1
|
Song XQ, Yu TJ, Ou-Yang Y, Ding JH, Jiang YZ, Shao ZM, Xiao Y. Copy number amplification of FLAD1 promotes the progression of triple-negative breast cancer through lipid metabolism. Nat Commun 2025; 16:1241. [PMID: 39890808 PMCID: PMC11785949 DOI: 10.1038/s41467-025-56458-w] [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: 04/24/2024] [Accepted: 01/20/2025] [Indexed: 02/03/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is known for frequent copy number alterations (CNAs) and metabolic reprogramming. However, the mechanism by which CNAs of metabolic genes drive distinct metabolic reprogramming and affect disease progression remains unclear. Through an integrated analysis of our TNBC multiomic dataset (n = 465) and subsequent experimental validation, we identify copy number amplification of the metabolic gene flavin-adenine dinucleotide synthetase 1 (FLAD1) as a crucial genetic event that drives TNBC progression. Mechanistically, FLAD1, but not its enzymatically inactive mutant, upregulates the enzymatic activity of FAD-dependent lysine-specific demethylase 1 (LSD1). LSD1 subsequently promotes the expression of sterol regulatory element-binding protein 1 (SREBP1) by demethylating dimethyl histone H3 lysine 9 (H3K9me2). The upregulation of SREBP1 enhances the expression of lipid biosynthesis genes, ultimately facilitating the progression of TNBC. Clinically, pharmacological inhibition of the FLAD1/LSD1/SREBP1 axis effectively suppresses FLAD1-induced tumor progression. Moreover, LSD1 inhibitor enhances the therapeutic effect of doxorubicin and sacituzumab govitecan (SG). In conclusion, our findings reveal the CNA-derived oncogenic signalling axis of FLAD1/LSD1/SREBP1 and present a promising treatment strategy for TNBC.
Collapse
Affiliation(s)
- Xiao-Qing Song
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P. R. China
| | - Tian-Jian Yu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P. R. China.
| | - Yang Ou-Yang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P. R. China
| | - Jia-Han Ding
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P. R. China
- Shanghai Key Laboratory of Medical Epigenetics, International Colaboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P. R. China
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P. R. China.
| | - Yi Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P. R. China.
| |
Collapse
|
2
|
Vaz FM, Ferdinandusse S, Salomons GS, Wanders RJA. Disorders of fatty acid homeostasis. J Inherit Metab Dis 2025; 48:e12734. [PMID: 38693715 PMCID: PMC11730842 DOI: 10.1002/jimd.12734] [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: 11/15/2023] [Revised: 03/12/2024] [Accepted: 03/18/2024] [Indexed: 05/03/2024]
Abstract
Humans derive fatty acids (FA) from exogenous dietary sources and/or endogenous synthesis from acetyl-CoA, although some FA are solely derived from exogenous sources ("essential FA"). Once inside cells, FA may undergo a wide variety of different modifications, which include their activation to their corresponding CoA ester, the introduction of double bonds, the 2- and ω-hydroxylation and chain elongation, thereby generating a cellular FA pool which can be used for the synthesis of more complex lipids. The biological properties of complex lipids are very much determined by their molecular composition in terms of the FA incorporated into these lipid species. This immediately explains the existence of a range of genetic diseases in man, often with severe clinical consequences caused by variants in one of the many genes coding for enzymes responsible for these FA modifications. It is the purpose of this review to describe the current state of knowledge about FA homeostasis and the genetic diseases involved. This includes the disorders of FA activation, desaturation, 2- and ω-hydroxylation, and chain elongation, but also the disorders of FA breakdown, including disorders of peroxisomal and mitochondrial α- and β-oxidation.
Collapse
Affiliation(s)
- Frédéric M. Vaz
- Department of Clinical Chemistry and Pediatrics, Laboratory Genetic Metabolic DiseasesEmma Children's Hospital, Amsterdam UMC location University of AmsterdamAmsterdamThe Netherlands
- Inborn Errors of Metabolism, Amsterdam Gastroenterology Endocrinology MetabolismAmsterdamThe Netherlands
- Core Facility MetabolomicsAmsterdam UMC location University of AmsterdamAmsterdamThe Netherlands
| | - Sacha Ferdinandusse
- Department of Clinical Chemistry and Pediatrics, Laboratory Genetic Metabolic DiseasesEmma Children's Hospital, Amsterdam UMC location University of AmsterdamAmsterdamThe Netherlands
- Inborn Errors of Metabolism, Amsterdam Gastroenterology Endocrinology MetabolismAmsterdamThe Netherlands
| | - Gajja S. Salomons
- Department of Clinical Chemistry and Pediatrics, Laboratory Genetic Metabolic DiseasesEmma Children's Hospital, Amsterdam UMC location University of AmsterdamAmsterdamThe Netherlands
- Inborn Errors of Metabolism, Amsterdam Gastroenterology Endocrinology MetabolismAmsterdamThe Netherlands
- Core Facility MetabolomicsAmsterdam UMC location University of AmsterdamAmsterdamThe Netherlands
| | - Ronald J. A. Wanders
- Department of Clinical Chemistry and Pediatrics, Laboratory Genetic Metabolic DiseasesEmma Children's Hospital, Amsterdam UMC location University of AmsterdamAmsterdamThe Netherlands
- Inborn Errors of Metabolism, Amsterdam Gastroenterology Endocrinology MetabolismAmsterdamThe Netherlands
| |
Collapse
|
3
|
Nisco A, Tolomeo M, Scalise M, Zanier K, Barile M. Exploring the impact of flavin homeostasis on cancer cell metabolism. Biochim Biophys Acta Rev Cancer 2024; 1879:189149. [PMID: 38971209 DOI: 10.1016/j.bbcan.2024.189149] [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: 04/24/2024] [Revised: 06/25/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
Flavins and their associated proteins have recently emerged as compelling players in the landscape of cancer biology. Flavins, encompassing flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD), serve as coenzymes in a multitude of cellular processes, such as metabolism, apoptosis, and cell proliferation. Their involvement in oxidative phosphorylation, redox homeostasis, and enzymatic reactions has long been recognized. However, recent research has unveiled an extended role for flavins in the context of cancer. In parallel, riboflavin transporters (RFVTs), FAD synthase (FADS), and riboflavin kinase (RFK) have gained prominence in cancer research. These proteins, responsible for riboflavin uptake, FAD biosynthesis, and FMN generation, are integral components of the cellular machinery that governs flavin homeostasis. Dysregulation in the expression/function of these proteins has been associated with various cancers, underscoring their potential as diagnostic markers, therapeutic targets, and key determinants of cancer cell behavior. This review embarks on a comprehensive exploration of the multifaceted role of flavins and of the flavoproteins involved in nucleus-mitochondria crosstalk in cancer. We journey through the influence of flavins on cancer cell energetics, the modulation of RFVTs in malignant transformation, the diagnostic and prognostic significance of FADS, and the implications of RFK in drug resistance and apoptosis. This review also underscores the potential of these molecules and processes as targets for novel diagnostic and therapeutic strategies, offering new avenues for the battle against this relentless disease.
Collapse
Affiliation(s)
- Alessia Nisco
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Italy
| | - Maria Tolomeo
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Italy; Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende, Italy
| | - Mariafrancesca Scalise
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende, Italy
| | - Katia Zanier
- Biotechnology and Cell Signaling (CNRS/Université de Strasbourg, UMR 7242), Ecole Superieure de Biotechnologie de Strasbourg, Illkirch, France
| | - Maria Barile
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Italy.
| |
Collapse
|
4
|
Li XX, Yang XN, Pan HD, Liu L. Fatal multiple acyl-CoA dehydrogenase deficiency caused by ETFDH gene mutation: A case report. World J Clin Cases 2024; 12:5422-5430. [PMID: 39156098 PMCID: PMC11238677 DOI: 10.12998/wjcc.v12.i23.5422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/28/2024] [Accepted: 06/17/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND Multiple acyl-CoA dehydrogenase deficiency (MADD) is a disease of rare autosomal recessive disorder. There are three types of MADD. Type I is a neonatal-onset form with congenital anomalies. Type II is a neonatal-onset form without congenital anomalies. Type III is considered to a milder form and usually responds to riboflavin. However, late-onset form could also be fatal and not responsive to treatments. CASE SUMMARY We report a severe case of a young man with onset type III MADD induced by drugs and strenuous exercise characterized by rhabdomyolysis and liver dysfunction. Urine analysis indicated 12 out of 70 kinds of organic acids like glutaric acid-2 were detected. Serum analysis in genetic metabolic diseases revealed 24 out of 43 tested items were abnormal, revealing the elevation of several acylcarnitines and the reduction of carnitine in the patient. By next generation sequencing technology for gene sequencing related to fatty acid oxidation and carnitine cycle defects, a rare ETFDH gene variant was identified: NM_004453:4:C.1448C>T(p.Pro483 Leu). The patient was diagnosed with late-onset GAII. He was not responsive to riboflavin and progressively worsened into multiple organ failure that finally led to death. CONCLUSION Type III MADD can also be fatal and not responsive to treatments.
Collapse
Affiliation(s)
- Xue-Xia Li
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Xiao-Nan Yang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Hu-Dan Pan
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong Province, China
| | - Liang Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong Province, China
| |
Collapse
|
5
|
Demetriou K, Nisbet J, Coman D, Ewing AD, Phillips L, Smith S, Lipke M, Inwood A, Spicer J, Atthow C, Wilgen U, Robertson T, McWhinney A, Swenson R, Espley B, Snowdon B, McGill JJ, Summers KM. Molecular genetic analysis of candidate genes for glutaric aciduria type II in a cohort of patients from Queensland, Australia. Mol Genet Metab 2024; 142:108516. [PMID: 38941880 DOI: 10.1016/j.ymgme.2024.108516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 06/11/2024] [Accepted: 06/14/2024] [Indexed: 06/30/2024]
Abstract
Glutaric aciduria type II (GAII) is a heterogeneous genetic disorder affecting mitochondrial fatty acid, amino acid and choline oxidation. Clinical manifestations vary across the lifespan and onset may occur at any time from the early neonatal period to advanced adulthood. Historically, some patients, in particular those with late onset disease, have experienced significant benefit from riboflavin supplementation. GAII has been considered an autosomal recessive condition caused by pathogenic variants in the gene encoding electron-transfer flavoprotein ubiquinone-oxidoreductase (ETFDH) or in the genes encoding electron-transfer flavoprotein subunits A and B (ETFA and ETFB respectively). Variants in genes involved in riboflavin metabolism have also been reported. However, in some patients, molecular analysis has failed to reveal diagnostic molecular results. In this study, we report the outcome of molecular analysis in 28 Australian patients across the lifespan, 10 paediatric and 18 adult, who had a diagnosis of glutaric aciduria type II based on both clinical and biochemical parameters. Whole genome sequencing was performed on 26 of the patients and two neonatal onset patients had targeted sequencing of candidate genes. The two patients who had targeted sequencing had biallelic pathogenic variants (in ETFA and ETFDH). None of the 26 patients whose whole genome was sequenced had biallelic variants in any of the primary candidate genes. Interestingly, nine of these patients (34.6%) had a monoallelic pathogenic or likely pathogenic variant in a single primary candidate gene and one patient (3.9%) had a monoallelic pathogenic or likely pathogenic variant in two separate genes within the same pathway. The frequencies of the damaging variants within ETFDH and FAD transporter gene SLC25A32 were significantly higher than expected when compared to the corresponding allele frequencies in the general population. The remaining 16 patients (61.5%) had no pathogenic or likely pathogenic variants in the candidate genes. Ten (56%) of the 18 adult patients were taking the selective serotonin reuptake inhibitor antidepressant sertraline, which has been shown to produce a GAII phenotype, and another two adults (11%) were taking a serotonin-norepinephrine reuptake inhibitor antidepressant, venlafaxine or duloxetine, which have a mechanism of action overlapping that of sertraline. Riboflavin deficiency can also mimic both the clinical and biochemical phenotype of GAII. Several patients on these antidepressants showed an initial response to riboflavin but then that response waned. These results suggest that the GAII phenotype can result from a complex interaction between monoallelic variants and the cellular environment. Whole genome or targeted gene panel analysis may not provide a clear molecular diagnosis.
Collapse
Affiliation(s)
- Kalliope Demetriou
- Queensland Lifespan Metabolic Medicine Service, Queensland Children's Hospital, South Brisbane, QLD 4101, Australia
| | - Janelle Nisbet
- Queensland Lifespan Metabolic Medicine Service, Mater Hospital Brisbane, South Brisbane, QLD 4101, Australia
| | - David Coman
- Queensland Lifespan Metabolic Medicine Service, Mater Hospital Brisbane, South Brisbane, QLD 4101, Australia; Wesley Medical Centre, Auchenflower, QLD 4066, Australia; University of Queensland, St Lucia, QLD 4072, Australia
| | - Adam D Ewing
- Mater Research Institute-University of Queensland, Translational Research Institute, 37 Kent St, Woolloongabba, QLD 4102, Australia
| | - Liza Phillips
- Queensland Lifespan Metabolic Medicine Service, Mater Hospital Brisbane, South Brisbane, QLD 4101, Australia
| | - Sally Smith
- Queensland Lifespan Metabolic Medicine Service, Queensland Children's Hospital, South Brisbane, QLD 4101, Australia; Queensland Lifespan Metabolic Medicine Service, Mater Hospital Brisbane, South Brisbane, QLD 4101, Australia
| | - Michelle Lipke
- Queensland Lifespan Metabolic Medicine Service, Queensland Children's Hospital, South Brisbane, QLD 4101, Australia; Queensland Lifespan Metabolic Medicine Service, Mater Hospital Brisbane, South Brisbane, QLD 4101, Australia
| | - Anita Inwood
- Queensland Lifespan Metabolic Medicine Service, Queensland Children's Hospital, South Brisbane, QLD 4101, Australia; Queensland Lifespan Metabolic Medicine Service, Mater Hospital Brisbane, South Brisbane, QLD 4101, Australia; University of Queensland, St Lucia, QLD 4072, Australia
| | - Janette Spicer
- Queensland Lifespan Metabolic Medicine Service, Queensland Children's Hospital, South Brisbane, QLD 4101, Australia
| | - Catherine Atthow
- Queensland Lifespan Metabolic Medicine Service, Queensland Children's Hospital, South Brisbane, QLD 4101, Australia
| | - Urs Wilgen
- University of Queensland, St Lucia, QLD 4072, Australia; Chemical Pathology, Pathology Queensland, Royal Brisbane and Women's Hospital, Herston, QLD 4029, Australia
| | - Thomas Robertson
- University of Queensland, St Lucia, QLD 4072, Australia; Anatomical Pathology, Pathology Queensland, Royal Brisbane and Women's Hospital, Herston, QLD 4029, Australia
| | - Avis McWhinney
- Chemical Pathology, Mater Pathology, Mater Hospital, Mater Hospital Brisbane, QLD 4101, Australia
| | - Rebecca Swenson
- Chemical Pathology, Pathology Queensland, Royal Brisbane and Women's Hospital, Herston, QLD 4029, Australia
| | - Brayden Espley
- Chemical Pathology, Pathology Queensland, Royal Brisbane and Women's Hospital, Herston, QLD 4029, Australia
| | - Brianna Snowdon
- Chemical Pathology, Pathology Queensland, Royal Brisbane and Women's Hospital, Herston, QLD 4029, Australia
| | - James J McGill
- Queensland Lifespan Metabolic Medicine Service, Queensland Children's Hospital, South Brisbane, QLD 4101, Australia; Queensland Lifespan Metabolic Medicine Service, Mater Hospital Brisbane, South Brisbane, QLD 4101, Australia; Chemical Pathology, Pathology Queensland, Royal Brisbane and Women's Hospital, Herston, QLD 4029, Australia; Chemical Pathology, Mater Pathology, Mater Hospital, Mater Hospital Brisbane, QLD 4101, Australia
| | - Kim M Summers
- Mater Research Institute-University of Queensland, Translational Research Institute, 37 Kent St, Woolloongabba, QLD 4102, Australia.
| |
Collapse
|
6
|
Aragão MÂ, Pires L, Santos-Buelga C, Barros L, Calhelha RC. Revitalising Riboflavin: Unveiling Its Timeless Significance in Human Physiology and Health. Foods 2024; 13:2255. [PMID: 39063339 PMCID: PMC11276209 DOI: 10.3390/foods13142255] [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: 06/19/2024] [Revised: 07/14/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Since the early twentieth century, research on vitamins has revealed their therapeutic potential beyond their role as essential micronutrients. Riboflavin, known as vitamin B2, stands out for its unique characteristics. Despite numerous studies, riboflavin remains vital, with implications for human health. Abundantly present in various foods, riboflavin acts as a coenzyme in numerous enzymatic reactions crucial for human metabolism. Its role in energy production, erythrocyte synthesis, and vitamin metabolism underscores its importance in maintaining homeostasis. The impact of riboflavin extends to neurological function, skin health, and cardiovascular well-being, with adequate levels linked to reduced risks of various ailments. However, inadequate intake or physiological stress can lead to deficiency, a condition that poses serious health risks, including severe complications. This underscores the importance of maintaining sufficient levels of riboflavin for general wellness. The essential role of riboflavin in immune function further emphasises its significance for human health and vitality. This paper examines the diverse effects of riboflavin on health and stresses the importance of maintaining sufficient levels for overall well-being.
Collapse
Affiliation(s)
- M. Ângela Aragão
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal; (M.Â.A.); (L.P.); (L.B.)
- Laboratório Associado para Sustentabilidade e Tecnologia em Regiões de Montanha (SusTEC), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal
- Grupo de Investigación en Polifenoles (GIP-USAL), Facultad de Farmacia, Campus Miguel de Unamuno, Universidad de Salamanca, s/n, 37007 Salamanca, Spain;
| | - Lara Pires
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal; (M.Â.A.); (L.P.); (L.B.)
- Laboratório Associado para Sustentabilidade e Tecnologia em Regiões de Montanha (SusTEC), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal
- Grupo de Investigación en Polifenoles (GIP-USAL), Facultad de Farmacia, Campus Miguel de Unamuno, Universidad de Salamanca, s/n, 37007 Salamanca, Spain;
| | - Celestino Santos-Buelga
- Grupo de Investigación en Polifenoles (GIP-USAL), Facultad de Farmacia, Campus Miguel de Unamuno, Universidad de Salamanca, s/n, 37007 Salamanca, Spain;
| | - Lillian Barros
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal; (M.Â.A.); (L.P.); (L.B.)
- Laboratório Associado para Sustentabilidade e Tecnologia em Regiões de Montanha (SusTEC), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal
| | - Ricardo C. Calhelha
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal; (M.Â.A.); (L.P.); (L.B.)
- Laboratório Associado para Sustentabilidade e Tecnologia em Regiões de Montanha (SusTEC), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal
| |
Collapse
|
7
|
Leo G, Leone P, Ataie Kachoie E, Tolomeo M, Galluccio M, Indiveri C, Barile M, Capaldi S. Structural insights into the bifunctional enzyme human FAD synthase. Structure 2024; 32:953-965.e5. [PMID: 38688286 DOI: 10.1016/j.str.2024.04.006] [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: 02/08/2024] [Revised: 03/20/2024] [Accepted: 04/03/2024] [Indexed: 05/02/2024]
Abstract
Human flavin adenine dinucleotide synthase (hFADS) is a bifunctional, multi-domain enzyme that exhibits both flavin mononucleotide adenylyltransferase and pyrophosphatase activities. Here we report the crystal structure of full-length hFADS2 and its C-terminal PAPS domain in complex with flavin adenine dinucleotide (FAD), and dissect the structural determinants underlying the contribution of each individual domain, within isoforms 1 and 2, to each of the two enzymatic activities. Structural and functional characterization performed on complete or truncated constructs confirmed that the C-terminal domain tightly binds FAD and catalyzes its synthesis, while the combination of the N-terminal molybdopterin-binding and KH domains is the minimal essential substructure required for the hydrolysis of FAD and other ADP-containing dinucleotides. hFADS2 associates in a stable C2-symmetric dimer, in which the packing of the KH domain of one protomer against the N-terminal domain of the other creates the adenosine-specific active site responsible for the hydrolytic activity.
Collapse
Affiliation(s)
- Giulia Leo
- Department of Biotechnology, University of Verona, Strada Le Grazie 15, 37134 Verona, Italy
| | - Piero Leone
- Department of Biosciences, Biotechnology and Environment, University of Bari, via Orabona 4, 70126 Bari, Italy
| | - Elham Ataie Kachoie
- Department of Biotechnology, University of Verona, Strada Le Grazie 15, 37134 Verona, Italy
| | - Maria Tolomeo
- Department of Biosciences, Biotechnology and Environment, University of Bari, via Orabona 4, 70126 Bari, Italy; Department of Biology, Ecology and Earth Sciences (DiBEST), Laboratory of Biochemistry, Molecular Biotechnology, and Molecular Biology, University of Calabria, via P. Bucci 4c, 6c, 87036 Arcavacata di Rende, Italy
| | - Michele Galluccio
- Department of Biology, Ecology and Earth Sciences (DiBEST), Laboratory of Biochemistry, Molecular Biotechnology, and Molecular Biology, University of Calabria, via P. Bucci 4c, 6c, 87036 Arcavacata di Rende, Italy
| | - Cesare Indiveri
- Department of Biology, Ecology and Earth Sciences (DiBEST), Laboratory of Biochemistry, Molecular Biotechnology, and Molecular Biology, University of Calabria, via P. Bucci 4c, 6c, 87036 Arcavacata di Rende, Italy; National Research Council (CNR), Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), via Amendola 122/O, 70126 Bari, Italy
| | - Maria Barile
- Department of Biosciences, Biotechnology and Environment, University of Bari, via Orabona 4, 70126 Bari, Italy.
| | - Stefano Capaldi
- Department of Biotechnology, University of Verona, Strada Le Grazie 15, 37134 Verona, Italy.
| |
Collapse
|
8
|
Ji Y, Zhao J, Gong J, Sedlazeck FJ, Fan S. Unveiling novel genetic variants in 370 challenging medically relevant genes using the long read sequencing data of 41 samples from 19 global populations. Mol Genet Genomics 2024; 299:65. [PMID: 38972030 PMCID: PMC11955097 DOI: 10.1007/s00438-024-02158-x] [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: 12/06/2023] [Accepted: 06/16/2024] [Indexed: 07/08/2024]
Abstract
BACKGROUND A large number of challenging medically relevant genes (CMRGs) are situated in complex or highly repetitive regions of the human genome, hindering comprehensive characterization of genetic variants using next-generation sequencing technologies. In this study, we employed long-read sequencing technology, extensively utilized in studying complex genomic regions, to characterize genetic alterations, including short variants (single nucleotide variants and short insertions and deletions) and copy number variations, in 370 CMRGs across 41 individuals from 19 global populations. RESULTS Our analysis revealed high levels of genetic variants in CMRGs, with 68.73% exhibiting copy number variations and 65.20% containing short variants that may disrupt protein function across individuals. Such variants can influence pharmacogenomics, genetic disease susceptibility, and other clinical outcomes. We observed significant differences in CMRG variation across populations, with individuals of African ancestry harboring the highest number of copy number variants and short variants compared to samples from other continents. Notably, 15.79% to 33.96% of short variants were exclusively detectable through long-read sequencing. While the T2T-CHM13 reference genome significantly improved the assembly of CMRG regions, thereby facilitating variant detection in these regions, some regions still lacked resolution. CONCLUSION Our results provide an important reference for future clinical and pharmacogenetic studies, highlighting the need for a comprehensive representation of global genetic diversity in the reference genome and improved variant calling techniques to fully resolve medically relevant genes.
Collapse
Affiliation(s)
- Yanfeng Ji
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, School of Life Science, Fudan University, Shanghai, 200438, China
| | - Junfan Zhao
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, School of Life Science, Fudan University, Shanghai, 200438, China
| | - Jiao Gong
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, School of Life Science, Fudan University, Shanghai, 200438, China
| | - Fritz J Sedlazeck
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA.
- Department of Computer Science, Rice University, 6100 Main Street, Houston, TX, 77005, USA.
| | - Shaohua Fan
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, School of Life Science, Fudan University, Shanghai, 200438, China.
| |
Collapse
|
9
|
Peng Y, Zhu M, Gong Y, Wang C. Identification and functional prediction of lncRNAs associated with intramuscular lipid deposition in Guangling donkeys. Front Vet Sci 2024; 11:1410109. [PMID: 39036793 PMCID: PMC11258529 DOI: 10.3389/fvets.2024.1410109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 05/30/2024] [Indexed: 07/23/2024] Open
Abstract
Many studies have shown that long non-coding RNAs (lncRNAs) play key regulatory roles in various biological processes. However, the importance and molecular regulatory mechanisms of lncRNAs in donkey intramuscular fat deposition remain to be further investigated. In this study, we used published transcriptomic data from the longissimus dorsi muscle of Guangling donkeys to identify lncRNAs and obtained 196 novel lncRNAs. Compared with the coding genes, the novel lncRNAs and the known lncRNAs exhibited some typical features, such as shorter transcript length and smaller exons. A total of 272 coding genes and 52 lncRNAs were differentially expressed between the longissimus dorsi muscles of the low-fat and high-fat groups. The differentially expressed genes were found to be involved in various biological processes related to lipid metabolism. The potential target genes of differentially expressed lncRNAs were predicted by cis and trans. Functional analysis of lncRNA targets showed that some lncRNAs may act on potential target genes involved in lipid metabolism processes and regulate lipid deposition in the longissimus dorsi muscle. This study provides valuable information for further investigation of the molecular mechanisms of lipid deposition traits in donkeys, which may improve meat traits and facilitate the selection process of donkeys in future breeding.
Collapse
Affiliation(s)
- Yongdong Peng
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Agricultural Science and Engineering School, Liaocheng University, Liaocheng, China
| | | | | | - Changfa Wang
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Agricultural Science and Engineering School, Liaocheng University, Liaocheng, China
| |
Collapse
|
10
|
Wen B, Tang R, Tang S, Sun Y, Xu J, Zhao D, Wang T, Yan C. A comparative study on riboflavin responsive multiple acyl-CoA dehydrogenation deficiency due to variants in FLAD1 and ETFDH gene. J Hum Genet 2024; 69:125-131. [PMID: 38228875 DOI: 10.1038/s10038-023-01216-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 12/02/2023] [Accepted: 12/13/2023] [Indexed: 01/18/2024]
Abstract
Lipid storage myopathy (LSM) is a heterogeneous group of lipid metabolism disorders predominantly affecting skeletal muscle by triglyceride accumulation in muscle fibers. Riboflavin therapy has been shown to ameliorate symptoms in some LSM patients who are essentially concerned with multiple acyl-CoA dehydrogenation deficiency (MADD). It is proved that riboflavin responsive LSM caused by MADD is mainly due to ETFDH gene variant (ETFDH-RRMADD). We described here a case with riboflavin responsive LSM and MADD resulting from FLAD1 gene variants (c.1588 C > T p.Arg530Cys and c.1589 G > C p.Arg530Pro, FLAD1-RRMADD). And we compared our patient together with 9 FLAD1-RRMADD cases from literature to 106 ETFDH-RRMADD cases in our neuromuscular center on clinical history, laboratory investigations and pathological features. Furthermore, the transcriptomics study on FLAD1-RRMADD and ETFDH-RRMADD were carried out. On muscle pathology, both FLAD1-RRMADD and ETFDH-RRMADD were proved with lipid storage myopathy in which atypical ragged red fibers were more frequent in ETFDH-RRMADD, while fibers with faint COX staining were more common in FLAD1-RRMADD. Molecular study revealed that the expression of GDF15 gene in muscle and GDF15 protein in both serum and muscle was significantly increased in FLAD1-RRMADD and ETFDH-RRMADD groups. Our data revealed that FLAD1-RRMADD (p.Arg530) has similar clinical, biochemical, and fatty acid metabolism changes to ETFDH-RRMADD except for muscle pathological features.
Collapse
Affiliation(s)
- Bing Wen
- Department of Neurology and Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China
| | - Runqi Tang
- Department of Pathology, Maternal and Child Health Hospital of Liaocheng, Liaocheng, 252000, Shandong, China
| | - Shuyao Tang
- Department of Neurology and Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China
| | - Yuan Sun
- Department of Neurology, Qilu Hospital (Qingdao), Shandong University, Qingdao, 266035, Shandong, China
| | - Jingwen Xu
- Department of Neurology and Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China
| | - Dandan Zhao
- Department of Neurology and Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China
| | - Tan Wang
- Department of Geriatric Medicine, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China.
| | - Chuanzhu Yan
- Department of Neurology and Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China.
- Brain Science Research Institute, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
11
|
Murgia C, Dehlia A, Guthridge MA. New insights into the nutritional genomics of adult-onset riboflavin-responsive diseases. Nutr Metab (Lond) 2023; 20:42. [PMID: 37845732 PMCID: PMC10580530 DOI: 10.1186/s12986-023-00764-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 10/04/2023] [Indexed: 10/18/2023] Open
Abstract
Riboflavin, or vitamin B2, is an essential nutrient that serves as a precursor to flavin adenine dinucleotide (FAD) and flavin mononucleotide (FMN). The binding of the FAD and/or FMN cofactors to flavoproteins is critical for regulating their assembly and activity. There are over 90 proteins in the human flavoproteome that regulate a diverse array of biochemical pathways including mitochondrial metabolism, riboflavin transport, ubiquinone and FAD synthesis, antioxidant signalling, one-carbon metabolism, nitric oxide signalling and peroxisome oxidative metabolism. The identification of patients with genetic variants in flavoprotein genes that lead to adult-onset pathologies remains a major diagnostic challenge. However, once identified, many patients with adult-onset inborn errors of metabolism demonstrate remarkable responses to riboflavin therapy. We review the structure:function relationships of mutant flavoproteins and propose new mechanistic insights into adult-onset riboflavin-responsive pathologies and metabolic dysregulations that apply to multiple biochemical pathways. We further address the vexing issue of how the inheritance of genetic variants in flavoprotein genes leads to an adult-onset disease with complex symptomologies and varying severities. We also propose a broad clinical framework that may not only improve the current diagnostic rates, but also facilitate a personalized approach to riboflavin therapy that is low cost, safe and lead to transformative outcomes in many patients.
Collapse
Affiliation(s)
- Chiara Murgia
- The School of Agriculture, Food and Ecosystem Sciences (SAFES), Faculty of Science, The University of Melbourne, Parkville, Australia.
| | - Ankush Dehlia
- School of Life and Environmental Sciences, Deakin University, Burwood, Australia
| | - Mark A Guthridge
- School of Life and Environmental Sciences, Deakin University, Burwood, Australia
| |
Collapse
|
12
|
Nisco A, Carvalho TMA, Tolomeo M, Di Molfetta D, Leone P, Galluccio M, Medina M, Indiveri C, Reshkin SJ, Cardone RA, Barile M. Increased demand for FAD synthesis in differentiated and stem pancreatic cancer cells is accomplished by modulating FLAD1 gene expression: the inhibitory effect of Chicago Sky Blue. FEBS J 2023; 290:4679-4694. [PMID: 37254652 DOI: 10.1111/febs.16881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/31/2023] [Accepted: 05/30/2023] [Indexed: 06/01/2023]
Abstract
FLAD1, along with its FAD synthase (FADS, EC 2.7.7.2) product, is crucial for flavin homeostasis and, due to its role in the mitochondrial respiratory chain and nuclear epigenetics, is closely related to cellular metabolism. Therefore, it is not surprising that it could be correlated with cancer. To our knowledge, no previous study has investigated FLAD1 prognostic significance in pancreatic ductal adenocarcinoma (PDAC). Thus, in the present work, the FAD synthesis process was evaluated in two PDAC cell lines: (a) PANC-1- and PANC-1-derived cancer stem cells (CSCs), presenting the R273H mutation in the oncosuppressor p53, and (b) MiaPaca2 and MiaPaca2-derived CSCs, presenting the R248W mutation in p53. As a control, HPDE cells expressing wt-p53 were used. FADS expression/activity increase was found with malignancy and even more with stemness. An increased FAD synthesis rate in cancer cell lines is presumably demanded by the increase in the FAD-dependent lysine demethylase 1 protein amount as well as by the increased expression levels of the flavoprotein subunit of complex II of the mitochondrial respiratory chain, namely succinate dehydrogenase. With the aim of proposing FADS as a novel target for cancer therapy, the inhibitory effect of Chicago Sky Blue on FADS enzymatic activity was tested on the recombinant 6His-hFADS2 (IC50 = 1.2 μm) and PANC-1-derived CSCs' lysate (IC50 = 2-10 μm). This molecule was found effective in inhibiting the growth of PANC-1 and even more of its derived CSC line, thus assessing its role as a potential chemotherapeutic drug.
Collapse
Affiliation(s)
- Alessia Nisco
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Italy
| | - Tiago M A Carvalho
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Italy
| | - Maria Tolomeo
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Italy
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende, Italy
| | - Daria Di Molfetta
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Italy
| | - Piero Leone
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Italy
| | - Michele Galluccio
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende, Italy
| | - Milagros Medina
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Instituto de Biocomputación y Física de Sistemas Complejos (BIFI) (GBsC-CSIC Joint Unit), University of Zaragoza, Spain
| | - Cesare Indiveri
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende, Italy
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), Bari, Italy
| | - Stephan Joel Reshkin
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Italy
| | - Rosa Angela Cardone
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Italy
| | - Maria Barile
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Italy
| |
Collapse
|
13
|
Vianey-Saban C, Guffon N, Fouilhoux A, Acquaviva C. Fifty years of research on mitochondrial fatty acid oxidation disorders: The remaining challenges. J Inherit Metab Dis 2023; 46:848-873. [PMID: 37530674 DOI: 10.1002/jimd.12664] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 07/19/2023] [Accepted: 07/26/2023] [Indexed: 08/03/2023]
Abstract
Since the identification of the first disorder of mitochondrial fatty acid oxidation defects (FAOD) in 1973, more than 20 defects have been identified. Although there are some differences, most FAOD have similar clinical signs, which are mainly due to energy depletion and toxicity of accumulated metabolites. However, some of them have an unusual clinical phenotype or specific clinical signs. This manuscript focuses on what we have learnt so far on the pathophysiology of these disorders, which present with clinical signs that are not typical of categorical FAOD. It also highlights that some disorders have not yet been identified and tries to make assumptions to explain why. It also deals with new treatments under consideration in FAOD, including triheptanoin and similar anaplerotic substrates, ketone body treatments, RNA and gene therapy approaches. Finally, it suggests challenges for the diagnosis of FAOD in the coming years, both for symptomatic patients and for those diagnosed through newborn screening. The ultimate goal would be to identify all the patients born with FAOD and ensure for them the best possible quality of life.
Collapse
Affiliation(s)
- Christine Vianey-Saban
- Biochemical and Molecular Biology Laboratory, Metabolic Inborn Errors of Metabolism Unit, Groupement Hospitalier Est, CHU de Lyon, Bron, France
| | - Nathalie Guffon
- National Reference Centre for Hereditary Metabolic Diseases, Groupement Hospitalier Est, CHU de Lyon, Bron, France
| | - Alain Fouilhoux
- National Reference Centre for Hereditary Metabolic Diseases, Groupement Hospitalier Est, CHU de Lyon, Bron, France
| | - Cécile Acquaviva
- Biochemical and Molecular Biology Laboratory, Metabolic Inborn Errors of Metabolism Unit, Groupement Hospitalier Est, CHU de Lyon, Bron, France
| |
Collapse
|
14
|
van de Leemkolk FEM, Lo Faro ML, Shaheed S, Mulvey JF, Huurman VAL, Alwayn IPJ, Putter H, Jochmans I, Lindeman JHN, Ploeg RJ. The role of flavin mononucleotide (FMN) as a potentially clinically relevant biomarker to predict the quality of kidney grafts during hypothermic (oxygenated) machine perfusion. PLoS One 2023; 18:e0287713. [PMID: 37352336 PMCID: PMC10289320 DOI: 10.1371/journal.pone.0287713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 05/21/2023] [Indexed: 06/25/2023] Open
Abstract
Hypothermic machine perfusion (HMP) provides preservation superior to cold storage and may allow for organ assessment prior to transplantation. Since flavin mononucleotide (FMN) in perfusate has been proposed as a biomarker of organ quality during HMP of donor livers, the aim of this study was to validate FMN as a biomarker for organ quality in the context of HMP preserved kidneys. Perfusate samples (n = 422) from the paired randomised controlled COPE-COMPARE-trial, comparing HMP with oxygenation (HMPO2) versus standard HMP in kidneys, were used. Fluorescence intensity (FI) was assessed using fluorescence spectroscopy (excitation 450nm; emission 500-600nm) and validated by fluorospectrophotometer and targeted liquid chromatography mass spectrometry (LC-MS/MS). Fluorescence intensity (FI)(ex450;em500-600) increased over time during machine perfusion in both groups (p<0.0001). This increase was similar for both groups (p = 0.83). No correlation, however, was found between FI(ex450;em500-600) and post-transplant outcomes, including day 5 or 7 serum creatinine (p = 0.11; p = 0.16), immediate graft function (p = 0.91), creatinine clearance and biopsy-proven rejection at one year (p = 0.14; p = 0.59). LC-MS/MS validation experiments of samples detected FMN in only one perfusate sample, whilst the majority of samples with the highest fluorescence (n = 37/38, 97.4%) remained negative. In the context of clinical kidney HMP, fluorescence spectroscopy unfortunately appears to be not specific and probably unsuitable for FMN. This study shows that FMN does not classify as a clinically relevant predictive biomarker of kidney graft function after transplantation.
Collapse
Affiliation(s)
- Fenna E. M. van de Leemkolk
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
- LUMC Transplant Center, Leiden University Medical Center, Leiden, The Netherlands
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - M. Letizia Lo Faro
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, United Kingdom
| | - Sadr Shaheed
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, United Kingdom
| | - John F. Mulvey
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Volkert A. L. Huurman
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
- LUMC Transplant Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Ian P. J. Alwayn
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
- LUMC Transplant Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Hein Putter
- LUMC Transplant Center, Leiden University Medical Center, Leiden, The Netherlands
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Ina Jochmans
- Department of Abdominal Transplant Surgery, University Hospitals Leuven, Leuven, Belgium
- Transplantation Research Group, Lab of Abdominal Transplantation, Department of Microbiology, Immunology, and Transplantation, KU Leuven, Belgium
| | - Jan H. N. Lindeman
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
- LUMC Transplant Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Rutger J. Ploeg
- LUMC Transplant Center, Leiden University Medical Center, Leiden, The Netherlands
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, United Kingdom
| | | |
Collapse
|
15
|
Prasun P, Evans A, Cork E, Houten SM, Webb BD. A novel deleterious ETFA promoter variant causative of multiple acyl-CoA dehydrogenase deficiency. Am J Med Genet A 2023; 191:1089-1093. [PMID: 36579410 DOI: 10.1002/ajmg.a.63104] [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: 10/21/2022] [Revised: 12/08/2022] [Accepted: 12/10/2022] [Indexed: 12/30/2022]
Abstract
Multiple acyl-CoA dehydrogenase deficiency (MADD) is an autosomal recessive disorder of fatty acid, amino acid, and choline metabolism. We describe a patient identified through newborn screening in which the diagnosis of MADD was confirmed based on metabolic profiling, but clinical molecular sequencing of ETFA, ETFB, and ETFDH was normal. In order to identify the genetic etiology of MADD, we performed whole genome sequencing and identified a novel homozygous promoter variant in ETFA (c.-85G > A). Subsequent studies showed decreased ETFA protein expression in lymphoblasts. A promoter luciferase assay confirmed decreased activity of the mutant promoter. In both assays, the variant displayed considerable residual activity, therefore we speculate that our patient may have a late onset form of MADD (Type III). Our findings may be helpful in establishing a molecular diagnosis in other MADD patients with a characteristic biochemical profile but apparently normal molecular studies.
Collapse
Affiliation(s)
- Pankaj Prasun
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Anthony Evans
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Emalyn Cork
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Sander M Houten
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Bryn D Webb
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Division of Genetics and Metabolism, Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
16
|
Baker MJ, Crameri JJ, Thorburn DR, Frazier AE, Stojanovski D. Mitochondrial biology and dysfunction in secondary mitochondrial disease. Open Biol 2022; 12:220274. [PMID: 36475414 PMCID: PMC9727669 DOI: 10.1098/rsob.220274] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial diseases are a broad, genetically heterogeneous class of metabolic disorders characterized by deficits in oxidative phosphorylation (OXPHOS). Primary mitochondrial disease (PMD) defines pathologies resulting from mutation of mitochondrial DNA (mtDNA) or nuclear genes affecting either mtDNA expression or the biogenesis and function of the respiratory chain. Secondary mitochondrial disease (SMD) arises due to mutation of nuclear-encoded genes independent of, or indirectly influencing OXPHOS assembly and operation. Despite instances of novel SMD increasing year-on-year, PMD is much more widely discussed in the literature. Indeed, since the implementation of next generation sequencing (NGS) techniques in 2010, many novel mitochondrial disease genes have been identified, approximately half of which are linked to SMD. This review will consolidate existing knowledge of SMDs and outline discrete categories within which to better understand the diversity of SMD phenotypes. By providing context to the biochemical and molecular pathways perturbed in SMD, we hope to further demonstrate the intricacies of SMD pathologies outside of their indirect contribution to mitochondrial energy generation.
Collapse
Affiliation(s)
- Megan J. Baker
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Jordan J. Crameri
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3052, Australia
| | - David R. Thorburn
- Murdoch Children's Research Institute, Royal Children's Hospital and Department of Paediatrics, University of Melbourne, Parkville, Victoria 3052, Australia,Victorian Clinical Genetics Services, Royal Children's Hospital, Parkville, Victoria 3052, Australia
| | - Ann E. Frazier
- Murdoch Children's Research Institute, Royal Children's Hospital and Department of Paediatrics, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Diana Stojanovski
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3052, Australia
| |
Collapse
|
17
|
Tummolo A, Leone P, Tolomeo M, Solito R, Mattiuzzo M, Lepri FR, Lorè T, Cardinali R, De Giovanni D, Simonetti S, Barile M. Combined isobutyryl-CoA and multiple acyl-CoA dehydrogenase deficiency in a boy with altered riboflavin homeostasis. JIMD Rep 2022; 63:276-291. [PMID: 35822092 PMCID: PMC9259400 DOI: 10.1002/jmd2.12292] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 04/07/2022] [Accepted: 04/12/2022] [Indexed: 11/24/2022] Open
Abstract
In this report, we describe the case of an 11-year-old boy, who came to our attention for myalgia and muscle weakness, associated with inappetence and vomiting. Hypertransaminasemia was also noted, with ultrasound evidence of hepatomegaly. Biochemical investigations revealed acylcarnitine and organic acid profiles resembling those seen in MADD, that is, multiple acyl-CoA dehydrogenase deficiencies (OMIM #231680) a rare inherited disorder of fatty acids, amino acids, and choline metabolism. The patient carried a single pathogenetic variant in the ETFDH gene (c.524G>A, p.Arg175His) and no pathogenetic variant in the riboflavin (Rf) homeostasis related genes (SLC52A1, SLC52A2, SLC52A3, SLC25A32, FLAD1). Instead, compound heterozygosity was found in the ACAD8 gene (c.512C>G, p.Ser171Cys; c.822C>A, p.Asn274Lys), coding for isobutyryl-CoA dehydrogenase (IBD), whose pathogenic variants are associated to IBD deficiency (OMIM #611283), a rare autosomal recessive disorder of valine catabolism. The c.822C>A was never previously described in a patient. Subsequent further analyses of Rf homeostasis showed reduced levels of flavins in plasma and altered FAD-dependent enzymatic activities in erythrocytes, as well as a significant reduction in the level of the plasma membrane Rf transporter 2 in erythrocytes. The observed Rf/flavin scarcity in this patient, possibly associated with a decreased ETF:QO efficiency might be responsible for the observed MADD-like phenotype. The patient's clinical picture improved after supplementation of Rf, l-carnitine, Coenzyme Q10, and also 3OH-butyrate. This report demonstrates that, even in the absence of genetic defects in genes involved in Rf homeostasis, further targeted molecular analysis may reveal secondary and possibly treatable biochemical alterations in this pattern.
Collapse
Affiliation(s)
- Albina Tummolo
- Metabolic Diseases and Clinical Genetics UnitChildren's Hospital “Giovanni XXIII”BariItaly
| | - Piero Leone
- Department of Biosciences, Biotechnology and BiopharmaceuticsUniversity of Bari “A. Moro”BariItaly
| | - Maria Tolomeo
- Department of Biosciences, Biotechnology and BiopharmaceuticsUniversity of Bari “A. Moro”BariItaly
| | - Rita Solito
- Department of Biosciences, Biotechnology and BiopharmaceuticsUniversity of Bari “A. Moro”BariItaly
| | - Matteo Mattiuzzo
- Laboratory of Medical GeneticsTranslational Cytogenomics Research Unit, Bambino Gesù Children HospitalRomeItaly
| | - Francesca Romana Lepri
- Laboratory of Medical GeneticsTranslational Cytogenomics Research Unit, Bambino Gesù Children HospitalRomeItaly
| | - Tania Lorè
- Regional Centre for Neonatal ScreeningChildren's Hospital “Giovanni XXIII”BariItaly
| | - Roberta Cardinali
- Regional Centre for Neonatal ScreeningChildren's Hospital “Giovanni XXIII”BariItaly
| | - Donatella De Giovanni
- Metabolic Diseases and Clinical Genetics UnitChildren's Hospital “Giovanni XXIII”BariItaly
| | - Simonetta Simonetti
- Regional Centre for Neonatal ScreeningChildren's Hospital “Giovanni XXIII”BariItaly
| | - Maria Barile
- Department of Biosciences, Biotechnology and BiopharmaceuticsUniversity of Bari “A. Moro”BariItaly
| |
Collapse
|
18
|
Tolomeo M, Chimienti G, Lanza M, Barbaro R, Nisco A, Latronico T, Leone P, Petrosillo G, Liuzzi GM, Ryder B, Inbar-Feigenberg M, Colella M, Lezza AMS, Olsen RKJ, Barile M. Retrograde response to mitochondrial dysfunctions associated to LOF variations in FLAD1 exon 2: unraveling the importance of RFVT2. Free Radic Res 2022; 56:511-525. [PMID: 36480241 DOI: 10.1080/10715762.2022.2146501] [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: 12/13/2022]
Abstract
Flavin adenine dinucleotide (FAD) synthase (EC 2.7.7.2), encoded by human flavin adenine dinucleotide synthetase 1 (FLAD1), catalyzes the last step of the pathway converting riboflavin (Rf) into FAD. FLAD1 variations were identified as a cause of LSMFLAD (lipid storage myopathy due to FAD synthase deficiency, OMIM #255100), resembling Multiple Acyl-CoA Dehydrogenase Deficiency, sometimes treatable with high doses of Rf; no alternative therapeutic strategies are available. We describe here cell morphological and mitochondrial alterations in dermal fibroblasts derived from a LSMFLAD patient carrying a homozygous truncating FLAD1 variant (c.745C > T) in exon 2. Despite a severe decrease in FAD synthesis rate, the patient had decreased cellular levels of Rf and flavin mononucleotide and responded to Rf treatment. We hypothesized that disturbed flavin homeostasis and Rf-responsiveness could be due to a secondary impairment in the expression of the Rf transporter 2 (RFVT2), encoded by SLC52A2, in the frame of an adaptive retrograde signaling to mitochondrial dysfunction. Interestingly, an antioxidant response element (ARE) is found in the region upstream of the transcriptional start site of SLC52A2. Accordingly, we found that abnormal mitochondrial morphology and impairments in bioenergetics were accompanied by increased cellular reactive oxygen species content and mtDNA oxidative damage. Concomitantly, an active response to mitochondrial stress is suggested by increased levels of PPARγ-co-activator-1α and Peroxiredoxin III. In this scenario, the treatment with high doses of Rf might compensate for the secondary RFVT2 molecular defect, providing a molecular rationale for the Rf responsiveness in patients with loss of function variants in FLAD1 exon 2.HIGHLIGHTSFAD synthase deficiency alters mitochondrial morphology and bioenergetics;FAD synthase deficiency triggers a mitochondrial retrograde response;FAD synthase deficiency evokes nuclear signals that adapt the expression of RFVT2.
Collapse
Affiliation(s)
- Maria Tolomeo
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Bari, Italy.,Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende, Italy
| | - Guglielmina Chimienti
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Martina Lanza
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Roberto Barbaro
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Alessia Nisco
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Tiziana Latronico
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Piero Leone
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Giuseppe Petrosillo
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council (CNR), Bari, Italy
| | - Grazia Maria Liuzzi
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Bryony Ryder
- National Metabolic Service, Starship Children's Hospital, Auckland, New Zealand
| | - Michal Inbar-Feigenberg
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Matilde Colella
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Angela M S Lezza
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Rikke K J Olsen
- Research Unit for Molecular Medicine, Department for Clinical Medicine, Aarhus University and Aarhus University Hospital, Aarhus, Denmark
| | - Maria Barile
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
19
|
Peng MZ, Shao YX, Li XZ, Zhang KD, Cai YN, Lin YT, Jiang MY, Liu ZC, Su XY, Zhang W, Jiang XL, Liu L. Mitochondrial FAD shortage in SLC25A32 deficiency affects folate-mediated one-carbon metabolism. Cell Mol Life Sci 2022; 79:375. [PMID: 35727412 PMCID: PMC11072207 DOI: 10.1007/s00018-022-04404-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/06/2022] [Accepted: 05/27/2022] [Indexed: 11/03/2022]
Abstract
The SLC25A32 dysfunction is associated with neural tube defects (NTDs) and exercise intolerance, but very little is known about disease-specific mechanisms due to a paucity of animal models. Here, we generated homozygous (Slc25a32Y174C/Y174C and Slc25a32K235R/K235R) and compound heterozygous (Slc25a32Y174C/K235R) knock-in mice by mimicking the missense mutations identified from our patient. A homozygous knock-out (Slc25a32-/-) mouse was also generated. The Slc25a32K235R/K235R and Slc25a32Y174C/K235R mice presented with mild motor impairment and recapitulated the biochemical disturbances of the patient. While Slc25a32-/- mice die in utero with NTDs. None of the Slc25a32 mutations hindered the mitochondrial uptake of folate. Instead, the mitochondrial uptake of flavin adenine dinucleotide (FAD) was specifically blocked by Slc25a32Y174C/K235R, Slc25a32K235R/K235R, and Slc25a32-/- mutations. A positive correlation between SLC25A32 dysfunction and flavoenzyme deficiency was observed. Besides the flavoenzymes involved in fatty acid β-oxidation and amino acid metabolism being impaired, Slc25a32-/- embryos also had a subunit of glycine cleavage system-dihydrolipoamide dehydrogenase damaged, resulting in glycine accumulation and glycine derived-formate reduction, which further disturbed folate-mediated one-carbon metabolism, leading to 5-methyltetrahydrofolate shortage and other folate intermediates accumulation. Maternal formate supplementation increased the 5-methyltetrahydrofolate levels and ameliorated the NTDs in Slc25a32-/- embryos. The Slc25a32K235R/K235R and Slc25a32Y174C/K235R mice had no glycine accumulation, but had another formate donor-dimethylglycine accumulated and formate deficiency. Meanwhile, they suffered from the absence of all folate intermediates in mitochondria. Formate supplementation increased the folate amounts, but this effect was not restricted to the Slc25a32 mutant mice only. In summary, we established novel animal models, which enabled us to understand the function of SLC25A32 better and to elucidate the role of SLC25A32 dysfunction in human disease development and progression.
Collapse
Affiliation(s)
- Min-Zhi Peng
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, the Affiliated Hospital of Guangzhou Medical University, 9 Jinsui Road, Guangzhou, China
| | - Yong-Xian Shao
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, the Affiliated Hospital of Guangzhou Medical University, 9 Jinsui Road, Guangzhou, China
| | - Xiu-Zhen Li
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, the Affiliated Hospital of Guangzhou Medical University, 9 Jinsui Road, Guangzhou, China
| | - Kang-Di Zhang
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, the Affiliated Hospital of Guangzhou Medical University, 9 Jinsui Road, Guangzhou, China
| | - Yan-Na Cai
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, the Affiliated Hospital of Guangzhou Medical University, 9 Jinsui Road, Guangzhou, China
| | - Yun-Ting Lin
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, the Affiliated Hospital of Guangzhou Medical University, 9 Jinsui Road, Guangzhou, China
| | - Min-Yan Jiang
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, the Affiliated Hospital of Guangzhou Medical University, 9 Jinsui Road, Guangzhou, China
| | - Zong-Cai Liu
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, the Affiliated Hospital of Guangzhou Medical University, 9 Jinsui Road, Guangzhou, China
| | - Xue-Ying Su
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, the Affiliated Hospital of Guangzhou Medical University, 9 Jinsui Road, Guangzhou, China
| | - Wen Zhang
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, the Affiliated Hospital of Guangzhou Medical University, 9 Jinsui Road, Guangzhou, China.
| | - Xiao-Ling Jiang
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, the Affiliated Hospital of Guangzhou Medical University, 9 Jinsui Road, Guangzhou, China.
| | - Li Liu
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, the Affiliated Hospital of Guangzhou Medical University, 9 Jinsui Road, Guangzhou, China.
| |
Collapse
|
20
|
Pan X, Yuan Y, Wu B, Zheng W, Tian M. Lipid-storage myopathy with glycogen storage disease gene mutations mimicking polymyositis: a case report and review of the literature. J Int Med Res 2022; 50:3000605221084873. [PMID: 35296144 PMCID: PMC8943314 DOI: 10.1177/03000605221084873] [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] [Indexed: 11/17/2022] Open
Abstract
A 26-year-old Asian woman with persistent muscle weakness was diagnosed with polymyositis based on biopsy findings at another hospital 11 years ago. However, her symptoms fluctuated repeatedly under treatment with prednisone and immunosuppressive agents, and worsened 2 months prior to the current presentation. A second muscle biopsy suggested metabolic myopathy, and genetic testing revealed a novel c.1074C > T variant in the glycogen synthase 1 gene (GYS1), which is implicated in muscle glycogen storage disease type 0. However, no abnormalities in glycogen deposition were found by biopsy; rather, muscle fibers exhibited large intracellular lipid droplets. Furthermore, muscle strength was greatly restored and circulating levels of creatine kinase indicative of muscle degeneration greatly reduced by vitamin B2 treatment. Therefore, the final diagnosis was lipid storage myopathy.
Collapse
Affiliation(s)
- Xiaoli Pan
- Department of Rheumatology and Immunology, 159358Affiliated Hospital of Zunyi Medical University, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P. R. China
| | - Yuan Yuan
- School of Foreign Languages of Zunyi Medical University, Zunyi, Guizhou 563003, P. R. China
| | - Bangcui Wu
- Department of Rheumatology and Immunology, 159358Affiliated Hospital of Zunyi Medical University, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P. R. China
| | - Wendan Zheng
- Department of Rheumatology and Immunology, 159358Affiliated Hospital of Zunyi Medical University, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P. R. China
| | - Mei Tian
- Department of Rheumatology and Immunology, 159358Affiliated Hospital of Zunyi Medical University, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P. R. China
| |
Collapse
|
21
|
Lupica A, Oteri R, Volta S, Ghezzi D, Drago SFA, Rodolico C, Musumeci O, Toscano A. Diagnostic Challenges in Late Onset Multiple Acyl-CoA Dehydrogenase Deficiency: Clinical, Morphological, and Genetic Aspects. Front Neurol 2022; 13:815523. [PMID: 35309592 PMCID: PMC8929684 DOI: 10.3389/fneur.2022.815523] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/18/2022] [Indexed: 12/04/2022] Open
Abstract
Background Multiple acyl-CoA dehydrogenase deficiency (MADD) is an autosomal recessive disorder of fatty acid oxidation due to deficiency of the mitochondrial electron transfer chain. The late-onset form is characterized by exercise intolerance, muscle weakness, and lipid storage in myofibers. Most MADD patients greatly benefit from riboflavin supplementation. Patients and methods A retrospective study was conducted on patients with a diagnosis of vacuolar myopathy with lipid storage followed in our neuromuscular unit in the last 20 years. We selected 10 unrelated patients with the diagnosis of MADD according to clinical, morphological, and biochemical aspects. Clinical features, blood tests including serum acylcarnitines, EMG, and ENG were revised. Muscle biopsy was performed in all, and one individual underwent also a sural nerve biopsy. Gene sequencing of ETFA, ETFB, and ETFDH was performed as a first-tier genetic analysis followed by next-generation sequencing of an hyperCKemia gene panel in patients with undefined genotypes. Results Clinical evaluation at onset in all our patients showed fatigue and muscle weakness; four patients showed difficulties in chewing, three patients complained of dysphagia, two patients had a dropped head, and a patient had an unexpected ataxia with numbness and dysesthesia. Laboratory blood tests revealed a variable increase in serum CK (266–6,500) and LDH levels (500–2,000). Plasma acylcarnitine profile evidenced increased levels of different chains intermediates. EMG was either normal or showed myogenic or neurogenic patterns. NCS demonstrated sensory neuropathy in two patients. Muscle biopsies showed a vacuolar myopathy with a variable increase in lipid content. Nerve biopsy evidenced an axonal degeneration with the loss of myelinated fibers. ETFDH genetic analysis identifies 14 pathogenic variants. Patients were treated with high doses of riboflavin (400 mg/die). All of them showed a rapid muscle strength improvement and normalization of abnormal values in laboratory tests. Neuropathic symptoms did not improve. Conclusion Our data confirmed that clinical features in MADD patients are extremely variable in terms of disease onset and symptoms making diagnosis difficult. Laboratory investigations, such as serum acylcarnitine profile and muscle biopsy evaluation, may strongly address to a correct diagnosis. The favorable response to riboflavin supplementation strengthens the importance of an early diagnosis of these disorders among the spectrum of metabolic myopathies.
Collapse
Affiliation(s)
- Antonino Lupica
- Department of Biomedicine, Neuroscience and Advanced Diagnostic (BIND), University of Palermo, Palermo, Italy
| | - Rosaria Oteri
- Unit of Neurology and Neuromuscular Disorders, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Sara Volta
- Department of Neurosciences, University of Padova, Padova, Italy
| | - Daniele Ghezzi
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) Istituto Neurologico Carlo Besta, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Selene Francesca Anna Drago
- Unit of Neurology and Neuromuscular Disorders, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Carmelo Rodolico
- Unit of Neurology and Neuromuscular Disorders, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Olimpia Musumeci
- Unit of Neurology and Neuromuscular Disorders, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
- *Correspondence: Olimpia Musumeci
| | - Antonio Toscano
- Unit of Neurology and Neuromuscular Disorders, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| |
Collapse
|
22
|
Curtabbi A, Enríquez JA. The ins and outs of the flavin mononucleotide cofactor of respiratory complex I. IUBMB Life 2022; 74:629-644. [PMID: 35166025 DOI: 10.1002/iub.2600] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/23/2022] [Accepted: 01/24/2022] [Indexed: 12/12/2022]
Abstract
The flavin mononucleotide (FMN) cofactor of respiratory complex I occupies a key position in the electron transport chain. Here, the electrons coming from NADH start the sequence of oxidoreduction reactions, which drives the generation of the proton-motive force necessary for ATP synthesis. The overall architecture and the general catalytic proprieties of the FMN site are mostly well established. However, several aspects regarding the complex I flavin cofactor are still unknown. For example, the flavin binding to the N-module, the NADH-oxidizing portion of complex I, lacks a molecular description. The dissociation of FMN from the enzyme is beginning to emerge as an important regulatory mechanism of complex I activity and ROS production. Finally, how mitochondria import and metabolize FMN is still uncertain. This review summarizes the current knowledge on complex I flavin cofactor and discusses the open questions for future research.
Collapse
Affiliation(s)
- Andrea Curtabbi
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - José Antonio Enríquez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain.,Centro de Investigación Biomédica en Red en Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
23
|
AlFakhri N, Sojar S, Bortcosh W. Hyperammonemia and Altered Mental Status in a 17-year-old Girl. Pediatr Rev 2022; 43:112-115. [PMID: 35102399 DOI: 10.1542/pir.2020-003061] [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)
- Nora AlFakhri
- Department of Pediatrics, University of Florida Health Shands Children's Hospital, University of Florida College of Medicine, Gainesville, FL
| | - Sakina Sojar
- Division of Pediatric Emergency Medicine, Alpert Medical School, Brown University, Providence, RI
| | - William Bortcosh
- Department of Pediatrics, University of Florida Health Shands Children's Hospital, University of Florida College of Medicine, Gainesville, FL.,Division of Pediatric Critical Care, University of Florida Health Shands Children's Hospital, University of Florida College of Medicine, Gainesville, FL
| |
Collapse
|
24
|
Nalini A, Vengalil S, Polavarapu K, Preethish-Kumar V, Nashi S, Arunachal G, Chawla T, Bardhan M, Mohan D, Christopher R, Bevinahalli N, Kulanthaivelu K, Nishino I, Faruq M. Mutation spectrum of primary lipid storage myopathies. Ann Indian Acad Neurol 2022; 25:106-113. [PMID: 35342266 PMCID: PMC8954319 DOI: 10.4103/aian.aian_333_21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 10/16/2021] [Accepted: 10/27/2021] [Indexed: 11/04/2022] Open
Abstract
Background: Lipid storage myopathies (LSM) constitute an important group of treatable myopathies. Genetic testing is essential for confirming the diagnosis and also helps in explaining phenotypic heterogeneity. The objective of this study was to describe the clinical features and genetic spectrum of LSM seen in a quaternary referral center in India. Methods: Eleven cases of suspected LSM underwent clinical, biochemical, histopathological and genetic evaluation. Tandem Mass Spectrometry and clinical exome sequencing with Sanger validation were performed. Results: All patients had exertion induced myalgia and either progressive or episodic limb girdle muscle weakness (LGMW). The age of onset ranged 10 to 31 years (mean- 21 ± 6.7y), age at presentation- 14 to 49 years (mean- 26.5 ± 9.5y). Mutations identified: ETFDH = 5, CPT2 = 3, FLAD1 = 1, ACADVL = 1, FLAD1 = 1. Dropped head syndrome was seen in two patients with ETFDH mutations. Bulbar symptoms and Beevor's sign were noted in a patient with FLAD1 variant. Novel variants were identified in seven patients. Conclusions: This is the first report on the genetic spectrum of LSM from India. LSM should be considered in patients with exertion induced myalgias, LGMW, cranial nerve involvement or dropped head syndrome. Genetic testing is essential for identification of these treatable disorders.
Collapse
|
25
|
Tang Z, Gao S, He M, Chen Q, Fang J, Luo Y, Yan W, Shi X, Huang H, Tang J. Clinical Presentations and Genetic Characteristics of Late-Onset MADD Due to ETFDH Mutations in Five Patients: A Case Series. Front Neurol 2021; 12:747360. [PMID: 34819910 PMCID: PMC8606537 DOI: 10.3389/fneur.2021.747360] [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: 07/26/2021] [Accepted: 10/05/2021] [Indexed: 01/24/2023] Open
Abstract
Background: Late-onset multiple acyl-CoA dehydrogenase deficiency (LO-MADD) describes a curable autosomal recessive genetic disease caused by ETFDH mutations that result in defects in ETF-ubiquinone oxidoreductase. Almost all patients are responsive to riboflavin. This study describes the clinical presentations and genetic characteristics of five LO-MADD patients. Methods: From 2018 to 2021, we collected clinical and genetic data on five patients diagnosed with LO-MADD at our hospital and retrospectively analyzed their clinical characteristics, laboratory examination, electromyography, muscle biopsy, genetic analysis, and outcome data. Results: This study included three males and two females with mean onset age of 37.8 years. Fluctuating exercise intolerance was the most common presentation. Serum creatine kinase (CK) levels were significantly elevated in all patients, and plasma acylcarnitine profiles revealed an increase in long-chain acylcarnitine species in three cases. The urinary organic acid study revealed a high level of hydroxyglutaric acid in all patients. Electrophysiology demonstrated myogenic impairment. Muscle biopsies revealed lipid storage myopathy. Molecular analysis identified nine mutations (three novels and six reported) in ETFDH. Exercise intolerance and muscle weakness were dramatically improved in all patients treated with riboflavin (100 mg) daily following diagnosis. Conclusions: LO-MADD is caused by ETFDH variants and responds well to riboflavin. Three novel ETFDH pathogenic variants were identified, expanding their spectrum in the Chinese population and facilitating future interpretation and analysis of ETFDH mutations.
Collapse
Affiliation(s)
- Zhenchu Tang
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, Second Xiangya Hospital, Central South University, Changsha, China
| | - Shan Gao
- Department of Gastroenterology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Miao He
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, Second Xiangya Hospital, Central South University, Changsha, China
| | - Qihua Chen
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Jia Fang
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yingying Luo
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Weiqian Yan
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoliu Shi
- Department of Medical Genetics, Second Xiangya Hospital, Central South University, Changsha, China
| | - Hui Huang
- Department of Medical Genetics, Second Xiangya Hospital, Central South University, Changsha, China
| | - Jianguang Tang
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
26
|
Jin C, Yonezawa A. Recent advances in riboflavin transporter RFVT and its genetic disease. Pharmacol Ther 2021; 233:108023. [PMID: 34662687 DOI: 10.1016/j.pharmthera.2021.108023] [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: 08/30/2021] [Revised: 09/30/2021] [Accepted: 10/12/2021] [Indexed: 12/20/2022]
Abstract
Riboflavin (vitamin B2) is essential for cellular growth and function. It is enzymatically converted to flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD), which participate in the metabolic oxidation-reduction reactions of carbohydrates, amino acids, and lipids. Human riboflavin transporters RFVT1, RFVT2, and RFVT3 have been identified and characterized since 2008. They are highly specific transporters of riboflavin. RFVT3 has functional characteristics different from those of RFVT1 and RFVT2. RFVT3 contributes to absorption in the small intestine, reabsorption in the kidney, and transport to the fetus in the placenta, while RFVT2 mediates the tissue distribution of riboflavin from the blood. Several mutations in the SLC52A2 gene encoding RFVT2 and the SLC52A3 gene encoding RFVT3 were found in patients with a rare neurological disorder known as Brown-Vialetto-Van Laere syndrome. These patients commonly present with bulbar palsy, hearing loss, muscle weakness, and respiratory symptoms in infancy or later in childhood. A decrease in plasma riboflavin levels has been observed in several cases. Recent studies on knockout mice and patient-derived cells have advanced the understanding of these mechanisms. Here, we summarize novel findings on RFVT1-3 and their genetic diseases and discuss their potential as therapeutic drugs.
Collapse
Affiliation(s)
- Congyun Jin
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Graduate School of Pharmaceutical Sciences, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Atsushi Yonezawa
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Graduate School of Pharmaceutical Sciences, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.
| |
Collapse
|
27
|
Gong J, Zhang W, Ding L, Zhang M, Zheng S, Ma R, Tang J, Yi W, Xu H, Zhang Y. 4,4'-Dimethoxychalcone regulates redox homeostasis by targeting riboflavin metabolism in Parkinson's disease therapy. Free Radic Biol Med 2021; 174:40-56. [PMID: 34332078 DOI: 10.1016/j.freeradbiomed.2021.07.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/25/2021] [Accepted: 07/27/2021] [Indexed: 02/08/2023]
Abstract
Oxidative stress damage plays a pivotal role in Parkinson's disease (PD) pathogenesis. Previously, we developed a blood brain barrier-penetrating peptide-based "Trojan Horse" strategy to deliver 4,4'-dimethoxychalcone (DMC) for PD therapy and revealed neuroprotective properties of DMC in a PD model; however, the underlying mechanisms remained unclear. Here, we report that DMC attenuated motor impairment, degeneration of DA neurons and α-synuclein aggregation in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and exogenous human α-synuclein-induced PD mouse models. Mechanistically, DMC increased the expression of two critical intermediates in riboflavin metabolism: riboflavin kinase (RFK) and its metabolic product, flavin mononucleotide (FMN). We provide the first direct evidence that FMN ameliorated oxidative stress damage and dopaminergic neuron degeneration both in vitro and in vivo and that riboflavin metabolism was required for DMC-mediated neuroprotection. DMC-induced restoration of redox homeostasis was mediated via the activation of protein kinase Cθ (PKCθ) signaling. Together, our findings reveal that DMC may serve as a novel antioxidant in PD intervention and also define a novel mechanism that underlies its therapeutic activity.
Collapse
Affiliation(s)
- Junwei Gong
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, China
| | - Wenlong Zhang
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Liuyan Ding
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Mengran Zhang
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, China
| | - Shaohui Zheng
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, China
| | - Runfang Ma
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, China
| | - Junyuan Tang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation & Molecular Target and Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Wei Yi
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation & Molecular Target and Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Huaxi Xu
- Center for Brain Sciences of the First Affiliated Hospital of Xiamen University, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361005, China
| | - Yunlong Zhang
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, China.
| |
Collapse
|
28
|
Leone P, Tolomeo M, Piancone E, Puzzovio PG, De Giorgi C, Indiveri C, Di Schiavi E, Barile M. Mimicking human riboflavin responsive neuromuscular disorders by silencing flad-1 gene in C. elegans: Alteration of vitamin transport and cholinergic transmission. IUBMB Life 2021; 74:672-683. [PMID: 34558787 PMCID: PMC9292511 DOI: 10.1002/iub.2553] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 08/14/2021] [Accepted: 08/30/2021] [Indexed: 01/01/2023]
Abstract
Riboflavin (Rf), or vitamin B2, is the precursor of FMN and FAD, redox cofactors of several dehydrogenases involved in energy metabolism, redox balance and other cell regulatory processes. FAD synthase, coded by FLAD1 gene in humans, is the last enzyme in the pathway converting Rf into FAD. Mutations in FLAD1 gene are responsible for neuromuscular disorders, in some cases treatable with Rf. In order to mimic these disorders, the Caenorhabditis elegans (C. elegans) gene orthologue of FLAD1 (flad‐1) was silenced in a model strain hypersensitive to RNA interference in nervous system. Silencing flad‐1 resulted in a significant decrease in total flavin content, paralleled by a decrease in the level of the FAD‐dependent ETFDH protein and by a secondary transcriptional down‐regulation of the Rf transporter 1 (rft‐1) possibly responsible for the total flavin content decrease. Conversely an increased ETFDH mRNA content was found. These biochemical changes were accompanied by significant phenotypical changes, including impairments of fertility and locomotion due to altered cholinergic transmission, as indicated by the increased sensitivity to aldicarb. A proposal is made that neuronal acetylcholine production/release is affected by alteration of Rf homeostasis. Rf supplementation restored flavin content, increased rft‐1 transcript levels and eliminated locomotion defects. In this aspect, C. elegans could provide a low‐cost animal model to elucidate the molecular rationale for Rf therapy in human Rf responsive neuromuscular disorders and to screen other molecules with therapeutic potential.
Collapse
Affiliation(s)
- Piero Leone
- Department of Biosciences, Biotechnology, and Biopharmaceutics, University of Bari, Bari, Italy
| | - Maria Tolomeo
- Department of Biosciences, Biotechnology, and Biopharmaceutics, University of Bari, Bari, Italy
| | - Elisabetta Piancone
- Department of Biosciences, Biotechnology, and Biopharmaceutics, University of Bari, Bari, Italy
| | - Pier Giorgio Puzzovio
- Faculty of Medicine, Pharmacology and Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Carla De Giorgi
- Department of Biosciences, Biotechnology, and Biopharmaceutics, University of Bari, Bari, Italy
| | - Cesare Indiveri
- Department DiBEST (Biologia, Ecologia, Scienze della Terra), Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Elia Di Schiavi
- Institute of Biosciences and Bioresources (IBBR) CNR, Naples, Italy
| | - Maria Barile
- Department of Biosciences, Biotechnology, and Biopharmaceutics, University of Bari, Bari, Italy
| |
Collapse
|
29
|
Averina OA, Permyakov OA, Grigorieva OO, Starshin AS, Mazur AM, Prokhortchouk EB, Dontsova OA, Sergiev PV. Comparative Analysis of Genome Editors Efficiency on a Model of Mice Zygotes Microinjection. Int J Mol Sci 2021; 22:10221. [PMID: 34638562 PMCID: PMC8508852 DOI: 10.3390/ijms221910221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/21/2021] [Accepted: 09/21/2021] [Indexed: 11/16/2022] Open
Abstract
Genome editing is an indispensable tool for functional genomics. The caveat of the genome-editing pipeline is a prevalence of error-prone non-homologous end joining over homologous recombination, while only the latter is suitable to introduce particularly desired genetic variants. To overcome this problem, a toolbox of genome engineering was appended by a variety of improved instruments. In this work, we compared the efficiency of a number of recently suggested improved systems for genome editing applied to the same genome regions on a murine zygote model via microinjection. As a result, we observed that homologous recombination utilizing an ssDNA template following sgRNA directed Cas9 cleavage is still the method of choice for the creation of animals with precise genome alterations.
Collapse
Affiliation(s)
- Olga A. Averina
- Institute of Functional Genomics, Lomonosov Moscow State University, 119991 Moscow, Russia; (O.A.A.); (O.A.P.); (O.O.G.)
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119899 Moscow, Russia
| | - Oleg A. Permyakov
- Institute of Functional Genomics, Lomonosov Moscow State University, 119991 Moscow, Russia; (O.A.A.); (O.A.P.); (O.O.G.)
| | - Olga O. Grigorieva
- Institute of Functional Genomics, Lomonosov Moscow State University, 119991 Moscow, Russia; (O.A.A.); (O.A.P.); (O.O.G.)
| | - Alexey S. Starshin
- Research Center of Biotechnology of the Russian Academy of Sciences, Institute of Bioengineering, 119071 Moscow, Russia; (A.S.S.); (A.M.M.); (E.B.P.)
| | - Alexander M. Mazur
- Research Center of Biotechnology of the Russian Academy of Sciences, Institute of Bioengineering, 119071 Moscow, Russia; (A.S.S.); (A.M.M.); (E.B.P.)
| | - Egor B. Prokhortchouk
- Research Center of Biotechnology of the Russian Academy of Sciences, Institute of Bioengineering, 119071 Moscow, Russia; (A.S.S.); (A.M.M.); (E.B.P.)
| | - Olga A. Dontsova
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia;
- Center of Life Sciences, Skolkovo Institute of Science and Technology, 121205 Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia
| | - Petr V. Sergiev
- Institute of Functional Genomics, Lomonosov Moscow State University, 119991 Moscow, Russia; (O.A.A.); (O.A.P.); (O.O.G.)
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia;
- Center of Life Sciences, Skolkovo Institute of Science and Technology, 121205 Moscow, Russia
| |
Collapse
|
30
|
Bruni F, Giancaspero TA, Oreb M, Tolomeo M, Leone P, Boles E, Roberti M, Caselle M, Barile M. Subcellular Localization of Fad1p in Saccharomyces cerevisiae: A Choice at Post-Transcriptional Level? Life (Basel) 2021; 11:967. [PMID: 34575116 PMCID: PMC8470081 DOI: 10.3390/life11090967] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/06/2021] [Accepted: 09/13/2021] [Indexed: 11/24/2022] Open
Abstract
FAD synthase is the last enzyme in the pathway that converts riboflavin into FAD. In Saccharomyces cerevisiae, the gene encoding for FAD synthase is FAD1, from which a sole protein product (Fad1p) is expected to be generated. In this work, we showed that a natural Fad1p exists in yeast mitochondria and that, in its recombinant form, the protein is able, per se, to both enter mitochondria and to be destined to cytosol. Thus, we propose that FAD1 generates two echoforms-that is, two identical proteins addressed to different subcellular compartments. To shed light on the mechanism underlying the subcellular destination of Fad1p, the 3' region of FAD1 mRNA was analyzed by 3'RACE experiments, which revealed the existence of (at least) two FAD1 transcripts with different 3'UTRs, the short one being 128 bp and the long one being 759 bp. Bioinformatic analysis on these 3'UTRs allowed us to predict the existence of a cis-acting mitochondrial localization motif, present in both the transcripts and, presumably, involved in protein targeting based on the 3'UTR context. Here, we propose that the long FAD1 transcript might be responsible for the generation of mitochondrial Fad1p echoform.
Collapse
Affiliation(s)
- Francesco Bruni
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Via Orabona 4, 70125 Bari, Italy; (F.B.); (T.A.G.); (M.T.); (P.L.); (M.R.)
| | - Teresa Anna Giancaspero
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Via Orabona 4, 70125 Bari, Italy; (F.B.); (T.A.G.); (M.T.); (P.L.); (M.R.)
| | - Mislav Oreb
- Institute of Molecular Biosciences, Goethe-University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany; (M.O.); (E.B.)
| | - Maria Tolomeo
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Via Orabona 4, 70125 Bari, Italy; (F.B.); (T.A.G.); (M.T.); (P.L.); (M.R.)
| | - Piero Leone
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Via Orabona 4, 70125 Bari, Italy; (F.B.); (T.A.G.); (M.T.); (P.L.); (M.R.)
| | - Eckhard Boles
- Institute of Molecular Biosciences, Goethe-University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany; (M.O.); (E.B.)
| | - Marina Roberti
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Via Orabona 4, 70125 Bari, Italy; (F.B.); (T.A.G.); (M.T.); (P.L.); (M.R.)
| | - Michele Caselle
- Physics Department, University of Turin and INFN, Via P. Giuria 1, 10125 Turin, Italy;
| | - Maria Barile
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Via Orabona 4, 70125 Bari, Italy; (F.B.); (T.A.G.); (M.T.); (P.L.); (M.R.)
| |
Collapse
|
31
|
Hagemeijer MC, Oussoren E, Ruijter GJG, Onkenhout W, Huidekoper HH, Ebberink MS, Waterham HR, Ferdinandusse S, de Vries MC, Huigen MCDG, Kluijtmans LAJ, Coene KLM, Blom HJ. Abnormal VLCADD newborn screening resembling MADD in four neonates with decreased riboflavin levels and VLCAD activity. JIMD Rep 2021; 61:12-18. [PMID: 34485012 PMCID: PMC8411102 DOI: 10.1002/jmd2.12223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/27/2021] [Accepted: 04/13/2021] [Indexed: 11/17/2022] Open
Abstract
Early detection of congenital disorders by newborn screening (NBS) programs is essential to prevent or limit disease manifestation in affected neonates. These programs balance between the detection of the highest number of true cases and the lowest number of false-positives. In this case report, we describe four unrelated cases with a false-positive NBS result for very-long-chain acyl-CoA dehydrogenase deficiency (VLCADD). Three neonates presented with decreased but not deficient VLCAD enzyme activity and two of them carried a single heterozygous ACADVL c.1844G>A mutation. Initial biochemical investigations after positive NBS referral in these infants revealed acylcarnitine and organic acid profiles resembling those seen in multiple acyl-CoA dehydrogenase deficiency (MADD). Genetic analysis did not reveal any pathogenic mutations in the genes encoding the electron transfer flavoprotein (ETF alpha and beta subunits) nor in ETF dehydrogenase. Subsequent further diagnostics revealed decreased levels of riboflavin in the newborns and oral riboflavin administration normalized the MADD-like biochemical profiles. During pregnancy, the mothers followed a vegan, vegetarian or lactose-free diet which probably caused alimentary riboflavin deficiency in the neonates. This report demonstrates that a secondary (alimentary) maternal riboflavin deficiency in combination with reduced VLCAD activity in the newborns can result in an abnormal VLCADD/MADD acylcarnitine profile and can cause false-positive NBS. We hypothesize that maternal riboflavin deficiency contributed to the false-positive VLCADD neonatal screening results.
Collapse
Affiliation(s)
- Marne C. Hagemeijer
- Center for Lysosomal and Metabolic Diseases, Department of Clinical GeneticsErasmus University Medical CenterRotterdamThe Netherlands
| | - Esmee Oussoren
- Center for Lysosomal and Metabolic Diseases, Department of PediatricsErasmus University Medical CenterRotterdamThe Netherlands
| | - George J. G. Ruijter
- Center for Lysosomal and Metabolic Diseases, Department of Clinical GeneticsErasmus University Medical CenterRotterdamThe Netherlands
| | - Willem Onkenhout
- Center for Lysosomal and Metabolic Diseases, Department of Clinical GeneticsErasmus University Medical CenterRotterdamThe Netherlands
| | - Hidde H. Huidekoper
- Center for Lysosomal and Metabolic Diseases, Department of PediatricsErasmus University Medical CenterRotterdamThe Netherlands
| | - Merel S. Ebberink
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam Gastroenterology Endocrinology MetabolismAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
| | - Hans R. Waterham
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam Gastroenterology Endocrinology MetabolismAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
| | - Sacha Ferdinandusse
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam Gastroenterology Endocrinology MetabolismAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
| | - Maaike C. de Vries
- Department of Laboratory Medicine, Translational Metabolic Laboratory (TML)Radboud University Medical CenterNijmegenThe Netherlands
| | - Marleen C. D. G. Huigen
- Department of Laboratory Medicine, Translational Metabolic Laboratory (TML)Radboud University Medical CenterNijmegenThe Netherlands
| | - Leo A. J. Kluijtmans
- Department of Laboratory Medicine, Translational Metabolic Laboratory (TML)Radboud University Medical CenterNijmegenThe Netherlands
| | - Karlien L. M. Coene
- Department of Laboratory Medicine, Translational Metabolic Laboratory (TML)Radboud University Medical CenterNijmegenThe Netherlands
| | - Henk J Blom
- Center for Lysosomal and Metabolic Diseases, Department of Clinical GeneticsErasmus University Medical CenterRotterdamThe Netherlands
| |
Collapse
|
32
|
Fogh S, Dipace G, Bie A, Veiga‐da‐Cunha M, Hansen J, Kjeldsen M, Mosegaard S, Ribes A, Gregersen N, Aagaard L, Van Schaftingen E, Olsen RKJ. Variants in the ethylmalonyl-CoA decarboxylase (ECHDC1) gene: a novel player in ethylmalonic aciduria? J Inherit Metab Dis 2021; 44:1215-1225. [PMID: 33973257 PMCID: PMC8518634 DOI: 10.1002/jimd.12394] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 05/03/2021] [Accepted: 05/07/2021] [Indexed: 12/13/2022]
Abstract
Ethylmalonic acid (EMA) is a major and potentially cytotoxic metabolite associated with short-chain acyl-CoA dehydrogenase (SCAD) deficiency, a condition whose status as a disease is uncertain. Unexplained high EMA is observed in some individuals with complex neurological symptoms, who carry the SCAD gene (ACADS) variants, c.625G>A and c.511C>T. The variants have a high allele frequency in the general population, but are significantly overrepresented in individuals with elevated EMA. This has led to the idea that these variants need to be associated with variants in other genes to cause hyperexcretion of ethylmalonic acid and possibly a diseased state. Ethylmalonyl-CoA decarboxylase (ECHDC1) has been described and characterized as an EMA metabolite repair enzyme, however, its clinical relevance has never been investigated. In this study, we sequenced the ECHDC1 gene (ECHDC1) in 82 individuals, who were reported with unexplained high EMA levels due to the presence of the common ACADS variants only. Three individuals with ACADS c.625G>A variants were found to be heterozygous for ECHDC1 loss-of-function variants. Knockdown experiments of ECHDC1, in healthy human cells with different ACADS c.625G>A genotypes, showed that ECHDC1 haploinsufficiency and homozygosity for the ACADS c.625G>A variant had a synergistic effect on cellular EMA excretion. This study reports the first cases of ECHDC1 gene defects in humans and suggests that ECHDC1 may be involved in elevated EMA excretion in only a small group of individuals with the common ACADS variants. However, a direct link between ECHDC1/ACADS deficiency, EMA and disease could not be proven.
Collapse
Affiliation(s)
- Sarah Fogh
- Research Unit for Molecular Medicine, Department for Clinical MedicineAarhus University and Aarhus University HospitalAarhusDenmark
- Department of BiomedicineAarhus UniversityAarhusDenmark
| | - Graziana Dipace
- Research Unit for Molecular Medicine, Department for Clinical MedicineAarhus University and Aarhus University HospitalAarhusDenmark
| | - Anne Bie
- Research Unit for Molecular Medicine, Department for Clinical MedicineAarhus University and Aarhus University HospitalAarhusDenmark
| | | | - Jakob Hansen
- Department of Forensic MedicineAarhus University HospitalAarhusDenmark
| | - Margrethe Kjeldsen
- Research Unit for Molecular Medicine, Department for Clinical MedicineAarhus University and Aarhus University HospitalAarhusDenmark
| | - Signe Mosegaard
- Research Unit for Molecular Medicine, Department for Clinical MedicineAarhus University and Aarhus University HospitalAarhusDenmark
| | - Antonia Ribes
- Secció d'Errors Congènits del Metabolisme‐IBC, Servei de Bioquímica i Genètica MolecularHospital Clínic, IDIBAPS, CIBERERBarcelonaSpain
| | - Niels Gregersen
- Research Unit for Molecular Medicine, Department for Clinical MedicineAarhus University and Aarhus University HospitalAarhusDenmark
| | - Lars Aagaard
- Department of BiomedicineAarhus UniversityAarhusDenmark
| | | | - Rikke K. J. Olsen
- Research Unit for Molecular Medicine, Department for Clinical MedicineAarhus University and Aarhus University HospitalAarhusDenmark
| |
Collapse
|
33
|
Infant with early onset bilateral facial and bulbar weakness: Successful treatment of riboflavin in multiple acyl-CoA dehydrogenase deficiency caused by biallelic nonsense FLAD1 variants. Neuromuscul Disord 2021; 31:1194-1198. [PMID: 34454814 DOI: 10.1016/j.nmd.2021.07.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 06/22/2021] [Accepted: 07/09/2021] [Indexed: 11/23/2022]
Abstract
Multiple acyl-CoA dehydrogenase deficiency (MADD) is a heterogeneous group of inborn error of metabolic disease affecting the oxidation of fatty acids and amino acids, and choline metabolism. Genes involved in electrons transfer to the mitochondrial respiratory chain typically induce MADD. Recently, FLAD1, which encodes flavin adenine dinucleotide synthase, has also been reported as a cause of MADD. Here, we present a case of a 28-month girl with progressive weakness in facial and bulbar muscle. She has been suffering from feeding difficulty and recurrent respiratory distress. Lipid storage myopathy was evident from muscle biopsy. Furthermore, whole exome sequencing identified homozygous variant of c.745C > T (p.Arg249*) in FLAD1, confirming the diagnosis of FLAD1-related MADD. The patient showed improvements in her symptoms and exhibited catch-up growth following the supplementation of riboflavin. Lipid storage myopathy with FLAD1-related MADD is potentially treatable. Therefore, we should have high clinical suspicion, even though the diagnosis is challenging.
Collapse
|
34
|
Console L, Tolomeo M, Cosco J, Massey K, Barile M, Indiveri C. Impact of natural mutations on the riboflavin transporter 2 and their relevance to human riboflavin transporter deficiency 2. IUBMB Life 2021; 74:618-628. [PMID: 34428344 DOI: 10.1002/iub.2541] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/07/2021] [Accepted: 07/29/2021] [Indexed: 12/28/2022]
Abstract
Riboflavin transporter deficiency 2 (RTD2) is a rare neurological disorder caused by mutations in the Solute carrier family 52 member 2 (Slc52a2) gene encoding human riboflavin transporter 2 (RFVT2). This transporter is ubiquitously expressed and mediates tissue distribution of riboflavin, a water-soluble vitamin that, after conversion into FMN and FAD, plays pivotal roles in carbohydrate, protein, and lipid metabolism. The 3D structure of RFVT2 has been constructed by homology modeling using three different templates that are equilibrative nucleoside transporter 1 (ENT1), Fucose: proton symporter, and glucose transporter type 5 (GLUT5). The structure has been validated by several approaches. All known point mutations of RFVT2, associated with RTD2, have been localized in the protein 3D model. Six of these mutations have been introduced in the recombinant protein for functional characterization. The mutants W31S, S52F, S128L, L312P, C325G, and M423V have been expressed in E. coli, purified, and reconstituted into proteoliposomes for transport assay. All the mutants showed impairment of function. The Km for riboflavin of the mutants increased from about 3 to 9 times with respect to that of WT, whereas Vmax was only marginally affected. This agrees with the improved outcome of most RTD2 patients after administration of high doses of riboflavin.
Collapse
Affiliation(s)
- Lara Console
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Maria Tolomeo
- Department of Biosciences, Biotechnology, and Biopharmaceutics, University of Bari, Bari, Italy
| | - Jessica Cosco
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | | | - Maria Barile
- Department of Biosciences, Biotechnology, and Biopharmaceutics, University of Bari, Bari, Italy
| | - Cesare Indiveri
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| |
Collapse
|
35
|
Mayr JA, Feichtinger RG, Achleitner MT, Brugger K, Kutsam K, Spenger J, Koch J, Hofbauer P, Lagler FB, Sperl W, Weghuber D, Wortmann SB. [Molecular medicine: pathobiochemistry as the key to personalized treatment of inherited diseases]. Monatsschr Kinderheilkd 2021; 169:828-836. [PMID: 34341617 PMCID: PMC8320310 DOI: 10.1007/s00112-021-01252-3] [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] [Accepted: 06/24/2021] [Indexed: 11/27/2022]
Abstract
Genetische Defekte werden vielfach noch als Schicksal empfunden, mit dem man sich Zeit seines Lebens abfinden muss. Es stimmt, dass vererbte Anlagen in vielen Fällen zu schweren Krankheiten führen, allerdings stimmt es auch, dass der Anteil von genetischen Defekten, bei denen eine Therapieoption besteht, stetig wächst und sich der Ausbruch von Krankheitssymptomen bei einigen davon bestenfalls gänzlich verhindern lässt. Die Kenntnis des genauen molekularen Krankheitsmechanismus liefert oft die Grundlage für einen Therapieansatz. Zum Auffinden des genetischen Defekts haben die Möglichkeiten der genomweiten Sequenzierung und ihr mittlerweile breiter Einsatz in der Diagnostik entscheidend beigetragen. Nach dem Nachweis einer genetischen Veränderung braucht es aber noch die Untersuchung der pathobiochemischen Konsequenzen auf zellulärer und systemischer Ebene. Dabei handelt es sich oft um einen längeren Prozess, da der volle Umfang von Funktionsausfällen nicht immer auf Anhieb erkennbar ist. Bei metabolischen Defekten kann die Therapie ein Auffüllen von fehlenden Produkten oder eine Reduktion von giftigen Substraten sein. Oft lässt sich auch die Restfunktion von betroffenen „pathways“ verbessern. Neuerdings haben Therapien mit direkter Korrektur des betroffenen Gendefekts Einzug in die therapeutische Anwendung gefunden. Da die ersten Krankheitssymptome in vielen Fällen früh im Leben auftreten, trifft die Kinderheilkunde eine Vorreiterrolle in der Entwicklung von Therapieansätzen.
Collapse
Affiliation(s)
- J A Mayr
- Universitätsklinik für Kinder- und Jugendheilkunde, Paracelsus Medizinische Privatuniversität, Müllner Hauptstr. 48, 5020 Salzburg, Österreich
| | - R G Feichtinger
- Universitätsklinik für Kinder- und Jugendheilkunde, Paracelsus Medizinische Privatuniversität, Müllner Hauptstr. 48, 5020 Salzburg, Österreich
| | - M T Achleitner
- Universitätsklinik für Kinder- und Jugendheilkunde, Paracelsus Medizinische Privatuniversität, Müllner Hauptstr. 48, 5020 Salzburg, Österreich
| | - K Brugger
- Universitätsklinik für Kinder- und Jugendheilkunde, Paracelsus Medizinische Privatuniversität, Müllner Hauptstr. 48, 5020 Salzburg, Österreich
| | - K Kutsam
- Universitätsklinik für Kinder- und Jugendheilkunde, Paracelsus Medizinische Privatuniversität, Müllner Hauptstr. 48, 5020 Salzburg, Österreich
| | - J Spenger
- Universitätsklinik für Kinder- und Jugendheilkunde, Paracelsus Medizinische Privatuniversität, Müllner Hauptstr. 48, 5020 Salzburg, Österreich
| | - J Koch
- Universitätsklinik für Kinder- und Jugendheilkunde, Paracelsus Medizinische Privatuniversität, Müllner Hauptstr. 48, 5020 Salzburg, Österreich
| | - P Hofbauer
- Arzneimittelproduktion, Landesapotheke Salzburg, Betrieb des Landes Salzburg, Müllner Hauptstr. 50, 5020 Salzburg, Österreich
| | - F B Lagler
- Universitätsklinik für Kinder- und Jugendheilkunde, Paracelsus Medizinische Privatuniversität, Müllner Hauptstr. 48, 5020 Salzburg, Österreich.,Institut für angeborene Stoffwechselerkrankungen, Paracelsus Medizinische Privatuniversität, Strubergasse 22, 5020 Salzburg, Österreich
| | - W Sperl
- Universitätsklinik für Kinder- und Jugendheilkunde, Paracelsus Medizinische Privatuniversität, Müllner Hauptstr. 48, 5020 Salzburg, Österreich
| | - D Weghuber
- Universitätsklinik für Kinder- und Jugendheilkunde, Paracelsus Medizinische Privatuniversität, Müllner Hauptstr. 48, 5020 Salzburg, Österreich
| | - S B Wortmann
- Universitätsklinik für Kinder- und Jugendheilkunde, Paracelsus Medizinische Privatuniversität, Müllner Hauptstr. 48, 5020 Salzburg, Österreich.,Amalia Children's Hospital, Radboudumc, Geert Grote Plein Zuid 10, 6525GA Nijmegen, Niederlande
| |
Collapse
|
36
|
Significant Diagnostic and Prognostic Value of FLAD1 and Related MicroRNAs in Breast Cancer after a Pan-Cancer Analysis. DISEASE MARKERS 2021; 2021:6962526. [PMID: 34336008 PMCID: PMC8321750 DOI: 10.1155/2021/6962526] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 05/30/2021] [Accepted: 06/24/2021] [Indexed: 12/09/2022]
Abstract
The identification of biomarkers plays an important role in the diagnosis and prognosis of cancers. In this study, we explored the diagnostic and prognostic value of the FLAD1 expression across pan-cancer analysis from online databases (Oncomine, cBioPortal, Breast Cancer Gene-Expression Miner, UALCAN, GEO, BCIP, TNMplot, ENCORI, Kaplan-Meier Plotter, and LinkedOmics). We found that FLAD1 was overexpressed in a number of different kinds of cancers, especially in breast cancer, and higher FLAD1 expression level was associated with the HER+, p53 mutant, node-involved, NPI stage 3, basal-like, and triple-negative groups compared with the other subgroups of breast cancer. The FLAD1 expression levels were higher in patients that were 21–40 years old than those in patients of other ages and were higher in the African-American group than in the Caucasian group. We also analyzed the FLAD1-related microRNAs and their prognostic values in breast cancer. This study highlights the significance of FLAD1 in cancers and provides evidence for its potential as a biomarker for the diagnosis and prognosis of cancers.
Collapse
|
37
|
Alteration of Flavin Cofactor Homeostasis in Human Neuromuscular Pathologies. Methods Mol Biol 2021; 2280:275-295. [PMID: 33751442 DOI: 10.1007/978-1-0716-1286-6_18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The aim of this short review chapter is to provide a brief summary of the relevance of riboflavin (Rf or vitamin B2) and its derived cofactors flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD) for human neuromuscular bioenergetics.Therefore, as a completion of this book we would like to summarize what kind of human pathologies could derive from genetic disturbances of Rf transport, flavin cofactor synthesis and delivery to nascent apoflavoproteins, as well as by alteration of vitamin recycling during protein turnover.
Collapse
|
38
|
Brandão SR, Ferreira R, Rocha H. Exploring the contribution of mitochondrial dynamics to multiple acyl-CoA dehydrogenase deficiency-related phenotype. Arch Physiol Biochem 2021; 127:210-216. [PMID: 31215835 DOI: 10.1080/13813455.2019.1628065] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Mitochondrial fatty acid β-oxidation disorders (FAOD) are among the diseases detected by newborn screening in most developed countries. Alterations of mitochondrial functionality are characteristic of these metabolic disorders. However, many questions remain to be clarified, namely how the interplay between the signaling pathways harbored in mitochondria contributes to the disease-related phenotype. Herein, we overview the role of mitochondria on the regulation of cell homeostasis through the production of ROS, mitophagy, apoptosis, and mitochondrial biogenesis. Emphasis is given to the signaling pathways involving MnSOD, sirtuins and PGC-1α, which seem to contribute to FAOD phenotype, namely to multiple acyl-CoA dehydrogenase deficiency (MADD). The association between phenotype and genotype is not straightforward, suggesting that specific molecular mechanisms may contribute to MADD pathogenesis, making MADD an interesting model to better understand this interplay. However, more work needs to be done envisioning the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Sofia R Brandão
- Mass Spectrometry Group, QOPNA, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Rita Ferreira
- Mass Spectrometry Group, QOPNA, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Hugo Rocha
- Newborn Screening, Metabolism and Genetics Unit, Human Genetics Department, National Institute of Health Ricardo Jorge, Porto, Portugal
| |
Collapse
|
39
|
Characterization of ETFDH and PHGDH Mutations in a Patient with Mild Glutaric Aciduria Type II and Serine Deficiency. Genes (Basel) 2021; 12:genes12050703. [PMID: 34066864 PMCID: PMC8150808 DOI: 10.3390/genes12050703] [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] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 04/29/2021] [Accepted: 05/05/2021] [Indexed: 12/20/2022] Open
Abstract
Glutaric aciduria type II (GA-II) is a rare autosomal recessive disease caused by defects in electron transfer flavoprotein (ETF), ultimately causing insufficiencies in multiple acyl-CoA dehydrogenase (MAD). 3-phosphoglycerate dehydrogenase (3-PHGDH) deficiency, is another rare autosomal disorder that appears due to a defect in the synthesis of L-serine amino acid. Several mutations of ETFDH and PHGDH genes have been associated with different forms of GA-II and serine deficiency, respectively. In this study, we report a unique case of GA-II with serine deficiency using biochemical, genetic, and in silico approaches. The proband of Syrian descent had positive newborn screening (NBS) for GA-II. At two years of age, the patient presented with developmental regression, ataxia, and intractable seizures. Results of amino acid profiling demonstrated extremely low levels of serine. Confirmatory tests for GA-II and whole exome sequencing (WES) were performed to determine the etiology of intractable seizure. Sequencing results indicated a previously reported homozygous missense mutation, c.679 C>A (p.Pro227Thr) in the ETFDH gene and a novel missense homozygous mutation c.1219 T>C (p.Ser407Pro) in the PHGDH gene. In silico tools predicted these mutations as deleterious. Here, the clinical and biochemical investigations indicate that ETFDH:p.Pro227Thr and PHGDH:p.Ser407Pro variants likely underlie the pathogenesis of GA-II and serine deficiency, respectively. This study indicates that two rare autosomal recessive disorders should be considered in consanguineous families, more specifically in those with atypical presentation.
Collapse
|
40
|
Missaglia S, Tavian D, Angelini C. ETF dehydrogenase advances in molecular genetics and impact on treatment. Crit Rev Biochem Mol Biol 2021; 56:360-372. [PMID: 33823724 DOI: 10.1080/10409238.2021.1908952] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Electron transfer flavoprotein dehydrogenase, also called ETF-ubiquinone oxidoreductase (ETF-QO), is a protein localized in the inner membrane of mitochondria, playing a central role in the electron-transfer system. Indeed, ETF-QO mediates electron transport from flavoprotein dehydrogenases to the ubiquinone pool. ETF-QO mutations are often associated with riboflavin-responsive multiple acyl-CoA dehydrogenase deficiency (RR-MADD, OMIM#231680), a multisystem genetic disease characterized by various clinical manifestations with different degrees of severity. In this review, we outline the clinical features correlated with ETF-QO deficiency and the benefits obtained from different treatments, such as riboflavin, L-carnitine and/or coenzyme Q10 supplementation, and a diet poor in fat and protein. Moreover, we provide a detailed summary of molecular and bioinformatic investigations, describing the mutations identified in ETFDH gene and highlighting their predicted impact on enzymatic structure and activity. In addition, we report biochemical and functional analysis, performed in HEK293 cells and patient fibroblasts and muscle cells, to show the relationship between the nature of ETFDH mutations, the variable impairment of enzyme function, and the different degrees of RR-MADD severity. Finally, we describe in detail 5 RR-MADD patients carrying different ETFDH mutations and presenting variable degrees of clinical symptom severity.
Collapse
Affiliation(s)
- Sara Missaglia
- Laboratory of Cellular Biochemistry and Molecular Biology, CRIBENS, Università Cattolica del Sacro Cuore, Milan, Italy.,Psychology Department, Università Cattolica del Sacro Cuore, Milan, Italy
| | - Daniela Tavian
- Laboratory of Cellular Biochemistry and Molecular Biology, CRIBENS, Università Cattolica del Sacro Cuore, Milan, Italy.,Psychology Department, Università Cattolica del Sacro Cuore, Milan, Italy
| | - Corrado Angelini
- Neuromuscular Laboratory, Department of Neurosciences, University of Padova, Padova, Italy
| |
Collapse
|
41
|
Alston CL, Stenton SL, Hudson G, Prokisch H, Taylor RW. The genetics of mitochondrial disease: dissecting mitochondrial pathology using multi-omic pipelines. J Pathol 2021; 254:430-442. [PMID: 33586140 PMCID: PMC8600955 DOI: 10.1002/path.5641] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/07/2021] [Accepted: 02/09/2021] [Indexed: 12/12/2022]
Abstract
Mitochondria play essential roles in numerous metabolic pathways including the synthesis of adenosine triphosphate through oxidative phosphorylation. Clinically, mitochondrial diseases occur when there is mitochondrial dysfunction – manifesting at any age and affecting any organ system; tissues with high energy requirements, such as muscle and the brain, are often affected. The clinical heterogeneity is parallel to the degree of genetic heterogeneity associated with mitochondrial dysfunction. Around 10% of human genes are predicted to have a mitochondrial function, and defects in over 300 genes are reported to cause mitochondrial disease. Some involve the mitochondrial genome (mtDNA), but the vast majority occur within the nuclear genome. Except for a few specific genetic defects, there remains no cure for mitochondrial diseases, which means that a genetic diagnosis is imperative for genetic counselling and the provision of reproductive options for at‐risk families. Next‐generation sequencing strategies, particularly exome and whole‐genome sequencing, have revolutionised mitochondrial diagnostics such that the traditional muscle biopsy has largely been replaced with a minimally‐invasive blood sample for an unbiased approach to genetic diagnosis. Where these genomic approaches have not identified a causative defect, or where there is insufficient support for pathogenicity, additional functional investigations are required. The application of supplementary ‘omics’ technologies, including transcriptomics, proteomics, and metabolomics, has the potential to greatly improve diagnostic strategies. This review aims to demonstrate that whilst a molecular diagnosis can be achieved for many cases through next‐generation sequencing of blood DNA, the use of patient tissues and an integrated, multidisciplinary multi‐omics approach is pivotal for the diagnosis of more challenging cases. Moreover, the analysis of clinically relevant tissues from affected individuals remains crucial for understanding the molecular mechanisms underlying mitochondrial pathology. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Charlotte L Alston
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.,NHS Highly Specialised Services for Rare Mitochondrial Disorders, Royal Victoria Infirmary, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Sarah L Stenton
- Institute of Human Genetics, Technische Universität München, München, Germany.,Institute of Neurogenomics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Gavin Hudson
- Wellcome Centre for Mitochondrial Research, Bioscience Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Holger Prokisch
- Institute of Human Genetics, Technische Universität München, München, Germany.,Institute of Neurogenomics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Robert W Taylor
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.,NHS Highly Specialised Services for Rare Mitochondrial Disorders, Royal Victoria Infirmary, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| |
Collapse
|
42
|
Liu XY, Chen XJ, Zhao M, Wang ZQ, Chen HZ, Li HF, Wang CJ, Wu SF, Peng C, Yin Y, Fu HX, Lin MT, Yu L, Xiong ZQ, Wu ZY, Wang N. CHIP control degradation of mutant ETF:QO through ubiquitylation in late-onset multiple acyl-CoA dehydrogenase deficiency. J Inherit Metab Dis 2021; 44:450-468. [PMID: 33438237 DOI: 10.1002/jimd.12361] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 12/24/2020] [Accepted: 01/11/2021] [Indexed: 11/12/2022]
Abstract
Late-onset multiple acyl-CoA dehydrogenase deficiency (MADD) is the most common form of lipid storage myopathy. The disease is mainly caused by mutations in electron-transfer flavoprotein dehydrogenase gene (ETFDH), which leads to decreased levels of ETF:QO in skeletal muscle. However, the specific underlying mechanisms triggering such degradation remain unknown. We constructed expression plasmids containing wild type ETF:QO and mutants ETF:QO-A84T, R175H, A215T, Y333C, and cultured patient-derived fibroblasts containing the following mutations in ETFDH: c.250G>A (p.A84T), c.998A>G (p.Y333C), c.770A>G (p.Y257C), c.1254_1257delAACT (p. L418TfsX10), c.524G>A (p.R175H), c.380T>A (p.L127P), and c.892C>T (p.P298S). We used in vitro expression systems and patient-derived fibroblasts to detect stability of ETF:QO mutants then evaluated their interaction with Hsp70 interacting protein CHIP with active/inactive ubiquitin E3 ligase carboxyl terminus using western blot and immunofluorescence staining. This interaction was confirmed in vitro and in vivo by co-immunoprecipitation and immunofluorescence staining. We confirmed the existence two ubiquitination sites in mutant ETF:QO using mass spectrometry (MS) analysis. We found that mutant ETF:QO proteins were unstable and easily degraded in patient fibroblasts and in vitro expression systems by ubiquitin-proteasome pathway, and identified the specific ubiquitin E3 ligase as CHIP, which forms complex to control mutant ETF:QO degradation through poly-ubiquitination. CHIP-dependent degradation of mutant ETF:QO proteins was confirmed by MS and site-directed mutagenesis of ubiquitination sites. Hsp70 is directly involved in this process as molecular chaperone of CHIP. CHIP plays an important role in ubiquitin-proteasome pathway dependent degradation of mutant ETF:QO by working as a chaperone-assisted E3 ligase, which reveals CHIP's potential role in pathological mechanisms of late-onset MADD.
Collapse
Affiliation(s)
- Xin-Yi Liu
- Department of Neurology, Fujian Institute of Neurology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Xue-Jiao Chen
- Department of Neurology, Fujian Institute of Neurology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Department of Neurology, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian, China
| | - Miao Zhao
- Department of Neurology, Fujian Institute of Neurology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhi-Qiang Wang
- Department of Neurology, Fujian Institute of Neurology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian, China
| | - Hai-Zhu Chen
- Department of Neurology, Fujian Institute of Neurology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Hong-Fu Li
- Department of Neurology and Research Center of Neurology in the Second Affiliated Hospital, and the Collaborative Innovation Center for Brain Science, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chen-Ji Wang
- State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Shi-Fei Wu
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Science, Shanghai, China
| | - Chao Peng
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Science, Shanghai, China
| | - Yue Yin
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Science, Shanghai, China
| | - Hong-Xia Fu
- Department of Neurology, Fujian Institute of Neurology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Min-Ting Lin
- Department of Neurology, Fujian Institute of Neurology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Long Yu
- State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Zhi-Qi Xiong
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhi-Ying Wu
- Department of Neurology and Research Center of Neurology in the Second Affiliated Hospital, and the Collaborative Innovation Center for Brain Science, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ning Wang
- Department of Neurology, Fujian Institute of Neurology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
43
|
Mereis M, Wanders RJA, Schoonen M, Dercksen M, Smuts I, van der Westhuizen FH. Disorders of flavin adenine dinucleotide metabolism: MADD and related deficiencies. Int J Biochem Cell Biol 2021; 132:105899. [PMID: 33279678 DOI: 10.1016/j.biocel.2020.105899] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 11/27/2020] [Accepted: 11/30/2020] [Indexed: 12/13/2022]
Abstract
Multiple acyl-coenzyme A dehydrogenase deficiency (MADD), or glutaric aciduria type II (GAII), is a group of clinically heterogeneous disorders caused by mutations in electron transfer flavoprotein (ETF) and ETF-ubiquinone oxidoreductase (ETFQO) - the two enzymes responsible for the re-oxidation of enzyme-bound flavin adenine dinucleotide (FADH2) via electron transfer to the respiratory chain at the level of coenzyme Q10. Over the past decade, an increasing body of evidence has further coupled mutations in FAD metabolism (including intercellular riboflavin transport, FAD biosynthesis and FAD transport) to MADD-like phenotypes. In this review we provide a detailed description of the overarching and specific metabolic pathways involved in MADD. We examine the eight associated genes (ETFA, ETFB, ETFDH, FLAD1, SLC25A32 and SLC52A1-3) and clinical phenotypes, and report ∼436 causative mutations following a systematic literature review. Finally, we focus attention on the value and shortcomings of current diagnostic approaches, as well as current and future therapeutic options for MADD and its phenotypic disorders.
Collapse
Affiliation(s)
- Michelle Mereis
- Human Metabolomics, North-West University, Potchefstroom, South Africa
| | - Ronald J A Wanders
- Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, the Netherlands
| | - Maryke Schoonen
- Human Metabolomics, North-West University, Potchefstroom, South Africa; Centre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa
| | - Marli Dercksen
- Human Metabolomics, North-West University, Potchefstroom, South Africa
| | - Izelle Smuts
- Department of Paediatrics, Steve Biko Academic Hospital, University of Pretoria, South Africa
| | | |
Collapse
|
44
|
Henriques BJ, Katrine Jentoft Olsen R, Gomes CM, Bross P. Electron transfer flavoprotein and its role in mitochondrial energy metabolism in health and disease. Gene 2021; 776:145407. [PMID: 33450351 DOI: 10.1016/j.gene.2021.145407] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 12/08/2020] [Accepted: 12/16/2020] [Indexed: 12/15/2022]
Abstract
Electron transfer flavoprotein (ETF) is an enzyme with orthologs from bacteria to humans. Human ETF is nuclear encoded by two separate genes, ETFA and ETFB, respectively. After translation, the two subunits are imported to the mitochondrial matrix space and assemble into a heterodimer containing one FAD and one AMP as cofactors. ETF functions as a hub taking up electrons from at least 14 flavoenzymes, feeding them into the respiratory chain. This represents a major source of reducing power for the electron transport chain from fatty acid oxidation and amino acid degradation. Transfer of electrons from the donor enzymes to ETF occurs by direct transfer between the enzyme bound flavins, a process that is tightly regulated by the polypeptide chain and by protein:protein interactions. ETF, in turn relays electrons to the iron sulfur cluster of the inner membrane protein ETF:QO, from where they travel via the FAD in ETF:QO to ubiquinone, entering the respiratory chain at the level of complex III. ETF recognizes its dehydrogenase partners via a recognition loop that anchors the protein on its partner followed by dynamic movements of the ETF flavin domain that bring redox cofactors in close proximity, thus promoting electron transfer. Genetic mutations in the ETFA or ETFB genes cause the Mendelian disorder multiple acyl-CoA dehydrogenase deficiency (MADD; OMIM #231680). We here review the knowledge on human ETF and investigations of the effects of disease-associated missense mutations in this protein that have promoted the understanding of the essential role that ETF plays in cellular metabolism and human disease.
Collapse
Affiliation(s)
- Bárbara J Henriques
- Biosystems and Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal; Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal.
| | - Rikke Katrine Jentoft Olsen
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, 8200 Aarhus, Denmark.
| | - Cláudio M Gomes
- Biosystems and Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal; Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal.
| | - Peter Bross
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, 8200 Aarhus, Denmark.
| |
Collapse
|
45
|
Leone P, Tolomeo M, Barile M. Continuous and Discontinuous Approaches to Study FAD Synthesis and Degradation Catalyzed by Purified Recombinant FAD Synthase or Cellular Fractions. Methods Mol Biol 2021; 2280:87-116. [PMID: 33751431 DOI: 10.1007/978-1-0716-1286-6_7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Riboflavin, or vitamin B2, is the precursor of flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD), essential redox (and sometimes non-redox) cofactors of a large number of flavoenzymes involved in energetic metabolism, protein folding, apoptosis, chromatin remodeling, and a number of other cell regulatory processes.The cellular and subcellular steady-state concentrations of flavin cofactors, which are available for flavoprotein biogenesis and assembly, depend on carrier-mediated transport processes and on coordinated synthesizing/destroying enzymatic activities, catalyzed by enzymes whose catalytic and structural properties are still matter of investigation.Alteration of flavin homeostasis has been recently correlated to human pathological conditions, such as neuromuscular disorders and cancer, and therefore we propose here protocols useful to detect metabolic processes involved in FAD forming and destroying.Our protocols exploit the chemical-structural differences between riboflavin, FMN , and FAD , which are responsible for differences in the spectroscopic properties (mainly fluorescence) of the two cofactors (FMN and FAD); therefore, in our opinion, when applicable measurements of fluorescence changes in continuo represent the elective techniques to follow FAD synthesis and degradation. Thus, after procedures able to calibrate flavin concentrations (Subheading 3.1), we describe simple continuous and rapid procedures, based on the peculiar optical properties of free flavins, useful to determine the rate of cofactor metabolism catalyzed by either recombinant enzymes or natural enzymes present in cellular lysates/subfractions (Subheading 3.2).Fluorescence properties of free flavins can also be useful in analytical determinations of the three molecular flavin forms, based on HPLC separation, with a quite high sensitivity. Assaying at different incubation times the molecular composition of the reaction mixture is a discontinuous experimental approach to measure the rate of FAD synthesis/degradation catalyzed by cell lysates or recombinant FAD synthase (Subheading 3.3). Continuous and discontinuous approaches can, when necessary, be performed in parallel.
Collapse
Affiliation(s)
- Piero Leone
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari "A. Moro", Bari, Italy
| | - Maria Tolomeo
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari "A. Moro", Bari, Italy
| | - Maria Barile
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari "A. Moro", Bari, Italy.
| |
Collapse
|
46
|
Galluccio M, Indiveri C. Heterologous Overexpression of Human FAD Synthase Isoforms 1 and 2. Methods Mol Biol 2021; 2280:55-67. [PMID: 33751429 DOI: 10.1007/978-1-0716-1286-6_5] [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] [Indexed: 06/12/2023]
Abstract
The study of human FAD synthase enzymes requires a recombinant strategy to produce large amount of purified proteins in a soluble form. E. coli was exploited to this aim. To achieve the production of FAD synthase in a large scale, E. coli strains, plasmids (promoter, tags), growth temperature, inducer concentration, medium composition, and osmotic pressure were optimized. To date there is no universal protocol for protein expression, but for each protein a specific combination of "expression parameters" can be selected in order to maximize the results. An experimental protocol for the expression of two isoforms of the human FAD synthase was set up. The final procedures are based on the use of E. coli Rosetta(DE3) strain. Two different plasmids were used to obtain optimal amount of the two protein isoforms. In both cases, following the addition of the IPTG inducer, the growth temperature was lowered to increase the solubility of the recombinant protein. The detailed procedures for FAD synthase isoform 1 and isoform 2 overproduction are described in this protocol.
Collapse
Affiliation(s)
- Michele Galluccio
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Cesare Indiveri
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy.
| |
Collapse
|
47
|
Pitceathly RD, Keshavan N, Rahman J, Rahman S. Moving towards clinical trials for mitochondrial diseases. J Inherit Metab Dis 2021; 44:22-41. [PMID: 32618366 PMCID: PMC8432143 DOI: 10.1002/jimd.12281] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 06/22/2020] [Accepted: 06/30/2020] [Indexed: 12/11/2022]
Abstract
Primary mitochondrial diseases represent some of the most common and severe inherited metabolic disorders, affecting ~1 in 4,300 live births. The clinical and molecular diversity typified by mitochondrial diseases has contributed to the lack of licensed disease-modifying therapies available. Management for the majority of patients is primarily supportive. The failure of clinical trials in mitochondrial diseases partly relates to the inefficacy of the compounds studied. However, it is also likely to be a consequence of the significant challenges faced by clinicians and researchers when designing trials for these disorders, which have historically been hampered by a lack of natural history data, biomarkers and outcome measures to detect a treatment effect. Encouragingly, over the past decade there have been significant advances in therapy development for mitochondrial diseases, with many small molecules now transitioning from preclinical to early phase human interventional studies. In this review, we present the treatments and management strategies currently available to people with mitochondrial disease. We evaluate the challenges and potential solutions to trial design and highlight the emerging pharmacological and genetic strategies that are moving from the laboratory to clinical trials for this group of disorders.
Collapse
Affiliation(s)
- Robert D.S. Pitceathly
- Department of Neuromuscular DiseasesUCL Queen Square Institute of Neurology and The National Hospital for Neurology and NeurosurgeryLondonUK
| | - Nandaki Keshavan
- Mitochondrial Research GroupUCL Great Ormond Street Institute of Child HealthLondonUK
- Metabolic UnitGreat Ormond Street Hospital for Children NHS Foundation TrustLondonUK
| | - Joyeeta Rahman
- Mitochondrial Research GroupUCL Great Ormond Street Institute of Child HealthLondonUK
| | - Shamima Rahman
- Mitochondrial Research GroupUCL Great Ormond Street Institute of Child HealthLondonUK
- Metabolic UnitGreat Ormond Street Hospital for Children NHS Foundation TrustLondonUK
| |
Collapse
|
48
|
Koňaříková E, Marković A, Korandová Z, Houštěk J, Mráček T. Current progress in the therapeutic options for mitochondrial disorders. Physiol Res 2020; 69:967-994. [PMID: 33129249 PMCID: PMC8549882 DOI: 10.33549/physiolres.934529] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 10/02/2020] [Indexed: 12/20/2022] Open
Abstract
Mitochondrial disorders manifest enormous genetic and clinical heterogeneity - they can appear at any age, present with various phenotypes affecting any organ, and display any mode of inheritance. What mitochondrial diseases do have in common, is impairment of respiratory chain activity, which is responsible for more than 90% of energy production within cells. While diagnostics of mitochondrial disorders has been accelerated by introducing Next-Generation Sequencing techniques in recent years, the treatment options are still very limited. For many patients only a supportive or symptomatic therapy is available at the moment. However, decades of basic and preclinical research have uncovered potential target points and numerous compounds or interventions are now subjects of clinical trials. In this review, we focus on current and emerging therapeutic approaches towards the treatment of mitochondrial disorders. We focus on small compounds, metabolic interference, such as endurance training or ketogenic diet and also on genomic approaches.
Collapse
Affiliation(s)
- E Koňaříková
- Laboratory of Bioenergetics, Institute of Physiology Czech Acad. Sci., Prague, Czech Republic. ,
| | | | | | | | | |
Collapse
|
49
|
Barcelos I, Shadiack E, Ganetzky RD, Falk MJ. Mitochondrial medicine therapies: rationale, evidence, and dosing guidelines. Curr Opin Pediatr 2020; 32:707-718. [PMID: 33105273 PMCID: PMC7774245 DOI: 10.1097/mop.0000000000000954] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Primary mitochondrial disease is a highly heterogeneous but collectively common inherited metabolic disorder, affecting at least one in 4300 individuals. Therapeutic management of mitochondrial disease typically involves empiric prescription of enzymatic cofactors, antioxidants, and amino acid and other nutrient supplements, based on biochemical reasoning, historical experience, and consensus expert opinion. As the field continues to rapidly advance, we review here the preclinical and clinical evidence, and specific dosing guidelines, for common mitochondrial medicine therapies to guide practitioners in their prescribing practices. RECENT FINDINGS Since publication of Mitochondrial Medicine Society guidelines for mitochondrial medicine therapies management in 2009, data has emerged to support consideration for using additional therapeutic agents and discontinuation of several previously used agents. Preclinical animal modeling data have indicated a lack of efficacy for vitamin C as an antioxidant for primary mitochondrial disease, but provided strong evidence for vitamin E and N-acetylcysteine. Clinical data have suggested L-carnitine may accelerate atherosclerotic disease. Long-term follow up on L-arginine use as prophylaxis against or acute treatment for metabolic strokes has provided more data supporting its clinical use in individuals with mitochondrial encephalopathy with lactic acidosis and stroke-like episodes (MELAS) syndrome and Leigh syndrome. Further, several precision therapies have been developed for specific molecular causes and/or shared clinical phenotypes of primary mitochondrial disease. SUMMARY We provide a comprehensive update on mitochondrial medicine therapies based on current evidence and our single-center clinical experience to support or refute their use, and provide detailed dosing guidelines, for the clinical management of mitochondrial disease. The overarching goal of empiric mitochondrial medicines is to utilize therapies with favorable benefit-to-risk profiles that may stabilize and enhance residual metabolic function to improve cellular resiliency and slow clinical disease progression and/or prevent acute decompensation.
Collapse
Affiliation(s)
- Isabella Barcelos
- Center for Applied Genomics, Division of Human Genetics, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Edward Shadiack
- Mitochondrial Medicine Frontier Program, Division of Human Genetics, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Rebecca D. Ganetzky
- Mitochondrial Medicine Frontier Program, Division of Human Genetics, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Marni J. Falk
- Mitochondrial Medicine Frontier Program, Division of Human Genetics, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
50
|
Zhang R, Engel AL, Wang Y, Li B, Shen W, Gillies MC, Chao JR, Du J. Inhibition of Mitochondrial Respiration Impairs Nutrient Consumption and Metabolite Transport in Human Retinal Pigment Epithelium. J Proteome Res 2020; 20:909-922. [PMID: 32975122 DOI: 10.1021/acs.jproteome.0c00690] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Mitochondrial respiration in mammalian cells not only generates ATP to meet their own energy needs but also couples with biosynthetic pathways to produce metabolites that can be exported to support neighboring cells. However, how defects in mitochondrial respiration influence these biosynthetic and exporting pathways remains poorly understood. Mitochondrial dysfunction in retinal pigment epithelium (RPE) cells is an emerging contributor to the death of their neighboring photoreceptors in degenerative retinal diseases including age-related macular degeneration. In this study, we used targeted-metabolomics and 13C tracing to investigate how inhibition of mitochondrial respiration influences the intracellular and extracellular metabolome. We found inhibition of mitochondrial respiration strikingly influenced both the intracellular and extracellular metabolome in primary RPE cells. Intriguingly, the extracellular metabolic changes sensitively reflected the intracellular changes. These changes included substantially enhanced glucose consumption and lactate production; reduced release of pyruvate, citrate, and ketone bodies; and massive accumulation of multiple amino acids and nucleosides. In conclusion, these findings reveal a metabolic signature of nutrient consumption and release in mitochondrial dysfunction in RPE cells. Testing medium metabolites provides a sensitive and noninvasive method to assess mitochondrial function in nutrient utilization and transport.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Ophthalmology, West Virginia University, Morgantown, West Virginia 26506, United States.,Department of Biochemistry, West Virginia University, Morgantown, West Virginia 26506, United States.,Save Sight Institute, Sydney Medical School, University of Sydney, Sydney, NSW 2000, Australia
| | - Abbi L Engel
- Department of Ophthalmology, University of Washington, Seattle, Washington 98109, United States
| | - Yekai Wang
- Department of Ophthalmology, West Virginia University, Morgantown, West Virginia 26506, United States.,Department of Biochemistry, West Virginia University, Morgantown, West Virginia 26506, United States
| | - Bo Li
- Department of Ophthalmology, West Virginia University, Morgantown, West Virginia 26506, United States.,Department of Biochemistry, West Virginia University, Morgantown, West Virginia 26506, United States
| | - Weiyong Shen
- Save Sight Institute, Sydney Medical School, University of Sydney, Sydney, NSW 2000, Australia
| | - Mark C Gillies
- Save Sight Institute, Sydney Medical School, University of Sydney, Sydney, NSW 2000, Australia
| | - Jennifer R Chao
- Department of Ophthalmology, University of Washington, Seattle, Washington 98109, United States
| | - Jianhai Du
- Department of Ophthalmology, West Virginia University, Morgantown, West Virginia 26506, United States.,Department of Biochemistry, West Virginia University, Morgantown, West Virginia 26506, United States
| |
Collapse
|