1
|
Shen MM, Rummey C, Lynch DR. Phenotypic variation of FXN compound heterozygotes in a Friedreich ataxia cohort. Ann Clin Transl Neurol 2024; 11:1110-1121. [PMID: 38396238 PMCID: PMC11093247 DOI: 10.1002/acn3.52027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/24/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
OBJECTIVE Most individuals with Friedreich ataxia (FRDA) have homozygous GAA triplet repeat expansions in the FXN gene, correlating with a typical phenotype of ataxia and cardiomyopathy. A minority are compound heterozygotes carrying a GAA expansion on one allele and a mutation on the other. The study aim was to examine phenotypic variation among compound heterozygotes. METHODS Data on FXN mutations were obtained from the Friedreich Ataxia Clinical Outcome Measures Study (FA-COMS). We compared clinical features in a single-site FA-COMS cohort of 51 compound heterozygous and 358 homozygous patients, including quantitative measures of cardiac, neurologic, and visual disease progression. RESULTS Non-GAA repeat mutations were associated with reduced cardiac disease, and patients with minimal/no function mutations otherwise had a typical FRDA phenotype but with significantly more severe progression. The partial function mutation group was characterized by relative sparing of bulbar and upper limb function, as well as particularly low cardiac involvement. Other clinical features in this group, including optic atrophy and diabetes mellitus, varied widely depending on the specific type of partial function mutation. INTERPRETATION These data support that the typical FRDA phenotype is driven by frataxin deficiency, especially severe in compound heterozygotes with minimal/no function mutations, whereas the heterogeneous presentations of those with partial function mutations may indicate other contributing factors to FRDA pathogenesis.
Collapse
Affiliation(s)
- Megan M. Shen
- Division of NeurologyThe Children's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
- Perelman School of Medicine, University of Pennsylvania.PhiladelphiaPennsylvaniaUSA
| | | | - David R. Lynch
- Division of NeurologyThe Children's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
- Perelman School of Medicine, University of Pennsylvania.PhiladelphiaPennsylvaniaUSA
| |
Collapse
|
2
|
Irigoyen P, Mansilla S, Castro L, Cassina A, Sapiro R. Mitochondrial function and reactive oxygen species production during human sperm capacitation: Unraveling key players. FASEB J 2024; 38:e23486. [PMID: 38407497 DOI: 10.1096/fj.202301957rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/22/2024] [Accepted: 01/30/2024] [Indexed: 02/27/2024]
Abstract
Sperm capacitation is a critical process for male fertility. It involves a series of biochemical and physiological changes that occur in the female reproductive tract, rendering the sperm competent for successful fertilization. The precise mechanisms and, specifically, the role of mitochondria, in sperm capacitation remain incompletely understood. Previously, we revealed that in mouse sperm mitochondrial activity (e.g., oxygen consumption, membrane potential, ATP/ADP exchange, and mitochondrial Ca2+ ) increases during capacitation. Herein, we studied mitochondrial function by high-resolution respirometry (HRR) and reactive oxygen species production in capacitated (CAP) and non-capacitated (NC) human spermatozoa. We found that in capacitated sperm from normozoospermic donors, the respiratory control ratio increased by 36%, accompanied by a double oxygen consumption rate (OCR) in the presence of antimycin A. Extracellular hydrogen peroxide (H2 O2 ) detection was three times higher in CAP than in NC sperm cells. To confirm that H2 O2 production depends on mitochondrial superoxide (O 2 · - $$ {\mathrm{O}}_2^{\cdotp -} $$ ) formation, we evaluated mitochondrial aconitase (ACO2) amount, activity, and role in the metabolic flux from the sperm tricarboxylic acid cycle. We estimated that CAP cells produce, on average by individual, (59 ± 22)% moreO 2 · - $$ {\mathrm{O}}_2^{\cdotp -} $$ in the steady-state compared to NC cells. Finally, we analyzed two targets of oxidative stress: lipid peroxidation by western blot against 4-hydroxynonenal and succinate dehydrogenase (SDH) activity by HRR. We did not observe modifications in lipoperoxidation nor the activity of SDH, suggesting that during capacitation, the increase in mitochondrial H2 O2 production does not damage sperm and it is necessary for the normal CAP process.
Collapse
Affiliation(s)
- Pilar Irigoyen
- Unidad Académica Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- Centro de Investigaciones Biomédicas (CEINBIO), Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Santiago Mansilla
- Centro de Investigaciones Biomédicas (CEINBIO), Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- Departamento de Métodos Cuantitativos, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Laura Castro
- Centro de Investigaciones Biomédicas (CEINBIO), Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Adriana Cassina
- Centro de Investigaciones Biomédicas (CEINBIO), Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Rossana Sapiro
- Unidad Académica Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- Centro de Investigaciones Biomédicas (CEINBIO), Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
3
|
Luffarelli R, Panarello L, Quatrana A, Tiano F, Fortuni S, Rufini A, Malisan F, Testi R, Condò I. Interferon Gamma Enhances Cytoprotective Pathways via Nrf2 and MnSOD Induction in Friedreich's Ataxia Cells. Int J Mol Sci 2023; 24:12687. [PMID: 37628866 PMCID: PMC10454386 DOI: 10.3390/ijms241612687] [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: 06/08/2023] [Revised: 07/27/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Friedreich's ataxia (FRDA) is a rare monogenic disease characterized by multisystem, slowly progressive degeneration. Because of the genetic defect in a non-coding region of FXN gene, FRDA cells exhibit severe deficit of frataxin protein levels. Hence, FRDA pathophysiology is characterized by a plethora of metabolic disruptions related to iron metabolism, mitochondrial homeostasis and oxidative stress. Importantly, an impairment of the antioxidant defences exacerbates the oxidative damage. This appears closely associated with the disablement of key antioxidant proteins, such as the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) and the mitochondrial superoxide dismutase (MnSOD). The cytokine interferon gamma (IFN-γ) has been shown to increase frataxin expression in FRDA cells and to improve functional deficits in FRDA mice. Currently, IFN-γ represents a potential therapy under clinical evaluation in FRDA patients. Here, we show that IFN-γ induces a rapid expression of Nrf2 and MnSOD in different cell types, including FRDA patient-derived fibroblasts. Our data indicate that IFN-γ signals two separate pathways to enhance Nrf2 and MnSOD levels in FRDA fibroblasts. MnSOD expression increased through an early transcriptional regulation, whereas the levels of Nrf2 are induced by a post-transcriptional mechanism. We demonstrate that the treatment of FRDA fibroblasts with IFN-γ stimulates a non-canonical Nrf2 activation pathway through p21 and potentiates antioxidant responses under exposure to hydrogen peroxide. Moreover, IFN-γ significantly reduced the sensitivity to hydrogen peroxide-induced cell death in FRDA fibroblasts. Collectively, these results indicate the presence of multiple pathways triggered by IFN-γ with therapeutic relevance to FRDA.
Collapse
Affiliation(s)
- Riccardo Luffarelli
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (R.L.); (L.P.); (A.Q.); (F.T.); (S.F.); (A.R.); (F.M.); (R.T.)
| | - Luca Panarello
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (R.L.); (L.P.); (A.Q.); (F.T.); (S.F.); (A.R.); (F.M.); (R.T.)
| | - Andrea Quatrana
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (R.L.); (L.P.); (A.Q.); (F.T.); (S.F.); (A.R.); (F.M.); (R.T.)
| | - Francesca Tiano
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (R.L.); (L.P.); (A.Q.); (F.T.); (S.F.); (A.R.); (F.M.); (R.T.)
| | - Silvia Fortuni
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (R.L.); (L.P.); (A.Q.); (F.T.); (S.F.); (A.R.); (F.M.); (R.T.)
| | - Alessandra Rufini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (R.L.); (L.P.); (A.Q.); (F.T.); (S.F.); (A.R.); (F.M.); (R.T.)
- Departmental Faculty of Medicine and Surgery, Saint Camillus International University of Health and Medical Sciences, 00131 Rome, Italy
| | - Florence Malisan
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (R.L.); (L.P.); (A.Q.); (F.T.); (S.F.); (A.R.); (F.M.); (R.T.)
| | - Roberto Testi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (R.L.); (L.P.); (A.Q.); (F.T.); (S.F.); (A.R.); (F.M.); (R.T.)
| | - Ivano Condò
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (R.L.); (L.P.); (A.Q.); (F.T.); (S.F.); (A.R.); (F.M.); (R.T.)
| |
Collapse
|
4
|
Rodden LN, Gilliam KM, Lam C, Rojsajjakul T, Mesaros C, Dionisi C, Pook M, Pandolfo M, Lynch DR, Blair IA, Bidichandani SI. DNA methylation in Friedreich ataxia silences expression of frataxin isoform E. Sci Rep 2022; 12:5031. [PMID: 35322126 PMCID: PMC8943190 DOI: 10.1038/s41598-022-09002-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 03/14/2022] [Indexed: 11/15/2022] Open
Abstract
Epigenetic silencing in Friedreich ataxia (FRDA), induced by an expanded GAA triplet-repeat in intron 1 of the FXN gene, results in deficiency of the mitochondrial protein, frataxin. A lesser known extramitochondrial isoform of frataxin detected in erythrocytes, frataxin-E, is encoded via an alternate transcript (FXN-E) originating in intron 1 that lacks a mitochondrial targeting sequence. We show that FXN-E is deficient in FRDA, including in patient-derived cell lines, iPS-derived proprioceptive neurons, and tissues from a humanized mouse model. In a series of FRDA patients, deficiency of frataxin-E protein correlated with the length of the expanded GAA triplet-repeat, and with repeat-induced DNA hypermethylation that occurs in close proximity to the intronic origin of FXN-E. CRISPR-induced epimodification to mimic DNA hypermethylation seen in FRDA reproduced FXN-E transcriptional deficiency. Deficiency of frataxin E is a consequence of FRDA-specific epigenetic silencing, and therapeutic strategies may need to address this deficiency.
Collapse
Affiliation(s)
- Layne N Rodden
- Department of Pediatrics, University of Oklahoma Health Sciences Center, OU Children's Physician Building, Suite 12100, 1200 Children's Avenue, Oklahoma City, OK, 73104, USA
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kaitlyn M Gilliam
- Department of Pediatrics, University of Oklahoma Health Sciences Center, OU Children's Physician Building, Suite 12100, 1200 Children's Avenue, Oklahoma City, OK, 73104, USA
| | - Christina Lam
- Department of Pediatrics, University of Oklahoma Health Sciences Center, OU Children's Physician Building, Suite 12100, 1200 Children's Avenue, Oklahoma City, OK, 73104, USA
| | - Teerapat Rojsajjakul
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Clementina Mesaros
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Mark Pook
- Division of Biosciences, Department of Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge, UK
| | - Massimo Pandolfo
- Université Libre de Bruxelles (ULB), Brussels, Belgium
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - David R Lynch
- Division of Neurology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ian A Blair
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sanjay I Bidichandani
- Department of Pediatrics, University of Oklahoma Health Sciences Center, OU Children's Physician Building, Suite 12100, 1200 Children's Avenue, Oklahoma City, OK, 73104, USA.
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
5
|
Rufini A, Malisan F, Condò I, Testi R. Drug Repositioning in Friedreich Ataxia. Front Neurosci 2022; 16:814445. [PMID: 35221903 PMCID: PMC8863941 DOI: 10.3389/fnins.2022.814445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 01/07/2022] [Indexed: 12/14/2022] Open
Abstract
Friedreich ataxia is a rare neurodegenerative disorder caused by insufficient levels of the essential mitochondrial protein frataxin. It is a severely debilitating disease that significantly impacts the quality of life of affected patients and reduces their life expectancy, however, an adequate cure is not yet available for patients. Frataxin function, although not thoroughly elucidated, is associated with assembly of iron-sulfur cluster and iron metabolism, therefore insufficient frataxin levels lead to reduced activity of many mitochondrial enzymes involved in the electron transport chain, impaired mitochondrial metabolism, reduced ATP production and inefficient anti-oxidant response. As a consequence, neurons progressively die and patients progressively lose their ability to coordinate movement and perform daily activities. Therapeutic strategies aim at restoring sufficient frataxin levels or at correcting some of the downstream consequences of frataxin deficiency. However, the classical pathways of drug discovery are challenging, require a significant amount of resources and time to reach the final approval, and present a high failure rate. Drug repositioning represents a viable alternative to boost the identification of a therapy, particularly for rare diseases where resources are often limited. In this review we will describe recent efforts aimed at the identification of a therapy for Friedreich ataxia through drug repositioning, and discuss the limitation of such strategies.
Collapse
Affiliation(s)
- Alessandra Rufini
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
- Fratagene Therapeutics, Rome, Italy
- Saint Camillus International University of Health and Medical Sciences, Rome, Italy
- *Correspondence: Alessandra Rufini,
| | - Florence Malisan
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
| | - Ivano Condò
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
| | - Roberto Testi
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
- Fratagene Therapeutics, Rome, Italy
| |
Collapse
|
6
|
Maio N, Rouault TA. Mammalian iron sulfur cluster biogenesis: From assembly to delivery to recipient proteins with a focus on novel targets of the chaperone and co‐chaperone proteins. IUBMB Life 2022; 74:684-704. [PMID: 35080107 PMCID: PMC10118776 DOI: 10.1002/iub.2593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/05/2021] [Accepted: 12/23/2021] [Indexed: 12/17/2022]
Affiliation(s)
- Nunziata Maio
- Molecular Medicine Branch Eunice Kennedy Shriver National Institute of Child Health and Human Development Bethesda Maryland USA
| | - Tracey A. Rouault
- Molecular Medicine Branch Eunice Kennedy Shriver National Institute of Child Health and Human Development Bethesda Maryland USA
| |
Collapse
|
7
|
Medina-Carbonero M, Sanz-Alcázar A, Britti E, Delaspre F, Cabiscol E, Ros J, Tamarit J. Mice harboring the FXN I151F pathological point mutation present decreased frataxin levels, a Friedreich ataxia-like phenotype, and mitochondrial alterations. Cell Mol Life Sci 2022; 79:74. [PMID: 35038030 PMCID: PMC8763788 DOI: 10.1007/s00018-021-04100-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 11/21/2021] [Accepted: 12/16/2021] [Indexed: 12/13/2022]
Abstract
Friedreich Ataxia (FA) is a rare neuro-cardiodegenerative disease caused by mutations in the frataxin (FXN) gene. The most prevalent mutation is a GAA expansion in the first intron of the gene causing decreased frataxin expression. Some patients present the GAA expansion in one allele and a missense mutation in the other allele. One of these mutations, FXNI154F, was reported to result in decreased content of mature frataxin and increased presence of an insoluble intermediate proteoform in cellular models. By introducing this mutation into the murine Fxn gene (I151F, equivalent to human I154F) we have now analyzed the consequences of this pathological point mutation in vivo. We have observed that FXNI151F homozygous mice present low frataxin levels in all tissues, with no evidence of insoluble proteoforms. Moreover, they display neurological deficits resembling those observed in FA patients. Biochemical analysis of heart, cerebrum and cerebellum have revealed decreased content of components from OXPHOS complexes I and II, decreased aconitase activity, and alterations in antioxidant defenses. These mitochondrial alterations are more marked in the nervous system than in heart, precede the appearance of neurological symptoms, and are similar to those observed in other FA models. We conclude that the primary pathological mechanism underlying the I151F mutation is frataxin deficiency, like in patients carrying GAA expansions. Therefore, patients carrying the I154F mutation would benefit from frataxin replacement therapies. Furthermore, our results also show that the FXNI151F mouse is an excellent tool for analyzing tissue-specific consequences of frataxin deficiency and for testing new therapies.
Collapse
Affiliation(s)
- Marta Medina-Carbonero
- Dept. Ciències Mèdiques Bàsiques, Fac. Medicina, IRBLleida, Universitat de Lleida, Av. Rovira Roure, 80, 25198, Lleida, Spain
| | - Arabela Sanz-Alcázar
- Dept. Ciències Mèdiques Bàsiques, Fac. Medicina, IRBLleida, Universitat de Lleida, Av. Rovira Roure, 80, 25198, Lleida, Spain
| | - Elena Britti
- Dept. Ciències Mèdiques Bàsiques, Fac. Medicina, IRBLleida, Universitat de Lleida, Av. Rovira Roure, 80, 25198, Lleida, Spain
| | - Fabien Delaspre
- Dept. Ciències Mèdiques Bàsiques, Fac. Medicina, IRBLleida, Universitat de Lleida, Av. Rovira Roure, 80, 25198, Lleida, Spain
| | - Elisa Cabiscol
- Dept. Ciències Mèdiques Bàsiques, Fac. Medicina, IRBLleida, Universitat de Lleida, Av. Rovira Roure, 80, 25198, Lleida, Spain
| | - Joaquim Ros
- Dept. Ciències Mèdiques Bàsiques, Fac. Medicina, IRBLleida, Universitat de Lleida, Av. Rovira Roure, 80, 25198, Lleida, Spain
| | - Jordi Tamarit
- Dept. Ciències Mèdiques Bàsiques, Fac. Medicina, IRBLleida, Universitat de Lleida, Av. Rovira Roure, 80, 25198, Lleida, Spain.
| |
Collapse
|
8
|
Shah S, Dooms MM, Amaral-Garcia S, Igoillo-Esteve M. Current Drug Repurposing Strategies for Rare Neurodegenerative Disorders. Front Pharmacol 2022; 12:768023. [PMID: 34992533 PMCID: PMC8724568 DOI: 10.3389/fphar.2021.768023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/10/2021] [Indexed: 12/12/2022] Open
Abstract
Rare diseases are life-threatening or chronically debilitating low-prevalent disorders caused by pathogenic mutations or particular environmental insults. Due to their high complexity and low frequency, important gaps still exist in their prevention, diagnosis, and treatment. Since new drug discovery is a very costly and time-consuming process, leading pharmaceutical companies show relatively low interest in orphan drug research and development due to the high cost of investments compared to the low market return of the product. Drug repurposing–based approaches appear then as cost- and time-saving strategies for the development of therapeutic opportunities for rare diseases. In this article, we discuss the scientific, regulatory, and economic aspects of the development of repurposed drugs for the treatment of rare neurodegenerative disorders with a particular focus on Huntington’s disease, Friedreich’s ataxia, Wolfram syndrome, and amyotrophic lateral sclerosis. The role of academia, pharmaceutical companies, patient associations, and foundations in the identification of candidate compounds and their preclinical and clinical evaluation will also be discussed.
Collapse
Affiliation(s)
- Sweta Shah
- Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | | | | | | |
Collapse
|
9
|
Sanchez Caballero L, Gorgogietas V, Arroyo MN, Igoillo-Esteve M. Molecular mechanisms of β-cell dysfunction and death in monogenic forms of diabetes. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 359:139-256. [PMID: 33832649 DOI: 10.1016/bs.ircmb.2021.02.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Monogenetic forms of diabetes represent 1%-5% of all diabetes cases and are caused by mutations in a single gene. These mutations, that affect genes involved in pancreatic β-cell development, function and survival, or insulin regulation, may be dominant or recessive, inherited or de novo. Most patients with monogenic diabetes are very commonly misdiagnosed as having type 1 or type 2 diabetes. The severity of their symptoms depends on the nature of the mutation, the function of the affected gene and, in some cases, the influence of additional genetic or environmental factors that modulate severity and penetrance. In some patients, diabetes is accompanied by other syndromic features such as deafness, blindness, microcephaly, liver and intestinal defects, among others. The age of diabetes onset may also vary from neonatal until early adulthood manifestations. Since the different mutations result in diverse clinical presentations, patients usually need different treatments that range from just diet and exercise, to the requirement of exogenous insulin or other hypoglycemic drugs, e.g., sulfonylureas or glucagon-like peptide 1 analogs to control their glycemia. As a consequence, awareness and correct diagnosis are crucial for the proper management and treatment of monogenic diabetes patients. In this chapter, we describe mutations causing different monogenic forms of diabetes associated with inadequate pancreas development or impaired β-cell function and survival, and discuss the molecular mechanisms involved in β-cell demise.
Collapse
Affiliation(s)
- Laura Sanchez Caballero
- ULB Center for Diabetes Research (UCDR), Université Libre de Bruxelles, Brussels, Belgium. http://www.ucdr.be/
| | - Vyron Gorgogietas
- ULB Center for Diabetes Research (UCDR), Université Libre de Bruxelles, Brussels, Belgium. http://www.ucdr.be/
| | - Maria Nicol Arroyo
- ULB Center for Diabetes Research (UCDR), Université Libre de Bruxelles, Brussels, Belgium. http://www.ucdr.be/
| | - Mariana Igoillo-Esteve
- ULB Center for Diabetes Research (UCDR), Université Libre de Bruxelles, Brussels, Belgium. http://www.ucdr.be/.
| |
Collapse
|
10
|
Pallardó FV, Pagano G, Rodríguez LR, Gonzalez-Cabo P, Lyakhovich A, Trifuoggi M. Friedreich Ataxia: current state-of-the-art, and future prospects for mitochondrial-focused therapies. Transl Res 2021; 229:135-141. [PMID: 32841735 DOI: 10.1016/j.trsl.2020.08.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 12/21/2022]
Abstract
Friedreich's Ataxia is an autosomal recessive genetic disease causing the defective gene product, frataxin. A body of literature has been focused on the attempts to counteract frataxin deficiency and the consequent iron imbalance, in order to mitigate the disease-associated pro-oxidant state and clinical course. The present mini review is aimed at evaluating the basic and clinical reports on the roles and the use of a set of iron chelators, antioxidants and some cofactors involved in the key mitochondrial functions. Extensive literature has focused on the protective roles of iron chelators, coenzyme Q10 and analogs, and vitamin E, altogether with varying outcomes in clinical studies. Other studies have suggested mitoprotective roles for other mitochondrial cofactors, involved in Krebs cycle, such as alpha-lipoic acid and carnitine, involved in acyl transport across the mitochondrial membrane. A body of evidence points to the strong antioxidant properties of these cofactors, and to their potential contribution in mitoprotective strategies in Friedreich's Ataxia clinical evolution. Thus, we suggest the rationale for planning combination strategies based on the 3 mitochondrial cofactors and of some antioxidants and iron binders as mitoprotective cocktails in Friedreich Ataxia patients, calling attention to clinical practitioners of the importance to implement clinical trials.
Collapse
Affiliation(s)
- Federico V Pallardó
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia-INCLIVA, Valencia, Spain; CIBER de Enfermedades Raras (CIBERER), Valencia, Spain.
| | - Giovanni Pagano
- Department of Chemical Sciences, Federico II Naples University, Naples, Italy
| | - Laura R Rodríguez
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia-INCLIVA, Valencia, Spain
| | - Pilar Gonzalez-Cabo
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia-INCLIVA, Valencia, Spain; CIBER de Enfermedades Raras (CIBERER), Valencia, Spain
| | - Alex Lyakhovich
- Vall d'Hebron Institut de Recerca, Barcelona, Catalunya, Spain; Institute of Molecular Biology and Biophysics of the "Federal Research Center of Fundamental and Translational Medicine", Novosibirsk, Russia
| | - Marco Trifuoggi
- Department of Chemical Sciences, Federico II Naples University, Naples, Italy
| |
Collapse
|
11
|
Tamarit J, Britti E, Delaspre F, Medina-Carbonero M, Sanz-Alcázar A, Cabiscol E, Ros J. Mitochondrial iron and calcium homeostasis in Friedreich ataxia. IUBMB Life 2021; 73:543-553. [PMID: 33675183 DOI: 10.1002/iub.2457] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/08/2021] [Accepted: 02/08/2021] [Indexed: 12/13/2022]
Abstract
Friedreich Ataxia is a neuro-cardiodegenerative disease caused by the deficiency of frataxin, a mitochondrial protein. Many evidences indicate that frataxin deficiency causes an unbalance of iron homeostasis. Nevertheless, in the last decade many results also highlighted the importance of calcium unbalance in the deleterious downstream effects caused by frataxin deficiency. In this review, the role of these two metals has been gathered to give a whole view of how iron and calcium dyshomeostasys impacts on cellular functions and, as a result, which strategies can be followed to find an effective therapy for the disease.
Collapse
Affiliation(s)
- Jordi Tamarit
- Dept. Ciències Mèdiques Bàsiques, Universitat de Lleida, IRBLleida, Lleida, Spain
| | - Elena Britti
- Dept. Ciències Mèdiques Bàsiques, Universitat de Lleida, IRBLleida, Lleida, Spain
| | - Fabien Delaspre
- Dept. Ciències Mèdiques Bàsiques, Universitat de Lleida, IRBLleida, Lleida, Spain
| | | | - Arabela Sanz-Alcázar
- Dept. Ciències Mèdiques Bàsiques, Universitat de Lleida, IRBLleida, Lleida, Spain
| | - Elisa Cabiscol
- Dept. Ciències Mèdiques Bàsiques, Universitat de Lleida, IRBLleida, Lleida, Spain
| | - Joaquim Ros
- Dept. Ciències Mèdiques Bàsiques, Universitat de Lleida, IRBLleida, Lleida, Spain
| |
Collapse
|
12
|
Rodríguez LR, Calap-Quintana P, Lapeña-Luzón T, Pallardó FV, Schneuwly S, Navarro JA, Gonzalez-Cabo P. Oxidative stress modulates rearrangement of endoplasmic reticulum-mitochondria contacts and calcium dysregulation in a Friedreich's ataxia model. Redox Biol 2020; 37:101762. [PMID: 33128998 PMCID: PMC7585950 DOI: 10.1016/j.redox.2020.101762] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 10/07/2020] [Accepted: 10/12/2020] [Indexed: 12/13/2022] Open
Abstract
Friedreich ataxia (FRDA) is a neurodegenerative disorder characterized by neuromuscular and neurological manifestations. It is caused by mutations in the FXN gene, which results in loss of the mitochondrial protein frataxin. Endoplasmic Reticulum-mitochondria associated membranes (MAMs) are inter-organelle structures involved in the regulation of essential cellular processes, including lipid metabolism and calcium signaling. In the present study, we have analyzed in both, unicellular and multicellular models of FRDA, calcium management and integrity of MAMs. We observed that function of MAMs is compromised in our cellular model of FRDA, which was improved upon treatment with antioxidants. In agreement, promoting mitochondrial calcium uptake was sufficient to restore several defects caused by frataxin deficiency in Drosophila Melanogaster. Remarkably, our findings describe for the first time frataxin as a member of the protein network of MAMs, where interacts with two of the main proteins implicated in endoplasmic reticulum-mitochondria communication. These results suggest a new role of frataxin, indicate that FRDA goes beyond mitochondrial defects and highlight MAMs as novel therapeutic candidates to improve patient's conditions.
Collapse
Affiliation(s)
- Laura R Rodríguez
- Department of Physiology, Faculty of Medicine and Dentistry. University of Valencia-INCLIVA, Valencia, 46010, Spain; Associated Unit for Rare Diseases INCLIVA-CIPF, Valencia, Spain
| | - Pablo Calap-Quintana
- Department of Physiology, Faculty of Medicine and Dentistry. University of Valencia-INCLIVA, Valencia, 46010, Spain; Associated Unit for Rare Diseases INCLIVA-CIPF, Valencia, Spain; CIBER de Enfermedades Raras (CIBERER), Valencia, Spain
| | - Tamara Lapeña-Luzón
- Department of Physiology, Faculty of Medicine and Dentistry. University of Valencia-INCLIVA, Valencia, 46010, Spain; Associated Unit for Rare Diseases INCLIVA-CIPF, Valencia, Spain; CIBER de Enfermedades Raras (CIBERER), Valencia, Spain
| | - Federico V Pallardó
- Department of Physiology, Faculty of Medicine and Dentistry. University of Valencia-INCLIVA, Valencia, 46010, Spain; Associated Unit for Rare Diseases INCLIVA-CIPF, Valencia, Spain; CIBER de Enfermedades Raras (CIBERER), Valencia, Spain
| | - Stephan Schneuwly
- Institute of Zoology, Universitaetsstrasse 31, University of Regensburg, 93040, Regensburg, Germany
| | - Juan A Navarro
- Institute of Zoology, Universitaetsstrasse 31, University of Regensburg, 93040, Regensburg, Germany; INCLIVA Biomedial Research Institute, Valencia, Spain
| | - Pilar Gonzalez-Cabo
- Department of Physiology, Faculty of Medicine and Dentistry. University of Valencia-INCLIVA, Valencia, 46010, Spain; Associated Unit for Rare Diseases INCLIVA-CIPF, Valencia, Spain; CIBER de Enfermedades Raras (CIBERER), Valencia, Spain.
| |
Collapse
|
13
|
Weng L, Laboureur L, Wang Q, Guo L, Xu P, Gottlieb L, Lynch DR, Mesaros C, Blair IA. Extra-mitochondrial mouse frataxin and its implications for mouse models of Friedreich's ataxia. Sci Rep 2020; 10:15788. [PMID: 32978498 PMCID: PMC7519113 DOI: 10.1038/s41598-020-72884-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 08/28/2020] [Indexed: 01/04/2023] Open
Abstract
Mature frataxin is essential for the assembly of iron-sulfur cluster proteins including a number of mitochondrial enzymes. Reduced levels of mature frataxin (81-20) in human subjects caused by the genetic disease Friedreich's ataxia results in decreased mitochondrial function, neurodegeneration, and cardiomyopathy. Numerous studies of mitochondrial dysfunction have been conducted using mouse models of frataxin deficiency. However, mouse frataxin that is reduced in these models, is assumed to be mature frataxin (78-207) by analogy with human mature frataxin (81-210). Using immunoaffinity purification coupled with liquid chromatography-high resolution tandem mass spectrometry, we have discovered that mature frataxin in mouse heart (77%), brain (86%), and liver (47%) is predominantly a 129-amino acid truncated mature frataxin (79-207) in which the N-terminal lysine residue has been lost. Mature mouse frataxin (78-207) only contributes 7-15% to the total frataxin protein present in mouse tissues. We have also found that truncated mature frataxin (79-207) is present primarily in the cytosol of mouse liver; whereas, frataxin (78-207) is primarily present in the mitochondria. These findings, which provide support for the role of extra-mitochondrial frataxin in the etiology of Friedreich's ataxia, also have important implications for studies of mitochondrial dysfunction conducted in mouse models of frataxin deficiency.
Collapse
Affiliation(s)
- Liwei Weng
- Penn Medicine/CHOP Center of Excellence in Friedreich's Ataxia, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Laurent Laboureur
- Penn Medicine/CHOP Center of Excellence in Friedreich's Ataxia, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Qingqing Wang
- Penn Medicine/CHOP Center of Excellence in Friedreich's Ataxia, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Lili Guo
- Penn Medicine/CHOP Center of Excellence in Friedreich's Ataxia, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Peining Xu
- Penn Medicine/CHOP Center of Excellence in Friedreich's Ataxia, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Leah Gottlieb
- Penn Medicine/CHOP Center of Excellence in Friedreich's Ataxia, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - David R Lynch
- Penn Medicine/CHOP Center of Excellence in Friedreich's Ataxia, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Departments of Pediatrics and Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Clementina Mesaros
- Penn Medicine/CHOP Center of Excellence in Friedreich's Ataxia, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ian A Blair
- Penn Medicine/CHOP Center of Excellence in Friedreich's Ataxia, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
14
|
Bon C, Luffarelli R, Russo R, Fortuni S, Pierattini B, Santulli C, Fimiani C, Persichetti F, Cotella D, Mallamaci A, Santoro C, Carninci P, Espinoza S, Testi R, Zucchelli S, Condò I, Gustincich S. SINEUP non-coding RNAs rescue defective frataxin expression and activity in a cellular model of Friedreich's Ataxia. Nucleic Acids Res 2019; 47:10728-10743. [PMID: 31584077 PMCID: PMC6847766 DOI: 10.1093/nar/gkz798] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 08/08/2019] [Accepted: 09/28/2019] [Indexed: 12/16/2022] Open
Abstract
Friedreich's ataxia (FRDA) is an untreatable disorder with neuro- and cardio-degenerative progression. This monogenic disease is caused by the hyper-expansion of naturally occurring GAA repeats in the first intron of the FXN gene, encoding for frataxin, a protein implicated in the biogenesis of iron-sulfur clusters. As the genetic defect interferes with FXN transcription, FRDA patients express a normal frataxin protein but at insufficient levels. Thus, current therapeutic strategies are mostly aimed to restore physiological FXN expression. We have previously described SINEUPs, natural and synthetic antisense long non-coding RNAs, which promote translation of partially overlapping mRNAs through the activity of an embedded SINEB2 domain. Here, by in vitro screening, we have identified a number of SINEUPs targeting human FXN mRNA and capable to up-regulate frataxin protein to physiological amounts acting at the post-transcriptional level. Furthermore, FXN-specific SINEUPs promote the recovery of disease-associated mitochondrial aconitase defects in FRDA-derived cells. In summary, we provide evidence that SINEUPs may be the first gene-specific therapeutic approach to activate FXN translation in FRDA and, more broadly, a novel scalable platform to develop new RNA-based therapies for haploinsufficient diseases.
Collapse
Affiliation(s)
- Carlotta Bon
- Central RNA Laboratory, Istituto Italiano di Tecnologia (IIT), Genova, Italy
- Area of Neuroscience, International School for Advanced Studies (SISSA), Italy
| | - Riccardo Luffarelli
- Department of Biomedicine and Prevention, Laboratory of Signal Transduction, University of Rome Tor Vergata, Rome, Italy
| | - Roberta Russo
- Area of Neuroscience, International School for Advanced Studies (SISSA), Italy
| | - Silvia Fortuni
- Department of Biomedicine and Prevention, Laboratory of Signal Transduction, University of Rome Tor Vergata, Rome, Italy
| | - Bianca Pierattini
- Central RNA Laboratory, Istituto Italiano di Tecnologia (IIT), Genova, Italy
- Area of Neuroscience, International School for Advanced Studies (SISSA), Italy
| | - Chiara Santulli
- Area of Neuroscience, International School for Advanced Studies (SISSA), Italy
| | - Cristina Fimiani
- Area of Neuroscience, International School for Advanced Studies (SISSA), Italy
| | - Francesca Persichetti
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale (UPO), Novara, Italy
| | - Diego Cotella
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale (UPO), Novara, Italy
| | - Antonello Mallamaci
- Area of Neuroscience, International School for Advanced Studies (SISSA), Italy
| | - Claudio Santoro
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale (UPO), Novara, Italy
| | - Piero Carninci
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Yokohama, Kanagawa, Japan
| | - Stefano Espinoza
- Central RNA Laboratory, Istituto Italiano di Tecnologia (IIT), Genova, Italy
| | - Roberto Testi
- Department of Biomedicine and Prevention, Laboratory of Signal Transduction, University of Rome Tor Vergata, Rome, Italy
| | - Silvia Zucchelli
- Area of Neuroscience, International School for Advanced Studies (SISSA), Italy
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale (UPO), Novara, Italy
| | - Ivano Condò
- Department of Biomedicine and Prevention, Laboratory of Signal Transduction, University of Rome Tor Vergata, Rome, Italy
| | - Stefano Gustincich
- Central RNA Laboratory, Istituto Italiano di Tecnologia (IIT), Genova, Italy
- Area of Neuroscience, International School for Advanced Studies (SISSA), Italy
| |
Collapse
|
15
|
Is brain iron trafficking part of the physiology of the amyloid precursor protein? J Biol Inorg Chem 2019; 24:1171-1177. [PMID: 31578640 DOI: 10.1007/s00775-019-01684-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 06/19/2019] [Indexed: 12/11/2022]
Abstract
The amyloid precursor protein is so named, because a proteolytic fragment of it was found associated with a neuropathic disorder now known as Alzheimer's disease. This fragment, Aβ, along with tau makes up the plaques and tangles that are the hallmark of AD. Iron (and other first-row transition metals) is found associated with these proteinaceous deposits. Much research has focused on the relationship of the plaques and iron to the etiology of the disease. This commentary asks another question, one only more recently addressed namely, what is the physiologic function of the amyloid precursor protein (APP) and of its secretase-generated soluble species? Overall, the data make clear that APP and its products have neurotrophic functions and some data indicate one of these may be to modulate the trafficking of iron in the brain.
Collapse
|
16
|
Han THL, Camadro JM, Barbault F, Santos R, El Hage Chahine JM, Ha-Duong NT. In Vitro interaction between yeast frataxin and superoxide dismutases: Influence of mitochondrial metals. Biochim Biophys Acta Gen Subj 2019; 1863:883-892. [PMID: 30797804 DOI: 10.1016/j.bbagen.2019.02.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 02/15/2019] [Accepted: 02/19/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND Friedreich's ataxia results from a decreased expression of the nuclear gene encoding the mitochondrial protein, frataxin. Frataxin participates in the biosynthesis of iron-sulfur clusters and heme cofactors, as well as in iron storage and protection against oxidative stress. How frataxin interacts with the antioxidant defence components is poorly understood. METHODS Therefore, we have investigated by kinetic, thermodynamic and modelling approaches the molecular interactions between yeast frataxin (Yfh1) and superoxide dismutases, Sod1 and Sod2, and the influence of Yfh1 on their enzymatic activities. RESULTS Yfh1 interacts with cytosolic Sod1 with a dissociation constant, Kd = 1.3 ± 0.3 μM, in two kinetic steps. The first step occurs in the 200 ms range and corresponds to the Yfh1-Sod1 interaction, whereas the second is slow and is assumed to be a change in the conformation of the protein-protein adduct. Furthermore, computational investigations confirm the stability of the Yfh1-Sod1 complex. Yfh1 forms two protein complexes with mitochondrial Sod2 with 1:1 and 2:1 Yfh1/Sod2 stoichiometry (Kd1 = 1.05 ± 0.05 and Kd2 = 6.6 ± 0.1 μM). Furthermore, Yfh1 increases the enzymatic activity of Sod1 while slightly affecting that of Sod2. Finally, the stabilities of the protein-protein adducts and the effect of Yfh1 on superoxide dismutase activities depend on the nature of the mitochondrial metal. CONCLUSIONS This work confirms the participation of Yfh1 in cellular defence against oxidative stress.
Collapse
Affiliation(s)
- Thi Hong Lien Han
- Interfaces, Traitements, Organisation et Dynamique des Systèmes (ITODYS), CNRS UMR 7086, Univ Paris Diderot, Sorbonne Paris Cité, 15 rue Jean-Antoine de Baïf, F-75205 Paris Cedex 13, France
| | - Jean-Michel Camadro
- Mitochondries, Métaux et Stress Oxydant, Institut Jacques Monod, CNRS UMR 7592, Université Paris Diderot, Sorbonne Paris Cité, 15 rue Hélène Brion, F-75205 Paris Cedex 13, France
| | - Florent Barbault
- Interfaces, Traitements, Organisation et Dynamique des Systèmes (ITODYS), CNRS UMR 7086, Univ Paris Diderot, Sorbonne Paris Cité, 15 rue Jean-Antoine de Baïf, F-75205 Paris Cedex 13, France
| | - Renata Santos
- Mitochondries, Métaux et Stress Oxydant, Institut Jacques Monod, CNRS UMR 7592, Université Paris Diderot, Sorbonne Paris Cité, 15 rue Hélène Brion, F-75205 Paris Cedex 13, France
| | - Jean-Michel El Hage Chahine
- Interfaces, Traitements, Organisation et Dynamique des Systèmes (ITODYS), CNRS UMR 7086, Univ Paris Diderot, Sorbonne Paris Cité, 15 rue Jean-Antoine de Baïf, F-75205 Paris Cedex 13, France
| | - Nguyet-Thanh Ha-Duong
- Interfaces, Traitements, Organisation et Dynamique des Systèmes (ITODYS), CNRS UMR 7086, Univ Paris Diderot, Sorbonne Paris Cité, 15 rue Jean-Antoine de Baïf, F-75205 Paris Cedex 13, France.
| |
Collapse
|
17
|
Alfedi G, Luffarelli R, Condò I, Pedini G, Mannucci L, Massaro DS, Benini M, Toschi N, Alaimo G, Panarello L, Pacini L, Fortuni S, Serio D, Malisan F, Testi R, Rufini A. Drug repositioning screening identifies etravirine as a potential therapeutic for friedreich's ataxia. Mov Disord 2019; 34:323-334. [PMID: 30624801 DOI: 10.1002/mds.27604] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 10/31/2018] [Accepted: 11/05/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Friedreich's ataxia is an autosomal-recessive cerebellar ataxia caused by mutation of the frataxin gene, resulting in decreased frataxin expression, mitochondrial dysfunction, and oxidative stress. Currently, no treatment is available for Friedreich's ataxia patients. Given that levels of residual frataxin critically affect disease severity, the main goal of a specific therapy for Friedreich's ataxia is to increase frataxin levels. OBJECTIVES With the aim to accelerate the development of a new therapy for Friedreich's ataxia, we took a drug repositioning approach to identify market-available drugs able to increase frataxin levels. METHODS Using a cell-based reporter assay to monitor variation in frataxin amount, we performed a high-throughput screening of a library containing 853 U.S. Food and Drug Administration-approved drugs. RESULTS Among the potentially interesting candidates isolated from the screening, we focused our attention on etravirine, an antiviral drug currently in use as an anti-human immunodeficiency virus therapy. Here, we show that etravirine can promote a significant increase in frataxin levels in cells derived from Friedreich's ataxia patients, by enhancing frataxin messenger RNA translation. Importantly, frataxin accumulation in treated patient cell lines is comparable to frataxin levels in unaffected carrier cells, suggesting that etravirine could be therapeutically relevant. Indeed, etravirine treatment restores the activity of the iron-sulphur cluster containing enzyme aconitase and confers resistance to oxidative stress in cells derived from Friedreich's ataxia patients. CONCLUSIONS Considering its excellent safety profile along with its ability to increase frataxin levels and correct some of the disease-related defects, etravirine represents a promising candidate as a therapeutic for Friedreich's ataxia. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Giulia Alfedi
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Riccardo Luffarelli
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Ivano Condò
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Giorgia Pedini
- Laboratory of Molecular Neurobiology, Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Liliana Mannucci
- Laboratory of Molecular Neurobiology, Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Damiano S Massaro
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Monica Benini
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
- Fratagene Therapeutics Srl, Rome, Italy
| | - Nicola Toschi
- Medical Physics Section, Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging and Harvard Medical School, Boston, Massachusetts, USA
| | - Giorgia Alaimo
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
- Fratagene Therapeutics Srl, Rome, Italy
| | - Luca Panarello
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Laura Pacini
- Laboratory of Molecular Neurobiology, Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Silvia Fortuni
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Dario Serio
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Florence Malisan
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Roberto Testi
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
- Fratagene Therapeutics Srl, Rome, Italy
| | - Alessandra Rufini
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
- Fratagene Therapeutics Srl, Rome, Italy
| |
Collapse
|
18
|
Llorens JV, Soriano S, Calap-Quintana P, Gonzalez-Cabo P, Moltó MD. The Role of Iron in Friedreich's Ataxia: Insights From Studies in Human Tissues and Cellular and Animal Models. Front Neurosci 2019; 13:75. [PMID: 30833885 PMCID: PMC6387962 DOI: 10.3389/fnins.2019.00075] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 01/23/2019] [Indexed: 12/12/2022] Open
Abstract
Friedreich's ataxia (FRDA) is a rare early-onset degenerative disease that affects both the central and peripheral nervous systems, and other extraneural tissues, mainly the heart and endocrine pancreas. This disorder progresses as a mixed sensory and cerebellar ataxia, primarily disturbing the proprioceptive pathways in the spinal cord, peripheral nerves and nuclei of the cerebellum. FRDA is an inherited disease with an autosomal recessive pattern caused by an insufficient amount of the nuclear-encoded mitochondrial protein frataxin, which is an essential and highly evolutionary conserved protein whose deficit results in iron metabolism dysregulation and mitochondrial dysfunction. The first experimental evidence connecting frataxin with iron homeostasis came from Saccharomyces cerevisiae; iron accumulates in the mitochondria of yeast with deletion of the frataxin ortholog gene. This finding was soon linked to previous observations of iron deposits in the hearts of FRDA patients and was later reported in animal models of the disease. Despite advances made in the understanding of FRDA pathophysiology, the role of iron in this disease has not yet been completely clarified. Some of the questions still unresolved include the molecular mechanisms responsible for the iron accumulation and iron-mediated toxicity. Here, we review the contribution of the cellular and animal models of FRDA and relevance of the studies using FRDA patient samples to gain knowledge about these issues. Mechanisms of mitochondrial iron overload are discussed considering the potential roles of frataxin in the major mitochondrial metabolic pathways that use iron. We also analyzed the effect of iron toxicity on neuronal degeneration in FRDA by reactive oxygen species (ROS)-dependent and ROS-independent mechanisms. Finally, therapeutic strategies based on the control of iron toxicity are considered.
Collapse
Affiliation(s)
- José Vicente Llorens
- Department of Genetics, Faculty of Biological Sciences, University of Valencia, Valencia, Spain
- Unit for Psychiatry and Neurodegenerative Diseases, Biomedical Research Institute INCLIVA, Valencia, Spain
| | - Sirena Soriano
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
| | - Pablo Calap-Quintana
- Department of Genetics, Faculty of Biological Sciences, University of Valencia, Valencia, Spain
- Unit for Psychiatry and Neurodegenerative Diseases, Biomedical Research Institute INCLIVA, Valencia, Spain
| | - Pilar Gonzalez-Cabo
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain
- Center of Biomedical Network Research on Rare Diseases CIBERER, Valencia, Spain
- Associated Unit for Rare Diseases INCLIVA-CIPF, Biomedical Research Institute INCLIVA, Valencia, Spain
| | - María Dolores Moltó
- Department of Genetics, Faculty of Biological Sciences, University of Valencia, Valencia, Spain
- Unit for Psychiatry and Neurodegenerative Diseases, Biomedical Research Institute INCLIVA, Valencia, Spain
- Center of Biomedical Network Research on Mental Health CIBERSAM, Valencia, Spain
| |
Collapse
|
19
|
Abstract
Friedreich's ataxia (FRDA) is a degenerative disease that affects both the central and the peripheral nervous systems and non-neural tissues including, mainly, heart, and endocrine pancreas. It is an autosomal recessive disease caused by a GAA triplet-repeat localized within an Alu sequence element in intron 1 of frataxin (FXN) gene, which encodes a mitochondrial protein FXN. This protein is essential for mitochondrial function by the involvement of iron-sulfur cluster biogenesis. The effects of its deficiency also include disruption of cellular, particularly mitochondrial, iron homeostasis, i.e., relatively more iron accumulated in mitochondria and less iron presented in cytosol. Though iron toxicity is commonly thought to be mediated via Fenton reaction, oxidative stress seems not to be the main problem to result in detrimental effects on cell survival, particularly neuron survival. Therefore, the basic research on FXN function is urgently demanded to understand the disease. This chapter focuses on the outcome of FXN expression, regulation, and function in cellular or animal models of FRDA and on iron pathophysiology in the affected tissues. Finally, therapeutic strategies based on the control of iron toxicity and iron cellular redistribution are considered. The combination of multiple therapeutic targets including iron, oxidative stress, mitochondrial function, and FXN regulation is also proposed.
Collapse
Affiliation(s)
- Kuanyu Li
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, People's Republic of China.
| |
Collapse
|
20
|
Guo L, Wang Q, Weng L, Hauser LA, Strawser CJ, Mesaros C, Lynch DR, Blair IA. Characterization of a new N-terminally acetylated extra-mitochondrial isoform of frataxin in human erythrocytes. Sci Rep 2018; 8:17043. [PMID: 30451920 PMCID: PMC6242848 DOI: 10.1038/s41598-018-35346-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 11/02/2018] [Indexed: 01/18/2023] Open
Abstract
Frataxin is a highly conserved protein encoded by the frataxin (FXN) gene. The full-length 210-amino acid form of protein frataxin (1-210; isoform A) expressed in the cytosol of cells rapidly translocates to the mitochondria, where it is converted to the mature form (81-210) by mitochondrial processing peptidase. Mature frataxin (81-210) is a critically important protein because it facilitates the assembly of mitochondrial iron-sulfur cluster protein complexes such as aconitase, lipoate synthase, and succinate dehydrogenases. Decreased expression of frataxin protein is responsible for the devastating rare genetic disease of Friedreich's ataxia. The mitochondrial form of frataxin has long been thought to be present in erythrocytes even though paradoxically, erythrocytes lack mitochondria. We have discovered that erythrocyte frataxin is in fact a novel isoform of frataxin (isoform E) with 135-amino acids and an N-terminally acetylated methionine residue. There is three times as much isoform E in erythrocytes (20.9 ± 6.4 ng/mL) from the whole blood of healthy volunteers (n = 10) when compared with the mature mitochondrial frataxin present in other blood cells (7.1 ± 1.0 ng/mL). Isoform E lacks a mitochondrial targeting sequence and so is distributed to both cytosol and the nucleus when expressed in cultured cells. When extra-mitochondrial frataxin isoform E is expressed in HEK 293 cells, it is converted to a shorter isoform identical to the mature frataxin found in mitochondria, which raises the possibility that it is involved in disease etiology. The ability to specifically quantify extra-mitochondrial and mitochondrial isoforms of frataxin in whole blood will make it possible to readily follow the natural history of diseases such as Friedreich's ataxia and monitor the efficacy of therapeutic interventions.
Collapse
Affiliation(s)
- Lili Guo
- Penn SRP Center and Center of Excellence in Environmental Toxicology Center, Department of Systems Pharmacology and Translational Therapeutics Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
- Penn/CHOP Center of Excellence in Friedreich's ataxia, Philadelphia, PA, 19104, United States
| | - Qingqing Wang
- Penn SRP Center and Center of Excellence in Environmental Toxicology Center, Department of Systems Pharmacology and Translational Therapeutics Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
- Penn/CHOP Center of Excellence in Friedreich's ataxia, Philadelphia, PA, 19104, United States
| | - Liwei Weng
- Penn SRP Center and Center of Excellence in Environmental Toxicology Center, Department of Systems Pharmacology and Translational Therapeutics Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Lauren A Hauser
- Penn/CHOP Center of Excellence in Friedreich's ataxia, Philadelphia, PA, 19104, United States
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, United States
- Departments of Pediatrics and Neurology Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Cassandra J Strawser
- Penn/CHOP Center of Excellence in Friedreich's ataxia, Philadelphia, PA, 19104, United States
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, United States
- Departments of Pediatrics and Neurology Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Clementina Mesaros
- Penn SRP Center and Center of Excellence in Environmental Toxicology Center, Department of Systems Pharmacology and Translational Therapeutics Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
- Penn/CHOP Center of Excellence in Friedreich's ataxia, Philadelphia, PA, 19104, United States
| | - David R Lynch
- Penn/CHOP Center of Excellence in Friedreich's ataxia, Philadelphia, PA, 19104, United States
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, United States
- Departments of Pediatrics and Neurology Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Ian A Blair
- Penn SRP Center and Center of Excellence in Environmental Toxicology Center, Department of Systems Pharmacology and Translational Therapeutics Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States.
- Penn/CHOP Center of Excellence in Friedreich's ataxia, Philadelphia, PA, 19104, United States.
| |
Collapse
|
21
|
Alsina D, Purroy R, Ros J, Tamarit J. Iron in Friedreich Ataxia: A Central Role in the Pathophysiology or an Epiphenomenon? Pharmaceuticals (Basel) 2018; 11:E89. [PMID: 30235822 PMCID: PMC6161073 DOI: 10.3390/ph11030089] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 09/15/2018] [Accepted: 09/17/2018] [Indexed: 12/16/2022] Open
Abstract
Friedreich ataxia is a neurodegenerative disease with an autosomal recessive inheritance. In most patients, the disease is caused by the presence of trinucleotide GAA expansions in the first intron of the frataxin gene. These expansions cause the decreased expression of this mitochondrial protein. Many evidences indicate that frataxin deficiency causes the deregulation of cellular iron homeostasis. In this review, we will discuss several hypotheses proposed for frataxin function, their caveats, and how they could provide an explanation for the deregulation of iron homeostasis found in frataxin-deficient cells. We will also focus on the potential mechanisms causing cellular dysfunction in Friedreich Ataxia and on the potential use of the iron chelator deferiprone as a therapeutic agent for this disease.
Collapse
Affiliation(s)
- David Alsina
- Departament de Ciències Mèdiques Bàsiques, IRBLleida, Universitat de Lleida, 25198 Lleida, Spain.
| | - Rosa Purroy
- Departament de Ciències Mèdiques Bàsiques, IRBLleida, Universitat de Lleida, 25198 Lleida, Spain.
| | - Joaquim Ros
- Departament de Ciències Mèdiques Bàsiques, IRBLleida, Universitat de Lleida, 25198 Lleida, Spain.
| | - Jordi Tamarit
- Departament de Ciències Mèdiques Bàsiques, IRBLleida, Universitat de Lleida, 25198 Lleida, Spain.
| |
Collapse
|
22
|
Abeti R, Baccaro A, Esteras N, Giunti P. Novel Nrf2-Inducer Prevents Mitochondrial Defects and Oxidative Stress in Friedreich's Ataxia Models. Front Cell Neurosci 2018; 12:188. [PMID: 30065630 PMCID: PMC6056642 DOI: 10.3389/fncel.2018.00188] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 06/11/2018] [Indexed: 12/30/2022] Open
Abstract
Friedreich’s Ataxia (FRDA) is an autosomal recessive neurodegenerative disorder, affecting dorsal root ganglia (DRG), cerebellar dentate nuclei and heart. It is caused by a GAA repeat expansion mutation within the frataxin gene (FXN). This impedes FXN transcription resulting in a progressive decrease of the mitochondrial protein, frataxin. Increased oxidative stress leading to a chronic depletion of endogenous antioxidants affects the survival of the cells and causes neurodegeneration. In particular, cerebellar granule neurons (CGNs) show a significant increase of reactive oxygen species (ROS), lipid peroxidation and lower level of reduced glutathione (GSH). In FRDA, one of the major pathways of oxidant scavengers, the Nrf2 antioxidant pathway, is defective. Previous studies on FRDA-like CGNs showed that the reduced level of frataxin and the oxidative stress induce mitochondrial impairments. By triggering the Nrf2 endogenous pathway pharmacologically we determined whether this could promote mitochondrial fitness and counteract oxidative stress. In this work, we sought to investigate the beneficial effect of a promising Nrf2-inducer, omaveloxolone (omav), in CGNs from two FRDA mouse models, KIKO and YG8R, and human fibroblasts from patients. We found that CGNs from both KIKO and YG8R presented Complex I deficiency and that omav was able to restore substrate availability and Complex I activity. This was also confirmed in human primary fibroblasts from FRDA patients. Although fibroblasts are not the major tissue affected, we found that they show significant differences recapitulating the disease; this is therefore an important tool to investigate patients’ pathophysiology. Interestingly, we found that patient fibroblasts had an increased level of endogenous lipid peroxidation and mitochondrial ROS (mROS), and lower GSH at rest. Omav was able to reverse this phenotype, protecting the cells against oxidative stress. By stimulating the cells with hydrogen peroxide (H2O2) and looking for potential mitochondrial pathophysiology, we found that fibroblasts could not maintain their mitochondrial membrane potential (ΔΨm). Remarkably, omav was protective to mitochondrial depolarization, promoting mitochondrial respiration and preventing cell death. Our results show that omav promotes Complex I activity and protect cells from oxidative stress. Omav could, therefore, be used as a novel therapeutic drug to ameliorate the pathophysiology of FRDA.
Collapse
Affiliation(s)
- Rosella Abeti
- Ataxia Centre, Department of Molecular Neuroscience, UCL Institute of Neurology, London, United Kingdom
| | - Annalisa Baccaro
- Ataxia Centre, Department of Molecular Neuroscience, UCL Institute of Neurology, London, United Kingdom
| | - Noemi Esteras
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, United Kingdom
| | - Paola Giunti
- Ataxia Centre, Department of Molecular Neuroscience, UCL Institute of Neurology, London, United Kingdom
| |
Collapse
|
23
|
Monnier V, Llorens JV, Navarro JA. Impact of Drosophila Models in the Study and Treatment of Friedreich's Ataxia. Int J Mol Sci 2018; 19:E1989. [PMID: 29986523 PMCID: PMC6073496 DOI: 10.3390/ijms19071989] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 06/26/2018] [Accepted: 07/03/2018] [Indexed: 02/07/2023] Open
Abstract
Drosophila melanogaster has been for over a century the model of choice of several neurobiologists to decipher the formation and development of the nervous system as well as to mirror the pathophysiological conditions of many human neurodegenerative diseases. The rare disease Friedreich’s ataxia (FRDA) is not an exception. Since the isolation of the responsible gene more than two decades ago, the analysis of the fly orthologue has proven to be an excellent avenue to understand the development and progression of the disease, to unravel pivotal mechanisms underpinning the pathology and to identify genes and molecules that might well be either disease biomarkers or promising targets for therapeutic interventions. In this review, we aim to summarize the collection of findings provided by the Drosophila models but also to go one step beyond and propose the implications of these discoveries for the study and cure of this disorder. We will present the physiological, cellular and molecular phenotypes described in the fly, highlighting those that have given insight into the pathology and we will show how the ability of Drosophila to perform genetic and pharmacological screens has provided valuable information that is not easily within reach of other cellular or mammalian models.
Collapse
Affiliation(s)
- Véronique Monnier
- Unité de Biologie Fonctionnelle et Adaptative (BFA), Sorbonne Paris Cité, Université Paris Diderot, UMR8251 CNRS, 75013 Paris, France.
| | - Jose Vicente Llorens
- Department of Genetics, University of Valencia, Campus of Burjassot, 96100 Valencia, Spain.
| | - Juan Antonio Navarro
- Lehrstuhl für Entwicklungsbiologie, Universität Regensburg, 93040 Regensburg, Germany.
| |
Collapse
|
24
|
Cherif K, Gérard C, Rousseau J, Ouellet DL, Chapdelaine P, Tremblay JP. Increased Frataxin Expression Induced in Friedreich Ataxia Cells by Platinum TALE-VP64s or Platinum TALE-SunTag. MOLECULAR THERAPY-NUCLEIC ACIDS 2018; 12:19-32. [PMID: 30195758 PMCID: PMC6019861 DOI: 10.1016/j.omtn.2018.04.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 04/24/2018] [Accepted: 04/24/2018] [Indexed: 11/28/2022]
Abstract
Frataxin gene (FXN) expression is reduced in Friedreich’s ataxia patients due to an increase in the number of GAA trinucleotides in intron 1. The frataxin protein, encoded by that gene, plays an important role in mitochondria’s iron metabolism. Platinum TALE (plTALE) proteins targeting the regulatory region of the FXN gene, fused with a transcriptional activator (TA) such as VP64 or P300, were used to increase the expression of that gene. Many effectors, plTALEVP64, plTALEp300, and plTALESunTag, targeting 14 sequences of the FXN gene promoter or intron 1 were produced. This permitted selection of 3 plTALEVP64s and 2 plTALESunTag that increased FXN gene expression by up to 19-fold in different Friedreich ataxia (FRDA) primary fibroblasts. Adeno-associated viruses were used to deliver the best effectors to the YG8R mouse model to validate their efficiencies in vivo. Our results showed that these selected plTALEVP64s or plTALESunTag induced transcriptional activity of the endogenous FXN gene as well as expression of the frataxin protein in YG8R mouse heart by 10-fold and in skeletal muscles by up to 35-fold. The aconitase activity was positively modulated by the frataxin level in mitochondria, and it was, thus, increased in vitro and in vivo by the increased frataxin expression.
Collapse
Affiliation(s)
- Khadija Cherif
- Centre de Recherche du CHU, Québec-Université Laval, Québec, QC, Canada; Département de Médecine Moléculaire, l'Université Laval Québec, Québec, QC, Canada
| | - Catherine Gérard
- Centre de Recherche du CHU, Québec-Université Laval, Québec, QC, Canada; Département de Médecine Moléculaire, l'Université Laval Québec, Québec, QC, Canada
| | - Joël Rousseau
- Centre de Recherche du CHU, Québec-Université Laval, Québec, QC, Canada; Département de Médecine Moléculaire, l'Université Laval Québec, Québec, QC, Canada
| | - Dominique L Ouellet
- Centre de Recherche du CHU, Québec-Université Laval, Québec, QC, Canada; Département de Médecine Moléculaire, l'Université Laval Québec, Québec, QC, Canada
| | - Pierre Chapdelaine
- Centre de Recherche du CHU, Québec-Université Laval, Québec, QC, Canada; Département de Médecine Moléculaire, l'Université Laval Québec, Québec, QC, Canada
| | - Jacques P Tremblay
- Centre de Recherche du CHU, Québec-Université Laval, Québec, QC, Canada; Département de Médecine Moléculaire, l'Université Laval Québec, Québec, QC, Canada.
| |
Collapse
|
25
|
Vögtle FN, Brändl B, Larson A, Pendziwiat M, Friederich MW, White SM, Basinger A, Kücükköse C, Muhle H, Jähn JA, Keminer O, Helbig KL, Delto CF, Myketin L, Mossmann D, Burger N, Miyake N, Burnett A, van Baalen A, Lovell MA, Matsumoto N, Walsh M, Yu HC, Shinde DN, Stephani U, Van Hove JLK, Müller FJ, Helbig I. Mutations in PMPCB Encoding the Catalytic Subunit of the Mitochondrial Presequence Protease Cause Neurodegeneration in Early Childhood. Am J Hum Genet 2018; 102:557-573. [PMID: 29576218 PMCID: PMC5985287 DOI: 10.1016/j.ajhg.2018.02.014] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 02/19/2018] [Indexed: 12/21/2022] Open
Abstract
Mitochondrial disorders causing neurodegeneration in childhood are genetically heterogeneous, and the underlying genetic etiology remains unknown in many affected individuals. We identified biallelic variants in PMPCB in individuals of four families including one family with two affected siblings with neurodegeneration and cerebellar atrophy. PMPCB encodes the catalytic subunit of the essential mitochondrial processing protease (MPP), which is required for maturation of the majority of mitochondrial precursor proteins. Mitochondria isolated from two fibroblast cell lines and induced pluripotent stem cells derived from one affected individual and differentiated neuroepithelial stem cells showed reduced PMPCB levels and accumulation of the processing intermediate of frataxin, a sensitive substrate for MPP dysfunction. Introduction of the identified PMPCB variants into the homologous S. cerevisiae Mas1 protein resulted in a severe growth and MPP processing defect leading to the accumulation of mitochondrial precursor proteins and early impairment of the biogenesis of iron-sulfur clusters, which are indispensable for a broad range of crucial cellular functions. Analysis of biopsy materials of an affected individual revealed changes and decreased activity in iron-sulfur cluster-containing respiratory chain complexes and dysfunction of mitochondrial and cytosolic Fe-S cluster-dependent enzymes. We conclude that biallelic mutations in PMPCB cause defects in MPP proteolytic activity leading to dysregulation of iron-sulfur cluster biogenesis and triggering a complex neurological phenotype of neurodegeneration in early childhood.
Collapse
Affiliation(s)
- F-Nora Vögtle
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany.
| | - Björn Brändl
- Department of Psychiatry and Psychotherapy, University Hospital Schleswig Holstein, Kiel 24105, Germany
| | - Austin Larson
- Department of Pediatrics, Section of Clinical Genetics and Metabolism, University of Colorado, Aurora, CO 80045, USA
| | - Manuela Pendziwiat
- Department of Neuropediatrics, Christian-Albrechts-University of Kiel, Kiel 24105, Germany
| | - Marisa W Friederich
- Department of Pediatrics, Section of Clinical Genetics and Metabolism, University of Colorado, Aurora, CO 80045, USA
| | - Susan M White
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia; Department of Paediatrics, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Alice Basinger
- Cook Children's Physician Network, Department of Genetics, Fort Worth, TX 76102, USA
| | - Cansu Kücükköse
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany; Faculty of Biology, University of Freiburg, Freiburg 79104, Germany
| | - Hiltrud Muhle
- Department of Neuropediatrics, Christian-Albrechts-University of Kiel, Kiel 24105, Germany
| | - Johanna A Jähn
- Department of Neuropediatrics, Christian-Albrechts-University of Kiel, Kiel 24105, Germany
| | - Oliver Keminer
- Fraunhofer-Institut für Molekularbiologie und Angewandte Ökologie IME, ScreeningPort, Hamburg 22525, Germany
| | - Katherine L Helbig
- Division of Neurology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Carolyn F Delto
- Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Würzburg 97080, Germany
| | - Lisa Myketin
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany
| | - Dirk Mossmann
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany
| | - Nils Burger
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany
| | - Noriko Miyake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Audrey Burnett
- Cook Children's Physician Network, Department of Genetics, Fort Worth, TX 76102, USA
| | - Andreas van Baalen
- Department of Neuropediatrics, Christian-Albrechts-University of Kiel, Kiel 24105, Germany
| | - Mark A Lovell
- Department of Pathology, University of Colorado, Aurora, CO 80045, USA
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Maie Walsh
- Adult Genetic Medicine, Royal Melbourne Hospital, Melbourne, VIC 3052, Australia
| | - Hung-Chun Yu
- Department of Pediatrics, Section of Clinical Genetics and Metabolism, University of Colorado, Aurora, CO 80045, USA
| | - Deepali N Shinde
- Division of Clinical Genomics, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Ulrich Stephani
- Department of Neuropediatrics, Christian-Albrechts-University of Kiel, Kiel 24105, Germany
| | - Johan L K Van Hove
- Department of Pediatrics, Section of Clinical Genetics and Metabolism, University of Colorado, Aurora, CO 80045, USA
| | - Franz-Josef Müller
- Department of Psychiatry and Psychotherapy, University Hospital Schleswig Holstein, Kiel 24105, Germany; Max Planck Institute for Molecular Genetics, Berlin 14195, Germany
| | - Ingo Helbig
- Department of Neuropediatrics, Christian-Albrechts-University of Kiel, Kiel 24105, Germany; Division of Neurology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| |
Collapse
|
26
|
Alsina D, Ros J, Tamarit J. Nitric oxide prevents Aft1 activation and metabolic remodeling in frataxin-deficient yeast. Redox Biol 2018; 14:131-141. [PMID: 28918000 PMCID: PMC5602528 DOI: 10.1016/j.redox.2017.09.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 08/25/2017] [Accepted: 09/02/2017] [Indexed: 11/30/2022] Open
Abstract
Yeast frataxin homolog (Yfh1) is the orthologue of human frataxin, a mitochondrial protein whose deficiency causes Friedreich Ataxia. Yfh1 deficiency activates Aft1, a transcription factor governing iron homeostasis in yeast cells. Although the mechanisms causing this activation are not completely understood, it is assumed that it may be caused by iron-sulfur deficiency. However, several evidences indicate that activation of Aft1 occurs in the absence of iron-sulfur deficiency. Besides, Yfh1 deficiency also leads to metabolic remodeling (mainly consisting in a shift from respiratory to fermentative metabolism) and to induction of Yhb1, a nitric oxide (NO) detoxifying enzyme. In this work, we have used conditional Yfh1 mutant yeast strains to investigate the relationship between NO, Aft1 activation and metabolic remodeling. We have observed that NO prevents Aft1 activation caused by Yfh1 deficiency. This phenomenon is not observed when Aft1 is activated by iron scarcity or impaired iron-sulfur biogenesis. In addition, analyzing key metabolic proteins by a targeted proteomics approach, we have observed that NO prevents the metabolic remodeling caused by Yfh1 deficiency. We conclude that Aft1 activation in Yfh1-deficient yeasts is not caused by iron-sulfur deficiency or iron scarcity. Our hypothesis is that Yfh1 deficiency leads to the presence of anomalous iron species that can compromise iron bioavailability and activate a signaling cascade that results in Aft1 activation and metabolic remodeling.
Collapse
Affiliation(s)
- David Alsina
- Departament de Ciències Mèdiques Bàsiques, IRBLleida, Universitat de Lleida, Lleida, Spain
| | - Joaquim Ros
- Departament de Ciències Mèdiques Bàsiques, IRBLleida, Universitat de Lleida, Lleida, Spain
| | - Jordi Tamarit
- Departament de Ciències Mèdiques Bàsiques, IRBLleida, Universitat de Lleida, Lleida, Spain.
| |
Collapse
|
27
|
Cook A, Giunti P. Friedreich's ataxia: clinical features, pathogenesis and management. Br Med Bull 2017; 124:19-30. [PMID: 29053830 PMCID: PMC5862303 DOI: 10.1093/bmb/ldx034] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 09/06/2017] [Accepted: 09/19/2017] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Friedreich's ataxia is the most common inherited ataxia. SOURCES OF DATA Literature search using PubMed with keywords Friedreich's ataxia together with published papers known to the authors. AREAS OF AGREEMENT The last decade has seen important advances in our understanding of the pathogenesis of disease. In particular, the genetic and epigenetic mechanisms underlying the disease now offer promising novel therapeutic targets. AREAS OF CONTROVERSY The search for effective disease-modifying agents continues. It remains to be determined whether the most effective approach to treatment lies with increasing frataxin protein levels or addressing the metabolic consequences of the disease, for example with antioxidants. AREAS TIMELY FOR DEVELOPING RESEARCH Management of Freidreich's ataxia is currently focussed on symptomatic management, delivered by the multidisciplinary team. Phase II clinical trials in agents that address the abberrant silencing of the frataxin gene need to be translated into large placebo-controlled Phase III trials to help establish their therapeutic potential.
Collapse
Affiliation(s)
- A Cook
- Department of Molecular Neuroscience, Ataxia Centre, UCL Institute of Neurology, Queen Square, London, UK
| | - P Giunti
- Department of Molecular Neuroscience, Ataxia Centre, UCL Institute of Neurology, Queen Square, London, UK
| |
Collapse
|
28
|
|
29
|
Benini M, Fortuni S, Condò I, Alfedi G, Malisan F, Toschi N, Serio D, Massaro DS, Arcuri G, Testi R, Rufini A. E3 Ligase RNF126 Directly Ubiquitinates Frataxin, Promoting Its Degradation: Identification of a Potential Therapeutic Target for Friedreich Ataxia. Cell Rep 2017; 18:2007-2017. [PMID: 28228265 PMCID: PMC5329121 DOI: 10.1016/j.celrep.2017.01.079] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 12/14/2016] [Accepted: 01/29/2017] [Indexed: 12/21/2022] Open
Abstract
Friedreich ataxia (FRDA) is a severe genetic neurodegenerative disease caused by reduced expression of the mitochondrial protein frataxin. To date, there is no therapy to treat this condition. The amount of residual frataxin critically affects the severity of the disease; thus, attempts to restore physiological frataxin levels are considered therapeutically relevant. Frataxin levels are controlled by the ubiquitin-proteasome system; therefore, inhibition of the frataxin E3 ligase may represent a strategy to achieve an increase in frataxin levels. Here, we report the identification of the RING E3 ligase RNF126 as the enzyme that specifically mediates frataxin ubiquitination and targets it for degradation. RNF126 interacts with frataxin and promotes its ubiquitination in a catalytic activity-dependent manner, both in vivo and in vitro. Most importantly, RNF126 depletion results in frataxin accumulation in cells derived from FRDA patients, highlighting the relevance of RNF126 as a new therapeutic target for Friedreich ataxia.
Collapse
Affiliation(s)
- Monica Benini
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," Via Montpellier 1, 00133 Rome, Italy; Fratagene Therapeutics Srl, Viale dei Campioni 8, 00144 Rome, Italy
| | - Silvia Fortuni
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," Via Montpellier 1, 00133 Rome, Italy
| | - Ivano Condò
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," Via Montpellier 1, 00133 Rome, Italy
| | - Giulia Alfedi
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," Via Montpellier 1, 00133 Rome, Italy
| | - Florence Malisan
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," Via Montpellier 1, 00133 Rome, Italy
| | - Nicola Toschi
- Medical Physics Section, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," Via Montpellier 1, 00133 Rome, Italy; Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging and Harvard Medical School, Boston, MA 02115, USA
| | - Dario Serio
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," Via Montpellier 1, 00133 Rome, Italy; Fratagene Therapeutics Srl, Viale dei Campioni 8, 00144 Rome, Italy
| | - Damiano Sergio Massaro
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," Via Montpellier 1, 00133 Rome, Italy
| | - Gaetano Arcuri
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," Via Montpellier 1, 00133 Rome, Italy
| | - Roberto Testi
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," Via Montpellier 1, 00133 Rome, Italy; Fratagene Therapeutics Srl, Viale dei Campioni 8, 00144 Rome, Italy
| | - Alessandra Rufini
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," Via Montpellier 1, 00133 Rome, Italy; Fratagene Therapeutics Srl, Viale dei Campioni 8, 00144 Rome, Italy.
| |
Collapse
|
30
|
Tamarit J, Obis È, Ros J. Oxidative stress and altered lipid metabolism in Friedreich ataxia. Free Radic Biol Med 2016; 100:138-146. [PMID: 27296838 DOI: 10.1016/j.freeradbiomed.2016.06.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 06/07/2016] [Accepted: 06/10/2016] [Indexed: 12/31/2022]
Abstract
Friedreich ataxia is a genetic disease caused by the deficiency of frataxin, a mitochondrial protein. Frataxin deficiency impacts in the cell physiology at several levels. One of them is oxidative stress with consequences in terms of protein dysfunctions and metabolic alterations. Among others, alterations in lipid metabolism have been observed in several models of the disease. In this review we summarize the current knowledge of the molecular basis of the disease, the relevance of oxidative stress and the therapeutic strategies based on reduction of mitochondrial reactive oxygen species production. Finally, we will focus the interest in alterations of lipid metabolism as a consequence of mitochondrial dysfunction and describe the therapeutic approaches based on targeting lipid metabolism.
Collapse
Affiliation(s)
- Jordi Tamarit
- Departament de Ciències Mèdiques Bàsiques, IRB-Lleida, Universitat de Lleida, Lleida, Spain
| | - Èlia Obis
- Departament de Ciències Mèdiques Bàsiques, IRB-Lleida, Universitat de Lleida, Lleida, Spain
| | - Joaquim Ros
- Departament de Ciències Mèdiques Bàsiques, IRB-Lleida, Universitat de Lleida, Lleida, Spain.
| |
Collapse
|
31
|
Skliarova EI, Popova TN, Shulgin KK. Effects of N-[Imino(1-Piperidinyl)Methyl] Guanidine on the Intensity of Free Radical Processes, Aconitase Activity, and Citrate Level in the Tissues of Rats with Experimental Type 2 Diabetes Mellitus. Bull Exp Biol Med 2016; 161:261-5. [PMID: 27383162 DOI: 10.1007/s10517-016-3391-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Indexed: 10/21/2022]
Abstract
Effects of a synthetic biguanide derivative N-[imino(1-piperidinyl)methyl] guanidine (NIPMG) on free radical homeostasis, aconitase activity, and citrate concentration were studied in the liver and blood serum of rats with type 2 diabetes mellitus. Analysis of biochemiluminescence parameters showed that administration of this agent (10 mg/kg body weight) to animals with diabetes reduced the intensity of free radical processes in study tissues relative to the increased values in untreated diabetic animals. Under these conditions, aconitase activity, a principal target of ROS effects, and citrate level in the liver and blood serum of rats approached the control levels. The results show that NIPMG can positively regulate free radical homeostasis and reduce the intensity of oxidative stress in type 2 diabetes mellitus, which was accompanied by normalization of the studied parameters.
Collapse
Affiliation(s)
| | - T N Popova
- Voronezh State University, Voronezh, Russia
| | | |
Collapse
|
32
|
Galea CA, Huq A, Lockhart PJ, Tai G, Corben LA, Yiu EM, Gurrin LC, Lynch DR, Gelbard S, Durr A, Pousset F, Parkinson M, Labrum R, Giunti P, Perlman SL, Delatycki MB, Evans-Galea MV. Compound heterozygous FXN mutations and clinical outcome in friedreich ataxia. Ann Neurol 2016; 79:485-95. [PMID: 26704351 DOI: 10.1002/ana.24595] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 12/16/2015] [Accepted: 12/17/2015] [Indexed: 02/01/2023]
Abstract
OBJECTIVE Friedreich ataxia (FRDA) is an inherited neurodegenerative disease characterized by ataxia and cardiomyopathy. Homozygous GAA trinucleotide repeat expansions in the first intron of FXN occur in 96% of affected individuals and reduce frataxin expression. Remaining individuals are compound heterozygous for a GAA expansion and a FXN point/insertion/deletion mutation. We examined disease-causing mutations and the impact on frataxin structure/function and clinical outcome in FRDA. METHODS We compared clinical information from 111 compound heterozygotes and 131 individuals with homozygous expansions. Frataxin mutations were examined using structural modeling, stability analyses and systematic literature review, and categorized into four groups: (1) homozygous expansions, and three compound heterozygote groups; (2) null (no frataxin produced); (3) moderate/strong impact; and (4) minimal impact. Mean age of onset and the presence of cardiomyopathy and diabetes mellitus were compared using regression analyses. RESULTS Mutations in the hydrophobic core of frataxin affected stability whereas surface residue mutations affected interactions with iron sulfur cluster assembly and heme biosynthetic proteins. The null group of compound heterozygotes had significantly earlier age of onset and increased diabetes mellitus, compared to the homozygous expansion group. There were no significant differences in mean age of onset between homozygotes and the minimal and moderate/strong impact groups. INTERPRETATION In compound heterozygotes, expression of partially functional mutant frataxin delays age of onset and reduces diabetes mellitus, compared to those with no frataxin expression from the non-expanded allele. This integrated analysis of categorized frataxin mutations and their correlation with clinical outcome provide a definitive resource for investigating disease pathogenesis in FRDA.
Collapse
Affiliation(s)
- Charles A Galea
- Medicinal Chemistry and Drug Delivery, Disposition and Dynamics (D4), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Bruce Lefroy Centre, Murdoch Childrens Research Institute, Parkville, Victoria, Australia
| | - Aamira Huq
- Bruce Lefroy Centre, Murdoch Childrens Research Institute, Parkville, Victoria, Australia
| | - Paul J Lockhart
- Bruce Lefroy Centre, Murdoch Childrens Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Geneieve Tai
- Bruce Lefroy Centre, Murdoch Childrens Research Institute, Parkville, Victoria, Australia
| | - Louise A Corben
- Bruce Lefroy Centre, Murdoch Childrens Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Royal Children's Hospital, Parkville, Victoria, Australia
- School of Psychological Sciences, Monash University, Clayton, Victoria, Australia
| | - Eppie M Yiu
- Bruce Lefroy Centre, Murdoch Childrens Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Neurology, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Lyle C Gurrin
- Center for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - David R Lynch
- Departments of Neurology and Pediatrics, University of Pennsylvania School of Medicine and The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Sarah Gelbard
- Departments of Neurology and Pediatrics, University of Pennsylvania School of Medicine and The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Alexandra Durr
- APHP, Department of Genetics and Cytogenetics, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
- Institut du Cerveau et de la Moelle épinière (ICM), Pitié-Salpêtrière University Hospital, Paris, France
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Université Paris 06 UMR S_1127, ICM, F-75013, France
| | - Francoise Pousset
- APHP, Cardiology Department, AP-HP Pitie-Salpétrière Hospital, Paris, France
| | - Michael Parkinson
- Department of Molecular Neuroscience, University College London Institute of Neurology, London, United Kingdom
| | - Robyn Labrum
- Department of Neurogenetics, University College London Hospital, Institute of Neurology, London, United Kingdom
| | - Paola Giunti
- Department of Molecular Neuroscience, University College London Institute of Neurology, London, United Kingdom
- Department of Neurogenetics, University College London Hospital, Institute of Neurology, London, United Kingdom
| | - Susan L Perlman
- Ataxia Center and Huntington Disease Center of Excellence, Department of Neurology, David Geffen School of Medicine at the University of California at Los Angeles, CA
| | - Martin B Delatycki
- Bruce Lefroy Centre, Murdoch Childrens Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Royal Children's Hospital, Parkville, Victoria, Australia
- School of Psychological Sciences, Monash University, Clayton, Victoria, Australia
- Clinical Genetics, Austin Health, Heidelberg, Victoria, Australia
| | - Marguerite V Evans-Galea
- Bruce Lefroy Centre, Murdoch Childrens Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Royal Children's Hospital, Parkville, Victoria, Australia
| |
Collapse
|
33
|
Cherubini F, Serio D, Guccini I, Fortuni S, Arcuri G, Condò I, Rufini A, Moiz S, Camerini S, Crescenzi M, Testi R, Malisan F. Src inhibitors modulate frataxin protein levels. Hum Mol Genet 2015; 24:4296-305. [PMID: 25948553 DOI: 10.1093/hmg/ddv162] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 04/30/2015] [Indexed: 12/19/2022] Open
Abstract
Defective expression of frataxin is responsible for the inherited, progressive degenerative disease Friedreich's Ataxia (FRDA). There is currently no effective approved treatment for FRDA and patients die prematurely. Defective frataxin expression causes critical metabolic changes, including redox imbalance and ATP deficiency. As these alterations are known to regulate the tyrosine kinase Src, we investigated whether Src might in turn affect frataxin expression. We found that frataxin can be phosphorylated by Src. Phosphorylation occurs primarily on Y118 and promotes frataxin ubiquitination, a signal for degradation. Accordingly, Src inhibitors induce accumulation of frataxin but are ineffective on a non-phosphorylatable frataxin-Y118F mutant. Importantly, all the Src inhibitors tested, some of them already in the clinic, increase frataxin expression and rescue the aconitase defect in frataxin-deficient cells derived from FRDA patients. Thus, Src inhibitors emerge as a new class of drugs able to promote frataxin accumulation, suggesting their possible use as therapeutics in FRDA.
Collapse
Affiliation(s)
- Fabio Cherubini
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', Via Montpellier 1, 00133 Rome, Italy
| | - Dario Serio
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', Via Montpellier 1, 00133 Rome, Italy
| | - Ilaria Guccini
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', Via Montpellier 1, 00133 Rome, Italy
| | - Silvia Fortuni
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', Via Montpellier 1, 00133 Rome, Italy, Fratagene Therapeutics Ltd, 22 Northumberland Rd, Dublin, Ireland and
| | - Gaetano Arcuri
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', Via Montpellier 1, 00133 Rome, Italy
| | - Ivano Condò
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', Via Montpellier 1, 00133 Rome, Italy
| | - Alessandra Rufini
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', Via Montpellier 1, 00133 Rome, Italy, Fratagene Therapeutics Ltd, 22 Northumberland Rd, Dublin, Ireland and
| | - Shadman Moiz
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', Via Montpellier 1, 00133 Rome, Italy
| | - Serena Camerini
- Department of Cell Biology and Neurosciences, Italian National Institute of Health, Viale Regina Elena, 299, 00161 Rome, Italy
| | - Marco Crescenzi
- Department of Cell Biology and Neurosciences, Italian National Institute of Health, Viale Regina Elena, 299, 00161 Rome, Italy
| | - Roberto Testi
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', Via Montpellier 1, 00133 Rome, Italy, Fratagene Therapeutics Ltd, 22 Northumberland Rd, Dublin, Ireland and
| | - Florence Malisan
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', Via Montpellier 1, 00133 Rome, Italy,
| |
Collapse
|
34
|
Zaidi A, Singh KP, Anwar S, Suman SS, Equbal A, Singh K, Dikhit MR, Bimal S, Pandey K, Das P, Ali V. Interaction of frataxin, an iron binding protein, with IscU of Fe-S clusters biogenesis pathway and its upregulation in AmpB resistant Leishmania donovani. Biochimie 2015; 115:120-35. [PMID: 26032732 DOI: 10.1016/j.biochi.2015.05.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 05/19/2015] [Indexed: 01/10/2023]
Abstract
Leishmania donovani is a unicellular protozoon parasite that causes visceral leishmaniasis (VL), which is a fatal disease if left untreated. Certain Fe-S proteins of the TCA cycle and respiratory chain have been found in the Leishmania parasite but the precise mechanisms for their biogenesis and the maturation of Fe-S clusters remains unknown. Fe-S clusters are ubiquitous cofactors of proteins that perform critical cellular functions. The clusters are biosynthesized by the mitochondrial Iron-Sulphur Cluster (ISC) machinery with core protein components that include the catalytic cysteine desulphurase IscS, the scaffold proteins IscU and IscA, and frataxin as an iron carrier/donor. However, no information regarding frataxin, its regulation, or its role in drug resistance is available for the Leishmania parasite. In this study, we characterized Ld-frataxin to investigate its role in the ISC machinery of L. donovani. We expressed and purified the recombinant Ld-frataxin protein and observed its interaction with Ld-IscU by co-purification and pull-down assay. Furthermore, we observed that the cysteine desulphurase activity of the purified Ld-IscS protein was stimulated in the presence of Ld-frataxin and Ld-IscU, particularly in the presence of iron; neither Ld-frataxin nor Ld-IscU alone had significant effects on Ld-IscS activity. Interestingly, RT-PCR and western blotting showed that Ld-frataxin is upregulated in AmpB-resistant isolates compared to sensitive strains, which may support higher Fe-S protein activity in AmpB-resistant L. donovani. Additionally, Ld-frataxin was localized in the mitochondria, as revealed by digitonin fractionation and indirect immunofluorescence. Thus, our results suggest the role of Ld-frataxin as an iron binding/carrier protein for Fe-S cluster biogenesis that physically interacts with other core components of the ISC machinery within the mitochondria.
Collapse
Affiliation(s)
- Amir Zaidi
- Laboratory of Molecular Biochemistry and Cell Biology, Department of Biochemistry, Rajendra Memorial Research Institute of Medical Sciences (RMRIMS), Agamkuan, Patna 800007, India
| | - Krishn Pratap Singh
- Laboratory of Molecular Biochemistry and Cell Biology, Department of Biochemistry, Rajendra Memorial Research Institute of Medical Sciences (RMRIMS), Agamkuan, Patna 800007, India
| | - Shadab Anwar
- Laboratory of Molecular Biochemistry and Cell Biology, Department of Biochemistry, Rajendra Memorial Research Institute of Medical Sciences (RMRIMS), Agamkuan, Patna 800007, India
| | - Shashi S Suman
- Laboratory of Molecular Biochemistry and Cell Biology, Department of Biochemistry, Rajendra Memorial Research Institute of Medical Sciences (RMRIMS), Agamkuan, Patna 800007, India
| | - Asif Equbal
- Laboratory of Molecular Biochemistry and Cell Biology, Department of Biochemistry, Rajendra Memorial Research Institute of Medical Sciences (RMRIMS), Agamkuan, Patna 800007, India
| | - Kuljit Singh
- Laboratory of Molecular Biochemistry and Cell Biology, Department of Biochemistry, Rajendra Memorial Research Institute of Medical Sciences (RMRIMS), Agamkuan, Patna 800007, India
| | - Manas R Dikhit
- Biomedical Informatic Centre, Rajendra Memorial Research Institute of Medical Sciences (RMRIMS), Agamkuan, Patna, India
| | - Sanjeeva Bimal
- Department of Immunology, Rajendra Memorial Research Institute of Medical Sciences (RMRIMS), Agamkuan, Patna, India
| | - Krishna Pandey
- Department of Clinical Medicine, Rajendra Memorial Research Institute of Medical Sciences (RMRIMS), Agamkuan, Patna, India
| | - Pradeep Das
- Department of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences (RMRIMS), Agamkuan, Patna, India
| | - Vahab Ali
- Laboratory of Molecular Biochemistry and Cell Biology, Department of Biochemistry, Rajendra Memorial Research Institute of Medical Sciences (RMRIMS), Agamkuan, Patna 800007, India.
| |
Collapse
|
35
|
Pérez-Luz S, Gimenez-Cassina A, Fernández-Frías I, Wade-Martins R, Díaz-Nido J. Delivery of the 135 kb human frataxin genomic DNA locus gives rise to different frataxin isoforms. Genomics 2015; 106:76-82. [PMID: 26027909 DOI: 10.1016/j.ygeno.2015.05.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 05/21/2015] [Accepted: 05/23/2015] [Indexed: 11/25/2022]
Abstract
Friedreich's ataxia (FRDA) is the most common form of hereditary ataxia caused by recessive mutations in the FXN gene. Recent results have indicated the presence of different frataxin isoforms due to alternative gene expression mechanisms. Our previous studies demonstrated the advantages of using high-capacity herpes simplex virus type 1 (HSV-1) amplicon vectors containing the entire FXN genomic locus (iBAC-FXN) as a gene-delivery vehicle capable of ensuring physiologically-regulated and long-term persistence. Here we describe how expression from the 135 kb human FXN genomic locus produces the three frataxin isoforms both in cultured neuronal cells and also in vivo. Moreover, we also observed the correct expression of these frataxin isoforms in patient-derived cells after delivery of the iBAC-FXN. These results lend further support to the potential use of HSV-1 vectors containing entire genomic loci whose expression is mediated by complex transcriptional and posttranscriptional mechanisms for gene therapy applications.
Collapse
Affiliation(s)
- S Pérez-Luz
- Departamento Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, 28049 Madrid, Spain; CIBER de Enfermedades Raras (CIBERER), Spain; Instituto de Investigación Sanitaria Puerta de Hierro-Majadahonda, Spain
| | | | - I Fernández-Frías
- Departamento Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, 28049 Madrid, Spain; CIBER de Enfermedades Raras (CIBERER), Spain; Instituto de Investigación Sanitaria Puerta de Hierro-Majadahonda, Spain
| | | | - J Díaz-Nido
- Departamento Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, 28049 Madrid, Spain; CIBER de Enfermedades Raras (CIBERER), Spain; Instituto de Investigación Sanitaria Puerta de Hierro-Majadahonda, Spain.
| |
Collapse
|
36
|
Rufini A, Cavallo F, Condò I, Fortuni S, De Martino G, Incani O, Di Venere A, Benini M, Massaro DS, Arcuri G, Serio D, Malisan F, Testi R. Highly specific ubiquitin-competing molecules effectively promote frataxin accumulation and partially rescue the aconitase defect in Friedreich ataxia cells. Neurobiol Dis 2015; 75:91-9. [PMID: 25549872 PMCID: PMC4358773 DOI: 10.1016/j.nbd.2014.12.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 12/09/2014] [Accepted: 12/11/2014] [Indexed: 11/30/2022] Open
Abstract
Friedreich ataxia is an inherited neurodegenerative disease that leads to progressive disability. There is currently no effective treatment and patients die prematurely. The underlying genetic defect leads to reduced expression of the mitochondrial protein frataxin. Frataxin insufficiency causes mitochondrial dysfunction and ultimately cell death, particularly in peripheral sensory ganglia. There is an inverse correlation between the amount of residual frataxin and the severity of disease progression; therefore, therapeutic approaches aiming at increasing frataxin levels are expected to improve patients' conditions. We previously discovered that a significant amount of frataxin precursor is degraded by the ubiquitin/proteasome system before its functional mitochondrial maturation. We also provided evidence for the therapeutic potential of small molecules that increase frataxin levels by docking on the frataxin ubiquitination site, thus preventing frataxin ubiquitination and degradation. We called these compounds ubiquitin-competing molecules (UCM). By extending our search for effective UCM, we identified a set of new and more potent compounds that more efficiently promote frataxin accumulation. Here we show that these compounds directly interact with frataxin and prevent its ubiquitination. Interestingly, these UCM are not effective on the ubiquitin-resistant frataxin mutant, indicating their specific action on preventing frataxin ubiquitination. Most importantly, these compounds are able to promote frataxin accumulation and aconitase rescue in cells derived from patients, strongly supporting their therapeutic potential.
Collapse
Affiliation(s)
- Alessandra Rufini
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," Via Montpellier 1, Rome 00133, Italy; Fratagene Therapeutics Ltd., 22 Northumberland Rd., Dublin, Ireland
| | - Francesca Cavallo
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," Via Montpellier 1, Rome 00133, Italy
| | - Ivano Condò
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," Via Montpellier 1, Rome 00133, Italy
| | - Silvia Fortuni
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," Via Montpellier 1, Rome 00133, Italy; Fratagene Therapeutics Ltd., 22 Northumberland Rd., Dublin, Ireland
| | - Gabriella De Martino
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," Via Montpellier 1, Rome 00133, Italy
| | - Ottaviano Incani
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," Via Montpellier 1, Rome 00133, Italy
| | - Almerinda Di Venere
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata," Via Montpellier 1, Rome 00133, Italy
| | - Monica Benini
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," Via Montpellier 1, Rome 00133, Italy
| | - Damiano Sergio Massaro
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," Via Montpellier 1, Rome 00133, Italy
| | - Gaetano Arcuri
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," Via Montpellier 1, Rome 00133, Italy
| | - Dario Serio
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," Via Montpellier 1, Rome 00133, Italy
| | - Florence Malisan
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," Via Montpellier 1, Rome 00133, Italy
| | - Roberto Testi
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome "Tor Vergata," Via Montpellier 1, Rome 00133, Italy; Fratagene Therapeutics Ltd., 22 Northumberland Rd., Dublin, Ireland.
| |
Collapse
|
37
|
Angerer H. Eukaryotic LYR Proteins Interact with Mitochondrial Protein Complexes. BIOLOGY 2015; 4:133-50. [PMID: 25686363 PMCID: PMC4381221 DOI: 10.3390/biology4010133] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 02/04/2015] [Indexed: 01/18/2023]
Abstract
In eukaryotic cells, mitochondria host ancient essential bioenergetic and biosynthetic pathways. LYR (leucine/tyrosine/arginine) motif proteins (LYRMs) of the Complex1_LYR-like superfamily interact with protein complexes of bacterial origin. Many LYR proteins function as extra subunits (LYRM3 and LYRM6) or novel assembly factors (LYRM7, LYRM8, ACN9 and FMC1) of the oxidative phosphorylation (OXPHOS) core complexes. Structural insights into complex I accessory subunits LYRM6 and LYRM3 have been provided by analyses of EM and X-ray structures of complex I from bovine and the yeast Yarrowia lipolytica, respectively. Combined structural and biochemical studies revealed that LYRM6 resides at the matrix arm close to the ubiquinone reduction site. For LYRM3, a position at the distal proton-pumping membrane arm facing the matrix space is suggested. Both LYRMs are supposed to anchor an acyl-carrier protein (ACPM) independently to complex I. The function of this duplicated protein interaction of ACPM with respiratory complex I is still unknown. Analysis of protein-protein interaction screens, genetic analyses and predicted multi-domain LYRMs offer further clues on an interaction network and adaptor-like function of LYR proteins in mitochondria.
Collapse
Affiliation(s)
- Heike Angerer
- Goethe University Frankfurt, Medical School, Institute of Biochemistry II, Structural Bioenergetics Group, Max-von-Laue Street 9, Frankfurt am Main 60438, Germany.
| |
Collapse
|
38
|
Lane DJR, Merlot AM, Huang MLH, Bae DH, Jansson PJ, Sahni S, Kalinowski DS, Richardson DR. Cellular iron uptake, trafficking and metabolism: Key molecules and mechanisms and their roles in disease. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:1130-44. [PMID: 25661197 DOI: 10.1016/j.bbamcr.2015.01.021] [Citation(s) in RCA: 253] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 01/09/2015] [Accepted: 01/28/2015] [Indexed: 01/08/2023]
Abstract
Iron is a crucial transition metal for virtually all life. Two major destinations of iron within mammalian cells are the cytosolic iron-storage protein, ferritin, and mitochondria. In mitochondria, iron is utilized in critical anabolic pathways, including: iron-storage in mitochondrial ferritin, heme synthesis, and iron-sulfur cluster (ISC) biogenesis. Although the pathways involved in ISC synthesis in the mitochondria and cytosol have begun to be characterized, many crucial details remain unknown. In this review, we discuss major aspects of the journey of iron from its initial cellular uptake, its modes of trafficking within cells, to an overview of its downstream utilization in the cytoplasm and within mitochondria. The understanding of mitochondrial iron processing and its communication with other organelles/subcellular locations, such as the cytosol, has been elucidated by the analysis of certain diseases e.g., Friedreich's ataxia. Increased knowledge of the molecules and their mechanisms of action in iron processing pathways (e.g., ISC biogenesis) will shape the investigation of iron metabolism in human health and disease.
Collapse
Affiliation(s)
- D J R Lane
- Department of Pathology and Bosch Institute, Molecular Pharmacology and Pathology Program, Blackburn Building, University of Sydney, Sydney, New South Wales 2006, Australia.
| | - A M Merlot
- Department of Pathology and Bosch Institute, Molecular Pharmacology and Pathology Program, Blackburn Building, University of Sydney, Sydney, New South Wales 2006, Australia
| | - M L-H Huang
- Department of Pathology and Bosch Institute, Molecular Pharmacology and Pathology Program, Blackburn Building, University of Sydney, Sydney, New South Wales 2006, Australia
| | - D-H Bae
- Department of Pathology and Bosch Institute, Molecular Pharmacology and Pathology Program, Blackburn Building, University of Sydney, Sydney, New South Wales 2006, Australia
| | - P J Jansson
- Department of Pathology and Bosch Institute, Molecular Pharmacology and Pathology Program, Blackburn Building, University of Sydney, Sydney, New South Wales 2006, Australia
| | - S Sahni
- Department of Pathology and Bosch Institute, Molecular Pharmacology and Pathology Program, Blackburn Building, University of Sydney, Sydney, New South Wales 2006, Australia
| | - D S Kalinowski
- Department of Pathology and Bosch Institute, Molecular Pharmacology and Pathology Program, Blackburn Building, University of Sydney, Sydney, New South Wales 2006, Australia
| | - D R Richardson
- Department of Pathology and Bosch Institute, Molecular Pharmacology and Pathology Program, Blackburn Building, University of Sydney, Sydney, New South Wales 2006, Australia.
| |
Collapse
|
39
|
Obis È, Irazusta V, Sanchís D, Ros J, Tamarit J. Frataxin deficiency in neonatal rat ventricular myocytes targets mitochondria and lipid metabolism. Free Radic Biol Med 2014; 73:21-33. [PMID: 24751525 DOI: 10.1016/j.freeradbiomed.2014.04.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 04/04/2014] [Accepted: 04/04/2014] [Indexed: 11/21/2022]
Abstract
Friedreich ataxia (FRDA) is a hereditary disease caused by deficient frataxin expression. This mitochondrial protein has been related to iron homeostasis, energy metabolism, and oxidative stress. Patients with FRDA experience neurologic alterations and cardiomyopathy, which is the leading cause of death. The specific effects of frataxin depletion on cardiomyocytes are poorly understood because no appropriate cardiac cellular model is available to researchers. To address this research need, we present a model based on primary cultures of neonatal rat ventricular myocytes (NRVMs) and short-hairpin RNA interference. Using this approach, frataxin was reduced down to 5 to 30% of control protein levels after 7 days of transduction. At this stage the activity and amount of the iron-sulfur protein aconitase, in vitro activities of several OXPHOS components, levels of iron-regulated mRNAs, and the ATP/ADP ratio were comparable to controls. However, NRVMs exhibited markers of oxidative stress and a disorganized mitochondrial network with enlarged mitochondria. Lipids, the main energy source of heart cells, also underwent a clear metabolic change, indicated by the increased presence of lipid droplets and induction of medium-chain acyl-CoA dehydrogenase. These results indicate that mitochondria and lipid metabolism are primary targets of frataxin deficiency in NRVMs. Therefore, they contribute to the understanding of cardiac-specific mechanisms occurring in FRDA and give clues for the design of cardiac-specific treatment strategies for FRDA.
Collapse
MESH Headings
- Aconitate Hydratase/metabolism
- Animals
- Cardiomyopathies/pathology
- Cells, Cultured
- Disease Models, Animal
- Friedreich Ataxia/pathology
- Heart Ventricles/cytology
- Heart Ventricles/metabolism
- Humans
- Iron-Binding Proteins/genetics
- Lipid Metabolism/genetics
- Membrane Potential, Mitochondrial/physiology
- Mitochondria, Heart/genetics
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Myocytes, Cardiac/cytology
- Myocytes, Cardiac/metabolism
- Oxidative Stress/physiology
- Peroxisome Proliferator-Activated Receptors/metabolism
- RNA Interference
- RNA, Small Interfering
- Rats
- Rats, Sprague-Dawley
- Frataxin
Collapse
Affiliation(s)
- Èlia Obis
- Departament de Ciències Mèdiques Bàsiques, IRB-Lleida, Universitat de Lleida, 25198 Lleida, Spain
| | - Verónica Irazusta
- Instituto de Investigación para la Industria Química, INIQUI-CONICET, Salta, Argentina
| | - Daniel Sanchís
- Departament de Ciències Mèdiques Bàsiques, IRB-Lleida, Universitat de Lleida, 25198 Lleida, Spain
| | - Joaquim Ros
- Departament de Ciències Mèdiques Bàsiques, IRB-Lleida, Universitat de Lleida, 25198 Lleida, Spain
| | - Jordi Tamarit
- Departament de Ciències Mèdiques Bàsiques, IRB-Lleida, Universitat de Lleida, 25198 Lleida, Spain.
| |
Collapse
|
40
|
Moreno-Cermeño A, Alsina D, Cabiscol E, Tamarit J, Ros J. Metabolic remodeling in frataxin-deficient yeast is mediated by Cth2 and Adr1. BIOCHIMICA ET BIOPHYSICA ACTA 2013; 1833:3326-3337. [PMID: 24100161 DOI: 10.1016/j.bbamcr.2013.09.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 09/10/2013] [Accepted: 09/27/2013] [Indexed: 10/26/2022]
Abstract
Frataxin is a mitochondrial protein involved in iron metabolism whose deficiency in humans causes Friedreich ataxia. We performed transcriptomic and proteomic analyses of conditional Yeast Frataxin Homologue (Yfh1) mutants (tetO7-YFH1) to investigate metabolic remodeling upon Yfh1 depletion. These studies revealed that Yfh1 depletion leads to downregulation of many glucose-repressed genes. Most of them were Adr1 targets, a key transcription factor required for growth in non-fermentable carbon sources. Using a GFP-tagged Adr1, we observed that Yfh1 depletion promotes the export of Adr1 from the nucleus to the cytosol without affecting its protein levels. This effect was also observed upon H2O2 treatment, but not by iron overload/starvation, indicating the presence of a regulatory pathway involved in Adr1 export and inactivation upon stress conditions. We also observed that CTH2, a gene involved in the mRNA degradation of several iron-containing enzymes, was induced upon Yfh1 depletion. Accordingly, decreased levels of aconitase and succinate dehydrogenase were observed. Nevertheless, their levels were maintained in a Δcth2 mutant even in the absence of Yfh1. From these results we can conclude that, in addition to altering iron homeostasis, frataxin depletion involves drastic metabolic remodeling governed by Adr1 and Cth2 that finally leads to downregulation of iron-sulfur proteins and other proteins involved in respiratory metabolism.
Collapse
Affiliation(s)
- Armando Moreno-Cermeño
- Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina, IRB-Lleida, Universitat de Lleida, Lleida, Spain.
| | - David Alsina
- Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina, IRB-Lleida, Universitat de Lleida, Lleida, Spain
| | - Elisa Cabiscol
- Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina, IRB-Lleida, Universitat de Lleida, Lleida, Spain
| | - Jordi Tamarit
- Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina, IRB-Lleida, Universitat de Lleida, Lleida, Spain
| | - Joaquim Ros
- Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina, IRB-Lleida, Universitat de Lleida, Lleida, Spain.
| |
Collapse
|
41
|
Marelja Z, Mullick Chowdhury M, Dosche C, Hille C, Baumann O, Löhmannsröben HG, Leimkühler S. The L-cysteine desulfurase NFS1 is localized in the cytosol where it provides the sulfur for molybdenum cofactor biosynthesis in humans. PLoS One 2013; 8:e60869. [PMID: 23593335 PMCID: PMC3625234 DOI: 10.1371/journal.pone.0060869] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 03/04/2013] [Indexed: 11/18/2022] Open
Abstract
In humans, the L-cysteine desulfurase NFS1 plays a crucial role in the mitochondrial iron-sulfur cluster biosynthesis and in the thiomodification of mitochondrial and cytosolic tRNAs. We have previously demonstrated that purified NFS1 is able to transfer sulfur to the C-terminal domain of MOCS3, a cytosolic protein involved in molybdenum cofactor biosynthesis and tRNA thiolation. However, no direct evidence existed so far for the interaction of NFS1 and MOCS3 in the cytosol of human cells. Here, we present direct data to show the interaction of NFS1 and MOCS3 in the cytosol of human cells using Förster resonance energy transfer and a split-EGFP system. The colocalization of NFS1 and MOCS3 in the cytosol was confirmed by immunodetection of fractionated cells and localization studies using confocal fluorescence microscopy. Purified NFS1 was used to reconstitute the lacking molybdoenzyme activity of the Neurospora crassa nit-1 mutant, giving additional evidence that NFS1 is the sulfur donor for Moco biosynthesis in eukaryotes in general.
Collapse
Affiliation(s)
- Zvonimir Marelja
- Department of Molecular Enzymology, Institute of Biochemistry, University of Potsdam, Potsdam, Germany
| | - Mita Mullick Chowdhury
- Department of Molecular Enzymology, Institute of Biochemistry, University of Potsdam, Potsdam, Germany
| | - Carsten Dosche
- Department of Physical Chemistry, Institute of Chemistry, University of Potsdam, Potsdam, Germany
| | - Carsten Hille
- Department of Physical Chemistry, Institute of Chemistry, University of Potsdam, Potsdam, Germany
| | - Otto Baumann
- Department of Animal Physiology, Institute of Biochemistry, University of Potsdam, Potsdam, Germany
| | - Hans-Gerd Löhmannsröben
- Department of Physical Chemistry, Institute of Chemistry, University of Potsdam, Potsdam, Germany
| | - Silke Leimkühler
- Department of Molecular Enzymology, Institute of Biochemistry, University of Potsdam, Potsdam, Germany
- * E-mail:
| |
Collapse
|
42
|
Lavecchia A, Di Giovanni C, Cerchia C, Russo A, Russo G, Novellino E. Discovery of a novel small molecule inhibitor targeting the frataxin/ubiquitin interaction via structure-based virtual screening and bioassays. J Med Chem 2013; 56:2861-73. [PMID: 23506486 DOI: 10.1021/jm3017199] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Friedreich's ataxia (FRDA) is an autosomal recessive neuro- and cardiodegenerative disorder for which there are no proven effective treatments. FRDA is caused by decreased expression and/or function of the mitochondrial protein frataxin. Here, we report findings that frataxin is degraded via the ubiquitin-proteasomal pathway and that it is ubiquitinated at residue K(147) in Calu-6 cells. A theoretical model of the frataxin-K(147)/Ub complex, constructed by combining bioinformatics interface predictions with information-driven docking, revealed a hitherto unnoticed, potential ubiquitin-binding domain in frataxin. Through structure-based virtual screening and cell-based assays, we discovered a novel small molecule (compound (+)-11) able to prevent frataxin ubiquitination and degradation. (+)-11 was synthesized and tested for specific binding to frataxin by an UF-LC/MS based ligand-binding assay. Follow-up scaffold-based searches resulted in the identification of a lead series with micromolar activity in disrupting the frataxin/Ub interaction. This study also suggests that frataxin could be a potential target for FRDA drug development.
Collapse
Affiliation(s)
- Antonio Lavecchia
- Dipartimento di Chimica Farmaceutica e Tossicologica, Drug Discovery Laboratory, Università di Napoli Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy.
| | | | | | | | | | | |
Collapse
|
43
|
Simcox JA, McClain DA. Iron and diabetes risk. Cell Metab 2013; 17:329-41. [PMID: 23473030 PMCID: PMC3648340 DOI: 10.1016/j.cmet.2013.02.007] [Citation(s) in RCA: 356] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Revised: 11/03/2012] [Accepted: 11/29/2012] [Indexed: 02/06/2023]
Abstract
Iron overload is a risk factor for diabetes. The link between iron and diabetes was first recognized in pathologic conditions-hereditary hemochromatosis and thalassemia-but high levels of dietary iron also impart diabetes risk. Iron plays a direct and causal role in diabetes pathogenesis mediated both by β cell failure and insulin resistance. Iron also regulates metabolism in most tissues involved in fuel homeostasis, with the adipocyte in particular serving an iron-sensing role. The underlying molecular mechanisms mediating these effects are numerous and incompletely understood but include oxidant stress and modulation of adipokines and intracellular signal transduction pathways.
Collapse
Affiliation(s)
- Judith A Simcox
- Departments of Medicine and Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | | |
Collapse
|
44
|
Li H, Gakh O, Smith DY, Ranatunga WK, Isaya G. Missense mutations linked to friedreich ataxia have different but synergistic effects on mitochondrial frataxin isoforms. J Biol Chem 2013; 288:4116-27. [PMID: 23269675 PMCID: PMC3567662 DOI: 10.1074/jbc.m112.435263] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Revised: 12/21/2012] [Indexed: 12/25/2022] Open
Abstract
Friedreich ataxia is an early-onset multisystemic disease linked to a variety of molecular defects in the nuclear gene FRDA. This gene normally encodes the iron-binding protein frataxin (FXN), which is critical for mitochondrial iron metabolism, global cellular iron homeostasis, and antioxidant protection. In most Friedreich ataxia patients, a large GAA-repeat expansion is present within the first intron of both FRDA alleles, that results in transcriptional silencing ultimately leading to insufficient levels of FXN protein in the mitochondrial matrix and probably other cellular compartments. The lack of FXN in turn impairs incorporation of iron into iron-sulfur cluster and heme cofactors, causing widespread enzymatic deficits and oxidative damage catalyzed by excess labile iron. In a minority of patients, a typical GAA expansion is present in only one FRDA allele, whereas a missense mutation is found in the other allele. Although it is known that the disease course for these patients can be as severe as for patients with two expanded FRDA alleles, the underlying pathophysiological mechanisms are not understood. Human cells normally contain two major mitochondrial isoforms of FXN (FXN(42-210) and FXN(81-210)) that have different biochemical properties and functional roles. Using cell-free systems and different cellular models, we show that two of the most clinically severe FXN point mutations, I154F and W155R, have unique direct and indirect effects on the stability, biogenesis, or catalytic activity of FXN(42-210) and FXN(81-210) under physiological conditions. Our data indicate that frataxin point mutations have complex biochemical effects that synergistically contribute to the pathophysiology of Friedreich ataxia.
Collapse
Affiliation(s)
- Hongqiao Li
- From the Department of Pediatric and Adolescent Medicine and the Department of Biochemistry and Molecular Biology and the Mayo Clinic Children's Center, Mayo Clinic, Rochester, Minnesota 55905
| | - Oleksandr Gakh
- From the Department of Pediatric and Adolescent Medicine and the Department of Biochemistry and Molecular Biology and the Mayo Clinic Children's Center, Mayo Clinic, Rochester, Minnesota 55905
| | - Douglas Y. Smith
- From the Department of Pediatric and Adolescent Medicine and the Department of Biochemistry and Molecular Biology and the Mayo Clinic Children's Center, Mayo Clinic, Rochester, Minnesota 55905
| | - Wasantha K. Ranatunga
- From the Department of Pediatric and Adolescent Medicine and the Department of Biochemistry and Molecular Biology and the Mayo Clinic Children's Center, Mayo Clinic, Rochester, Minnesota 55905
| | - Grazia Isaya
- From the Department of Pediatric and Adolescent Medicine and the Department of Biochemistry and Molecular Biology and the Mayo Clinic Children's Center, Mayo Clinic, Rochester, Minnesota 55905
| |
Collapse
|
45
|
Xia H, Cao Y, Dai X, Marelja Z, Zhou D, Mo R, Al-Mahdawi S, Pook MA, Leimkühler S, Rouault TA, Li K. Novel frataxin isoforms may contribute to the pathological mechanism of Friedreich ataxia. PLoS One 2012; 7:e47847. [PMID: 23082224 PMCID: PMC3474739 DOI: 10.1371/journal.pone.0047847] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 09/21/2012] [Indexed: 12/12/2022] Open
Abstract
Friedreich ataxia (FRDA) is an inherited neurodegenerative disease caused by frataxin (FXN) deficiency. The nervous system and heart are the most severely affected tissues. However, highly mitochondria-dependent tissues, such as kidney and liver, are not obviously affected, although the abundance of FXN is normally high in these tissues. In this study we have revealed two novel FXN isoforms (II and III), which are specifically expressed in affected cerebellum and heart tissues, respectively, and are functional in vitro and in vivo. Increasing the abundance of the heart-specific isoform III significantly increased the mitochondrial aconitase activity, while over-expression of the cerebellum-specific isoform II protected against oxidative damage of Fe-S cluster-containing aconitase. Further, we observed that the protein level of isoform III decreased in FRDA patient heart, while the mRNA level of isoform II decreased more in FRDA patient cerebellum compared to total FXN mRNA. Our novel findings are highly relevant to understanding the mechanism of tissue-specific pathology in FRDA.
Collapse
Affiliation(s)
- Haiyan Xia
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Yun Cao
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Xiaoman Dai
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Zvonimir Marelja
- Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| | - Di Zhou
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Ran Mo
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Sahar Al-Mahdawi
- Division of Biosciences, School of Health Sciences and Social Care, Brunel University, Uxbridge, United Kingdom
| | - Mark A. Pook
- Division of Biosciences, School of Health Sciences and Social Care, Brunel University, Uxbridge, United Kingdom
| | - Silke Leimkühler
- Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| | - Tracey A. Rouault
- Molecular Medicine Program, National Institute of Child Health and Human Development, Bethesda, Maryland, United States of America
| | - Kuanyu Li
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| |
Collapse
|
46
|
Anderson CP, Shen M, Eisenstein RS, Leibold EA. Mammalian iron metabolism and its control by iron regulatory proteins. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1823:1468-83. [PMID: 22610083 DOI: 10.1016/j.bbamcr.2012.05.010] [Citation(s) in RCA: 346] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Revised: 05/07/2012] [Accepted: 05/11/2012] [Indexed: 02/06/2023]
Abstract
Cellular iron homeostasis is maintained by iron regulatory proteins 1 and 2 (IRP1 and IRP2). IRPs bind to iron-responsive elements (IREs) located in the untranslated regions of mRNAs encoding protein involved in iron uptake, storage, utilization and export. Over the past decade, significant progress has been made in understanding how IRPs are regulated by iron-dependent and iron-independent mechanisms and the pathological consequences of IRP2 deficiency in mice. The identification of novel IREs involved in diverse cellular pathways has revealed that the IRP-IRE network extends to processes other than iron homeostasis. A mechanistic understanding of IRP regulation will likely yield important insights into the basis of disorders of iron metabolism. This article is part of a Special Issue entitled: Cell Biology of Metals.
Collapse
Affiliation(s)
- Cole P Anderson
- Department of Oncological Sciences, University of Utah, 15 N. 2030 E., Salt Lake City, UT 84112, USA
| | | | | | | |
Collapse
|
47
|
Shan Y, Cortopassi G. HSC20 interacts with frataxin and is involved in iron-sulfur cluster biogenesis and iron homeostasis. Hum Mol Genet 2012; 21:1457-69. [PMID: 22171070 PMCID: PMC3298274 DOI: 10.1093/hmg/ddr582] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 11/30/2011] [Accepted: 12/06/2011] [Indexed: 12/20/2022] Open
Abstract
Friedreich's ataxia is a neurodegenerative disorder caused by mutations in the frataxin gene that produces a predominantly mitochondrial protein whose primary function appears to be mitochondrial iron-sulfur cluster (ISC) biosynthesis. Previously we demonstrated that frataxin interacts with multiple components of the mammalian ISC assembly machinery. Here we demonstrate that frataxin interacts with the mammalian mitochondrial chaperone HSC20. We show that this interaction is iron-dependent. We also show that like frataxin, HSC20 interacts with multiple proteins involved in ISC biogenesis including the ISCU/Nfs1 ISC biogenesis complex and the GRP75 ISC chaperone. Furthermore, knockdown of HSC20 caused functional defects in activity of mitochondrial ISC-containing enzymes and also defects in ISC protein expression. Alterations up or down of frataxin expression caused compensatory changes in HSC20 expression inversely, as expected of two cooperating proteins operating in the same pathway and suggesting a potential therapeutic strategy for the disease. Knockdown of HSC20 altered cytosolic and mitochondrial iron pools and increased the expression of transferrin receptor 1 and iron regulatory protein 2 consistent with decreased iron bioavailability. These results indicate that HSC20 interacts with frataxin structurally and functionally and is important for ISC biogenesis and iron homeostasis in mammals. Furthermore, they suggest that HSC20 may act late in the ISC pathway as a chaperone in ISC delivery to apoproteins and that HSC20 should be included in multi-protein complex studies of mammalian ISC biogenesis.
Collapse
Affiliation(s)
| | - Gino Cortopassi
- School of Veterinary Medicine, Department of Molecular Biosciences, University of California, 1120 Haring Hall, Davis, CA 95616, USA
| |
Collapse
|
48
|
Brigelius-Flohé R, Flohé L. Basic principles and emerging concepts in the redox control of transcription factors. Antioxid Redox Signal 2011; 15:2335-81. [PMID: 21194351 PMCID: PMC3166203 DOI: 10.1089/ars.2010.3534] [Citation(s) in RCA: 422] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Convincing concepts of redox control of gene transcription have been worked out for prokaryotes and lower eukaryotes, whereas the knowledge on complex mammalian systems still resembles a patchwork of poorly connected findings. The article, therefore, reviews principles of redox regulation with special emphasis on chemical feasibility, kinetic requirements, specificity, and physiological context, taking well investigated mammalian transcription factor systems, nuclear transcription factor of bone marrow-derived lymphocytes (NF-κB), and kelch-like ECH-associated protein-1 (Keap1)/Nrf2, as paradigms. Major conclusions are that (i) direct signaling by free radicals is restricted to O(2)•- and •NO and can be excluded for fast reacting radicals such as •OH, •OR, or Cl•; (ii) oxidant signals are H(2)O(2), enzymatically generated lipid hydroperoxides, and peroxynitrite; (iii) free radical damage is sensed via generation of Michael acceptors; (iv) protein thiol oxidation/alkylation is the prominent mechanism to modulate function; (v) redox sensors must be thiol peroxidases by themselves or proteins with similarly reactive cysteine or selenocysteine (Sec) residues to kinetically compete with glutathione peroxidase (GPx)- and peroxiredoxin (Prx)-type peroxidases or glutathione-S-transferases, respectively, a postulate that still has to be verified for putative mammalian sensors. S-transferases and Prxs are considered for system complementation. The impact of NF-κB and Nrf2 on hormesis, management of inflammatory diseases, and cancer prevention is critically discussed.
Collapse
Affiliation(s)
- Regina Brigelius-Flohé
- Department Biochemistry of Micronutrients, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, Nuthetal, Germany.
| | | |
Collapse
|
49
|
Bayot A, Santos R, Camadro JM, Rustin P. Friedreich's ataxia: the vicious circle hypothesis revisited. BMC Med 2011; 9:112. [PMID: 21985033 PMCID: PMC3198887 DOI: 10.1186/1741-7015-9-112] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Accepted: 10/11/2011] [Indexed: 12/18/2022] Open
Abstract
Friedreich's ataxia, the most frequent progressive autosomal recessive disorder involving the central and peripheral nervous systems, is mostly associated with unstable expansion of GAA trinucleotide repeats in the first intron of the FXN gene, which encodes the mitochondrial frataxin protein. Since FXN was shown to be involved in Friedreich's ataxia in the late 1990s, the consequence of frataxin loss of function has generated vigorous debate. Very early on we suggested a unifying hypothesis according to which frataxin deficiency leads to a vicious circle of faulty iron handling, impaired iron-sulphur cluster synthesis and increased oxygen radical production. However, data from cell and animal models now indicate that iron accumulation is an inconsistent and late event and that frataxin deficiency does not always impair the activity of iron-sulphur cluster-containing proteins. In contrast, frataxin deficiency appears to be consistently associated with increased sensitivity to reactive oxygen species as opposed to increased oxygen radical production. By compiling the findings of fundamental research and clinical observations we defend here the opinion that the very first consequence of frataxin depletion is indeed an abnormal oxidative status which initiates the pathogenic mechanism underlying Friedreich's ataxia.
Collapse
Affiliation(s)
- Aurélien Bayot
- Inserm, U676, Physiopathology and Therapy of Mitochondrial Diseases Laboratory, CHU - Hôpital Robert Debré, 48, boulevard Sérurier, F-75019 Paris, France
- Faculté de médecine Denis Diderot, Université Paris-Diderot, IFR02, 16, rue Henri Huchard, F-75018, Paris, France
| | - Renata Santos
- Institut Jacques Monod (UMR 7592 CNRS-Université Paris-Diderot), Mitochondria, Metals and Oxidative Stress Laboratory, Bâtiment Buffon - 15, rue Hélène Brion, F-75205 Paris, Cedex 13, France
| | - Jean-Michel Camadro
- Institut Jacques Monod (UMR 7592 CNRS-Université Paris-Diderot), Mitochondria, Metals and Oxidative Stress Laboratory, Bâtiment Buffon - 15, rue Hélène Brion, F-75205 Paris, Cedex 13, France
| | - Pierre Rustin
- Inserm, U676, Physiopathology and Therapy of Mitochondrial Diseases Laboratory, CHU - Hôpital Robert Debré, 48, boulevard Sérurier, F-75019 Paris, France
- Faculté de médecine Denis Diderot, Université Paris-Diderot, IFR02, 16, rue Henri Huchard, F-75018, Paris, France
| |
Collapse
|
50
|
Boehm T, Scheiber-Mojdehkar B, Kluge B, Goldenberg H, Laccone F, Sturm B. Variations of frataxin protein levels in normal individuals. Neurol Sci 2011; 32:327-30. [PMID: 20506029 DOI: 10.1007/s10072-010-0326-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2009] [Accepted: 04/13/2010] [Indexed: 10/19/2022]
Abstract
Friedreich's ataxia (FRDA) is the most common of the inherited ataxias and is associated with GAA trinucleotide repeat expansions within the first intron of the frataxin (FXN) gene. There are expanded FXN alleles from 66 to 1,700 GAA·TTC repeats in FRDA patients and correlations between number of GAA repeats and frataxin protein levels are assumed. Here, we present for the first time frataxin protein levels as well as analysis of GAA triplet repeats in the FXN gene in a population of 50 healthy Austrian people. Frataxin protein levels were measured in lymphocytes from blood samples by ELISA and GAA repeats were analyzed by capillary electrophoresis. Rather unexpectedly, we found a high variation of frataxin protein levels among the individuals. In addition, there was no correlation between frataxin levels, GAA repeats, age and sex in this group. However, these findings are of great importance for better characterization of the disease.
Collapse
Affiliation(s)
- Therese Boehm
- Department of Medical Chemistry, Medical University of Vienna, Waehringerstr. 10, 1090, Vienna, Austria
| | | | | | | | | | | |
Collapse
|