1
|
Kumari V, Antony S, Panchal K, Vaidya S, Vaswani RK, David JJE, Marandi S. Pancytopenia due to Transcobalamin II Deficiency (TCN2D). Indian J Pediatr 2025:10.1007/s12098-025-05514-w. [PMID: 40148655 DOI: 10.1007/s12098-025-05514-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025]
Affiliation(s)
- Vimlesh Kumari
- Department of Pediatrics, Seth G.S. Medical College and K.E.M Hospital, Mumbai, India
| | - Sonu Antony
- Department of Pediatrics, Seth G.S. Medical College and K.E.M Hospital, Mumbai, India
| | - Kunj Panchal
- Department of Pediatrics, Seth G.S. Medical College and K.E.M Hospital, Mumbai, India.
| | - Sneha Vaidya
- Department of Pediatrics, Seth G.S. Medical College and K.E.M Hospital, Mumbai, India
| | - Rajwanti K Vaswani
- Department of Pediatrics, Seth G.S. Medical College and K.E.M Hospital, Mumbai, India
| | - Jane J E David
- Department of Pediatrics, Seth G.S. Medical College and K.E.M Hospital, Mumbai, India
| | - Shweta Marandi
- Department of Pediatrics, Seth G.S. Medical College and K.E.M Hospital, Mumbai, India
| |
Collapse
|
2
|
Griffith CM, Conrotte JF, Paydar P, Xie X, Heins-Marroquin U, Gavotto F, Jäger C, Ellens KW, Linster CL. CLYBL averts vitamin B 12 depletion by repairing malyl-CoA. Nat Chem Biol 2025:10.1038/s41589-025-01857-9. [PMID: 40108300 DOI: 10.1038/s41589-025-01857-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 02/12/2025] [Indexed: 03/22/2025]
Abstract
Citrate lyase beta-like protein (CLYBL) is a ubiquitously expressed mammalian enzyme known for its role in the degradation of itaconate, a bactericidal immunometabolite produced in activated macrophages. The association of CLYBL loss of function with reduced circulating vitamin B12 levels was proposed to result from inhibition of the B12-dependent enzyme methylmalonyl-CoA mutase by itaconyl-CoA. The discrepancy between the highly inducible and locally confined production of itaconate and the broad expression profile of CLYBL across tissues suggested a role for this enzyme beyond itaconate catabolism. Here we discover that CLYBL additionally functions as a metabolite repair enzyme for malyl-CoA, a side product of promiscuous citric acid cycle enzymes. We found that CLYBL knockout cells, accumulating malyl-CoA but not itaconyl-CoA, show decreased levels of adenosylcobalamin and that malyl-CoA is a more potent inhibitor of methylmalonyl-CoA mutase than itaconyl-CoA. Our work thus suggests that malyl-CoA plays a role in the B12 deficiency observed in individuals with CLYBL loss of function.
Collapse
Affiliation(s)
- Corey M Griffith
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Jean-François Conrotte
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Parisa Paydar
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Xinqiang Xie
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Ursula Heins-Marroquin
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Floriane Gavotto
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Christian Jäger
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Kenneth W Ellens
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Carole L Linster
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
3
|
Plecko B. Inherited disorders of vitamin metabolism. Eur J Paediatr Neurol 2025; 55:18-32. [PMID: 40096763 DOI: 10.1016/j.ejpn.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 02/14/2025] [Accepted: 02/15/2025] [Indexed: 03/19/2025]
Abstract
Vitamins are essential cofactors of various enzyme reactions in amino acid, neurotransmitter, nucleotide and energy metabolism. Over the past decade a number of inborn errors of metabolism have been identified, that affect different steps in vitamin absorption, transport, activation or recycling and repair of active vitamin cofactors. According to the respective cofactor function this may result in acute or chronic multisystem disease or in disorders that selectively affect the nervous system. Most of these disorders are amenable to specific treatment with excellent results, but diagnostic delay can lead to rapid, irreversible damage or even death. Therefore, especially in case of acute and severe neurologic presentations compatible with one of the here discused disorders, a vitamin trial should be considered while awaiting results of biochemical and genetic testing. Diagnosis of these disorders is especially rewarding, as treatment is often per oral, available worldwide and comparably cheap. This article will review current knowledge of the clinical presentation, biomarkers and specific treatment of inborn errors of vitamin metabolism and illustrates why child neurologists should have vitamins in their pockets.
Collapse
Affiliation(s)
- Barbara Plecko
- Department of Pediatrics and Adolescent Medicine Division of General Pediatrics Medical University of Graz, Auenbruggerplatz 34/2, 8036, Graz, Austria.
| |
Collapse
|
4
|
Belhaj R, Maaloul I, Kolsi R, Rekik T, Chabchoub I, Aloulou H, Kamoun T. Study of clinical manifestations and etiologies of megaloblastic anemia in children. Transfus Clin Biol 2025:S1246-7820(25)00006-0. [PMID: 39842709 DOI: 10.1016/j.tracli.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 01/24/2025]
Abstract
BACKGROUND AND AIM Megaloblastic anemia (MA) is a rare pathology in childhood due, in the majority of cases, to a deficiency of folic acid and/or vitamin B12 (cobalamin). This study aims to determine the epidemiological, clinical, and paraclinical profiles of MA in children and to specify its etiologies, therapeutic modalities, and treatment responses. METHODS This is a retrospective descriptive study of MA cases in children carried out in the General Pediatrics Department of the Hedi Chaker University Hospital of Sfax over a period of 42 years, from January 1979 to December 2021. We included all the patients under 16 years old with a myelogram showing megaloblastosis. The selected patients' demographic characteristics, physical signs, laboratory findings, and treatment responses were recorded. RESULTS Twenty cases of MA were collected, including 11 boys and 9 girls. The incidence of MA in children was 0.014%. The median age at diagnosis was 3.37 years. The clinical presentation was anemic syndrome with pallor and asthenia in all the cases. Neurological manifestations were noted in 2 cases and digestive disorders in 10 cases. Seven infants had psychomotor delays. On admission, all our patients had anemia with an average value of 5.6 g/dl. It was macrocytic in 19 cases. The blood count also revealed leukopenia (7 cases), thrombocytopenia (10 cases), and pancytopenia (5 cases). The myelogram showed megaloblastosis in all the cases and sideroblasts in one. A brain MRI was performed on five patients, and it showed abnormalities in three cases. The etiological investigations revealed a vitamin B12 deficiency secondary to a maternal Biermer's disease (6 cases), malnutrition (3 cases), Imerslund's disease (3 cases), congenital deficiency in transcobalamin II (3 cases), Biermer's disease (1 case), giardiasis (1 case), folic acid deficiency secondary to a poor dietary intake (1 case), mitochondrial cytopathy with vitamin B12-Folic acid deficiency (1 case), and Pearson syndrome (1 case). Our treatment included symptomatic measures, replacement therapy, and etiological treatment. Favorable evolution was noted in 11 cases. Five patients had neurological sequelae, and one patient died. CONCLUSION Our study highlights the rarity and heterogeneity of the etiological contexts of MA in children. Early diagnosis and therapeutic support can improve the long-term neurological prognosis.
Collapse
Affiliation(s)
- Rim Belhaj
- Department of Pediatrics, Hedi Chaker University Hospital of Sfax, Tunisia; Faculty of Medecine, University of Sfax, Tunisia
| | - Ines Maaloul
- Department of Pediatrics, Hedi Chaker University Hospital of Sfax, Tunisia; Faculty of Medecine, University of Sfax, Tunisia.
| | - Roeya Kolsi
- Department of Pediatrics, Hedi Chaker University Hospital of Sfax, Tunisia; Faculty of Medecine, University of Sfax, Tunisia
| | - Taicir Rekik
- Faculty of Medecine, University of Sfax, Tunisia; Regional Center of Transfusion of Sfax, Tunisia
| | - Imen Chabchoub
- Department of Pediatrics, Hedi Chaker University Hospital of Sfax, Tunisia; Faculty of Medecine, University of Sfax, Tunisia
| | - Hajer Aloulou
- Department of Pediatrics, Hedi Chaker University Hospital of Sfax, Tunisia; Faculty of Medecine, University of Sfax, Tunisia
| | - Thouraya Kamoun
- Department of Pediatrics, Hedi Chaker University Hospital of Sfax, Tunisia; Faculty of Medecine, University of Sfax, Tunisia
| |
Collapse
|
5
|
Habib A, Idrus H, Malik NAA, Nor AM, Nasohah SM, Moey LH, Hian LS, Hock NL, Azize NAA. Clinical, biochemical, molecular characteristics and clinical outcome of hyperhomocysteinemia in Malaysian children. Clin Biochem 2024; 133-134:110828. [PMID: 39322052 DOI: 10.1016/j.clinbiochem.2024.110828] [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: 05/14/2024] [Revised: 09/20/2024] [Accepted: 09/22/2024] [Indexed: 09/27/2024]
Abstract
BACKGROUND Hyperhomocysteinemia can be due to various abnormalities of the complex interaction of methionine, folate and vitamin B12. It has been known to be a cardiovascular risk factor. This study aims to review the clinical presentation, underlying causes and clinical outcome in paediatric patients diagnosed with significant hyperhomocysteinemia in Malaysia. DESIGN AND METHODS Data were obtained from the medical records and the laboratory information system. Paediatric patients with significant hyperhomocysteinemia were identified from a selective high-risk screening of 96,721 patients, performed between 2010 and 2022. Inclusion criteria for the study were paediatric patients with significant hyperhomocysteinemia (>40 µmol/L). RESULTS Sixteen patients were identified. The average total homocysteine (tHcy) and methionine were 269 µmol/L and 499 µmol/L in cystathionine β-synthase deficiency (CBS), 127 µmol/L and 29 µmol/L in patients with remethylation defects and 390 µmol/L and 4 µmol/L in congenital B12 deficiency. We found c.609G>A as the most prevalent mutation in MMACHC gene and possible novel mutations for CBS (c.402del, c.1333C>T and c.1031T>G) and MTHFR genes (c.266T>A and c.1249del). Further subclassification revealed CBS was 5/16 patients (31 %), remethylation defects was 9/16 (56 %) and congenital B12 deficiency was 2/16 (13 %). All patients received standard treatment and regular monitoring of the main biomarkers. The average age at the time of diagnosis were 9.2 years (CBS) and 1.2 years (remethylation defects). Congenital B12 deficiency had slight delay in milestones, remethylation defects had mild to moderate learning disabilities, CBS had variable degree of intellectual disability, delayed milestones, ophthalmological abnormalities, and thrombosis at an early adolescent/adulthood. CONCLUSIONS The majority of significant hyperhomocysteinemia in Malaysian children was due to remethylation defects. Screening for hyperhomocysteinemia in Malaysian children is recommended for earlier treatment and improved clinical outcome.
Collapse
Affiliation(s)
- Anasufiza Habib
- Biochemistry Unit, Biochemistry & Genomics Research Centre, Institute for Medical Research, National Institutes of Health, Kuala Lumpur, Malaysia.
| | - Hamizah Idrus
- Biochemistry Unit, Biochemistry & Genomics Research Centre, Institute for Medical Research, National Institutes of Health, Kuala Lumpur, Malaysia
| | - Nur Aisyah Abdul Malik
- Biochemistry Unit, Biochemistry & Genomics Research Centre, Institute for Medical Research, National Institutes of Health, Kuala Lumpur, Malaysia
| | - Ainna Mohd Nor
- Biochemistry Unit, Biochemistry & Genomics Research Centre, Institute for Medical Research, National Institutes of Health, Kuala Lumpur, Malaysia
| | - Sofwatul Muktaroh Nasohah
- Biochemistry Unit, Biochemistry & Genomics Research Centre, Institute for Medical Research, National Institutes of Health, Kuala Lumpur, Malaysia
| | - Lip Hen Moey
- Department of Genetic, Hospital Pulau Pinang, Ministry of Health, Pulau Pinang, Malaysia
| | - Lua Seok Hian
- Molecular Diagnostics Unit, Biochemistry & Genomics Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health, Kuala Lumpur, Malaysia
| | - Ngu Lock Hock
- Department of Genetic, Hospital Kuala Lumpur, Ministry of Health, Kuala Lumpur, Malaysia
| | - Nor Azimah Abdul Azize
- Molecular Diagnostics Unit, Biochemistry & Genomics Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health, Kuala Lumpur, Malaysia
| |
Collapse
|
6
|
Georgiou T, Grafakou O, Malekkou A, Athanasiou E, Ioannou I, Choleva V, Dionysiou M, Mavrikiou G, Demetriadou A, Anastasiadou V, Drousiotou A, Petrou PP. A case series of Cypriot patients with CblC defect: Clinical, biochemical and molecular characteristics. Mol Genet Metab Rep 2024; 41:101158. [PMID: 39584041 PMCID: PMC11585715 DOI: 10.1016/j.ymgmr.2024.101158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/05/2024] [Accepted: 11/05/2024] [Indexed: 11/26/2024] Open
Abstract
Methylmalonic aciduria and homocystinuria, CblC type, is an inborn error of intracellular vitamin B12 (cobalamin) metabolism caused, in the majority of cases, by mutations in the MMACHC gene. Five Cypriot patients (four males and one female) were diagnosed with a CblC defect. Age at diagnosis ranged from 10 days to 9 months. We present here the clinical, biochemical and molecular findings of these patients. Our retrospective study indicates that all patients were carriers of the known p.Arg91LysfsTer14 variant in either a homozygous or compound heterozygous state with other known MMACHC pathogenic variants. Out of three patients sharing the same genotype the one diagnosed and initiated treatment in the neonatal period displayed an improved clinical outcome.
Collapse
Affiliation(s)
- Theodoros Georgiou
- Biochemical Genetics Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Olga Grafakou
- Inborn Errors of Metabolism Clinic, Archbishop Makarios III Hospital, Nicosia, Cyprus
| | - Anna Malekkou
- Biochemical Genetics Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Emilia Athanasiou
- Clinical Genetics Department, Archbishop Makarios III Hospital, Nicosia, Cyprus
| | - Ioannis Ioannou
- Paediatric Neurology Clinic, Archbishop Makarios III Hospital, Nicosia, Cyprus
| | - Vivi Choleva
- Ophthalmology Clinic, Archbishop Makarios III Hospital, Nicosia, Cyprus
| | - Maria Dionysiou
- Biochemical Genetics Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Gabriella Mavrikiou
- Biochemical Genetics Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Anthi Demetriadou
- Biochemical Genetics Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | | | - Anthi Drousiotou
- Biochemical Genetics Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Petros P. Petrou
- Biochemical Genetics Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| |
Collapse
|
7
|
Ahmed S, Cai L, Akbar F, Siddiqui A, DeBerardinis RJ, Ni M, Vu H, Afroze B. Evaluation of the clinical, biochemical, and molecular spectrum of Cobalamin C (CblC) defect in 33 patients from Pakistan. Scand J Clin Lab Invest 2024; 84:391-397. [PMID: 39225018 DOI: 10.1080/00365513.2024.2394983] [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: 05/18/2024] [Revised: 08/13/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Cobalamin C is the most common inborn error of intracellular cobalamin metabolism caused by biallelic pathogenic variants in the MMACHC gene, leading to impaired conversion of dietary vitamin B12 into its two metabolically active forms, methylcobalamin and adenosylcobalamin. Biochemical hallmarks are elevated plasma total homocysteine (HCYs) and low methionine accompanied by methylmalonic aciduria. This study aimed to evaluate the clinical, biochemical, and molecular analysis of Pakistani patients with CblC defect. METHODS Medical charts, urine organic acid (UOA) chromatograms, plasma amino acid levels, plasma tHcy and MMACHC gene results of patients presenting at the Biochemical Genetics Clinic, AKUH from 2013-2021 were reviewed. Details were collected on a pre-structured questionnaire. SPSS 22 was used for data analysis. RESULTS CblC was found in 33 cases (Male:Female 19:14). The median age of symptoms onset and diagnosis were 300 (IQR:135-1800) and 1380 (IQR: 240-2730) days. The most common clinical features were cognitive impairment (n = 29), seizures (n = 23), motor developmental delay (n = 20), hypotonia (n = 17), and sparse/hypopigmented scalp hair (n = 16). The MMACHC gene sequencing revealed homozygous pathogenic variant c.394C > T, (p.Arg132*) in 32 patients, whereas c.609G > A, (p.TRP203*) in one patient whose ancestors had settled in Pakistan from China decades ago. The median age of treatment initiation was 1530 (IQR: 240-2790). The median pre-treatment HCYs levels were 134 (IQR:87.2-155.5) compared to post-treatment levels of 33.3 (IQR: 27.3-44.95) umol/L. CONCLUSIONS Thirty-three cases of CblC defect from a single center underscores a significant number of the disorder within Pakistan. Late diagnosis emphasizes the need for increased clinical awareness and adequate diagnostic facilities.
Collapse
Affiliation(s)
- Sibtain Ahmed
- Section of Chemical Pathology, Department of Pathology and Laboratory Medicine, Aga Khan University
| | - Ling Cai
- Quantitative Biomedical Research Center, Peter O'Donnell School of Public Health, UT Southwestern, TX, USA
| | - Fizza Akbar
- Department of Paediatrics & Child Health, Aga Khan University Hospital, Karachi
| | - Ayra Siddiqui
- Medical College, Aga Khan University Hospital, Karachi
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute at UT Southwestern, Texas, USA
- Howard Hughes Medical Institute, UT Southwestern, Texas, USA
| | - Min Ni
- Children's Medical Center Research Institute at UT Southwestern, Texas, USA
| | - Hieu Vu
- Children's Medical Center Research Institute at UT Southwestern, Texas, USA
| | - Bushra Afroze
- Quantitative Biomedical Research Center, Peter O'Donnell School of Public Health, UT Southwestern, TX, USA
| |
Collapse
|
8
|
Demaret T, Bédard K, Soucy JF, Watkins D, Allard P, Levtova A, O'Brien A, Brunel-Guitton C, Rosenblatt DS, Mitchell GA. The MMACHC variant c.158T>C: Mild clinical and biochemical phenotypes and marked hydroxocobalamin response in cblC patients. Mol Genet Metab 2024; 142:108345. [PMID: 38387306 DOI: 10.1016/j.ymgme.2024.108345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 02/24/2024]
Abstract
Mutations in MMACHC cause cobalamin C disease (cblC, OMIM 277400), the commonest inborn error of vitamin B12 metabolism. In cblC, deficient activation of cobalamin results in methylcobalamin and adenosylcobalamin deficiency, elevating methylmalonic acid (MMA) and total plasma homocysteine (tHcy). We retrospectively reviewed the medical files of seven cblC patients: three compound heterozygotes for the MMACHC (NM_015506.3) missense variant c.158T>C p.(Leu53Pro) in trans with the common pathogenic mutation c.271dupA (p.(Arg91Lysfs*14), "compounds"), and four c.271dupA homozygotes ("homozygotes"). Compounds receiving hydroxocobalamin intramuscular injection monotherapy had age-appropriate psychomotor performance and normal ophthalmological examinations. In contrast, c.271dupA homozygotes showed marked psychomotor retardation, retinopathy and feeding problems despite penta-therapy (hydroxocobalamin, betaine, folinic acid, l-carnitine and acetylsalicylic acid). Pretreatment levels of plasma and urine MMA and tHcy were higher in c.271dupA homozygotes than in compounds. Under treatment, levels of the compounds approached or entered the reference range but not those of c.271dupA homozygotes (tHcy: compounds 9.8-32.9 μM, homozygotes 41.6-106.8 (normal (N) < 14); plasma MMA: compounds 0.14-0.81 μM, homozygotes, 10.4-61 (N < 0.4); urine MMA: compounds 1.75-48 mmol/mol creatinine, homozygotes 143-493 (N < 10)). Patient skin fibroblasts all had low cobalamin uptake, but this was milder in compound cells. Also, the distribution pattern of cobalamin species was qualitatively different between cells from compounds and from homozygotes. Compared to the classic cblC phenotype presented by c.271dupA homozygous patients, c.[158T>C];[271dupA] compounds had mild clinical and biochemical phenotypes and responded strikingly to hydroxocobalamin monotherapy.
Collapse
Affiliation(s)
- Tanguy Demaret
- Medical Genetics Division, Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montréal, Québec, Canada; Centre de Génétique Humaine, Institut de Pathologie et Génétique, Gosselies, Belgium
| | - Karine Bédard
- Medical Genetics Division, Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montréal, Québec, Canada; Laboratoire de Diagnostic Moléculaire, Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada; Département de Pathologie et Biologie Cellulaire, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Jean-François Soucy
- Medical Genetics Division, Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montréal, Québec, Canada
| | - David Watkins
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada; Department of Medical Genetics, McGill University Health Centre, Montreal, Quebec, Canada
| | - Pierre Allard
- Medical Genetics Division, Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montréal, Québec, Canada; Department of Biochemistry, CHU Sainte-Justine, Montréal, Québec, Canada
| | - Alina Levtova
- Service de Médecine Génique, Département de Médecine, Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada
| | - Alan O'Brien
- Service de Médecine Génique, Département de Médecine, Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada
| | - Catherine Brunel-Guitton
- Medical Genetics Division, Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montréal, Québec, Canada; Division of Biochemical Genetics, Department of Pediatrics, University of British Columbia, BC Children's Hospital, Vancouver, British Columbia, Canada
| | - David S Rosenblatt
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada; Department of Medical Genetics, McGill University Health Centre, Montreal, Quebec, Canada
| | - Grant A Mitchell
- Medical Genetics Division, Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
9
|
Mathew AR, Di Matteo G, La Rosa P, Barbati SA, Mannina L, Moreno S, Tata AM, Cavallucci V, Fidaleo M. Vitamin B12 Deficiency and the Nervous System: Beyond Metabolic Decompensation-Comparing Biological Models and Gaining New Insights into Molecular and Cellular Mechanisms. Int J Mol Sci 2024; 25:590. [PMID: 38203763 PMCID: PMC10778862 DOI: 10.3390/ijms25010590] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/16/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Vitamin B12 (VitB12) is a micronutrient and acts as a cofactor for fundamental biochemical reactions: the synthesis of succinyl-CoA from methylmalonyl-CoA and biotin, and the synthesis of methionine from folic acid and homocysteine. VitB12 deficiency can determine a wide range of diseases, including nervous system impairments. Although clinical evidence shows a direct role of VitB12 in neuronal homeostasis, the molecular mechanisms are yet to be characterized in depth. Earlier investigations focused on exploring the biochemical shifts resulting from a deficiency in the function of VitB12 as a coenzyme, while more recent studies propose a broader mechanism, encompassing changes at the molecular/cellular levels. Here, we explore existing study models employed to investigate the role of VitB12 in the nervous system, including the challenges inherent in replicating deficiency/supplementation in experimental settings. Moreover, we discuss the potential biochemical alterations and ensuing mechanisms that might be modified at the molecular/cellular level (such as epigenetic modifications or changes in lysosomal activity). We also address the role of VitB12 deficiency in initiating processes that contribute to nervous system deterioration, including ROS accumulation, inflammation, and demyelination. Consequently, a complex biological landscape emerges, requiring further investigative efforts to grasp the intricacies involved and identify potential therapeutic targets.
Collapse
Affiliation(s)
- Aimee Rachel Mathew
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (A.R.M.); (A.M.T.)
| | - Giacomo Di Matteo
- Department of Chemistry and Technology of Drugs, Sapienza University of Rome, 00185 Rome, Italy; (G.D.M.); (L.M.)
| | - Piergiorgio La Rosa
- Division of Neuroscience, Department of Psychology, Sapienza University of Rome, 00185 Rome, Italy;
- European Center for Brain Research, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy
| | - Saviana Antonella Barbati
- Departmental Faculty of Medicine and Surgery, UniCamillus-Saint Camillus International University of Health Sciences, 00131 Rome, Italy;
| | - Luisa Mannina
- Department of Chemistry and Technology of Drugs, Sapienza University of Rome, 00185 Rome, Italy; (G.D.M.); (L.M.)
| | - Sandra Moreno
- Department of Science, University Roma Tre, 00146 Rome, Italy;
- Laboratory of Neurodevelopment, Neurogenetics and Neuromolecular Biology, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy
| | - Ada Maria Tata
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (A.R.M.); (A.M.T.)
- Research Centre of Neurobiology “Daniel Bovet”, Sapienza University of Rome, 00185 Rome, Italy
| | - Virve Cavallucci
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Marco Fidaleo
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (A.R.M.); (A.M.T.)
- Research Center for Nanotechnology Applied to Engineering (CNIS), Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
10
|
Heinken A, El Kouche S, Guéant-Rodriguez RM, Guéant JL. Towards personalized genome-scale modeling of inborn errors of metabolism for systems medicine applications. Metabolism 2024; 150:155738. [PMID: 37981189 DOI: 10.1016/j.metabol.2023.155738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/09/2023] [Accepted: 11/12/2023] [Indexed: 11/21/2023]
Abstract
Inborn errors of metabolism (IEMs) are a group of more than 1000 inherited diseases that are individually rare but have a cumulative global prevalence of 50 per 100,000 births. Recently, it has been recognized that like common diseases, patients with rare diseases can greatly vary in the manifestation and severity of symptoms. Here, we review omics-driven approaches that enable an integrated, holistic view of metabolic phenotypes in IEM patients. We focus on applications of Constraint-based Reconstruction and Analysis (COBRA), a widely used mechanistic systems biology approach, to model the effects of inherited diseases. Moreover, we review evidence that the gut microbiome is also altered in rare diseases. Finally, we outline an approach using personalized metabolic models of IEM patients for the prediction of biomarkers and tailored therapeutic or dietary interventions. Such applications could pave the way towards personalized medicine not just for common, but also for rare diseases.
Collapse
Affiliation(s)
- Almut Heinken
- Inserm UMRS 1256 NGERE - Nutrition, Genetics, and Environmental Risk Exposure, University of Lorraine, Nancy F-54000, France.
| | - Sandra El Kouche
- Inserm UMRS 1256 NGERE - Nutrition, Genetics, and Environmental Risk Exposure, University of Lorraine, Nancy F-54000, France
| | - Rosa-Maria Guéant-Rodriguez
- Inserm UMRS 1256 NGERE - Nutrition, Genetics, and Environmental Risk Exposure, University of Lorraine, Nancy F-54000, France; National Center of Inborn Errors of Metabolism, University Regional Hospital Center of Nancy, Nancy F-54000, France
| | - Jean-Louis Guéant
- Inserm UMRS 1256 NGERE - Nutrition, Genetics, and Environmental Risk Exposure, University of Lorraine, Nancy F-54000, France; National Center of Inborn Errors of Metabolism, University Regional Hospital Center of Nancy, Nancy F-54000, France
| |
Collapse
|
11
|
Bjørke-Monsen AL, Lysne V. Vitamin B 12 - a scoping review for Nordic Nutrition Recommendations 2023. Food Nutr Res 2023; 67:10257. [PMID: 38084149 PMCID: PMC10710864 DOI: 10.29219/fnr.v67.10257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 08/02/2022] [Accepted: 09/27/2023] [Indexed: 01/31/2025] Open
Abstract
Vitamin B12 (cobalamin) is essential for normal metabolic function, and even moderate deficiency of this vitamin has negative health effects. Vitamin B12 is found in animal foods, and as vegetarian diets are increasingly popular in Western countries, one might expect a higher prevalence of vitamin B12 deficiency in the Nordic population. Setting recommendations for vitamin B12 intake has proven to be difficult, as uptake of vitamin B12 varies substantially, the clinical deficiency symptoms are often diffuse, and there is no clear agreement on the decision limits for vitamin B12 deficiency. Vitamin B12 deficiency is reported to be particularly common among pregnant women and infants, despite the fact that less than 1% of Norwegian pregnant women have a cobalamin intake below the Nordic Nutrition Recommendations 2012-recommended level of 2.0 µg/day. In addition, the assumption that breast milk contains sufficient vitamin B12 for optimal health and neurodevelopment during the first 6 months of life does not comply with the high prevalence of insufficient vitamin B12 status in this age group. Recommended intakes of vitamin B12 vary among age groups and must be based on markers of cobalamin status, indicating an optimal intracellular biochemical status, and not merely absence of clinical signs of vitamin B12 deficiency.
Collapse
Affiliation(s)
- Anne-Lise Bjørke-Monsen
- Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Vegard Lysne
- Norwegian Institute of Public Health, Oslo, Norway
| |
Collapse
|
12
|
Mascarenhas R, Guha A, Li Z, Ruetz M, An S, Seravalli J, Banerjee R. Cobalt-Sulfur Coordination Chemistry Drives B 12 Loading onto Methionine Synthase. J Am Chem Soc 2023:10.1021/jacs.3c07941. [PMID: 37916782 PMCID: PMC11063128 DOI: 10.1021/jacs.3c07941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
Cobalt-sulfur (Co-S) coordination is labile to both oxidation and reduction chemistry and is rarely seen in nature. Cobalamin (or vitamin B12) is an essential cobalt-containing organometallic cofactor in mammals and is escorted via an intricate network of chaperones to a single cytoplasmic target, methionine synthase. In this study, we report that the human cobalamin trafficking protein, MMADHC, exploits the chemical lability of Co-S coordination for cofactor off-loading onto methionine synthase. Cys-261 on MMADHC serves as the β-axial ligand to cobalamin. Complex formation between MMADHC and methionine synthase is signaled by loss of the lower axial nitrogen ligand, leading to five-coordinate thiolato-cobalamin. Nucleophilic displacement by the vicinal thiolate, Cys-262, completes cofactor transfer to methionine synthase and release of a cysteine disulfide-containing MMADHC. The physiological relevance of this mechanism is supported by clinical variants of MMADHC, which impair cofactor binding and off-loading, explaining the molecular basis of the associated homocystinuria.
Collapse
Affiliation(s)
- Romila Mascarenhas
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Arkajit Guha
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Zhu Li
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Markus Ruetz
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Sojin An
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Javier Seravalli
- Department of Biological Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska 68588, United States
| | - Ruma Banerjee
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
13
|
Mascarenhas R, Guha A, Li Z, Ruetz M, An S, Seravalli J, Banerjee R. Cobalt-sulfur coordination chemistry drives B 12 loading onto methionine synthase. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.25.550549. [PMID: 37546824 PMCID: PMC10402061 DOI: 10.1101/2023.07.25.550549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Cobalt-sulfur (Co-S) coordination is labile to both oxidation and reduction chemistry and is rarely seen in Nature. Cobalamin (or vitamin B 12 ) is an essential cobalt-containing organometallic cofactor in mammals, and is escorted via an intricate network of chaperones to a single cytoplasmic target, methionine synthase. In this study, we report that the human cobalamin trafficking protein, MMADHC, exploits the chemical lability of Co-S coordination, for cofactor off-loading onto methionine synthase. Cys-261 on MMADHC serves as the β-axial ligand to cobalamin. Complex formation between MMADHC and methionine synthase is signaled by loss of the lower axial nitrogen ligand, leading to five-coordinate thiolato-cobalamin. Nucleophilic displacement by the vicinal thiolate, Cys-262, completes cofactor transfer to methionine synthase and release of a cysteine disulfide-containing MMADHC. The physiological relevance of this mechanism is supported by clinical variants of MMADHC, which impair cofactor binding and off-loading, explaining the molecular basis of the associated homocystinuria.
Collapse
|
14
|
Mascarenhas R, Ruetz M, Gouda H, Heitman N, Yaw M, Banerjee R. Architecture of the human G-protein-methylmalonyl-CoA mutase nanoassembly for B 12 delivery and repair. Nat Commun 2023; 14:4332. [PMID: 37468522 PMCID: PMC10356863 DOI: 10.1038/s41467-023-40077-4] [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: 04/22/2023] [Accepted: 07/05/2023] [Indexed: 07/21/2023] Open
Abstract
G-proteins function as molecular switches to power cofactor translocation and confer fidelity in metal trafficking. The G-protein, MMAA, together with MMAB, an adenosyltransferase, orchestrate cofactor delivery and repair of B12-dependent human methylmalonyl-CoA mutase (MMUT). The mechanism by which the complex assembles and moves a >1300 Da cargo, or fails in disease, are poorly understood. Herein, we report the crystal structure of the human MMUT-MMAA nano-assembly, which reveals a dramatic 180° rotation of the B12 domain, exposing it to solvent. The complex, stabilized by MMAA wedging between two MMUT domains, leads to ordering of the switch I and III loops, revealing the molecular basis of mutase-dependent GTPase activation. The structure explains the biochemical penalties incurred by methylmalonic aciduria-causing mutations that reside at the MMAA-MMUT interfaces we identify here.
Collapse
Affiliation(s)
- Romila Mascarenhas
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Markus Ruetz
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Harsha Gouda
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Natalie Heitman
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Madeline Yaw
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Ruma Banerjee
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
15
|
Mascarenhas R, Ruetz M, Gouda H, Heitman N, Yaw M, Banerjee R. Architecture of the human G-protein-methylmalonyl-CoA mutase nanoassembly for B 12 delivery and repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.23.533963. [PMID: 36993209 PMCID: PMC10055420 DOI: 10.1101/2023.03.23.533963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
G-proteins function as molecular switches to power cofactor translocation and confer fidelity in metal trafficking. MMAA, a G-protein motor, together with MMAB, an adenosyltransferase, orchestrate cofactor delivery and repair of B 12 -dependent human methylmalonyl-CoA mutase (MMUT). The mechanism by which the motor assembles and moves a >1300 Da cargo, or fails in disease, are poorly understood. Herein, we report the crystal structure of the human MMUT-MMAA nanomotor assembly, which reveals a dramatic 180° rotation of the B 12 domain, exposing it to solvent. The nanomotor complex, stabilized by MMAA wedging between two MMUT domains, leads to ordering of the switch I and III loops, revealing the molecular basis of mutase-dependent GTPase activation. The structure explains the biochemical penalties incurred by methylmalonic aciduria-causing mutations that reside at the newly identified MMAA-MMUT interfaces.
Collapse
|
16
|
Mathew AR, Cavallucci V, Fidaleo M. Altered vitamin B12 metabolism in the central nervous system is associated with the modification of ribosomal gene expression: new insights from comparative RNA dataset analysis. Funct Integr Genomics 2023; 23:45. [PMID: 36683116 PMCID: PMC9868042 DOI: 10.1007/s10142-023-00969-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 01/05/2023] [Accepted: 01/06/2023] [Indexed: 01/24/2023]
Abstract
Recent studies have confirmed the direct role of vitamin B12 (VitB12) in the central nervous system (CNS) homeostasis; nevertheless, the detailed mechanisms are poorly understood. By analyzing RNA-Seq and microarray datasets obtained from databanks, this study aims to identify possible basic mechanisms, related to the brain, involved in altering the gene expression under VitB12 deficiency mimicking conditions. The database inquiry returned datasets generated from distinctly heterogeneous experimental sets and considering the quality and relevance requirements, two datasets from mouse and one from rat models were selected. The analyses of individual datasets highlighted a change in ribosomal gene expression in VitB12 deficiency mimicking conditions within each system. Specifically, a divergent regulation was observed depending on the animal model: mice showed a down regulation of the ribosomal gene expression, while rats an upregulation. Interestingly, E2f1 was significantly upregulated under VitB12 deficiency mimicking conditions in the animal models, with a greater upregulation in rats. The rat model also revealed putative E2F1 Transcription Factor Binding Sites (TFBSs) in the promoter of the differently regulated genes involved in ribosomal gene expression. This suggested the possibility that E2F1, being greater expressed in rats, could activate the ribosomal genes having E2F1 TFBSs, thus giving a plausible explication to the divergent regulation observed in animal models. Despite the great diversity of the experimental sets used to generate the datasets considered, a common alteration of the ribosomes exists, thereby indicating a possible basic and conserved response to VitB12 deficiency. Moreover, these findings could provide new insights on E2F1 and its association with CNS homeostasis and VitB12 deficiency.
Collapse
Affiliation(s)
- Aimee Rachel Mathew
- Department of Biology and Biotechnology Charles Darwin, University of Rome Sapienza, 00185, Rome, Italy
| | - Virve Cavallucci
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
- Institute of General Pathology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Marco Fidaleo
- Department of Biology and Biotechnology Charles Darwin, University of Rome Sapienza, 00185, Rome, Italy.
- Research Center for Nanotechnology for Engineering of Sapienza (CNIS), University of Rome Sapienza, 00185, Rome, Italy.
| |
Collapse
|
17
|
Luo J, Guo H, Feng L, Yang L, Chen X, Du T, Hu M, Yao H, Chen X. Case report: Novel compound-heterozygous mutations in the TCN2 gene identified in a chinese girl with transcobalamin deficiency. Front Genet 2022; 13:951007. [PMID: 36035190 PMCID: PMC9411981 DOI: 10.3389/fgene.2022.951007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/12/2022] [Indexed: 11/22/2022] Open
Abstract
Transcobalamin (TC) deficiency is a rare autosomal recessive disease characterized by megaloblastic anemia. It is caused by cellular vitamin B12 depletion, which subsequently results in elevated levels of homocysteine and methylmalonic acid. This disease is usually diagnosed by genetic analysis of the TCN2 gene. Here, we described a 2.2-month-old Chinese girl with TC deficiency presenting with diarrhea, fever and poor feeding. Whole-exome sequencing detected a pair of compound-heterozygous mutations in TCN2 gene, c.754-12C>G and c.1031_1032delGA (p.R344Tfs*20). To our knowledge, it is the first time that they were identified and reported in TC deficiency. This study contributes to a better understanding of the TC deficiency, expanding the spectrum of TCN2 mutations in this disorder and also supporting the early diagnosis and proper treatment of similar cases in the future.
Collapse
Affiliation(s)
- Juan Luo
- Department of Endocrinology and Metabolism, Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongxi Guo
- Department of General Surgery, Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lifang Feng
- Department of Endocrinology and Metabolism, Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Luhong Yang
- Department of Endocrinology and Metabolism, Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoqian Chen
- Department of Endocrinology and Metabolism, Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tingting Du
- Department of Endocrinology and Metabolism, Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Man Hu
- Department of Endocrinology and Metabolism, Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Yao
- Department of Endocrinology and Metabolism, Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Hui Yao, ; Xiaohong Chen,
| | - Xiaohong Chen
- Department of Endocrinology and Metabolism, Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Hui Yao, ; Xiaohong Chen,
| |
Collapse
|
18
|
Bioprocess Strategies for Vitamin B12 Production by Microbial Fermentation and Its Market Applications. Bioengineering (Basel) 2022; 9:bioengineering9080365. [PMID: 36004890 PMCID: PMC9405231 DOI: 10.3390/bioengineering9080365] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 11/16/2022] Open
Abstract
Vitamin B12 is a widely used compound in the feed and food, healthcare and medical industries that can only be produced by fermentation because of the complexity of its chemical synthesis. For this reason, finding better producer strains and optimizing their bioprocesses have been the main focus of industrial producers over the last few decades. In this review, we initially provide a historical overview of vitamin B12 research and the main biosynthetic characteristics of the two microorganism families typically used for its industrial production: several strains of Propionibacterium freudenreichii and strains related to Pseudomonas denitrificans. Later, a complete summary of the current state of vitamin B12 industrial production as well as the main advances and challenges for improving it is detailed, with a special focus on bioprocess optimization, which aims not only to increase production but also sustainability. In addition, a comprehensive list of the most important and relevant patents for the present industrial strains is provided. Finally, the potential applications of vitamin B12 in different markets are discussed.
Collapse
|
19
|
Orlowska K, Fling RR, Nault R, Sink WJ, Schilmiller AL, Zacharewski T. Dioxin-elicited decrease in cobalamin redirects propionyl-CoA metabolism to the β-oxidation-like pathway resulting in acrylyl-CoA conjugate buildup. J Biol Chem 2022; 298:102301. [PMID: 35931118 PMCID: PMC9418907 DOI: 10.1016/j.jbc.2022.102301] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/30/2022] Open
Abstract
2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is a persistent environmental contaminant that induces diverse biological and toxic effects, including reprogramming intermediate metabolism, mediated by the aryl hydrocarbon receptor. However, the specific reprogramming effects of TCDD are unclear. Here, we performed targeted LC-MS analysis of hepatic extracts from mice gavaged with TCDD. We detected an increase in S-(2-carboxyethyl)-L-cysteine, a conjugate from the spontaneous reaction between the cysteine sulfhydryl group and highly reactive acrylyl-CoA, an intermediate in the cobalamin (Cbl)-independent β-oxidation-like metabolism of propionyl-CoA. TCDD repressed genes in both the canonical Cbl-dependent carboxylase and the alternate Cbl-independent β-oxidation-like pathways as well as inhibited methylmalonyl-CoA mutase (MUT) at lower doses. Moreover, TCDD decreased serum Cbl levels and hepatic cobalt levels while eliciting negligible effects on gene expression associated with Cbl absorption, transport, trafficking, or derivatization to 5'-deoxy-adenosylcobalamin (AdoCbl), the required MUT cofactor. Additionally, TCDD induced the gene encoding aconitate decarboxylase 1 (Acod1), the enzyme responsible for decarboxylation of cis-aconitate to itaconate, and dose-dependently increased itaconate levels in hepatic extracts. Our results indicate MUT inhibition is consistent with itaconate activation to itaconyl-CoA, a MUT suicide inactivator that forms an adduct with adenosylcobalamin. This adduct in turn inhibits MUT activity and reduces Cbl levels. Collectively, these results suggest the decrease in MUT activity is due to Cbl depletion following TCDD treatment, which redirects propionyl-CoA metabolism to the alternate Cbl-independent β-oxidation-like pathway. The resulting hepatic accumulation of acrylyl-CoA likely contributes to TCDD-elicited hepatotoxicity and the multihit progression of steatosis to steatohepatitis with fibrosis.
Collapse
Affiliation(s)
- Karina Orlowska
- Biochemistry & Molecular Biology, Michigan State University, East Lansing, Michigan, USA,Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Russ R. Fling
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA,Microbiology & Molecular Genetics, Michigan Sptate University, East Lansing, Michigan, USA
| | - Rance Nault
- Biochemistry & Molecular Biology, Michigan State University, East Lansing, Michigan, USA,Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Warren J. Sink
- Biochemistry & Molecular Biology, Michigan State University, East Lansing, Michigan, USA,Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Anthony L. Schilmiller
- Mass Spectrometry and Metabolomics Core, Michigan State University, East Lansing, Michigan, USA
| | - Tim Zacharewski
- Biochemistry & Molecular Biology, Michigan State University, East Lansing, Michigan, USA; Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA.
| |
Collapse
|
20
|
Joseph FD, Campbell DR, Cohn S, Roach ES. A Child with Falls, Fatigue, and Dementia. Pediatr Rev 2022; 43:466-468. [PMID: 35909134 DOI: 10.1542/pir.2020-003053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Freddie D Joseph
- Division of Child Neurology, Dell Children's Medical Center, Austin, TX
| | - Damian R Campbell
- Division of Child Neurology, University of Texas Dell Medical School, Austin, TX
| | - Shannon Cohn
- Division of Hematology/Oncology, Dell Children's Medical Center, Austin, TX
| | - E Steve Roach
- Department of Neurology, University of Texas Dell Medical School, Austin, TX
| |
Collapse
|
21
|
Wang F, Liang L, Ling S, Yu Y, Chen T, Xu F, Gong Z, Han L. Clinical characteristics and genotype analysis of five infants with cblX type of methylmalonic acidemia. Zhejiang Da Xue Xue Bao Yi Xue Ban 2022; 51:298-305. [PMID: 36207831 PMCID: PMC9511482 DOI: 10.3724/zdxbyxb-2022-0194] [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: 04/20/2022] [Accepted: 05/30/2022] [Indexed: 06/16/2023]
Abstract
OBJECTIVE To investigate the clinical and genetic characteristics of infants with cobalamin (cbl) X type of methylmalonic acidemia (MMA). METHODS The clinical data of 5 infants with cblX type of MMA diagnosed in Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine and Shanghai Children's Hospital from the year 2016 to 2020 were collected. The levels of blood acylcarnitines were detected by tandem mass spectrometry, the levels of urinary organic acids were detected by gas-chromatography mass spectrometry, the pathogenic genes were detected by whole exon gene sequencing, and the effect of new pathogenic mutations on three-dimensional protein structure was predicted by bioinformatics analysis. RESULTS Five infants with cblX type were diagnosed, including 4 males and 1 female, and the onset age was 0-6 months. The main clinical manifestations of 4 males were intractable epilepsy, mental and motor retardation, metabolic abnormalities presented mild increase of blood homocysteine level. Among them, 3 cases were accompanied by slight increase of urinary methylmalonic acid, and 1 case was accompanied by increase of blood propionylcarnitine (C3) and C3/acetylcarnitine (C2). Gene detection found that 2 cases carried a same hemizygous mutation c.344C>T (p.A115V) of HCFC1 gene, which was the most reported mutation, and the other 2 cases carried novel pathogenic mutations, c.92G>A (p.R31Q) and c.166G>C (p.V56L). These 3 gene mutations located in the Kelch domain of HCFC1 protein. One female infant carried a benign mutation of c.3731G>T (p.R1244L). Her clinical symptoms were mild, and only the urinary methylmalonic acid was slightly increased. CONCLUSIONS The clinical manifestations of children with cblX type of MMA are intractable epilepsy, mental and motor retardation, and other serious neurological symptoms. Their metabolic abnormalities present the increase of blood homocysteine with methylmalonic acid (urinary methylmalonic acid or/and blood C3, C3/C2). The clinical and biochemical phenotypes are separated, so the diagnosis should be in combination with the results of gene testing.
Collapse
Affiliation(s)
- Fei Wang
- 1. Department of Endocrinology, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200062, China
| | - Lili Liang
- 2. Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Institute for Pediatric Research, Shanghai 200092, China
| | - Shiying Ling
- 2. Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Institute for Pediatric Research, Shanghai 200092, China
| | - Yue Yu
- 2. Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Institute for Pediatric Research, Shanghai 200092, China
| | - Ting Chen
- 2. Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Institute for Pediatric Research, Shanghai 200092, China
| | - Feng Xu
- 2. Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Institute for Pediatric Research, Shanghai 200092, China
| | - Zhuwen Gong
- 2. Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Institute for Pediatric Research, Shanghai 200092, China
| | - Lianshu Han
- 2. Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Institute for Pediatric Research, Shanghai 200092, China
| |
Collapse
|
22
|
Waisbren SE. Review of neuropsychological outcomes in isolated methylmalonic acidemia: recommendations for assessing impact of treatments. Metab Brain Dis 2022; 37:1317-1335. [PMID: 35348993 DOI: 10.1007/s11011-022-00954-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 03/09/2022] [Indexed: 11/29/2022]
Abstract
Methylmalonic acidemia (MMA) due to methylmalonyl-CoA mutase deficiency (OMIM #251,000) is an autosomal recessive disorder of organic acid metabolism associated with life-threatening acute metabolic decompensations and significant neuropsychological deficits. "Isolated" MMA refers to the presence of excess methylmalonic acid without homocysteine elevation. Belonging to this class of disorders are those that involve complete deficiency (mut0) and partial deficiency (mut-) of the methylmalonyl-CoA mutase enzyme and other disorders causing excess methylmalonic acid excretion. These other disorders include enzymatic subtypes related to cobalamin A defect (cblA) (OMIM #25,110), cobalamin B defect (cblB) (OMIM #251,110) and related conditions. Neuropsychological attributes associated with isolated MMA have become more relevant as survival rates increased following improved diagnostic and treatment strategies. Children with this disorder still are at risk for developmental delay, cognitive difficulties and progressive declines in functioning. Mean IQ for all types apart from cblA defect enzymatic subtype is rarely above 85 and much lower for mut0 enzymatic subtype. Identifying psychological domains responsive to improvements in biochemical status is important. This review suggests that processing speed, working memory, language, attention, and quality of life may be sensitive to fluctuations in metabolite levels while IQ and motor skills may be less amenable to change. Due to slower developmental trajectories, Growth Scale Values, Projected Retained Ability Scores and other indices of change need to be incorporated into clinical trial study protocols. Neuropsychologists are uniquely qualified to provide a differentiated picture of cognitive, behavioral and emotional consequences of MMA and analyze benefits or shortcomings of novel treatments.
Collapse
Affiliation(s)
- Susan E Waisbren
- Boston Children's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
23
|
Hannibal L, Jacobsen DW. Intracellular processing of vitamin B 12 by MMACHC (CblC). VITAMINS AND HORMONES 2022; 119:275-298. [PMID: 35337623 DOI: 10.1016/bs.vh.2022.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Vitamin B12 (cobalamin, Cbl, B12) is a water-soluble micronutrient synthesized exclusively by a group of microorganisms. Human beings are unable to make B12 and thus obtain the vitamin via intake of animal products, fermented plant-based foods or supplements. Vitamin B12 obtained from the diet comprises three major chemical forms, namely hydroxocobalamin (HOCbl), methylcobalamin (MeCbl) and adenosylcobalamin (AdoCbl). The most common form of B12 present in supplements is cyanocobalamin (CNCbl). Yet, these chemical forms cannot be utilized directly as they come, but instead, they undergo chemical processing by the MMACHC protein, also known as CblC. Processing of dietary B12 by CblC involves removal of the upper-axial ligand (beta-ligand) yielding the one-electron reduced intermediate cob(II)alamin. Newly formed cob(II)alamin undergoes trafficking and delivery to the two B12-dependent enzymes, cytosolic methionine synthase (MS) and mitochondrial methylmalonyl-CoA mutase (MUT). The catalytic cycles of MS and MUT incorporate cob(II)alamin as a precursor to regenerate the coenzyme forms MeCbl and AdoCbl, respectively. Mutations and epimutations in the MMACHC gene result in cblC disease, the most common inborn error of B12 metabolism, which manifests with combined homocystinuria and methylmalonic aciduria. Elevation of metabolites homocysteine and methylmalonic acid occurs because the lack of an active CblC blocks formation of the indispensable precursor cob(II)alamin that is necessary to activate MS and MUT. Thus, in patients with cblC disease, vitamin B12 is absorbed and present in circulation in normal to high concentrations, yet, cells are unable to make use of it. Mutations in seemingly unrelated genes that modify MMACHC gene expression also result in clinical phenotypes that resemble cblC disease. We review current knowledge on structural and functional aspects of intracellular processing of vitamin B12 by the versatile protein CblC, its partners and possible regulators.
Collapse
Affiliation(s)
- Luciana Hannibal
- Laboratory of Clinical Biochemistry and Metabolism, Department of General Pediatrics, Adolescent Medicine and Neonatology, Faculty of Medicine, Medical Center-University of Freiburg, Freiburg, Germany.
| | - Donald W Jacobsen
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
24
|
Ling S, Wu S, Shuai R, Yu Y, Qiu W, Wei H, Yang C, Xu P, Zou H, Feng J, Niu T, Hu H, Zhang H, Liang L, Lu D, Gong Z, Zhan X, Ji W, Gu X, Han L. The Follow-Up of Chinese Patients in cblC Type Methylmalonic Acidemia Identified Through Expanded Newborn Screening. Front Genet 2022; 13:805599. [PMID: 35242167 PMCID: PMC8886223 DOI: 10.3389/fgene.2022.805599] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 01/21/2022] [Indexed: 11/18/2022] Open
Abstract
Objective: The cblC type of combined methylmalonic acidemia and homocystinuria, an inherited disorder with variable phenotypes, is included in newborn screening (NBS) programs at multiple newborn screening centers in China. The present study aimed to investigate the long-term clinical benefits of screening individual. Methods: A national, retrospective multi-center study of infants with confirmed cblC defect identified by NBS between 2004 and 2020 was conducted. We collected a large cohort of 538 patients and investigated their clinical data in detail, including disease onset, biochemical metabolites, and gene variation, and explored different factors on the prognosis. Results: The long-term outcomes of all patients were evaluated, representing 44.6% for poor outcomes. In our comparison of patients with already occurring clinical signs before treatment to asymptomatic ones, the incidence of intellectual impairment, movement disorders, ocular complications, hydrocephalus, and death were significantly different (p < 0.01). The presence of disease onset [Odd ratio (OR) 12.39, 95% CI 5.15–29.81; p = 0.000], variants of c.609G>A (OR 2.55, 95% CI 1.49–4.35; p = 0.001), and c.567dupT (OR 2.28, 95% CI 1.03–5.05; p = 0.042) were independently associated with poor outcomes, especially for neurodevelopmental deterioration. Conclusion: NBS, avoiding major disease-related events and allowing an earlier treatment initiation, appeared to have protective effects on the prognosis of infants with cblC defect.
Collapse
Affiliation(s)
- Shiying Ling
- Department of Pediatric Endocrinology/Genetics, Shanghai Institute for Pediatric Research, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shengnan Wu
- Department of Endocrinology and Metabolism, Henan Key Laboratory of Children’s Genetics and Metabolic Diseases, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Ruixue Shuai
- Department of Pediatric Endocrinology/Genetics, Shanghai Institute for Pediatric Research, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yue Yu
- Department of Pediatric Endocrinology/Genetics, Shanghai Institute for Pediatric Research, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wenjuan Qiu
- Department of Pediatric Endocrinology/Genetics, Shanghai Institute for Pediatric Research, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Haiyan Wei
- Department of Endocrinology and Metabolism, Henan Key Laboratory of Children’s Genetics and Metabolic Diseases, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Chiju Yang
- Center of Neonatal Disease Screening, Jining Maternal and Child Health Care Hospital, Jining, China
| | - Peng Xu
- Center of Neonatal Disease Screening, Jining Maternal and Child Health Care Hospital, Jining, China
| | - Hui Zou
- Center of Neonatal Disease Screening, Jinan Maternal and Child Health Care Hospital, Jinan, China
| | - Jizhen Feng
- Center of Neonatal Disease Screening, Shijiazhuang Maternal and Child Health Care Hospital, Shijiazhuang, China
| | - Tingting Niu
- Center of Neonatal Disease Screening, Shandong Maternal and Child Health Care Hospital, Jinan, China
| | - Haili Hu
- Center of Neonatal Disease Screening, Hefei Maternal and Child Health Care Hospital, Hefei, China
| | - Huiwen Zhang
- Department of Pediatric Endocrinology/Genetics, Shanghai Institute for Pediatric Research, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lili Liang
- Department of Pediatric Endocrinology/Genetics, Shanghai Institute for Pediatric Research, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Deyun Lu
- Department of Pediatric Endocrinology/Genetics, Shanghai Institute for Pediatric Research, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhuwen Gong
- Department of Pediatric Endocrinology/Genetics, Shanghai Institute for Pediatric Research, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xia Zhan
- Department of Pediatric Endocrinology/Genetics, Shanghai Institute for Pediatric Research, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wenjun Ji
- Department of Pediatric Endocrinology/Genetics, Shanghai Institute for Pediatric Research, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xuefan Gu
- Department of Pediatric Endocrinology/Genetics, Shanghai Institute for Pediatric Research, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lianshu Han
- Department of Pediatric Endocrinology/Genetics, Shanghai Institute for Pediatric Research, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Lianshu Han,
| |
Collapse
|
25
|
Pangilinan F, Watkins D, Bernard D, Chen Y, Dong N, Wu Q, Ozel-Abaan H, Kaur M, Caggana M, Morrissey M, Browne ML, Mills JL, Van Ryzin C, Shchelochkov O, Sloan J, Venditti CP, Sarafoglou K, Rosenblatt DS, Kay DM, Brody LC. Probing the functional consequence and clinical relevance of CD320 p.E88del, a variant in the transcobalamin receptor gene. Am J Med Genet A 2022; 188:1124-1141. [PMID: 35107211 DOI: 10.1002/ajmg.a.62627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/12/2021] [Accepted: 11/20/2021] [Indexed: 11/06/2022]
Abstract
The biological and clinical significance of the p.E88del variant in the transcobalamin receptor, CD320, is unknown. This allele is annotated in ClinVar as likely benign, pathogenic, and of uncertain significance. To determine functional consequence and clinical relevance of this allele, we employed cell culture and genetic association studies. Fibroblasts from 16 CD320 p.E88del homozygotes exhibited reduced binding and uptake of cobalamin. Complete ascertainment of newborns with transiently elevated C3 (propionylcarnitine) in New York State demonstrated that homozygosity for CD320 p.E88del was over-represented (7/348, p < 6 × 10-5 ). Using population data, we estimate that ~85% of the p.E88del homozygotes born in the same period did not have elevated C3, suggesting that cobalamin metabolism in the majority of these infants with this genotype is unaffected. Clinical follow-up of 4/9 homozygous individuals uncovered neuropsychological findings, mostly in speech and language development. None of these nine individuals exhibited perturbation of cobalamin metabolism beyond the newborn stage even during periods of acute illness. Newborns homozygous for this allele in the absence of other factors are at low risk of requiring clinical intervention, although more studies are required to clarify the natural history of various CD320 variants across patient populations.
Collapse
Affiliation(s)
- Faith Pangilinan
- Genetics and Environment Interaction Section, National Human Genome Research Institute, National Institutes of Health, Maryland, USA
| | - David Watkins
- Department of Human Genetics, McGill University and Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - David Bernard
- Genetics and Environment Interaction Section, National Human Genome Research Institute, National Institutes of Health, Maryland, USA
| | - Yue Chen
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Ningzheng Dong
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China.,MOH Key Laboratory of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qingyu Wu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Hatice Ozel-Abaan
- Genetics and Environment Interaction Section, National Human Genome Research Institute, National Institutes of Health, Maryland, USA
| | - Manjit Kaur
- Genetics and Environment Interaction Section, National Human Genome Research Institute, National Institutes of Health, Maryland, USA
| | - Michele Caggana
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, New York, USA
| | - Mark Morrissey
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, New York, USA
| | - Marilyn L Browne
- Birth Defects Registry, New York State Department of Health, Albany, New York and University at Albany School of Public Health, Rensselaer, New York, USA
| | - James L Mills
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Carol Van Ryzin
- Organic Acid Research Section, National Human Genome Research Institute, National Institutes of Health, Maryland, USA
| | - Oleg Shchelochkov
- Organic Acid Research Section, National Human Genome Research Institute, National Institutes of Health, Maryland, USA
| | - Jennifer Sloan
- Organic Acid Research Section, National Human Genome Research Institute, National Institutes of Health, Maryland, USA
| | - Charles P Venditti
- Organic Acid Research Section, National Human Genome Research Institute, National Institutes of Health, Maryland, USA
| | - Kyriakie Sarafoglou
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - David S Rosenblatt
- Department of Human Genetics, McGill University and Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.,Division of Medical Biochemistry, Department of Specialized Medicine, McGill University Health Centre, Montreal, Quebec, Canada.,Division of Medical Genetics, Department of Specialized Medicine, McGill University Health Centre, Montreal, Quebec, Canada
| | - Denise M Kay
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, New York, USA
| | - Lawrence C Brody
- Genetics and Environment Interaction Section, National Human Genome Research Institute, National Institutes of Health, Maryland, USA
| |
Collapse
|
26
|
Pappas KB, Younan M, Conway R. Transcobalamin receptor deficiency in seven asymptomatic patients ascertained through newborn screening. Am J Med Genet A 2022; 188:1102-1108. [PMID: 34978764 DOI: 10.1002/ajmg.a.62623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/03/2021] [Accepted: 12/13/2021] [Indexed: 11/08/2022]
Abstract
We report seven cases from our clinic with transcobalamin receptor deficiency (TCRD). None of our cases have experienced health issues or metabolic decompensation. All have experienced typical growth and development throughout childhood, with our oldest case now 10 years old. Every case has had normalization of initial biochemical abnormalities following parenteral hydroxocobalamin administration. Several cases had trace elevations of methylmalonic acid throughout childhood, all which normalized without further hydroxocobalamin administration. Population data from our state's newborn screening program suggest the incidence of TCRD is comparable to other metabolic disorders associated with elevations of C3 acylcarnitine including propionic academia, isolated methylmalonic academia and combined methylmalonic academia and hyperhomocysteinemia due to cobalamin metabolism disorders. Based on the generally benign nature of this condition, we assert that TCRD may be considered an incidental finding on newborn screen. However, additional long-term data are needed to ascertain the long term outcomes of children identified with TCRD.
Collapse
Affiliation(s)
- Kara B Pappas
- Department of Genetics, Genomics, and Metabolic Disorders, Children's Hospital of Michigan, Detroit, Michigan, USA.,Department of Pediatrics, Central Michigan University School of Medicine, Mt Pleasant, Michigan, USA
| | - Marissa Younan
- Department of Genetics, Genomics, and Metabolic Disorders, Children's Hospital of Michigan, Detroit, Michigan, USA
| | - Robert Conway
- Department of Genetics, Genomics, and Metabolic Disorders, Children's Hospital of Michigan, Detroit, Michigan, USA
| |
Collapse
|
27
|
Mascarenhas R, Gouda H, Ruetz M, Banerjee R. Human B 12-dependent enzymes: Methionine synthase and Methylmalonyl-CoA mutase. Methods Enzymol 2022; 668:309-326. [PMID: 35589199 PMCID: PMC9420401 DOI: 10.1016/bs.mie.2021.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Humans have only two known cobalamin or B12-dependent enzymes: cytoplasmic methionine synthase and mitochondrial methylmalonyl-CoA mutase. A complex intracellular B12 trafficking pathway, comprising a multitude of chaperones, process and deliver cobalamin to the two target enzymes. Methionine synthase catalyzes the transfer of a methyl group from N5-methytetrahydrofolate to homocysteine, generating tetrahydrofolate and methionine. Cobalamin serves as an intermediate methyl group carrier and cycles between methylcobalamin and cob(I)alamin. Methylmalonyl-CoA mutase uses the 5'-deoxyadenosylcobalamin form of the cofactor and catalyzes the 1,2 rearrangement of methylmalonyl-CoA to succinyl-CoA. Two chaperones, CblA (or MMAA) and CblB (or MMAB, also known as adenosyltransferase), serve the mutase and ensure that the fidelity of the cofactor loading and unloading processes is maintained. This chapter focuses on assays for purifying and measuring the activities of methionine synthase and methylmalonyl-CoA mutase.
Collapse
|
28
|
Gouda H, Mascarenhas R, Pillay S, Ruetz M, Koutmos M, Banerjee R. Patient mutations in human ATP:cob(I)alamin adenosyltransferase differentially affect its catalytic versus chaperone functions. J Biol Chem 2021; 297:101373. [PMID: 34757128 PMCID: PMC8633584 DOI: 10.1016/j.jbc.2021.101373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/25/2021] [Accepted: 10/27/2021] [Indexed: 11/26/2022] Open
Abstract
Human ATP:cob(I)alamin adenosyltransferase (ATR) is a mitochondrial enzyme that catalyzes an adenosyl transfer to cob(I)alamin, synthesizing 5′-deoxyadenosylcobalamin (AdoCbl) or coenzyme B12. ATR is also a chaperone that escorts AdoCbl, transferring it to methylmalonyl-CoA mutase, which is important in propionate metabolism. Mutations in ATR lead to methylmalonic aciduria type B, an inborn error of B12 metabolism. Our previous studies have furnished insights into how ATR protein dynamics influence redox-linked cobalt coordination chemistry, controlling its catalytic versus chaperone functions. In this study, we have characterized three patient mutations at two conserved active site residues in human ATR, R190C/H, and E193K and obtained crystal structures of R190C and E193K variants, which display only subtle structural changes. All three mutations were found to weaken affinities for the cob(II)alamin substrate and the AdoCbl product and increase KM(ATP). 31P NMR studies show that binding of the triphosphate product, formed during the adenosylation reaction, is also weakened. However, although the kcat of this reaction is significantly diminished for the R190C/H mutants, it is comparable with the WT enzyme for the E193K variant, revealing the catalytic importance of Arg-190. Furthermore, although the E193K mutation selectively impairs the chaperone function by promoting product release into solution, its catalytic function might be unaffected at physiological ATP concentrations. In contrast, the R190C/H mutations affect both the catalytic and chaperoning activities of ATR. Because the E193K mutation spares the catalytic activity of ATR, our data suggest that the patients carrying this mutation are more likely to be responsive to cobalamin therapy.
Collapse
Affiliation(s)
- Harsha Gouda
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Romila Mascarenhas
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Shubhadra Pillay
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Markus Ruetz
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Markos Koutmos
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan, USA; Department of Biophysics, University of Michigan, Ann Arbor, Michigan, USA
| | - Ruma Banerjee
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan, USA.
| |
Collapse
|
29
|
Ghergurovich JM, Xu X, Wang JZ, Yang L, Ryseck RP, Wang L, Rabinowitz JD. Methionine synthase supports tumour tetrahydrofolate pools. Nat Metab 2021; 3:1512-1520. [PMID: 34799699 PMCID: PMC9284419 DOI: 10.1038/s42255-021-00465-w] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 09/01/2021] [Indexed: 01/02/2023]
Abstract
Mammalian cells require activated folates to generate nucleotides for growth and division. The most abundant circulating folate species is 5-methyl tetrahydrofolate (5-methyl-THF), which is used to synthesize methionine from homocysteine via the cobalamin-dependent enzyme methionine synthase (MTR). Cobalamin deficiency traps folates as 5-methyl-THF. Here, we show using isotope tracing that MTR is only a minor source of methionine in cell culture, tissues or xenografted tumours. Instead, MTR is required for cells to avoid folate trapping and assimilate 5-methyl-THF into other folate species. Under conditions of physiological extracellular folates, genetic MTR knockout in tumour cells leads to folate trapping, purine synthesis stalling, nucleotide depletion and impaired growth in cell culture and as xenografts. These defects are rescued by free folate but not one-carbon unit supplementation. Thus, MTR plays a crucial role in liberating THF for use in one-carbon metabolism.
Collapse
Affiliation(s)
- Jonathan M Ghergurovich
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Xincheng Xu
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Joshua Z Wang
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Lifeng Yang
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Rolf-Peter Ryseck
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Lin Wang
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Joshua D Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA.
- Department of Chemistry, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
30
|
Kobalamina – właściwości biomedyczne i niedobór w ujęciu biochemicznym. POSTEP HIG MED DOSW 2021. [DOI: 10.2478/ahem-2021-0004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstrakt
Kobalamina (witamina B12) jest rozpuszczalnym w wodzie związkiem organicznym, zaliczanym do witamin grupy B. Złożona budowa i polarność cząsteczki witaminy B12 sprawiają, że do jej prawidłowego wykorzystania i przemian w organizmie człowieka niezbędny jest udział wyspecjalizowanych białek. Głównym źródłem kobalaminy człowieka jest pokarm pochodzenia zwierzęcego. Ze względu na rezerwy tkankowe tej witaminy, jej niedobór ujawnia się dopiero po kilku latach niewystarczającej podaży z pożywieniem. Badania przesiewowe pod kątem deficytu kobalaminy są jednak uzasadnione u osób z czynnikami ryzyka hipokobalaminemii, takimi jak: stan po resekcji żołądka lub jelita cienkiego, dieta wegańska, długotrwałe stosowanie metforminy, antagonistów receptora histaminowego H2 oraz leków z grupy inhibitorów pompy protonowej, a także podeszły wiek.
Witamina B12 jako kofaktor enzymatyczny uczestniczy w licznych przemianach wewnątrzmitochondrialnych oraz w syntezie metioniny, niezbędnej do powstania S-adenozylometioniny istotnej w procesie metylacji cząsteczek biologicznie czynnych. Przez powiązania metaboliczne z kwasem foliowym kobalamina wpływa na proces syntezy DNA i podział komórki. Obecnie coraz częściej zwraca się uwagę na potencjalny udział niedoboru witaminy B12 w patogenezie chorób neurodegeneracyjnych, a także nowotworowych. Niedobór kobalaminy na poziomie molekularnym destabilizuje genom komórek, zwiększając ryzyko ich złośliwej transformacji. Jednak u osób z chorobą nowotworową lub obciążonych ryzykiem jej rozwoju witamina B12 może nasilać ekspansję komórek neoplastycznych.
Collapse
|
31
|
Matmat K, Guéant-Rodriguez RM, Oussalah A, Wiedemann-Fodé A, Dionisi-Vici C, Coelho D, Guéant JL, Conart JB. Ocular manifestations in patients with inborn errors of intracellular cobalamin metabolism: a systematic review. Hum Genet 2021; 141:1239-1251. [PMID: 34652574 DOI: 10.1007/s00439-021-02350-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 08/19/2021] [Indexed: 01/26/2023]
Abstract
Inherited disorders of cobalamin (cbl) metabolism (cblA-J) result in accumulation of methylmalonic acid (MMA) and/or homocystinuria (HCU). Clinical presentation includes ophthalmological manifestations related to retina, optic nerve and posterior visual alterations, mainly reported in cblC and sporadically in other cbl inborn errors.We searched MEDLINE EMBASE and Cochrane Library, and analyzed articles reporting ocular manifestations in cbl inborn errors. Out of 166 studies a total of 52 studies reporting 163 cbl and 24 mut cases were included. Ocular manifestations were found in all cbl defects except for cblB and cblD-MMA; cblC was the most frequent disorder affecting 137 (84.0%) patients. The c.271dupA was the most common pathogenic variant, accounting for 70/105 (66.7%) cases. One hundred and thirty-seven out of 154 (88.9%) patients presented with early-onset disease (0-12 months). Nystagmus and strabismus were observed in all groups with the exception of MMA patients while maculopathy and peripheral retinal degeneration were almost exclusively found in MMA-HCU patients. Optic nerve damage ranging from mild temporal disc pallor to complete atrophy was prevalent in MMA-HCU.and MMA groups. Nystagmus was frequent in early-onset patients. Retinal and macular degeneration worsened despite early treatment and stabilized systemic function in these patients. The functional prognosis remains poor with final visual acuity < 20/200 in 55.6% (25/45) of cases. In conclusion, the spectrum of eye disease in Cbl patients depends on metabolic severity and age of onset. The development of visual manifestations over time despite early metabolic treatment point out the need for specific innovative therapies.
Collapse
Affiliation(s)
- Karim Matmat
- UMR_S 1256, NGERE - Nutrition, Genetics, and Environmental Risk Exposure, INSERM, University of Lorraine, 54000, Nancy, France
| | - Rosa-Maria Guéant-Rodriguez
- UMR_S 1256, NGERE - Nutrition, Genetics, and Environmental Risk Exposure, INSERM, University of Lorraine, 54000, Nancy, France.
- National Center of Inborn Errors of Metabolism, University Regional Hospital Center of Nancy, 54000, Nancy, France.
| | - Abderrahim Oussalah
- UMR_S 1256, NGERE - Nutrition, Genetics, and Environmental Risk Exposure, INSERM, University of Lorraine, 54000, Nancy, France
- National Center of Inborn Errors of Metabolism, University Regional Hospital Center of Nancy, 54000, Nancy, France
| | - Arnaud Wiedemann-Fodé
- UMR_S 1256, NGERE - Nutrition, Genetics, and Environmental Risk Exposure, INSERM, University of Lorraine, 54000, Nancy, France
| | - Carlo Dionisi-Vici
- Division of Metabolism, Bambino Gesù Children's Hospital IRCCS, Piazza S. Onofrio 4, 00165, Rome, Italy
| | - David Coelho
- UMR_S 1256, NGERE - Nutrition, Genetics, and Environmental Risk Exposure, INSERM, University of Lorraine, 54000, Nancy, France
| | - Jean-Louis Guéant
- UMR_S 1256, NGERE - Nutrition, Genetics, and Environmental Risk Exposure, INSERM, University of Lorraine, 54000, Nancy, France
- National Center of Inborn Errors of Metabolism, University Regional Hospital Center of Nancy, 54000, Nancy, France
| | - Jean-Baptiste Conart
- UMR_S 1256, NGERE - Nutrition, Genetics, and Environmental Risk Exposure, INSERM, University of Lorraine, 54000, Nancy, France.
- Department of Ophthalmology, Nancy University Hospital, 54500, Vandœuvre-lès-Nancy, France.
| |
Collapse
|
32
|
Guéant JL, Guéant-Rodriguez RM, Kosgei VJ, Coelho D. Causes and consequences of impaired methionine synthase activity in acquired and inherited disorders of vitamin B 12 metabolism. Crit Rev Biochem Mol Biol 2021; 57:133-155. [PMID: 34608838 DOI: 10.1080/10409238.2021.1979459] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Methyl-Cobalamin (Cbl) derives from dietary vitamin B12 and acts as a cofactor of methionine synthase (MS) in mammals. MS encoded by MTR catalyzes the remethylation of homocysteine to generate methionine and tetrahydrofolate, which fuel methionine and cytoplasmic folate cycles, respectively. Methionine is the precursor of S-adenosyl methionine (SAM), the universal methyl donor of transmethylation reactions. Impaired MS activity results from inadequate dietary intake or malabsorption of B12 and inborn errors of Cbl metabolism (IECM). The mechanisms at the origin of the high variability of clinical presentation of impaired MS activity are classically considered as the consequence of the disruption of the folate cycle and related synthesis of purines and pyrimidines and the decreased synthesis of endogenous methionine and SAM. For one decade, data on cellular and animal models of B12 deficiency and IECM have highlighted other key pathomechanisms, including altered interactome of MS with methionine synthase reductase, MMACHC, and MMADHC, endoplasmic reticulum stress, altered cell signaling, and genomic/epigenomic dysregulations. Decreased MS activity increases catalytic protein phosphatase 2A (PP2A) and produces imbalanced phosphorylation/methylation of nucleocytoplasmic RNA binding proteins, including ELAVL1/HuR protein, with subsequent nuclear sequestration of mRNAs and dramatic alteration of gene expression, including SIRT1. Decreased SAM and SIRT1 activity induce ER stress through impaired SIRT1-deacetylation of HSF1 and hypomethylation/hyperacetylation of peroxisome proliferator-activated receptor-γ coactivator-1α (PGC1α), which deactivate nuclear receptors and lead to impaired energy metabolism and neuroplasticity. The reversibility of these pathomechanisms by SIRT1 agonists opens promising perspectives in the treatment of IECM outcomes resistant to conventional supplementation therapies.
Collapse
Affiliation(s)
- Jean-Louis Guéant
- UMR Inserm 1256 N-GERE (Nutrition, Génetique et Exposition aux Risques Environmentaux), Université de Lorraine, Vandoeuvre-lès-Nancy, France.,Departments of Digestive Diseases and Molecular Medicine and National Center of Inborn Errors of Metabolism, University Hospital Center, Université de Lorraine, Vandoeuvre-lès-Nancy, France
| | - Rosa-Maria Guéant-Rodriguez
- UMR Inserm 1256 N-GERE (Nutrition, Génetique et Exposition aux Risques Environmentaux), Université de Lorraine, Vandoeuvre-lès-Nancy, France.,Departments of Digestive Diseases and Molecular Medicine and National Center of Inborn Errors of Metabolism, University Hospital Center, Université de Lorraine, Vandoeuvre-lès-Nancy, France
| | - Viola J Kosgei
- UMR Inserm 1256 N-GERE (Nutrition, Génetique et Exposition aux Risques Environmentaux), Université de Lorraine, Vandoeuvre-lès-Nancy, France
| | - David Coelho
- UMR Inserm 1256 N-GERE (Nutrition, Génetique et Exposition aux Risques Environmentaux), Université de Lorraine, Vandoeuvre-lès-Nancy, France
| |
Collapse
|
33
|
Ruiz de la Cruz M, de la Cruz Montoya AH, Rojas Jiménez EA, Martínez Gregorio H, Díaz Velásquez CE, Paredes de la Vega J, de la Cruz Hernández-Hernández F, Vaca Paniagua F. Cis-Acting Factors Causing Secondary Epimutations: Impact on the Risk for Cancer and Other Diseases. Cancers (Basel) 2021; 13:cancers13194807. [PMID: 34638292 PMCID: PMC8508567 DOI: 10.3390/cancers13194807] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/09/2021] [Accepted: 08/15/2021] [Indexed: 12/25/2022] Open
Abstract
Epigenetics affects gene expression and contributes to disease development by alterations known as epimutations. Hypermethylation that results in transcriptional silencing of tumor suppressor genes has been described in patients with hereditary cancers and without pathogenic variants in the coding region of cancer susceptibility genes. Although somatic promoter hypermethylation of these genes can occur in later stages of the carcinogenic process, constitutional methylation can be a crucial event during the first steps of tumorigenesis, accelerating tumor development. Primary epimutations originate independently of changes in the DNA sequence, while secondary epimutations are a consequence of a mutation in a cis or trans-acting factor. Secondary epimutations have a genetic basis in cis of the promoter regions of genes involved in familial cancers. This highlights epimutations as a novel carcinogenic mechanism whose contribution to human diseases is underestimated by the scarcity of the variants described. In this review, we provide an overview of secondary epimutations and present evidence of their impact on cancer. We propose the necessity for genetic screening of loci associated with secondary epimutations in familial cancer as part of prevention programs to improve molecular diagnosis, secondary prevention, and reduce the mortality of these diseases.
Collapse
Affiliation(s)
- Miguel Ruiz de la Cruz
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla 54090, Mexico; (M.R.d.l.C.); (E.A.R.J.); (H.M.G.); (C.E.D.V.); (J.P.d.l.V.)
- Avenida Instituto Politécnico Nacional # 2508, Colonia San Pedro Zacatenco, Delegación Gustavo A. Madero, C.P. Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City 07360, Mexico;
| | | | - Ernesto Arturo Rojas Jiménez
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla 54090, Mexico; (M.R.d.l.C.); (E.A.R.J.); (H.M.G.); (C.E.D.V.); (J.P.d.l.V.)
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla 54090, Mexico;
| | - Héctor Martínez Gregorio
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla 54090, Mexico; (M.R.d.l.C.); (E.A.R.J.); (H.M.G.); (C.E.D.V.); (J.P.d.l.V.)
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla 54090, Mexico;
| | - Clara Estela Díaz Velásquez
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla 54090, Mexico; (M.R.d.l.C.); (E.A.R.J.); (H.M.G.); (C.E.D.V.); (J.P.d.l.V.)
| | - Jimena Paredes de la Vega
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla 54090, Mexico; (M.R.d.l.C.); (E.A.R.J.); (H.M.G.); (C.E.D.V.); (J.P.d.l.V.)
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla 54090, Mexico;
| | - Fidel de la Cruz Hernández-Hernández
- Avenida Instituto Politécnico Nacional # 2508, Colonia San Pedro Zacatenco, Delegación Gustavo A. Madero, C.P. Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City 07360, Mexico;
| | - Felipe Vaca Paniagua
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla 54090, Mexico; (M.R.d.l.C.); (E.A.R.J.); (H.M.G.); (C.E.D.V.); (J.P.d.l.V.)
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla 54090, Mexico;
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Ciudad de México 14080, Mexico
- Correspondence: ; Tel.: +52-55-5623-1333 (ext. 39788)
| |
Collapse
|
34
|
Diagnosis of Classic Homocystinuria in Two Boys Presenting with Acute Cerebral Venous Thrombosis and Neurologic Dysfunction after Normal Newborn Screening. Int J Neonatal Screen 2021; 7:ijns7030048. [PMID: 34449521 PMCID: PMC8395908 DOI: 10.3390/ijns7030048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/15/2021] [Accepted: 07/16/2021] [Indexed: 11/17/2022] Open
Abstract
Homocystinuria, caused by cystathionine β-synthase deficiency, is a rare inherited disorder involving metabolism of methionine. Impaired synthesis of cystathionine leads to accumulation of homocysteine that affects several organ systems leading to abnormalities in the skeletal, cardiovascular, ophthalmic and central nervous systems. We report a 14-month-old and a 7-year-old boy who presented with neurologic dysfunction and were found to have cerebral venous sinus thromboses on brain magnetic resonance imaging (MRI)/magnetic resonance venogram (MRV) and metabolic and hypercoagulable work-up were consistent with classic homocystinuria. The 14-month-old boy had normal newborn screening. The 7-year-old boy initially had an abnormal newborn screen for homocystinuria but second tier test that consisted of total homocysteine was normal, so his newborn screen was reported as normal. With the advent of expanded newborn screening many treatable metabolic disorders are detected before affected infants and children become symptomatic. Methionine is the primary target in newborn screening for homocystinuria and total homocysteine is a secondary target. Screening is usually performed after 24-48 h of life in most states in the US and some states perform a second screen as a policy on all tested newborns or based on when the initial newborn screen was performed. This is done in hopes of detecting infants who may have been missed on their first screen. In the United Kingdom, NBS using dried blood spot is performed at 5 to 8 days after birth. It is universally known that methionine is a poor target and newborn screening laboratories have used different cutoffs for a positive screen. Reducing the methionine cutoff increases the sensitivity but not necessarily the specificity of the test and increasing the cutoff will miss babies who may have HCU whose levels may not be high enough to be detected at their age of ascertainment. It is not clear whether adjusting methionine level to decrease the false negative rates combined with total homocysteine as a second-tier test can be used effectively and feasibly to detect newborns with HCU. Between December 2005 and December 2020, 827,083 newborns were screened in Kentucky by MS/MS. Kentucky NBS program uses the postanalytical tools offered by the Collaborative Laboratory Integrated Reports (CLIR) project which considers gestational age and birthweight. One case of classical homocystinuria was detected and two were missed on first and second tier tests respectively. The newborn that had confirmed classical homocystinuria was one of twenty-three newborns that were referred for second tier test because of elevated methionine (cutoff is >60 µmol/L) and/or Met/Phe ratio (cutoff is >1.0); all 23 dried blood spots had elevated total homocysteine. One of the subjects of this case report had a normal methionine on initial screen and the other had a normal second-tier total homocysteine level. The performance of methionine and total homocysteine as screening analytes for homocystinuria suggest that it may be time for newborn screening programs to consider adopting next generation sequencing (NGS) platforms as alternate modality of metabolic newborn screening. Because of cost considerations, newborn screening programs may not want to adopt NGS, but the downstream healthcare cost incurred due to missed cases and the associated morbidity of affected persons far exceed costs to newborn screen programs. Since NGS is becoming more widely available and inexpensive, it may be feasible to change testing algorithms to use Newborn Metabolic NGS as the primary mode of testing on dry blood specimens with confirmation with biochemical testing. Some commercial laboratories have Newborn Screening Metabolic gene panel that includes all metabolic disorders on the most comprehensive newborn screening panel in addition to many other conditions that are not on the panel. A more targeted NGS panel can be designed that may not cost much and eventually help avoid the pitfalls associated with delayed diagnosis and cost of screening.
Collapse
|
35
|
Zhang X, Chen Q, Song Y, Guo P, Wang Y, Luo S, Zhang Y, Zhou C, Li D, Chen Y, Wei H. Epimutation of MMACHC compound to a genetic mutation in cblC cases. Mol Genet Genomic Med 2021; 9:e1625. [PMID: 33982424 PMCID: PMC8222841 DOI: 10.1002/mgg3.1625] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 01/05/2021] [Accepted: 02/10/2021] [Indexed: 12/22/2022] Open
Abstract
Background Methylmalonic aciduria (MMA) combined with homocystinuria, cobalamin(cbl)C deficiency type (OMIM 277400), is the most common autosomal recessive inherited disorder of intracellular cobalamin metabolism caused by mutations in the MMACHC gene (OMIM 609831), of which more than 100 mutations have been identified to date. In this study, we only identified a coding mutation in one allele at the MMACHC gene locus, and no large fragments deletion or duplication were found. Up to now, only three epimutation cblC cases were reported. We hypothesized whether the MMACHC was hypermethylated. Methods To address this hypothesis, the entire coding region and adjacent splice sites of the panel genes involved in metabolic diseases were sequenced using the Illumina HiSeq X platform, followed by confirmation via Sanger sequencing in their parents and brothers. Methylation analysis of the MMACHC was performed using an EpiTect Bisulfite Kit and methylation‐specific PCR (MSP) to investigate the role of epimutations in cblC disease. Results We identified a clearly pathogenic single heterozygous c.658_660del, p. (K220del) mutation, which was also identified in the mother. Analysis of the MMACHC indicated a heterozygous epimutation consisting of 34 hypermethylated CpG sites in a CpG island encompassing the promoter and first exon of the MMACHC, which was also identified in the father. Furthermore, we identified a single heterozygous c.*2C>T mutation in the sixth exon of the PRDX1 (OMIM 176763) in patients and their fathers, which was the only sequence variation that segregated with the MMACHC methylation. Neither c.658_660del and epimutation in MMACHC nor c.*2C>T in PRDX1 was discovered in her brother. Conclusion We report compound heterozygotes in MMACHC for a genetic mutation and an epimutation in cblC cases. To our best knowledge, this is the first report of two cblC cases from China caused by compound heterozygous mutations with a coding mutation in one allele and an epimutation in the other at the MMACHC locus.
Collapse
Affiliation(s)
- Xiaoman Zhang
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Qiong Chen
- Department of Pediatric Endocrinology and Genetic Metabolism, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Yinsen Song
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Pengbo Guo
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Yanhong Wang
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Shuying Luo
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Yaodong Zhang
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Chongchen Zhou
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Dongxiao Li
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Yongxing Chen
- Department of Pediatric Endocrinology and Genetic Metabolism, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Haiyan Wei
- Department of Pediatric Endocrinology and Genetic Metabolism, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| |
Collapse
|
36
|
Banerjee R, Gouda H, Pillay S. Redox-Linked Coordination Chemistry Directs Vitamin B 12 Trafficking. Acc Chem Res 2021; 54:2003-2013. [PMID: 33797888 DOI: 10.1021/acs.accounts.1c00083] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Metals are partners for an estimated one-third of the proteome and vary in complexity from mononuclear centers to organometallic cofactors. Vitamin B12 or cobalamin represents the epitome of this complexity and is the product of an assembly line comprising some 30 enzymes. Unable to biosynthesize cobalamin, mammals rely on dietary provision of this essential cofactor, which is needed by just two enzymes, one each in the cytoplasm (methionine synthase) and the mitochondrion (methylmalonyl-CoA mutase). Brilliant clinical genetics studies on patients with inborn errors of cobalamin metabolism spanning several decades had identified at least seven genetic loci in addition to the two encoding B12 enzymes. While cells are known to house a cadre of chaperones dedicated to metal trafficking pathways that contain metal reactivity and confer targeting specificity, the seemingly supernumerary chaperones in the B12 pathway had raised obvious questions as to the rationale for their existence.With the discovery of the genes underlying cobalamin disorders, our laboratory has been at the forefront of ascribing functions to B12 chaperones and elucidating the intricate redox-linked coordination chemistry and protein-linked cofactor conformational dynamics that orchestrate the processing and translocation of cargo along the trafficking pathway. These studies have uncovered novel chemistry that exploits the innate chemical versatility of alkylcobalamins, i.e., the ability to form and dismantle the cobalt-carbon bond using homolytic or heterolytic chemistry. In addition, they have revealed the practical utility of the dimethylbenzimidazole tail, an appendage unique to cobalamins and absent in the structural cousins, porphyrin, chlorin, and corphin, as an instrument for facilitating cofactor transfer between active sites.In this Account, we navigate the chemistry of the B12 trafficking pathway from its point of entry into cells, through lysosomes, and into the cytoplasm, where incoming cobalamin derivatives with a diversity of upper ligands are denuded by the β-ligand transferase activity of CblC to the common cob(II)alamin intermediate. The broad reaction and lax substrate specificity of CblC also enables conversion of cyanocobalamin (technically, vitamin B12, i.e., the form of the cofactor in one-a-day supplements), to cob(II)alamin. CblD then hitches up with CblC via a unique Co-sulfur bond to cob(II)alamin at a bifurcation point, leading to the cytoplasmic methylcobalamin or mitochondrial 5'-deoxyadenosylcobalamin branch. Mutations at loci upstream of the junction point typically affect both branches, leading to homocystinuria and methylmalonic aciduria, whereas mutations in downstream loci lead to one or the other disease. Elucidation of the biochemical penalties associated with individual mutations is providing molecular insights into the clinical data and, in some instances, identifying which cobalamin derivative(s) might be therapeutically beneficial.Our studies on B12 trafficking are revealing strategies for cofactor sequestration and mobilization from low- to high-affinity and low- to high-coordination-number sites, which in turn are regulated by protein dynamics that constructs ergonomic cofactor binding pockets. While these B12 lessons might be broadly relevant to other metal trafficking pathways, much remains to be learned. This Account concludes by identifying some of the major gaps and challenges that are needed to complete our understanding of B12 trafficking.
Collapse
Affiliation(s)
- Ruma Banerjee
- Department of Biological Chemistry, Michigan Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Harsha Gouda
- Department of Biological Chemistry, Michigan Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Shubhadra Pillay
- Department of Biological Chemistry, Michigan Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
37
|
Pillai NR, Miller D, Pierpont EI, Berry SA, Aggarwal A. Cobalamin J disease detected on newborn screening: Novel variant and normal neurodevelopmental course. Am J Med Genet A 2021; 185:1870-1874. [PMID: 33729671 DOI: 10.1002/ajmg.a.62170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 03/02/2021] [Indexed: 11/07/2022]
Abstract
Cobalamin J disease (CblJ) is an ultra-rare autosomal recessive disorder of intracellular cobalamin metabolism associated with combined methylmalonic acidemia and homocystinuria. It is caused by pathogenic variants in ABCD4, which encodes an ATP-binding cassette (ABC) transporter that affects the lysosomal release of cobalamin (Cbl) into the cytoplasm. Only six cases of CblJ have been reported in the literature. Described clinical features include feeding difficulties, failure to thrive, hypotonia, seizures, developmental delay, and hematological abnormalities. Information on clinical outcomes is extremely limited, and no cases of presymptomatic diagnosis have been reported. We describe a now 17-month-old male with CblJ detected by newborn screening and confirmed by biochemical, molecular, and complementation studies. With early detection and initiation of treatment, this patient has remained asymptomatic with normal growth parameters and neurodevelopmental function. To the best of our knowledge, this report represents the first asymptomatic and neurotypical patient with CblJ.
Collapse
Affiliation(s)
- Nishitha R Pillai
- Division of Genetics and Metabolism, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Dana Miller
- M Health Fairview, Minneapolis, Minnesota, USA
| | - Elizabeth I Pierpont
- Division of Clinical Behavioral Neuroscience, Department of Pediatrics, University of Minnesota, Minneapolis, Minneapolis, Minnesota, USA
| | - Susan A Berry
- Division of Genetics and Metabolism, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Anjali Aggarwal
- Division of Genetics and Metabolism, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
38
|
Current Nanocarrier Strategies Improve Vitamin B12 Pharmacokinetics, Ameliorate Patients' Lives, and Reduce Costs. NANOMATERIALS 2021; 11:nano11030743. [PMID: 33809596 PMCID: PMC8001893 DOI: 10.3390/nano11030743] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/17/2022]
Abstract
Vitamin B12 (VitB12) is a naturally occurring compound produced by microorganisms and an essential nutrient for humans. Several papers highlight the role of VitB12 deficiency in bone and heart health, depression, memory performance, fertility, embryo development, and cancer, while VitB12 treatment is crucial for survival in inborn errors of VitB12 metabolism. VitB12 is administrated through intramuscular injection, thus impacting the patients’ lifestyle, although it is known that oral administration may meet the specific requirement even in the case of malabsorption. Furthermore, the high-dose injection of VitB12 does not ensure a constant dosage, while the oral route allows only 1.2% of the vitamin to be absorbed in human beings. Nanocarriers are promising nanotechnology that can enable therapies to be improved, reducing side effects. Today, nanocarrier strategies applied at VitB12 delivery are at the initial phase and aim to simplify administration, reduce costs, improve pharmacokinetics, and ameliorate the quality of patients’ lives. The safety of nanotechnologies is still under investigation and few treatments involving nanocarriers have been approved, so far. Here, we highlight the role of VitB12 in human metabolism and diseases, and the issues linked to its molecule properties, and discuss how nanocarriers can improve the therapy and supplementation of the vitamin and reduce possible side effects and limits.
Collapse
|
39
|
Martino F, Magenta A, Troccoli ML, Martino E, Torromeo C, Putotto C, Barillà F. Long-term outcome of a patient with Transcobalamin deficiency caused by the homozygous c.1115_1116delCA mutation in TCN2 gene: a case report. Ital J Pediatr 2021; 47:54. [PMID: 33685478 PMCID: PMC7941906 DOI: 10.1186/s13052-021-01007-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 02/22/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Transcobalamin deficiency is a rare autosomal recessive inborn error of cobalamin transport (prevalence: < 1/1000000) which clinically manifests in early infancy. CASE PRESENTATION We describe the case of a 31 years old woman who at the age of 30 days presented with the classical clinical and laboratory signs of an inborn error of vitamin B12 metabolism. Family history revealed a sister who died at the age of 3 months with a similar clinical syndrome and with pancytopenia. She was started on empirical intramuscular (IM) cobalamin supplements (injections of hydroxocobalamin 1 mg/day for 1 week and then 1 mg twice a week) and several transfusions of washed and concentrated red blood cells. With these treatments a clear improvement in symptoms was observed, with the disappearance of vomiting, diarrhea and normalization of the full blood count. At 8 years of age injections were stopped for about two and a half months causing the appearance of pancytopenia. IM hydroxocobalamin was then restarted sine die. The definitive diagnosis could only be established at 29 years of age when a genetic evaluation revealed the homozygous c.1115_1116delCA mutation of TCN2 gene (p.Q373GfsX38). Currently she is healthy and she is taking 1 mg of IM hydroxocobalamin once a week. CONCLUSIONS Our case report highlights that early detection of TC deficiency and early initiation of aggressive IM treatment is likely associated with disease control and an overall favorable outcome.
Collapse
Affiliation(s)
- Francesco Martino
- Department of Pediatrics Gynecology and Obstetrics, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.
| | - Alessandra Magenta
- National Research Council of Italy (CNR), Institute of Translational Pharmacology IFT, Via Fosso del Cavaliere 100, 00133, Rome, Italy
| | - Maria Letizia Troccoli
- Clinical Analysis and Biochemistry Laboratory, Sant'Andrea Hospital, Via di Grottarossa, 1035/1039, 00189, Rome, Italy
| | - Eliana Martino
- Department of Pediatrics Gynecology and Obstetrics, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Concetta Torromeo
- Department of Cardiovascular, Respiratory, Nephrology, Anesthesiology and Geriatric Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - Carolina Putotto
- Department of Pediatrics Gynecology and Obstetrics, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Francesco Barillà
- Department of Cardiovascular, Respiratory, Nephrology, Anesthesiology and Geriatric Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| |
Collapse
|
40
|
Kaur R, Attri SV, Saini AG, Sankhyan N. A high frequency and geographical distribution of MMACHC R132* mutation in children with cobalamin C defect. Amino Acids 2021; 53:253-264. [PMID: 33515116 DOI: 10.1007/s00726-021-02942-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 12/19/2020] [Indexed: 12/16/2022]
Abstract
Cobalamin C defect is caused by pathogenic variants in the MMACHC gene leading to impaired conversion of dietary vitamin B12 into methylcobalamin and adenosylcobalamin. Variants in the MMACHC gene cause accumulation of methylmalonic acid and homocysteine along with decreased methionine synthesis. The spectrum of MMACHC gene variants differs in various populations. A total of 19 North Indian children (age 0-18 years) with elevated methylmalonic acid and homocysteine were included in the study, and their DNA samples were subjected to Sanger sequencing of coding exons with flanking intronic regions of MMACHC gene. The genetic analysis resulted in the identification of a common pathogenic nonsense mutation, c.394C > T (R132*) in 85.7% of the unrelated cases with suspected cobalamin C defect. Two other known mutations c.347T > C (7%) and c.316G > A were also detected. Plasma homocysteine was significantly elevated (> 100 µmol/L) in 75% of the cases and methionine was decreased in 81% of the cases. Propionyl (C3)-carnitine, the primary marker for cobalamin C defect, was found to be elevated in only 43.75% of cases. However, the secondary markers such as C3/C2 and C3/C16 ratios were elevated in 87.5% and 100% of the cases, respectively. Neurological manifestations were the most common in our cohort. Our findings of the high frequency of a single MMACHC R132* mutation in cases with combined homocystinuria and methylmalonic aciduria may be proven helpful in designing a cost-effective and time-saving diagnostic strategy for resource-constraint settings. Since the R132* mutation is located near the last exon-exon junction, this is a potential target for the read-through therapeutics.
Collapse
Affiliation(s)
- Rajdeep Kaur
- Pediatric Biochemistry Unit, Department of Pediatrics, PGIMER, Chandigarh, 160012, India
| | - Savita Verma Attri
- Pediatric Biochemistry Unit, Department of Pediatrics, PGIMER, Chandigarh, 160012, India.
| | - Arushi Gahlot Saini
- Pediatric Neurology Unit, Department of Pediatrics, PGIMER, Chandigarh, India
| | - Naveen Sankhyan
- Pediatric Neurology Unit, Department of Pediatrics, PGIMER, Chandigarh, India
| |
Collapse
|
41
|
Bourque DK, Mellin‐Sanchez LE, Bullivant G, Cruz V, Feigenbaum A, Hewson S, Raiman J, Schulze A, Siriwardena K, Mercimek‐Andrews S. Outcomes of patients with cobalamin C deficiency: A single center experience. JIMD Rep 2021; 57:102-114. [PMID: 33473346 PMCID: PMC7802631 DOI: 10.1002/jmd2.12179] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/16/2020] [Accepted: 10/21/2020] [Indexed: 12/29/2022] Open
Abstract
Biallelic variants in MMACHC results in the combined methylmalonic aciduria and homocystinuria, called cobalamin (cbl) C (cblC) deficiency. We report 26 patients with cblC deficiency with their phenotypes, genotypes, biochemical parameters, and treatment outcomes, who were diagnosed and treated at our center. We divided all cblC patients into two groups: group 1: SX group: identified after manifestations of symptoms (n = 11) and group 2: NB group: identified during the asymptomatic period via newborn screening (NBS) or positive family history of cblC deficiency (n = 15). All patients in the SX group had global developmental delay and/or cognitive dysfunction at the time of the diagnosis and at the last assessment. Seizure, stroke, retinopathy, anemia, cerebral atrophy, and thin corpus callosum in brain magnetic resonance imaging (MRI) were common in patients in the SX group. Global developmental delay and cognitive dysfunction was present in nine patients in the NB group at the last assessment. Retinopathy, anemia, and cerebral atrophy and thin corpus callosum in brain MRI were less frequent. We report favorable outcomes in patients identified in the neonatal period and treated pre-symptomatically. Identification of cblC deficiency by NBS is crucial to improve neurodevelopmental outcomes.
Collapse
Affiliation(s)
- Danielle K. Bourque
- Division of Clinical and Metabolic Genetics, Department of PediatricsThe Hospital for Sick ChildrenTorontoOntarioCanada
| | - Lizbeth E. Mellin‐Sanchez
- Division of Clinical and Metabolic Genetics, Department of PediatricsThe Hospital for Sick ChildrenTorontoOntarioCanada
| | - Garrett Bullivant
- Division of Clinical and Metabolic Genetics, Department of PediatricsThe Hospital for Sick ChildrenTorontoOntarioCanada
| | - Vivian Cruz
- Division of Clinical and Metabolic Genetics, Department of PediatricsThe Hospital for Sick ChildrenTorontoOntarioCanada
| | - Anette Feigenbaum
- Division of Clinical and Metabolic Genetics, Department of PediatricsThe Hospital for Sick ChildrenTorontoOntarioCanada
| | - Stacy Hewson
- Division of Clinical and Metabolic Genetics, Department of PediatricsThe Hospital for Sick ChildrenTorontoOntarioCanada
| | - Julian Raiman
- Division of Clinical and Metabolic Genetics, Department of PediatricsThe Hospital for Sick ChildrenTorontoOntarioCanada
| | - Andreas Schulze
- Division of Clinical and Metabolic Genetics, Department of PediatricsThe Hospital for Sick ChildrenTorontoOntarioCanada
- Department of PediatricsUniversity of TorontoTorontoOntarioCanada
| | - Komudi Siriwardena
- Division of Clinical and Metabolic Genetics, Department of PediatricsThe Hospital for Sick ChildrenTorontoOntarioCanada
| | - Saadet Mercimek‐Andrews
- Division of Clinical and Metabolic Genetics, Department of PediatricsThe Hospital for Sick ChildrenTorontoOntarioCanada
- Department of PediatricsUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
42
|
Li Q, Jin H, Liu Y, Rong Y, Yang T, Nie X, Song W. Determination of Cytokines and Oxidative Stress Biomarkers in Cognitive Impairment Induced by Methylmalonic Acidemia. Neuroimmunomodulation 2021; 28:178-186. [PMID: 34340239 DOI: 10.1159/000511590] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 09/06/2020] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Methylmalonic acidemia (MMA) is the most common organic acidemia in children. Many patients with MMA suffered from cognitive impairments. The aim of this study was to identify the significance of cytokines and oxidative stress biomarkers in MMA-induced cognitive impairment. METHODS We enrolled 64 children with combined MMA and homocystinuria and 64 age- and sex-matched healthy volunteers. Participants were subsequently classified as with or without cognitive impairments using a uniform neuropsychological assessment test. Serum samples were collected. The serum levels of cytokines and oxidative stress biomarkers were measured using the ELISA or chemical methods. RESULTS Compared to control group, the serum levels of tumor necrosis factor (TNF)-α, interleukin (IL)-6, malondialdehyde (MDA), and nitric oxide (NO) in the MMA patients increased markedly (p < 0.05); glutathione (GSH) and superoxide dismutase (SOD) decreased obviously (p < 0.01). The levels of IL-6, TNF-α, NO, and MDA in the serum were negatively associated with DQ or IQ scores. The levels of GSH and SOD in the serum were positively correlated with DQ or IQ scores. After receiver operating characteristic curve analysis, NO was the most useful individual marker for distinguishing the cognitive dysfunction, corresponding to the area under ROC curve (AUC) of 0.82 (95% CI, 0.74-0.91), sensitivity of 76.60%, and specificity of 80.25%. GSH and MDA were also useful for diagnosis of MMA-induced cognitive dysfunction, corresponding to the AUC of 0.80 (95% CI, 0.70-0.89), and 0.73 (95% CI, 0.63-0.82), respectively. The sensitivity and specificity of GSH were 72.34 and 80.25%, respectively. The sensitivity and specificity of MDA were 85.11 and 51.85%, respectively. CONCLUSIONS The high-concentration methylmalonic acid in the blood induced immune cells to release pro-inflammatory cytokines such as TNF-α and IL-6. These cytokines and high-concentration methylmalonic acid stimulated the immune cells to produce reactive oxygen species (ROS) and reactive nitrogen species (RNS). The serum methylmalonic acid, cytokines, ROS, and RNS were across the blood-brain barrier and induced cognitive impairment. The small molecule substances such as serum NO, MDA, and GSH participated in the process of neuroinflammation and oxidative stress injury induced by MMA and could be useful for distinguishing the cognitive impairment.
Collapse
Affiliation(s)
- Qiliang Li
- Department of Medical Laboratory, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Hong Jin
- Department of Neurology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Ying Liu
- Department of Medical Laboratory, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yu Rong
- Department of Rehabilitation, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Tana Yang
- Department of Medical Laboratory, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Xiaolu Nie
- Center for Clinical Epidemiology and Evidence-Based Medicine, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Wenqi Song
- Department of Medical Laboratory, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| |
Collapse
|
43
|
He R, Zhang H, Kang L, Li H, Shen M, Zhang Y, Mo R, Liu Y, Song J, Chen Z, Liu Y, Jin Y, Li M, Dong H, Zheng H, Li D, Qin J, Zhang H, Huang M, Liang D, Tian Y, Yao H, Yang Y. Analysis of 70 patients with hydrocephalus due to cobalamin C deficiency. Neurology 2020; 95:e3129-e3137. [PMID: 32943488 DOI: 10.1212/wnl.0000000000010912] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 07/23/2020] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVE To analyze the clinical characteristics of patients with hydrocephalus secondary to cobalamin C (cblC) deficiency and to discuss the optimal strategies for assessing and treating such patients by performing clinical and laboratory studies in 70 patients. METHODS A total of 1,211 patients were clinically diagnosed with methylmalonic acidemia (MMA) from 1998 to 2019. Among them, cblC deficiency was confirmed in 70 patients with hydrocephalus by brain imaging and biochemical and genetic analysis. RESULTS Of the 70 patients, 67 (95.7%) had early-onset MMA and homocystinuria. The patients typically had high blood propionylcarnitine and total homocysteine, low methionine, and methylmalonic aciduria. Signs of intracranial hypertension were relatively rare. We measured ventricular dilatation early in the disease by cranial ultrasound and MRI and/or CT. Eighteen different MMACHC mutations, including 4 novel mutations (c.427C>T, c.568insT, c.599G>A, and c.615C>A), were identified biallelically in all 70 patients. c.609G>A was the most frequent mutation, followed by c.658_660del, c.217C>T, and c.567dupT. Three cases were diagnosed by postmortem study. Metabolic therapy, including cobalamin injections supplemented with oral l-carnitine and betaine, was administered in the remaining 67 cases. A ventriculoperitoneal shunt was performed in 36 cases. During the follow-up, psychomotor development, nystagmus, impaired vision, and sunset eyes improved gradually. CONCLUSION Hydrocephalus is a severe condition with several different causes. In this study, ventriculomegaly was found in 70 patients with cblC deficiency. Early diagnosis, etiologic treatment, and prompt surgical intervention are crucial to improve the prognosis of patients.
Collapse
Affiliation(s)
- Ruxuan He
- From the Departments of Pediatrics (R.H., L.K., Y.Z., R.M., J.S., Z.C., Yi Liu, Y.J., M.L., H.D., Y.Y.) and Pediatric Surgery (H.Z., H.L., H.Y.), Peking University First Hospital; Translational Medicine Center (M.S., Y.T.), Chinese PLA General Hospital; Department of Pediatrics (Yupeng Liu, J.Q.), People's Hospital of Peking University, Beijing; Department of Pediatrics (H.Z.), First Affiliated Hospital of Henan University of Traditional Chinese Medicine; Department of Endocrinology and Genetic (D. Li), Henan Children's Hospital, Zhengzhou; Department of Pediatrics (H.Z.), Hebei Medical University Second Hospital, Shijiazhuang; Similan Clinic, (M.H.) Beijing; and School of Life Sciences (D. Liang), Central South University, Changsha, China
| | - Hongwu Zhang
- From the Departments of Pediatrics (R.H., L.K., Y.Z., R.M., J.S., Z.C., Yi Liu, Y.J., M.L., H.D., Y.Y.) and Pediatric Surgery (H.Z., H.L., H.Y.), Peking University First Hospital; Translational Medicine Center (M.S., Y.T.), Chinese PLA General Hospital; Department of Pediatrics (Yupeng Liu, J.Q.), People's Hospital of Peking University, Beijing; Department of Pediatrics (H.Z.), First Affiliated Hospital of Henan University of Traditional Chinese Medicine; Department of Endocrinology and Genetic (D. Li), Henan Children's Hospital, Zhengzhou; Department of Pediatrics (H.Z.), Hebei Medical University Second Hospital, Shijiazhuang; Similan Clinic, (M.H.) Beijing; and School of Life Sciences (D. Liang), Central South University, Changsha, China
| | - Lulu Kang
- From the Departments of Pediatrics (R.H., L.K., Y.Z., R.M., J.S., Z.C., Yi Liu, Y.J., M.L., H.D., Y.Y.) and Pediatric Surgery (H.Z., H.L., H.Y.), Peking University First Hospital; Translational Medicine Center (M.S., Y.T.), Chinese PLA General Hospital; Department of Pediatrics (Yupeng Liu, J.Q.), People's Hospital of Peking University, Beijing; Department of Pediatrics (H.Z.), First Affiliated Hospital of Henan University of Traditional Chinese Medicine; Department of Endocrinology and Genetic (D. Li), Henan Children's Hospital, Zhengzhou; Department of Pediatrics (H.Z.), Hebei Medical University Second Hospital, Shijiazhuang; Similan Clinic, (M.H.) Beijing; and School of Life Sciences (D. Liang), Central South University, Changsha, China
| | - Hui Li
- From the Departments of Pediatrics (R.H., L.K., Y.Z., R.M., J.S., Z.C., Yi Liu, Y.J., M.L., H.D., Y.Y.) and Pediatric Surgery (H.Z., H.L., H.Y.), Peking University First Hospital; Translational Medicine Center (M.S., Y.T.), Chinese PLA General Hospital; Department of Pediatrics (Yupeng Liu, J.Q.), People's Hospital of Peking University, Beijing; Department of Pediatrics (H.Z.), First Affiliated Hospital of Henan University of Traditional Chinese Medicine; Department of Endocrinology and Genetic (D. Li), Henan Children's Hospital, Zhengzhou; Department of Pediatrics (H.Z.), Hebei Medical University Second Hospital, Shijiazhuang; Similan Clinic, (M.H.) Beijing; and School of Life Sciences (D. Liang), Central South University, Changsha, China
| | - Ming Shen
- From the Departments of Pediatrics (R.H., L.K., Y.Z., R.M., J.S., Z.C., Yi Liu, Y.J., M.L., H.D., Y.Y.) and Pediatric Surgery (H.Z., H.L., H.Y.), Peking University First Hospital; Translational Medicine Center (M.S., Y.T.), Chinese PLA General Hospital; Department of Pediatrics (Yupeng Liu, J.Q.), People's Hospital of Peking University, Beijing; Department of Pediatrics (H.Z.), First Affiliated Hospital of Henan University of Traditional Chinese Medicine; Department of Endocrinology and Genetic (D. Li), Henan Children's Hospital, Zhengzhou; Department of Pediatrics (H.Z.), Hebei Medical University Second Hospital, Shijiazhuang; Similan Clinic, (M.H.) Beijing; and School of Life Sciences (D. Liang), Central South University, Changsha, China
| | - Yao Zhang
- From the Departments of Pediatrics (R.H., L.K., Y.Z., R.M., J.S., Z.C., Yi Liu, Y.J., M.L., H.D., Y.Y.) and Pediatric Surgery (H.Z., H.L., H.Y.), Peking University First Hospital; Translational Medicine Center (M.S., Y.T.), Chinese PLA General Hospital; Department of Pediatrics (Yupeng Liu, J.Q.), People's Hospital of Peking University, Beijing; Department of Pediatrics (H.Z.), First Affiliated Hospital of Henan University of Traditional Chinese Medicine; Department of Endocrinology and Genetic (D. Li), Henan Children's Hospital, Zhengzhou; Department of Pediatrics (H.Z.), Hebei Medical University Second Hospital, Shijiazhuang; Similan Clinic, (M.H.) Beijing; and School of Life Sciences (D. Liang), Central South University, Changsha, China
| | - Ruo Mo
- From the Departments of Pediatrics (R.H., L.K., Y.Z., R.M., J.S., Z.C., Yi Liu, Y.J., M.L., H.D., Y.Y.) and Pediatric Surgery (H.Z., H.L., H.Y.), Peking University First Hospital; Translational Medicine Center (M.S., Y.T.), Chinese PLA General Hospital; Department of Pediatrics (Yupeng Liu, J.Q.), People's Hospital of Peking University, Beijing; Department of Pediatrics (H.Z.), First Affiliated Hospital of Henan University of Traditional Chinese Medicine; Department of Endocrinology and Genetic (D. Li), Henan Children's Hospital, Zhengzhou; Department of Pediatrics (H.Z.), Hebei Medical University Second Hospital, Shijiazhuang; Similan Clinic, (M.H.) Beijing; and School of Life Sciences (D. Liang), Central South University, Changsha, China
| | - Yupeng Liu
- From the Departments of Pediatrics (R.H., L.K., Y.Z., R.M., J.S., Z.C., Yi Liu, Y.J., M.L., H.D., Y.Y.) and Pediatric Surgery (H.Z., H.L., H.Y.), Peking University First Hospital; Translational Medicine Center (M.S., Y.T.), Chinese PLA General Hospital; Department of Pediatrics (Yupeng Liu, J.Q.), People's Hospital of Peking University, Beijing; Department of Pediatrics (H.Z.), First Affiliated Hospital of Henan University of Traditional Chinese Medicine; Department of Endocrinology and Genetic (D. Li), Henan Children's Hospital, Zhengzhou; Department of Pediatrics (H.Z.), Hebei Medical University Second Hospital, Shijiazhuang; Similan Clinic, (M.H.) Beijing; and School of Life Sciences (D. Liang), Central South University, Changsha, China
| | - Jinqing Song
- From the Departments of Pediatrics (R.H., L.K., Y.Z., R.M., J.S., Z.C., Yi Liu, Y.J., M.L., H.D., Y.Y.) and Pediatric Surgery (H.Z., H.L., H.Y.), Peking University First Hospital; Translational Medicine Center (M.S., Y.T.), Chinese PLA General Hospital; Department of Pediatrics (Yupeng Liu, J.Q.), People's Hospital of Peking University, Beijing; Department of Pediatrics (H.Z.), First Affiliated Hospital of Henan University of Traditional Chinese Medicine; Department of Endocrinology and Genetic (D. Li), Henan Children's Hospital, Zhengzhou; Department of Pediatrics (H.Z.), Hebei Medical University Second Hospital, Shijiazhuang; Similan Clinic, (M.H.) Beijing; and School of Life Sciences (D. Liang), Central South University, Changsha, China
| | - Zhehui Chen
- From the Departments of Pediatrics (R.H., L.K., Y.Z., R.M., J.S., Z.C., Yi Liu, Y.J., M.L., H.D., Y.Y.) and Pediatric Surgery (H.Z., H.L., H.Y.), Peking University First Hospital; Translational Medicine Center (M.S., Y.T.), Chinese PLA General Hospital; Department of Pediatrics (Yupeng Liu, J.Q.), People's Hospital of Peking University, Beijing; Department of Pediatrics (H.Z.), First Affiliated Hospital of Henan University of Traditional Chinese Medicine; Department of Endocrinology and Genetic (D. Li), Henan Children's Hospital, Zhengzhou; Department of Pediatrics (H.Z.), Hebei Medical University Second Hospital, Shijiazhuang; Similan Clinic, (M.H.) Beijing; and School of Life Sciences (D. Liang), Central South University, Changsha, China
| | - Yi Liu
- From the Departments of Pediatrics (R.H., L.K., Y.Z., R.M., J.S., Z.C., Yi Liu, Y.J., M.L., H.D., Y.Y.) and Pediatric Surgery (H.Z., H.L., H.Y.), Peking University First Hospital; Translational Medicine Center (M.S., Y.T.), Chinese PLA General Hospital; Department of Pediatrics (Yupeng Liu, J.Q.), People's Hospital of Peking University, Beijing; Department of Pediatrics (H.Z.), First Affiliated Hospital of Henan University of Traditional Chinese Medicine; Department of Endocrinology and Genetic (D. Li), Henan Children's Hospital, Zhengzhou; Department of Pediatrics (H.Z.), Hebei Medical University Second Hospital, Shijiazhuang; Similan Clinic, (M.H.) Beijing; and School of Life Sciences (D. Liang), Central South University, Changsha, China
| | - Ying Jin
- From the Departments of Pediatrics (R.H., L.K., Y.Z., R.M., J.S., Z.C., Yi Liu, Y.J., M.L., H.D., Y.Y.) and Pediatric Surgery (H.Z., H.L., H.Y.), Peking University First Hospital; Translational Medicine Center (M.S., Y.T.), Chinese PLA General Hospital; Department of Pediatrics (Yupeng Liu, J.Q.), People's Hospital of Peking University, Beijing; Department of Pediatrics (H.Z.), First Affiliated Hospital of Henan University of Traditional Chinese Medicine; Department of Endocrinology and Genetic (D. Li), Henan Children's Hospital, Zhengzhou; Department of Pediatrics (H.Z.), Hebei Medical University Second Hospital, Shijiazhuang; Similan Clinic, (M.H.) Beijing; and School of Life Sciences (D. Liang), Central South University, Changsha, China
| | - Mengqiu Li
- From the Departments of Pediatrics (R.H., L.K., Y.Z., R.M., J.S., Z.C., Yi Liu, Y.J., M.L., H.D., Y.Y.) and Pediatric Surgery (H.Z., H.L., H.Y.), Peking University First Hospital; Translational Medicine Center (M.S., Y.T.), Chinese PLA General Hospital; Department of Pediatrics (Yupeng Liu, J.Q.), People's Hospital of Peking University, Beijing; Department of Pediatrics (H.Z.), First Affiliated Hospital of Henan University of Traditional Chinese Medicine; Department of Endocrinology and Genetic (D. Li), Henan Children's Hospital, Zhengzhou; Department of Pediatrics (H.Z.), Hebei Medical University Second Hospital, Shijiazhuang; Similan Clinic, (M.H.) Beijing; and School of Life Sciences (D. Liang), Central South University, Changsha, China
| | - Hui Dong
- From the Departments of Pediatrics (R.H., L.K., Y.Z., R.M., J.S., Z.C., Yi Liu, Y.J., M.L., H.D., Y.Y.) and Pediatric Surgery (H.Z., H.L., H.Y.), Peking University First Hospital; Translational Medicine Center (M.S., Y.T.), Chinese PLA General Hospital; Department of Pediatrics (Yupeng Liu, J.Q.), People's Hospital of Peking University, Beijing; Department of Pediatrics (H.Z.), First Affiliated Hospital of Henan University of Traditional Chinese Medicine; Department of Endocrinology and Genetic (D. Li), Henan Children's Hospital, Zhengzhou; Department of Pediatrics (H.Z.), Hebei Medical University Second Hospital, Shijiazhuang; Similan Clinic, (M.H.) Beijing; and School of Life Sciences (D. Liang), Central South University, Changsha, China
| | - Hong Zheng
- From the Departments of Pediatrics (R.H., L.K., Y.Z., R.M., J.S., Z.C., Yi Liu, Y.J., M.L., H.D., Y.Y.) and Pediatric Surgery (H.Z., H.L., H.Y.), Peking University First Hospital; Translational Medicine Center (M.S., Y.T.), Chinese PLA General Hospital; Department of Pediatrics (Yupeng Liu, J.Q.), People's Hospital of Peking University, Beijing; Department of Pediatrics (H.Z.), First Affiliated Hospital of Henan University of Traditional Chinese Medicine; Department of Endocrinology and Genetic (D. Li), Henan Children's Hospital, Zhengzhou; Department of Pediatrics (H.Z.), Hebei Medical University Second Hospital, Shijiazhuang; Similan Clinic, (M.H.) Beijing; and School of Life Sciences (D. Liang), Central South University, Changsha, China
| | - Dongxiao Li
- From the Departments of Pediatrics (R.H., L.K., Y.Z., R.M., J.S., Z.C., Yi Liu, Y.J., M.L., H.D., Y.Y.) and Pediatric Surgery (H.Z., H.L., H.Y.), Peking University First Hospital; Translational Medicine Center (M.S., Y.T.), Chinese PLA General Hospital; Department of Pediatrics (Yupeng Liu, J.Q.), People's Hospital of Peking University, Beijing; Department of Pediatrics (H.Z.), First Affiliated Hospital of Henan University of Traditional Chinese Medicine; Department of Endocrinology and Genetic (D. Li), Henan Children's Hospital, Zhengzhou; Department of Pediatrics (H.Z.), Hebei Medical University Second Hospital, Shijiazhuang; Similan Clinic, (M.H.) Beijing; and School of Life Sciences (D. Liang), Central South University, Changsha, China
| | - Jiong Qin
- From the Departments of Pediatrics (R.H., L.K., Y.Z., R.M., J.S., Z.C., Yi Liu, Y.J., M.L., H.D., Y.Y.) and Pediatric Surgery (H.Z., H.L., H.Y.), Peking University First Hospital; Translational Medicine Center (M.S., Y.T.), Chinese PLA General Hospital; Department of Pediatrics (Yupeng Liu, J.Q.), People's Hospital of Peking University, Beijing; Department of Pediatrics (H.Z.), First Affiliated Hospital of Henan University of Traditional Chinese Medicine; Department of Endocrinology and Genetic (D. Li), Henan Children's Hospital, Zhengzhou; Department of Pediatrics (H.Z.), Hebei Medical University Second Hospital, Shijiazhuang; Similan Clinic, (M.H.) Beijing; and School of Life Sciences (D. Liang), Central South University, Changsha, China
| | - Huifeng Zhang
- From the Departments of Pediatrics (R.H., L.K., Y.Z., R.M., J.S., Z.C., Yi Liu, Y.J., M.L., H.D., Y.Y.) and Pediatric Surgery (H.Z., H.L., H.Y.), Peking University First Hospital; Translational Medicine Center (M.S., Y.T.), Chinese PLA General Hospital; Department of Pediatrics (Yupeng Liu, J.Q.), People's Hospital of Peking University, Beijing; Department of Pediatrics (H.Z.), First Affiliated Hospital of Henan University of Traditional Chinese Medicine; Department of Endocrinology and Genetic (D. Li), Henan Children's Hospital, Zhengzhou; Department of Pediatrics (H.Z.), Hebei Medical University Second Hospital, Shijiazhuang; Similan Clinic, (M.H.) Beijing; and School of Life Sciences (D. Liang), Central South University, Changsha, China
| | - Min Huang
- From the Departments of Pediatrics (R.H., L.K., Y.Z., R.M., J.S., Z.C., Yi Liu, Y.J., M.L., H.D., Y.Y.) and Pediatric Surgery (H.Z., H.L., H.Y.), Peking University First Hospital; Translational Medicine Center (M.S., Y.T.), Chinese PLA General Hospital; Department of Pediatrics (Yupeng Liu, J.Q.), People's Hospital of Peking University, Beijing; Department of Pediatrics (H.Z.), First Affiliated Hospital of Henan University of Traditional Chinese Medicine; Department of Endocrinology and Genetic (D. Li), Henan Children's Hospital, Zhengzhou; Department of Pediatrics (H.Z.), Hebei Medical University Second Hospital, Shijiazhuang; Similan Clinic, (M.H.) Beijing; and School of Life Sciences (D. Liang), Central South University, Changsha, China
| | - Desheng Liang
- From the Departments of Pediatrics (R.H., L.K., Y.Z., R.M., J.S., Z.C., Yi Liu, Y.J., M.L., H.D., Y.Y.) and Pediatric Surgery (H.Z., H.L., H.Y.), Peking University First Hospital; Translational Medicine Center (M.S., Y.T.), Chinese PLA General Hospital; Department of Pediatrics (Yupeng Liu, J.Q.), People's Hospital of Peking University, Beijing; Department of Pediatrics (H.Z.), First Affiliated Hospital of Henan University of Traditional Chinese Medicine; Department of Endocrinology and Genetic (D. Li), Henan Children's Hospital, Zhengzhou; Department of Pediatrics (H.Z.), Hebei Medical University Second Hospital, Shijiazhuang; Similan Clinic, (M.H.) Beijing; and School of Life Sciences (D. Liang), Central South University, Changsha, China
| | - Yaping Tian
- From the Departments of Pediatrics (R.H., L.K., Y.Z., R.M., J.S., Z.C., Yi Liu, Y.J., M.L., H.D., Y.Y.) and Pediatric Surgery (H.Z., H.L., H.Y.), Peking University First Hospital; Translational Medicine Center (M.S., Y.T.), Chinese PLA General Hospital; Department of Pediatrics (Yupeng Liu, J.Q.), People's Hospital of Peking University, Beijing; Department of Pediatrics (H.Z.), First Affiliated Hospital of Henan University of Traditional Chinese Medicine; Department of Endocrinology and Genetic (D. Li), Henan Children's Hospital, Zhengzhou; Department of Pediatrics (H.Z.), Hebei Medical University Second Hospital, Shijiazhuang; Similan Clinic, (M.H.) Beijing; and School of Life Sciences (D. Liang), Central South University, Changsha, China
| | - Hongxin Yao
- From the Departments of Pediatrics (R.H., L.K., Y.Z., R.M., J.S., Z.C., Yi Liu, Y.J., M.L., H.D., Y.Y.) and Pediatric Surgery (H.Z., H.L., H.Y.), Peking University First Hospital; Translational Medicine Center (M.S., Y.T.), Chinese PLA General Hospital; Department of Pediatrics (Yupeng Liu, J.Q.), People's Hospital of Peking University, Beijing; Department of Pediatrics (H.Z.), First Affiliated Hospital of Henan University of Traditional Chinese Medicine; Department of Endocrinology and Genetic (D. Li), Henan Children's Hospital, Zhengzhou; Department of Pediatrics (H.Z.), Hebei Medical University Second Hospital, Shijiazhuang; Similan Clinic, (M.H.) Beijing; and School of Life Sciences (D. Liang), Central South University, Changsha, China.
| | - Yanling Yang
- From the Departments of Pediatrics (R.H., L.K., Y.Z., R.M., J.S., Z.C., Yi Liu, Y.J., M.L., H.D., Y.Y.) and Pediatric Surgery (H.Z., H.L., H.Y.), Peking University First Hospital; Translational Medicine Center (M.S., Y.T.), Chinese PLA General Hospital; Department of Pediatrics (Yupeng Liu, J.Q.), People's Hospital of Peking University, Beijing; Department of Pediatrics (H.Z.), First Affiliated Hospital of Henan University of Traditional Chinese Medicine; Department of Endocrinology and Genetic (D. Li), Henan Children's Hospital, Zhengzhou; Department of Pediatrics (H.Z.), Hebei Medical University Second Hospital, Shijiazhuang; Similan Clinic, (M.H.) Beijing; and School of Life Sciences (D. Liang), Central South University, Changsha, China.
| |
Collapse
|
44
|
Abstract
The recently delineated structure- and reactivity-based concept of antivitamins B12 has begun to bear fruit by the generation, and study, of a range of such B12 -dummies, either vitamin B12 -derived, or transition metal analogues that also represent potential antivitamins B12 or specific B12 -antimetabolites. As reviewed here, this has opened up new research avenues in organometallic B12 -chemistry and bioinorganic coordination chemistry. Exploratory studies with antivitamins B12 have, furthermore, revealed some of their potential, as pharmacologically interesting compounds, for inducing B12 -deficiency in a range of organisms, from hospital resistant bacteria to laboratory mice. The derived capacity of antivitamins B12 to induce functional B12 -deficiency in mammalian cells and organs also suggest their valuable potential as growth inhibitors of cancerous human and animal cells.
Collapse
Affiliation(s)
- Bernhard Kräutler
- Institute of Organic Chemistry and Center for Molecular Biosciences (CMBI)University of Innsbruck6020InnsbruckAustria
| |
Collapse
|
45
|
Ruan J, Han B, Zhuang J, Chen M, Chen F, Huang Y, Zhou W. Hereditary intrinsic factor deficiency in China caused by a novel mutation in the intrinsic factor gene-a case report. BMC MEDICAL GENETICS 2020; 21:221. [PMID: 33172407 PMCID: PMC7654184 DOI: 10.1186/s12881-020-01158-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 10/27/2020] [Indexed: 11/10/2022]
Abstract
Background Hereditary intrinsic factor deficiency is a rare disease characterized by cobalamin deficiency with the lack of gastric intrinsic factor because of gastric intrinsic factor (GIF) mutations. Patients usually present with cobalamin deficiency without gastroscopy abnormality and intrinsic factor antibodies. Case presentation A Chinese patient presented with recurrent severe anemia since age 2 with low cobalamin level and a mild elevation of indirect bilirubin. The hemoglobin level normalized each time after intramuscular vitamin B12 injection. Gene test verified a c.776delA frame shift mutation in exon 6 combined with c.585C > A nonsense early termination mutation in exon 5 of GIF which result in the dysfunction of gastric intrinsic factor protein. The hereditary intrinsic factor deficiency in literature was further reviewed and the ancestry of different mutation sites were discussed. Conclusions A novel compound heterozygous mutation of GIF in a Chinese patient of hereditary intrinsic factor deficiency was reported. It was the first identified mutation of GIF in East-Asia and may indicate a new ancestry.
Collapse
Affiliation(s)
- Jing Ruan
- Department of Hematology, Peking Union Medical College, Hospital, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Bing Han
- Department of Hematology, Peking Union Medical College, Hospital, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China.
| | - Junling Zhuang
- Department of Hematology, Peking Union Medical College, Hospital, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Miao Chen
- Department of Hematology, Peking Union Medical College, Hospital, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Fangfei Chen
- Department of Hematology, Peking Union Medical College, Hospital, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Yuzhou Huang
- Department of Hematology, Peking Union Medical College, Hospital, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Wenzhe Zhou
- Department of Hematology, Peking Union Medical College, Hospital, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| |
Collapse
|
46
|
Rzepka Z, Maszczyk M, Wrześniok D. Biological function of cobalamin: causes and effects of
hypocobalaminemia at the molecular, cellular, tissue
and organism level. POSTEP HIG MED DOSW 2020. [DOI: 10.5604/01.3001.0014.4741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Cobalamin (vitamin B12) is a complex compound, which is classified as a water-soluble
vitamin. Absorption of cobalamin in the gut and its transport to cells is a unique process,
in which many proteins are involved. The loss of function of these proteins causes serious
cell homeostasis disturbance, which may result in the dysfunction of many tissues and
organs. Vitamin B12, a cofactor of methionine synthase, provides methylation process and
nucleic acid synthesis. Cobalamin is also necessary for methylmalonyl-CoA mutase activity.
The enzyme synthesizes succinyl-CoA, an intermediate in tricarboxylic acid cycle.
Vitamin B12 deficiency is an important and current health problem. It may be caused by
insufficient dietary intake, age, or disease-related malabsorption and genetic defects of
mechanisms involved in the absorption, transport and metabolism of cobalamin. Hypocobalaminemia can also result from long-term pharmacotherapy with medicines:
metformin, proton pump inhibitors (e.g. omeprazole) and H2-receptor antagonists
(e.g. ranitidine).
Significant clinical symptoms of cobalamin deficiency include hematological abnormalities,
mainly megaloblastic anemia, as well as neurological disorders resulting from degeneration
within the nervous system. Early diagnosis and starting treatment with vitamin B12 increase
chances for a complete cure. Therefore, the diagnostically important symptom of hypocobalaminemia
may be skin manifestations, mainly hyperpigmentations, but also premature
graying of hair.
The aim of this review article was to summarize the current state of knowledge on the
biological function of cobalamin, as well as the causes and consequences of its deficiency
at the molecular, cellular, tissue and organism level.
Collapse
Affiliation(s)
- Zuzanna Rzepka
- Katedra i Zakład Chemii i Analizy Leków, Wydział Nauk Farmaceutycznych w Sosnowcu, Śląski Uniwersytet Medyczny w Katowicach
| | - Mateusz Maszczyk
- Katedra i Zakład Chemii i Analizy Leków, Wydział Nauk Farmaceutycznych w Sosnowcu, Śląski Uniwersytet Medyczny w Katowicach
| | - Dorota Wrześniok
- Katedra i Zakład Chemii i Analizy Leków, Wydział Nauk Farmaceutycznych w Sosnowcu, Śląski Uniwersytet Medyczny w Katowicach
| |
Collapse
|
47
|
Yekedüz MK, Ince EU, İleri T, Ertem M, Eminoğlu FT. Delayed Diagnosis of Cobalamin E Defect in an Adolescent Patient. J Pediatr Neurosci 2020; 15:140-144. [PMID: 33042249 PMCID: PMC7519755 DOI: 10.4103/jpn.jpn_132_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 02/03/2020] [Accepted: 01/16/2020] [Indexed: 12/26/2022] Open
Abstract
Cobalamin and its metabolites play a critical role in deoxyribonucleic acid synthesis. Disorders of cobalamin metabolism are rare and related with neurological and hematological problems. We report an adolescent patient with cobalamin E (CblE) defect presenting with megaloblastic anemia, mental retardation, cerebral atrophy, cortical visual impairment, white matter changes on brain magnetic resonance imaging, and hyperhomocysteinemia. Homozygous mutation at the c.245C>T in exon 3 of the MTRR gene was identified, which had been found to be related to CblE defect. He was treated with betaine, folic acid, vitamin B6, riboflavin, hydroxycobalamin (OH-B12), and carnitine. During treatment, homocysteine levels decreased over time.
Collapse
Affiliation(s)
- Merve Koç Yekedüz
- Department of Pediatric Metabolism, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Elif Unal Ince
- Department of Pediatric Hematology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Talia İleri
- Department of Pediatric Hematology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Mehmet Ertem
- Department of Pediatric Hematology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Fatma Tuba Eminoğlu
- Department of Pediatric Metabolism, Faculty of Medicine, Ankara University, Ankara, Turkey
| |
Collapse
|
48
|
Zhan S, Cheng F, He H, Hu S, Feng X. Identification of transcobalamin deficiency with two novel mutations in the TCN2 gene in a Chinese girl with abnormal immunity: a case report. BMC Pediatr 2020; 20:460. [PMID: 33023511 PMCID: PMC7537950 DOI: 10.1186/s12887-020-02357-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 09/22/2020] [Indexed: 12/22/2022] Open
Abstract
Background Transcobalamin (TC) transports vitamin B12 from blood into cells. TC II deficiency is a rare autosomal recessive disorder. It is characterized by failure to thrive, diarrhoea, pallor, anaemia, pancytopenia or agammaglobulinemia. It is usually confirmed by molecular analysis of the TCN2 gene. We report a 2-month-old girl with two novel mutations, which were first reported in humans. Case presentation We present a 2-month-old Chinese girl with pancytopenia, severe combined immunodeficiency disease, and megaloblastic anaemia. Targeted next-generation sequencing (NGS) was performed, which detected compound heterozygous variants in exon 7 of the TCN2 gene (Mutation 1: c.1033 C > T; Mutation 2: c.1017-1031delinsGTAACAGAGATGGTT). These mutations result in stop codons in TCN2. The c.1033C > T mutation causes a stop at codon 345 (p.Gln345Ter), and the c.1017-1031delinsGTAACAGAGATGGTT mutation causes a stop at codon 340 (p.Leu340Ter). After being diagnosed, she was treated with intramuscular 1 mg hydroxycobalamin (OH-Cbl) every day for 2 months. The CBC value returned to normal after half a month. The peripheral blood lymphocyte subsets and immunoglobulin recovered after 2 months. Then, the dosage of OH-Cbl was gradually reduced. Conclusions TC II deficiency is a serious complication that requires lifelong treatment. Its diagnosis is difficult due to the lack of clearly identifiable symptoms. Genetic testing should be performed as early as possible if this disease is suspected. The specific observations of this case report make a considerable contribution to the literature and provide a reference for the diagnosis and treatment of future cases.
Collapse
Affiliation(s)
- Shihong Zhan
- The Neonatal Department, Children's Hospital of Soochow University, No. 92 Zhongnan Road, 215000, Suzhou, Jiangsu Province, China
| | - Fangfang Cheng
- Infectious Diseases Department, Children's Hospital of Soochow University, No. 92 Zhongnan Road, 215000, Suzhou, Jiangsu Province, China
| | - Hailong He
- Hematology-Oncology Department, Children's Hospital of Soochow University, No. 92 Zhongnan Road, 215000, Suzhou, Jiangsu Province, China
| | - Shaoyan Hu
- Hematology-Oncology Department, Children's Hospital of Soochow University, No. 92 Zhongnan Road, 215000, Suzhou, Jiangsu Province, China.
| | - Xing Feng
- The Neonatal Department, Children's Hospital of Soochow University, No. 92 Zhongnan Road, 215000, Suzhou, Jiangsu Province, China.
| |
Collapse
|
49
|
Kožich V, Stabler S. Lessons Learned from Inherited Metabolic Disorders of Sulfur-Containing Amino Acids Metabolism. J Nutr 2020; 150:2506S-2517S. [PMID: 33000152 DOI: 10.1093/jn/nxaa134] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 03/12/2020] [Accepted: 04/17/2020] [Indexed: 12/16/2022] Open
Abstract
The metabolism of sulfur-containing amino acids (SAAs) requires an orchestrated interplay among several dozen enzymes and transporters, and an adequate dietary intake of methionine (Met), cysteine (Cys), and B vitamins. Known human genetic disorders are due to defects in Met demethylation, homocysteine (Hcy) remethylation, or cobalamin and folate metabolism, in Hcy transsulfuration, and Cys and hydrogen sulfide (H2S) catabolism. These disorders may manifest between the newborn period and late adulthood by a combination of neuropsychiatric abnormalities, thromboembolism, megaloblastic anemia, hepatopathy, myopathy, and bone and connective tissue abnormalities. Biochemical features include metabolite deficiencies (e.g. Met, S-adenosylmethionine (AdoMet), intermediates in 1-carbon metabolism, Cys, or glutathione) and/or their accumulation (e.g. S-adenosylhomocysteine, Hcy, H2S, or sulfite). Treatment should be started as early as possible and may include a low-protein/low-Met diet with Cys-enriched amino acid supplements, pharmacological doses of B vitamins, betaine to stimulate Hcy remethylation, the provision of N-acetylcysteine or AdoMet, or experimental approaches such as liver transplantation or enzyme replacement therapy. In several disorders, patients are exposed to long-term markedly elevated Met concentrations. Although these conditions may inform on Met toxicity, interpretation is difficult due to the presence of additional metabolic changes. Two disorders seem to exhibit Met-associated toxicity in the brain. An increased risk of demyelination in patients with Met adenosyltransferase I/III (MATI/III) deficiency due to biallelic mutations in the MATIA gene has been attributed to very high blood Met concentrations (typically >800 μmol/L) and possibly also to decreased liver AdoMet synthesis. An excessively high Met concentration in some patients with cystathionine β-synthase deficiency has been associated with encephalopathy and brain edema, and direct toxicity of Met has been postulated. In summary, studies in patients with various disorders of SAA metabolism showed complex metabolic changes with distant cellular consequences, most of which are not attributable to direct Met toxicity.
Collapse
Affiliation(s)
- Viktor Kožich
- Department of Pediatrics and Adolescent Medicine, Charles University-First Faculty of Medicine and General University Hospital, Prague, Czech Republic
| | - Sally Stabler
- Department of Medicine, University of Colorado School of Medicine Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
50
|
Li Z, Mascarenhas R, Twahir UT, Kallon A, Deb A, Yaw M, Penner-Hahn J, Koutmos M, Warncke K, Banerjee R. An Interprotein Co-S Coordination Complex in the B 12-Trafficking Pathway. J Am Chem Soc 2020; 142:16334-16345. [PMID: 32871076 DOI: 10.1021/jacs.0c06590] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The CblC and CblD chaperones are involved in early steps in the cobalamin trafficking pathway. Cobalamin derivatives entering the cytoplasm are converted by CblC to a common cob(II)alamin intermediate via glutathione-dependent alkyltransferase or reductive elimination activities. Cob(II)alamin is subsequently converted to one of two biologically active alkylcobalamins by downstream chaperones. The function of CblD has been elusive although it is known to form a complex with CblC under certain conditions. Here, we report that CblD provides a sulfur ligand to cob(II)alamin bound to CblC, forming an interprotein coordination complex that rapidly oxidizes to thiolato-cob(III)alamin. Cysteine scanning mutagenesis and EPR spectroscopy identified Cys-261 on CblD as the sulfur donor. The unusual interprotein Co-S bond was characterized by X-ray absorption spectroscopy and visualized in the crystal structure of the human CblD thiolato-cob(III)alamin complex. Our study provides insights into how cobalamin coordination chemistry could be utilized for cofactor translocation in the trafficking pathway.
Collapse
Affiliation(s)
- Zhu Li
- Department of Biological Chemistry, University of Michigan Medical Center, Ann Arbor, Michigan 48109-0600, United States
| | - Romila Mascarenhas
- Department of Biological Chemistry, University of Michigan Medical Center, Ann Arbor, Michigan 48109-0600, United States
| | - Umar T Twahir
- Department of Physics, Emory University, Atlanta, Georgia 30322-2430, United States
| | - Albert Kallon
- Department of Biological Chemistry, University of Michigan Medical Center, Ann Arbor, Michigan 48109-0600, United States
| | - Aniruddha Deb
- Departments of Chemistry and Biophysics, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Madeline Yaw
- Department of Biological Chemistry, University of Michigan Medical Center, Ann Arbor, Michigan 48109-0600, United States
| | - James Penner-Hahn
- Departments of Chemistry and Biophysics, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Markos Koutmos
- Departments of Chemistry and Biophysics, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Kurt Warncke
- Department of Physics, Emory University, Atlanta, Georgia 30322-2430, United States
| | - Ruma Banerjee
- Department of Biological Chemistry, University of Michigan Medical Center, Ann Arbor, Michigan 48109-0600, United States
| |
Collapse
|