Biology, Maastricht UniversityMaastrichtThe Netherlands
| |
Collapse
|
17
|
Affiliation(s)
- David J. Timson
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, UK
| |
Collapse
|
18
|
Canson DM, Silao CLT, Caoili SEC. Functional analysis of GALT variants found in classic galactosemia patients using a novel cell-free translation method. JIMD Rep 2019; 48:60-66. [PMID: 31392114 PMCID: PMC6606980 DOI: 10.1002/jmd2.12037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 04/04/2019] [Accepted: 04/18/2019] [Indexed: 11/15/2022] Open
Abstract
Classic galactosemia is an autosomal recessive disorder caused by deleterious variants in the galactose-1-phosphate uridylyltransferase (GALT) gene. GALT enzyme deficiency leads to an increase in the levels of galactose and its metabolites in the blood causing neurodevelopmental and other clinical complications in affected individuals. Two GALT variants NM_000155.3:c.347T>C (p.Leu116Pro) and NM_000155.3:c.533T>G (p.Met178Arg) were previously detected in Filipino patients. Here, we determine their functional effects on the GALT enzyme through in silico analysis and a novel experimental approach using a HeLa-based cell-free protein expression system. Enzyme activity was not detected for the p.Leu116Pro protein variant, while only 4.5% of wild-type activity was detected for the p.Met178Arg protein variant. Computational analysis of the variants revealed destabilizing structural effects and suggested protein misfolding as the potential mechanism of enzymological impairment. Biochemical and computational data support the classification of p.Leu116Pro and p.Met178Arg variants as pathogenic. Moreover, the protein expression method developed has utility for future studies of GALT variants.
Collapse
Affiliation(s)
- Daffodil M. Canson
- Institute of Human Genetics, National Institutes of HealthUniversity of the Philippines ManilaManilaPhilippines
- Department of Biochemistry and Molecular Biology, College of MedicineUniversity of the Philippines ManilaManilaPhilippines
| | - Catherine Lynn T. Silao
- Institute of Human Genetics, National Institutes of HealthUniversity of the Philippines ManilaManilaPhilippines
| | - Salvador Eugenio C. Caoili
- Department of Biochemistry and Molecular Biology, College of MedicineUniversity of the Philippines ManilaManilaPhilippines
| |
Collapse
|
19
|
Haskovic M, Derks B, van der Ploeg L, Trommelen J, Nyakayiru J, van Loon LJC, Mackinnon S, Yue WW, Peake RWA, Zha L, Demirbas D, Qi W, Huang X, Berry GT, Achten J, Bierau J, Rubio-Gozalbo ME, Coelho AI. Arginine does not rescue p.Q188R mutation deleterious effect in classic galactosemia. Orphanet J Rare Dis 2018; 13:212. [PMID: 30477550 PMCID: PMC6260575 DOI: 10.1186/s13023-018-0954-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 11/14/2018] [Indexed: 12/22/2022] Open
Abstract
Background Classic galactosemia is a rare genetic metabolic disease with an unmet treatment need. Current standard of care fails to prevent chronically-debilitating brain and gonadal complications. Many mutations in the GALT gene responsible for classic galactosemia have been described to give rise to variants with conformational abnormalities. This pathogenic mechanism is highly amenable to a therapeutic strategy based on chemical/pharmacological chaperones. Arginine, a chemical chaperone, has shown beneficial effect in other inherited metabolic disorders, as well as in a prokaryotic model of classic galactosemia. The p.Q188R mutation presents a high prevalence in the Caucasian population, making it a very clinically relevant mutation. This mutation gives rise to a protein with lower conformational stability and lower catalytic activity. The aim of this study is to assess the potential therapeutic role of arginine for this mutation. Methods Arginine aspartate administration to four patients with the p.Q188R/p.Q188R mutation, in vitro studies with three fibroblast cell lines derived from classic galactosemia patients as well as recombinant protein experiments were used to evaluate the effect of arginine in galactose metabolism. This study has been registered at https://clinicaltrials.gov (NCT03580122) on 09 July 2018. Retrospectively registered. Results Following a month of arginine administration, patients did not show a significant improvement of whole-body galactose oxidative capacity (p = 0.22), erythrocyte GALT activity (p = 0.87), urinary galactose (p = 0.52) and urinary galactitol levels (p = 0.41). Patients’ fibroblasts exposed to arginine did not show changes in GALT activity. Thermal shift analysis of recombinant p.Q188R GALT protein in the presence of arginine did not exhibit a positive effect. Conclusions This short pilot study in four patients homozygous for the p.Q188R/p.Q188R mutation reveals that arginine has no potential therapeutic role for galactosemia patients homozygous for the p.Q188R mutation. Electronic supplementary material The online version of this article (10.1186/s13023-018-0954-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Minela Haskovic
- Department of Clinical Genetics, Maastricht University Medical Center +, Maastricht, The Netherlands.,Department of Pediatrics, Maastricht University Medical Center +, Maastricht, The Netherlands.,GROW-School for Oncology and Developmental Biology, University of Maastricht, Maastricht, The Netherlands
| | - Britt Derks
- Department of Clinical Genetics, Maastricht University Medical Center +, Maastricht, The Netherlands.,Department of Pediatrics, Maastricht University Medical Center +, Maastricht, The Netherlands.,GROW-School for Oncology and Developmental Biology, University of Maastricht, Maastricht, The Netherlands
| | - Liesbeth van der Ploeg
- Department of Dietetics, Maastricht University Medical Center +, Maastricht, The Netherlands
| | - Jorn Trommelen
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center +, Maastricht, The Netherlands
| | - Jean Nyakayiru
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center +, Maastricht, The Netherlands
| | - Luc J C van Loon
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center +, Maastricht, The Netherlands
| | - Sabrina Mackinnon
- Structural Genomics Consortium, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Wyatt W Yue
- Structural Genomics Consortium, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Roy W A Peake
- Department of Laboratory Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Li Zha
- Department of Laboratory Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Didem Demirbas
- The Manton Center for Orphan Disease Research, Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wanshu Qi
- The Manton Center for Orphan Disease Research, Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xiaoping Huang
- The Manton Center for Orphan Disease Research, Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Gerard T Berry
- The Manton Center for Orphan Disease Research, Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jelle Achten
- Department of Clinical Genetics, Maastricht University Medical Center +, Maastricht, The Netherlands
| | - Jörgen Bierau
- Department of Clinical Genetics, Maastricht University Medical Center +, Maastricht, The Netherlands
| | - M Estela Rubio-Gozalbo
- Department of Clinical Genetics, Maastricht University Medical Center +, Maastricht, The Netherlands. .,Department of Pediatrics, Maastricht University Medical Center +, Maastricht, The Netherlands. .,GROW-School for Oncology and Developmental Biology, University of Maastricht, Maastricht, The Netherlands.
| | - Ana I Coelho
- Department of Clinical Genetics, Maastricht University Medical Center +, Maastricht, The Netherlands.,Department of Pediatrics, Maastricht University Medical Center +, Maastricht, The Netherlands.,GROW-School for Oncology and Developmental Biology, University of Maastricht, Maastricht, The Netherlands
| |
Collapse
|
20
|
Yuzyuk T, Balakrishnan B, Schwarz EL, De Biase I, Hobert J, Longo N, Mao R, Lai K, Pasquali M. Effect of genotype on galactose-1-phosphate in classic galactosemia patients. Mol Genet Metab 2018; 125:258-265. [PMID: 30172461 DOI: 10.1016/j.ymgme.2018.08.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/21/2018] [Accepted: 08/22/2018] [Indexed: 11/27/2022]
Abstract
Impaired activity of galactose-1-phosphate uridyltransferase (GALT) causes classic galactosemia (OMIM 230400), characterized by the accumulation of galactose-1-phosphate (GAL1P) in patients' red blood cells (RBCs). Our recent study demonstrated a correlation between RBC GAL1P and long-term outcomes in galactosemia patients. Here, we analyze biochemical and molecular results in 77 classic galactosemia patients to evaluate the association between GALT genotypes and GAL1P concentration in RBCs. Experimental data from model organisms were also included to assess the correlation between GAL1P and predicted residual activity of each genotype. Although all individuals in this study showed markedly reduced RBC GALT activity, we observed significant differences in RBC GAL1P concentrations among galactosemia genotypes. While levels of GAL1P on treatment did not correlate with RBC GALT activities (p = 0.166), there was a negative nonlinear correlation between mean GAL1P concentrations and predicted residual enzyme activity of genotype (p = 0.004). These studies suggest that GAL1P levels in RBCs on treatment likely reflect the overall functional impairment of GALT in patients with galactosemia.
Collapse
Affiliation(s)
- Tatiana Yuzyuk
- Department of Pathology, University of Utah, Salt Lake City, UT, USA; ARUP Laboratories, Salt Lake City, UT, USA.
| | - Bijina Balakrishnan
- Division of Medical Genetics/Pediatrics, University of Utah, Salt Lake City, UT, USA
| | | | - Irene De Biase
- Department of Pathology, University of Utah, Salt Lake City, UT, USA; ARUP Laboratories, Salt Lake City, UT, USA
| | - Judith Hobert
- Department of Pathology, University of Utah, Salt Lake City, UT, USA; ARUP Laboratories, Salt Lake City, UT, USA
| | - Nicola Longo
- Department of Pathology, University of Utah, Salt Lake City, UT, USA; ARUP Laboratories, Salt Lake City, UT, USA; Division of Medical Genetics/Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Rong Mao
- Department of Pathology, University of Utah, Salt Lake City, UT, USA; ARUP Laboratories, Salt Lake City, UT, USA
| | - Kent Lai
- Division of Medical Genetics/Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Marzia Pasquali
- Department of Pathology, University of Utah, Salt Lake City, UT, USA; ARUP Laboratories, Salt Lake City, UT, USA
| |
Collapse
|
21
|
Pey AL. Biophysical and functional perturbation analyses at cancer-associated P187 and K240 sites of the multifunctional NADP(H):quinone oxidoreductase 1. Int J Biol Macromol 2018; 118:1912-1923. [PMID: 30009918 DOI: 10.1016/j.ijbiomac.2018.07.051] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/10/2018] [Accepted: 07/11/2018] [Indexed: 12/14/2022]
Abstract
Once whole-genome sequencing has reached the clinical practice, a main challenge ahead is the high-throughput and accurate prediction of the pathogenicity of genetic variants. However, current prediction tools do not consider explicitly a well-known property of disease-causing mutations: their ability to affect multiple functional sites distant in the protein structure. Here we carried out an extensive biophysical characterization of fourteen mutant variants at two cancer-associated sites of the enzyme NQO1, a paradigm of multi-functional protein. We showed that the magnitude of destabilizing effects, their molecular origins (structural vs. dynamic) and their efficient propagation through the protein structure gradually led to functional perturbations at different sites. Modulation of these structural perturbations also led to switches between molecular phenotypes. Our work supports that experimental and computational perturbation analyses would improve our understanding of the molecular basis of many loss-of-function genetic diseases as well as our ability to accurately predict the pathogenicity of genetic variants in a high-throughput fashion.
Collapse
Affiliation(s)
- Angel L Pey
- Department of Physical Chemistry, University of Granada, Av. Fuentenueva S/N, 18071 Granada, Spain.
| |
Collapse
|
22
|
Vanoevelen JM, van Erven B, Bierau J, Huang X, Berry GT, Vos R, Coelho AI, Rubio-Gozalbo ME. Impaired fertility and motor function in a zebrafish model for classic galactosemia. J Inherit Metab Dis 2018; 41:117-127. [PMID: 28913702 PMCID: PMC5786655 DOI: 10.1007/s10545-017-0071-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 06/16/2017] [Accepted: 07/03/2017] [Indexed: 01/28/2023]
Abstract
Classic galactosemia is a genetic disorder of galactose metabolism, caused by severe deficiency of galactose-1-phosphate uridylyltransferase (GALT) enzyme activity due to mutations of the GALT gene. Its pathogenesis is still not fully elucidated, and a therapy that prevents chronic impairments is lacking. In order to move research forward, there is a high need for a novel animal model, which allows organ studies throughout development and high-throughput screening of pharmacologic compounds. Here, we describe the generation of a galt knockout zebrafish model and present its phenotypical characterization. Using a TALEN approach, a galt knockout line was successfully created. Accordingly, biochemical assays confirm essentially undetectable galt enzyme activity in homozygotes. Analogous to humans, galt knockout fish accumulate galactose-1-phosphate upon exposure to exogenous galactose. Furthermore, without prior exposure to exogenous galactose, they exhibit reduced motor activity and impaired fertility (lower egg quantity per mating, higher number of unsuccessful crossings), resembling the human phenotype(s) of neurological sequelae and subfertility. In conclusion, our galt knockout zebrafish model for classic galactosemia mimics the human phenotype(s) at biochemical and clinical levels. Future studies in our model will contribute to improved understanding and management of this disorder.
Collapse
Affiliation(s)
- Jo M Vanoevelen
- Department of Clinical Genetics, Maastricht University Medical Centre, Universiteitssingel 50, P.O. Box 616, box 16, 6200 MD, Maastricht, The Netherlands.
- GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands.
| | - Britt van Erven
- Department of Clinical Genetics, Maastricht University Medical Centre, Universiteitssingel 50, P.O. Box 616, box 16, 6200 MD, Maastricht, The Netherlands
- GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
- Department of Pediatrics, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Jörgen Bierau
- Department of Clinical Genetics, Maastricht University Medical Centre, Universiteitssingel 50, P.O. Box 616, box 16, 6200 MD, Maastricht, The Netherlands
| | - Xiaoping Huang
- The Manton Center for Orphan Disease Research, Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Gerard T Berry
- The Manton Center for Orphan Disease Research, Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rein Vos
- Department of Methodology and Statistics, School for Public Health and Primary Care (CAPHRI), Maastricht University, Maastricht, The Netherlands
| | - Ana I Coelho
- Department of Clinical Genetics, Maastricht University Medical Centre, Universiteitssingel 50, P.O. Box 616, box 16, 6200 MD, Maastricht, The Netherlands
- GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
- Department of Pediatrics, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - M Estela Rubio-Gozalbo
- Department of Clinical Genetics, Maastricht University Medical Centre, Universiteitssingel 50, P.O. Box 616, box 16, 6200 MD, Maastricht, The Netherlands.
- GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands.
- Department of Pediatrics, Maastricht University Medical Centre, Maastricht, The Netherlands.
| |
Collapse
|
23
|
Coelho AI, Bierau J, Lindhout M, Achten J, Kramer BW, Rubio-Gozalbo ME. Classic Galactosemia: Study on the Late Prenatal Development of GALT Specific Activity in a Sheep Model. Anat Rec (Hoboken) 2017; 300:1570-1575. [PMID: 28545161 DOI: 10.1002/ar.23616] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 11/14/2016] [Accepted: 12/27/2016] [Indexed: 11/06/2022]
Abstract
Classic galactosemia results from deficient activity of galactose-1-phosphate uridylyltransferase (GALT), a key enzyme of galactose metabolism. Despite early diagnosis and early postnatal therapeutic intervention, patients still develop neurologic and fertility impairments. Prenatal developmental toxicity has been hypothesized as a determinant factor of disease. In order to shed light on the importance of prenatal GALT activity, several studies have examined GALT activity throughout development. GALT was shown to increase with gestational age in 7-28 weeks human fetuses; later stages were not investigated. Prenatal studies in animals focused exclusively on brain and hepatic GALT activity. In this study, we aim to examine GALT specific activity in late prenatal and adult stages, using a sheep model. Galactosemia acute target-organs-liver, small intestine and kidney-had the highest late prenatal activity, whereas the chronic target-organs-brain and ovary-did not exhibit a noticeable pre- or postnatal different activity compared with nontarget organs. This is the first study on GALT specific activity in the late prenatal stage for a wide variety of organs. Our findings suggest that GALT activity cannot be the sole pathogenic factor accounting for galactosemia long-term complications, and that some organs/cells might have a greater susceptibility to galactose toxicity. Anat Rec, 300:1570-1575, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ana I Coelho
- Department of Pediatrics, Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Clinical Genetics, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Jörgen Bierau
- Department of Clinical Genetics, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Martijn Lindhout
- Department of Clinical Genetics, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Jelle Achten
- Department of Clinical Genetics, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Boris W Kramer
- Department of Pediatrics/Neonatology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - M Estela Rubio-Gozalbo
- Department of Pediatrics, Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Clinical Genetics, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
24
|
Coelho AI, Rubio-Gozalbo ME, Vicente JB, Rivera I. Sweet and sour: an update on classic galactosemia. J Inherit Metab Dis 2017; 40:325-342. [PMID: 28281081 PMCID: PMC5391384 DOI: 10.1007/s10545-017-0029-3] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 02/14/2017] [Accepted: 02/20/2017] [Indexed: 02/03/2023]
Abstract
Classic galactosemia is a rare inherited disorder of galactose metabolism caused by deficient activity of galactose-1-phosphate uridylyltransferase (GALT), the second enzyme of the Leloir pathway. It presents in the newborn period as a life-threatening disease, whose clinical picture can be resolved by a galactose-restricted diet. The dietary treatment proves, however, insufficient in preventing severe long-term complications, such as cognitive, social and reproductive impairments. Classic galactosemia represents a heavy burden on patients' and their families' lives. After its first description in 1908 and despite intense research in the past century, the exact pathogenic mechanisms underlying galactosemia are still not fully understood. Recently, new important insights on molecular and cellular aspects of galactosemia have been gained, and should open new avenues for the development of novel therapeutic strategies. Moreover, an international galactosemia network has been established, which shall act as a platform for expertise and research in galactosemia. Herein are reviewed some of the latest developments in clinical practice and research findings on classic galactosemia, an enigmatic disorder with many unanswered questions warranting dedicated research.
Collapse
Affiliation(s)
- Ana I Coelho
- Department of Pediatrics and Department of Clinical Genetics, Maastricht University Medical Centre, P. Debyelaan 25, PO Box 5800, 6202 AZ, Maastricht, The Netherlands.
| | - M Estela Rubio-Gozalbo
- Department of Pediatrics and Department of Clinical Genetics, Maastricht University Medical Centre, P. Debyelaan 25, PO Box 5800, 6202 AZ, Maastricht, The Netherlands
| | - João B Vicente
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Isabel Rivera
- Metabolism & Genetics Group, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
- Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
25
|
Timson DJ. The molecular basis of galactosemia — Past, present and future. Gene 2016; 589:133-41. [DOI: 10.1016/j.gene.2015.06.077] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 06/18/2015] [Accepted: 06/29/2015] [Indexed: 12/19/2022]
|
26
|
Modulating Mobility: a Paradigm for Protein Engineering? Appl Biochem Biotechnol 2016; 181:83-90. [PMID: 27449223 DOI: 10.1007/s12010-016-2200-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 07/17/2016] [Indexed: 12/30/2022]
Abstract
Proteins are highly mobile structures. In addition to gross conformational changes occurring on, for example, ligand binding, they are also subject to constant thermal motion. The mobility of a protein varies through its structure and can be modulated by ligand binding and other events. It is becoming increasingly clear that this mobility plays an important role in key functions of proteins including catalysis, allostery, cooperativity, and regulation. Thus, in addition to an optimum structure, proteins most likely also require an optimal dynamic state. Alteration of this dynamic state through protein engineering will affect protein function. A dramatic example of this is seen in some inherited metabolic diseases where alternation of residues distant from the active site affects the mobility of the protein and impairs function. We postulate that using molecular dynamics simulations, experimental data or a combination of the two, it should be possible to engineer the mobility of active sites. This may be useful in, for example, increasing the promiscuity of enzymes. Thus, a paradigm for protein engineering is suggested in which the mobility of the active site is rationally modified. This might be combined with more "traditional" approaches such as altering functional groups in the active site.
Collapse
|
27
|
McCorvie TJ, Kopec J, Pey AL, Fitzpatrick F, Patel D, Chalk R, Shrestha L, Yue WW. Molecular basis of classic galactosemia from the structure of human galactose 1-phosphate uridylyltransferase. Hum Mol Genet 2016; 25:2234-2244. [PMID: 27005423 PMCID: PMC5081055 DOI: 10.1093/hmg/ddw091] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 03/14/2016] [Indexed: 01/22/2023] Open
Abstract
Classic galactosemia is a potentially lethal disease caused by the dysfunction of galactose 1-phosphate uridylyltransferase (GALT). Over 300 disease-associated GALT mutations have been reported, with the majority being missense changes, although a better understanding of their underlying molecular effects has been hindered by the lack of structural information for the human enzyme. Here, we present the 1.9 Å resolution crystal structure of human GALT (hGALT) ternary complex, revealing a homodimer arrangement that contains a covalent uridylylated intermediate and glucose-1-phosphate in the active site, as well as a structural zinc-binding site, per monomer. hGALT reveals significant structural differences from bacterial GALT homologues in metal ligation and dimer interactions, and therefore is a zbetter model for understanding the molecular consequences of disease mutations. Both uridylylation and zinc binding influence the stability and aggregation tendency of hGALT. This has implications for disease-associated variants where p.Gln188Arg, the most commonly detected, increases the rate of aggregation in the absence of zinc likely due to its reduced ability to form the uridylylated intermediate. As such our structure serves as a template in the future design of pharmacological chaperone therapies and opens new concepts about the roles of metal binding and activity in protein misfolding by disease-associated mutants.
Collapse
Affiliation(s)
- Thomas J McCorvie
- Structural Genomics Consortium, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ , UK
| | - Jolanta Kopec
- Structural Genomics Consortium, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ , UK
| | - Angel L Pey
- Department of Physical Chemistry, Faculty of Sciences, University of Granada, Granada E-18071, Spain
| | - Fiona Fitzpatrick
- Structural Genomics Consortium, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ , UK
| | - Dipali Patel
- Structural Genomics Consortium, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ , UK
| | - Rod Chalk
- Structural Genomics Consortium, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ , UK
| | - Leela Shrestha
- Structural Genomics Consortium, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ , UK
| | - Wyatt W Yue
- Structural Genomics Consortium, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ , UK
| |
Collapse
|
28
|
Timson DJ. Value of predictive bioinformatics in inherited metabolic diseases. World J Med Genet 2015; 5:46-51. [DOI: 10.5496/wjmg.v5.i3.46] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 04/28/2015] [Accepted: 05/18/2015] [Indexed: 02/06/2023] Open
Abstract
Typically, inherited metabolic diseases arise from point mutations in genes encoding metabolic enzymes. Although some of these mutations directly affect amino acid residues in the active sites of these enzymes, the majority do not. It is now well accepted that the majority of these disease-associated mutations exert their effects through alteration of protein stability, which causes a reduction in enzymatic activity. This finding suggests a way to predict the severity of newly discovered mutations. In silico prediction of the effects of amino acid sequence alterations on protein stability often correlates with disease severity. However, no stability prediction tool is perfect and, in general, better results are obtained if the predictions from a variety of tools are combined and then interpreted. In addition to predicted alterations to stability, the degree of conservation of a particular residue can also be a factor which needs to be taken into account: alterations to highly conserved residues are more likely to be associated with severe forms of the disease. The approach has been successfully applied in a variety of inherited metabolic diseases, but further improvements are necessary to enable robust translation into clinically useful tools.
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW Galactose - a key source of energy and a crucial structural element in complex molecules - is particularly important for early human development. However, galactose metabolism might be important not only for fetal and neonatal development but also for adulthood, as evidenced by the inherited disorders of galactose metabolism. The purpose of this review is to summarize the current evidence of galactose metabolism in health and disease. RECENT FINDINGS The biological importance of galactose goes beyond its importance as a nutrient and a metabolite. Galactose has been selected by evolutionary pressure to exert also a crucial structural role in macromolecules. Additionally, galactose has recently been reported as beneficial in a number of diseases, particularly in those affecting the brain. SUMMARY Galactose is crucial for human metabolism, with an established role in energy delivery and galactosylation of complex molecules, and evidence for other roles is emerging.
Collapse
Affiliation(s)
- Ana I Coelho
- aDepartment of Pediatrics and Laboratory Genetic Metabolic Diseases, Maastricht University Medical Center, Maastricht, The Netherlands bThe Manton Center for Orphan Disease Research, Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA *Gerard T. Berry and M. Estela Rubio-Gozalbo contributed equally to the writing of this article
| | | | | |
Collapse
|
30
|
Arginine Functionally Improves Clinically Relevant Human Galactose-1-Phosphate Uridylyltransferase (GALT) Variants Expressed in a Prokaryotic Model. JIMD Rep 2015; 23:1-6. [PMID: 25814382 DOI: 10.1007/8904_2015_420] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 01/28/2015] [Accepted: 02/09/2015] [Indexed: 12/13/2022] Open
Abstract
Classic galactosemia is a rare genetic disease of the galactose metabolism, resulting from deficient activity of galactose-1-phosphate uridylyltransferase (GALT). The current standard of care is lifelong dietary restriction of galactose, which however fails to prevent the development of long-term complications. Structural-functional studies demonstrated that the most prevalent GALT mutations give rise to proteins with increased propensity to aggregate in solution. Arginine is a known stabilizer of aggregation-prone proteins, having already shown a beneficial effect in other inherited metabolic disorders.Herein we developed a prokaryotic model of galactose sensitivity that allows evaluating in a cellular context the mutations' impact on GALT function, as well as the potential effect of arginine in functionally rescuing clinically relevant variants.This study revealed that some hGALT variants, previously described to exhibit no detectable activity in vitro, actually present residual activity when determined in vivo. Furthermore, it revealed that arginine presents a mutation-specific beneficial effect, particularly on the prevalent p.Q188R and p.K285N variants, which led us to hypothesize that it might constitute a promising therapeutic agent in classic galactosemia.
Collapse
|
31
|
Cocanougher B, Aypar U, McDonald A, Hasadsri L, Bennett MJ, Edward Highsmith W, D'Aco K. Compound heterozygosity with a novel S222N GALT mutation leads to atypical galactosemia with loss of GALT activity in erythrocytes but little evidence of clinical disease. Mol Genet Metab Rep 2015. [PMID: 28649529 PMCID: PMC5471153 DOI: 10.1016/j.ymgmr.2014.12.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Galactosemia is an inborn error of galactose metabolism caused by mutations in the GALT gene. Though early detection and galactose restriction prevent severe liver disease, affected individuals have persistently elevated biomarkers and often neuro-developmental symptoms. We present a teenage compound heterozygote for a known pathogenic mutation (H132Q) and a novel variant of unknown significance (S222N), with nearly absent erythrocyte GALT enzyme activity but normal biomarkers and only mild anxiety despite diet non-adherence. This case is similar to a previously reported S135L mutation. In this report we investigate the novel S222N variant and critically evaluate a clinically puzzling case.
Collapse
Affiliation(s)
- Benjamin Cocanougher
- University of Rochester School of Medicine and Dentistry, Department of Pediatrics, Division of Genetics, Rochester, NY, USA
| | - Umut Aypar
- Molecular Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Amber McDonald
- Molecular Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Linda Hasadsri
- Molecular Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Michael J Bennett
- Metabolic Disease Laboratory, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - W Edward Highsmith
- Molecular Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Kristin D'Aco
- University of Rochester School of Medicine and Dentistry, Department of Pediatrics, Division of Genetics, Rochester, NY, USA
| |
Collapse
|
32
|
UDP-galactose 4'-epimerase from the liver fluke, Fasciola hepatica: biochemical characterization of the enzyme and identification of inhibitors. Parasitology 2014; 142:463-72. [PMID: 25124392 DOI: 10.1017/s003118201400136x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Leloir pathway enzyme uridine diphosphate (UDP)-galactose 4'-epimerase from the common liver fluke Fasciola hepatica (FhGALE) was identified and characterized. The enzyme can be expressed in, and purified from, Escherichia coli. The recombinant enzyme is active: the K(m) (470 μM) is higher than the corresponding human enzyme (HsGALE), whereas the k(cat) (2.3 s(-1)) is substantially lower. FhGALE binds NAD(+) and has shown to be dimeric by analytical gel filtration. Like the human and yeast GALEs, FhGALE is stabilized by the substrate UDP-galactose. Molecular modelling predicted that FhGALE adopts a similar overall fold to HsGALE and that tyrosine 155 is likely to be the catalytically critical residue in the active site. In silico screening of the National Cancer Institute Developmental Therapeutics Program library identified 40 potential inhibitors of FhGALE which were tested in vitro. Of these, 6 showed concentration-dependent inhibition of FhGALE, some with nanomolar IC50 values. Two inhibitors (5-fluoroorotate and N-[(benzyloxy)carbonyl]leucyltryptophan) demonstrated selectivity for FhGALE over HsGALE. These compounds also thermally destabilized FhGALE in a concentration-dependent manner. Interestingly, the selectivity of 5-fluoroorotate was not shown by orotic acid, which differs in structure by 1 fluorine atom. These results demonstrate that, despite the structural and biochemical similarities of FhGALE and HsGALE, it is possible to discover compounds which preferentially inhibit FhGALE.
Collapse
|
33
|
d'Acierno A, Facchiano A, Marabotti A. GALT protein database: querying structural and functional features of GALT enzyme. Hum Mutat 2014; 35:1060-7. [PMID: 24990533 DOI: 10.1002/humu.22613] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 06/24/2014] [Indexed: 01/19/2023]
Abstract
Knowledge of the impact of variations on protein structure can enhance the comprehension of the mechanisms of genetic diseases related to that protein. Here, we present a new version of GALT Protein Database, a Web-accessible data repository for the storage and interrogation of structural effects of variations of the enzyme galactose-1-phosphate uridylyltransferase (GALT), the impairment of which leads to classic Galactosemia, a rare genetic disease. This new version of this database now contains the models of 201 missense variants of GALT enzyme, including heterozygous variants, and it allows users not only to retrieve information about the missense variations affecting this protein, but also to investigate their impact on substrate binding, intersubunit interactions, stability, and other structural features. In addition, it allows the interactive visualization of the models of variants collected into the database. We have developed additional tools to improve the use of the database by nonspecialized users. This Web-accessible database (http://bioinformatica.isa.cnr.it/GALT/GALT2.0) represents a model of tools potentially suitable for application to other proteins that are involved in human pathologies and that are subjected to genetic variations.
Collapse
|
34
|
Coelho AI, Trabuco M, Ramos R, Silva MJ, Tavares de Almeida I, Leandro P, Rivera I, Vicente JB. Functional and structural impact of the most prevalent missense mutations in classic galactosemia. Mol Genet Genomic Med 2014; 2:484-96. [PMID: 25614870 PMCID: PMC4303218 DOI: 10.1002/mgg3.94] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 05/20/2014] [Accepted: 05/22/2014] [Indexed: 11/10/2022] Open
Abstract
Galactose-1-phosphate uridylyltransferase (GALT) is a key enzyme in galactose metabolism, particularly important in the neonatal period due to ingestion of galactose-containing milk. GALT deficiency results in the genetic disorder classic galactosemia, whose pathophysiology is still not fully elucidated. Whereas classic galactosemia has been hypothesized to result from GALT misfolding, a thorough functional-structural characterization of GALT most prevalent variants was still lacking, hampering the development of alternative therapeutic approaches. The aim of this study was to investigate the structural-functional effects of nine GALT mutations, four of which account for the vast majority of the mutations identified in galactosemic patients. Several methodologies were employed to evaluate the mutations' impact on GALT function, on the protein secondary and tertiary structures, and on the aggregation propensity. The major structural effect concerns disturbed propensity for aggregation, particularly striking for the p.Q188R variant, resulting from the most frequent (∼60%) allele at a worldwide scale. The absence of major effects at the secondary and tertiary structure levels suggests that the disturbed aggregation results from subtle perturbations causing a higher and/or longer exposure of hydrophobic residues in the variants as compared to WT GALT. The results herein described indicate a possible benefit from introducing proteostasis regulators and/or chemical/pharmacological chaperones to prevent the accumulation of protein aggregates, in new avenues of therapeutic research for classic galactosemia.
Collapse
Affiliation(s)
- Ana I Coelho
- Metabolism and Genetics Group, Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, University of Lisbon 1649-003, Lisbon, Portugal
| | - Matilde Trabuco
- Metabolism and Genetics Group, Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, University of Lisbon 1649-003, Lisbon, Portugal
| | - Ruben Ramos
- Metabolism and Genetics Group, Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, University of Lisbon 1649-003, Lisbon, Portugal
| | - Maria João Silva
- Metabolism and Genetics Group, Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, University of Lisbon 1649-003, Lisbon, Portugal ; Department of Biochemistry and Human Biology, Faculty of Pharmacy, University of Lisbon 1649-003, Lisbon, Portugal
| | - Isabel Tavares de Almeida
- Metabolism and Genetics Group, Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, University of Lisbon 1649-003, Lisbon, Portugal ; Department of Biochemistry and Human Biology, Faculty of Pharmacy, University of Lisbon 1649-003, Lisbon, Portugal
| | - Paula Leandro
- Metabolism and Genetics Group, Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, University of Lisbon 1649-003, Lisbon, Portugal ; Department of Biochemistry and Human Biology, Faculty of Pharmacy, University of Lisbon 1649-003, Lisbon, Portugal
| | - Isabel Rivera
- Metabolism and Genetics Group, Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, University of Lisbon 1649-003, Lisbon, Portugal ; Department of Biochemistry and Human Biology, Faculty of Pharmacy, University of Lisbon 1649-003, Lisbon, Portugal
| | - João B Vicente
- Metabolism and Genetics Group, Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, University of Lisbon 1649-003, Lisbon, Portugal ; Department of Biochemistry and Human Biology, Faculty of Pharmacy, University of Lisbon 1649-003, Lisbon, Portugal
| |
Collapse
|
35
|
Megarity CF, Gill JRE, Caraher MC, Stratford IJ, Nolan KA, Timson DJ. The two common polymorphic forms of human NRH-quinone oxidoreductase 2 (NQO2) have different biochemical properties. FEBS Lett 2014; 588:1666-72. [PMID: 24631540 PMCID: PMC4045209 DOI: 10.1016/j.febslet.2014.02.063] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 02/18/2014] [Accepted: 02/21/2014] [Indexed: 10/25/2022]
Abstract
There are two common forms of NRH-quinone oxidoreductase 2 (NQO2) in the human population resulting from SNP rs1143684. One has phenylalanine at position 47 (NQO2-F47) and the other leucine (NQO2-L47). Using recombinant proteins, we show that these variants have similar steady state kinetic parameters, although NQO2-L47 has a slightly lower specificity constant. NQO2-L47 is less stable towards proteolytic digestion and thermal denaturation than NQO2-F47. Both forms are inhibited by resveratrol, but NQO2-F47 shows negative cooperativity with this inhibitor. Thus these data demonstrate, for the first time, clear biochemical differences between the variants which help explain previous biomedical and epidemiological findings.
Collapse
Affiliation(s)
- Clare F Megarity
- School of Biological Sciences, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - James R E Gill
- School of Biological Sciences, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - M Clare Caraher
- School of Biological Sciences, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK; Manchester Pharmacy School, The University of Manchester, Stopford Building, Oxford Road, Manchester M13 9PT, UK
| | - Ian J Stratford
- Manchester Pharmacy School, The University of Manchester, Stopford Building, Oxford Road, Manchester M13 9PT, UK
| | - Karen A Nolan
- Manchester Pharmacy School, The University of Manchester, Stopford Building, Oxford Road, Manchester M13 9PT, UK
| | - David J Timson
- School of Biological Sciences, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK.
| |
Collapse
|
36
|
Biochemical characterisation of glyceraldehyde 3-phosphate dehydrogenase (GAPDH) from the liver fluke, Fasciola hepatica. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2014; 1844:744-9. [PMID: 24566472 DOI: 10.1016/j.bbapap.2014.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 02/07/2014] [Accepted: 02/12/2014] [Indexed: 11/22/2022]
Abstract
Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) catalyses one of the two steps in glycolysis which generate the reduced coenzyme NADH. This reaction precedes the two ATP generating steps. Thus, inhibition of GAPDH will lead to substantially reduced energy generation. Consequently, there has been considerable interest in developing GAPDH inhibitors as anti-cancer and anti-parasitic agents. Here, we describe the biochemical characterisation of GAPDH from the common liver fluke Fasciola hepatica (FhGAPDH). The primary sequence of FhGAPDH is similar to that from other trematodes and the predicted structure shows high similarity to those from other animals including the mammalian hosts. FhGAPDH lacks a binding pocket which has been exploited in the design of novel antitrypanosomal compounds. The protein can be expressed in, and purified from Escherichia coli; the recombinant protein was active and showed no cooperativity towards glyceraldehyde 3-phosphate as a substrate. In the absence of ligands, FhGAPDH was a mixture of homodimers and tetramers, as judged by protein-protein crosslinking and analytical gel filtration. The addition of either NAD⁺ or glyceraldehyde 3-phosphate shifted this equilibrium towards a compact dimer. Thermal scanning fluorimetry demonstrated that this form was considerably more stable than the unliganded one. These responses to ligand binding differ from those seen in mammalian enzymes. These differences could be exploited in the discovery of reagents which selectively disrupt the function of FhGAPDH.
Collapse
|
37
|
SUZUKI Y. Emerging novel concept of chaperone therapies for protein misfolding diseases. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2014; 90:145-62. [PMID: 24814990 PMCID: PMC4104511 DOI: 10.2183/pjab.90.145] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 03/14/2014] [Indexed: 06/03/2023]
Abstract
Chaperone therapy is a newly developed molecular therapeutic approach to protein misfolding diseases. Among them we found unstable mutant enzyme proteins in a few lysosomal diseases, resulting in rapid intracellular degradation and loss of function. Active-site binding low molecular competitive inhibitors (chemical chaperones) paradoxically stabilized and enhanced the enzyme activity in somatic cells by correction of the misfolding of enzyme protein. They reached the brain through the blood-brain barrier after oral administration, and corrected pathophysiology of the disease. In addition to these inhibitory chaperones, non-competitive chaperones without inhibitory bioactivity are being developed. Furthermore molecular chaperone therapy utilizing the heat shock protein and other chaperone proteins induced by small molecules has been experimentally tried to handle abnormally accumulated proteins as a new approach particularly to neurodegenerative diseases. These three types of chaperones are promising candidates for various types of diseases, genetic or non-genetic, and neurological or non-neurological, in addition to lysosomal diseases.
Collapse
Affiliation(s)
- Yoshiyuki SUZUKI
- Special Visiting Scientist, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.
| |
Collapse
|
38
|
Abstract
Galactosemia is an inherited metabolic disease in which galactose is not properly metabolised. There are various theories to explain the molecular pathology, and recent experimental evidence strongly suggests that oxidative stress plays a key role. High galactose diets are damaging to experimental animals and oxidative stress also plays a role in this toxicity which can be alleviated by purple sweet potato colour (PSPC). This plant extract is rich in acetylated anthocyanins which have been shown to quench free radical production. The objective of this Commentary is to advance the hypothesis that PSPC, or compounds therefrom, may be a viable basis for a novel therapy for galactosemia.
Collapse
Affiliation(s)
- David J Timson
- School of Biological Sciences, Medical Biology Centre, Institute for Global Food Security, Queen's University Belfast , Belfast , UK
| |
Collapse
|
39
|
Zinsser VL, Hoey EM, Trudgett A, Timson DJ. Biochemical characterisation of triose phosphate isomerase from the liver fluke Fasciola hepatica. Biochimie 2013; 95:2182-9. [DOI: 10.1016/j.biochi.2013.08.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 08/07/2013] [Indexed: 11/29/2022]
|
40
|
De-Souza EA, Pimentel FSA, Machado CM, Martins LS, da-Silva WS, Montero-Lomelí M, Masuda CA. The unfolded protein response has a protective role in yeast models of classic galactosemia. Dis Model Mech 2013; 7:55-61. [PMID: 24077966 PMCID: PMC3882048 DOI: 10.1242/dmm.012641] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Classic galactosemia is a human autosomal recessive disorder caused by mutations in the GALT gene (GAL7 in yeast), which encodes the enzyme galactose-1-phosphate uridyltransferase. Here we show that the unfolded protein response pathway is triggered by galactose in two yeast models of galactosemia: lithium-treated cells and the gal7Δ mutant. The synthesis of galactose-1-phosphate is essential to trigger the unfolded protein response under these conditions because the deletion of the galactokinase-encoding gene GAL1 completely abolishes unfolded protein response activation and galactose toxicity. Impairment of the unfolded protein response in both yeast models makes cells even more sensitive to galactose, unmasking its cytotoxic effect. These results indicate that endoplasmic reticulum stress is induced under galactosemic conditions and underscores the importance of the unfolded protein response pathway to cellular adaptation in these models of classic galactosemia.
Collapse
Affiliation(s)
- Evandro A De-Souza
- Instituto de Bioquímica Médica Leopoldo de Meis, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-590, Brazil
| | | | | | | | | | | | | |
Collapse
|
41
|
Zinsser VL, Farnell E, Dunne DW, Timson DJ. Triose phosphate isomerase from the blood flukeSchistosoma mansoni: Biochemical characterisation of a potential drug and vaccine target. FEBS Lett 2013; 587:3422-7. [DOI: 10.1016/j.febslet.2013.09.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 09/04/2013] [Accepted: 09/11/2013] [Indexed: 11/26/2022]
|