1
|
Donoghue S, Wright J, Voss AK, Lockhart PJ, Amor DJ. The Mendelian disorders of chromatin machinery: Harnessing metabolic pathways and therapies for treatment. Mol Genet Metab 2024; 142:108360. [PMID: 38428378 DOI: 10.1016/j.ymgme.2024.108360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 02/25/2024] [Accepted: 02/26/2024] [Indexed: 03/03/2024]
Abstract
The Mendelian disorders of chromatin machinery (MDCMs) represent a distinct subgroup of disorders that present with neurodevelopmental disability. The chromatin machinery regulates gene expression by a range of mechanisms, including by post-translational modification of histones, responding to histone marks, and remodelling nucleosomes. Some of the MDCMs that impact on histone modification may have potential therapeutic interventions. Two potential treatment strategies are to enhance the intracellular pool of metabolites that can act as substrates for histone modifiers and the use of medications that may inhibit or promote the modification of histone residues to influence gene expression. In this article we discuss the influence and potential treatments of histone modifications involving histone acetylation and histone methylation. Genomic technologies are facilitating earlier diagnosis of many Mendelian disorders, providing potential opportunities for early treatment from infancy. This has parallels with how inborn errors of metabolism have been afforded early treatment with newborn screening. Before this promise can be fulfilled, we require greater understanding of the biochemical fingerprint of these conditions, which may provide opportunities to supplement metabolites that can act as substrates for chromatin modifying enzymes. Importantly, understanding the metabolomic profile of affected individuals may also provide disorder-specific biomarkers that will be critical for demonstrating efficacy of treatment, as treatment response may not be able to be accurately assessed by clinical measures.
Collapse
Affiliation(s)
- Sarah Donoghue
- Murdoch Children's Research Institute, Parkville 3052, Australia; Department of Biochemical Genetics, Victorian Clinical Genetics Services, Parkville 3052, Australia; Department of Paediatrics, The University of Melbourne, Parkville 3052, Australia.
| | - Jordan Wright
- Murdoch Children's Research Institute, Parkville 3052, Australia; Department of Paediatrics, The University of Melbourne, Parkville 3052, Australia
| | - Anne K Voss
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Melbourne 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3052, Australia
| | - Paul J Lockhart
- Murdoch Children's Research Institute, Parkville 3052, Australia; Department of Paediatrics, The University of Melbourne, Parkville 3052, Australia
| | - David J Amor
- Murdoch Children's Research Institute, Parkville 3052, Australia; Department of Paediatrics, The University of Melbourne, Parkville 3052, Australia
| |
Collapse
|
2
|
Levi S, Ripamonti M, Moro AS, Cozzi A. Iron imbalance in neurodegeneration. Mol Psychiatry 2024; 29:1139-1152. [PMID: 38212377 PMCID: PMC11176077 DOI: 10.1038/s41380-023-02399-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/13/2024]
Abstract
Iron is an essential element for the development and functionality of the brain, and anomalies in its distribution and concentration in brain tissue have been found to be associated with the most frequent neurodegenerative diseases. When magnetic resonance techniques allowed iron quantification in vivo, it was confirmed that the alteration of brain iron homeostasis is a common feature of many neurodegenerative diseases. However, whether iron is the main actor in the neurodegenerative process, or its alteration is a consequence of the degenerative process is still an open question. Because the different iron-related pathogenic mechanisms are specific for distinctive diseases, identifying the molecular mechanisms common to the various pathologies could represent a way to clarify this complex topic. Indeed, both iron overload and iron deficiency have profound consequences on cellular functioning, and both contribute to neuronal death processes in different manners, such as promoting oxidative damage, a loss of membrane integrity, a loss of proteostasis, and mitochondrial dysfunction. In this review, with the attempt to elucidate the consequences of iron dyshomeostasis for brain health, we summarize the main pathological molecular mechanisms that couple iron and neuronal death.
Collapse
Affiliation(s)
- Sonia Levi
- Vita-Salute San Raffaele University, Milano, Italy.
- IRCCS San Raffaele Scientific Institute, Milano, Italy.
| | | | - Andrea Stefano Moro
- Vita-Salute San Raffaele University, Milano, Italy
- Department of Psychology, Sigmund Freud University, Milan, Italy
| | - Anna Cozzi
- IRCCS San Raffaele Scientific Institute, Milano, Italy
| |
Collapse
|
3
|
Suárez-Carrillo A, Álvarez-Córdoba M, Romero-González A, Talaverón-Rey M, Povea-Cabello S, Cilleros-Holgado P, Piñero-Pérez R, Reche-López D, Gómez-Fernández D, Romero-Domínguez JM, Munuera-Cabeza M, Díaz A, González-Granero S, García-Verdugo JM, Sánchez-Alcázar JA. Antioxidants Prevent Iron Accumulation and Lipid Peroxidation, but Do Not Correct Autophagy Dysfunction or Mitochondrial Bioenergetics in Cellular Models of BPAN. Int J Mol Sci 2023; 24:14576. [PMID: 37834028 PMCID: PMC11340724 DOI: 10.3390/ijms241914576] [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: 05/25/2023] [Revised: 09/06/2023] [Accepted: 09/08/2023] [Indexed: 10/15/2023] Open
Abstract
Neurodegeneration with brain iron accumulation (NBIA) is a group of rare neurogenetic disorders frequently associated with iron accumulation in the basal nuclei of the brain. Among NBIA subtypes, β-propeller protein-associated neurodegeneration (BPAN) is associated with mutations in the autophagy gene WDR45. The aim of this study was to demonstrate the autophagic defects and secondary pathological consequences in cellular models derived from two patients harboring WDR45 mutations. Both protein and mRNA expression levels of WDR45 were decreased in patient-derived fibroblasts. In addition, the increase of LC3B upon treatments with autophagy inducers or inhibitors was lower in mutant cells compared to control cells, suggesting decreased autophagosome formation and impaired autophagic flux. A transmission electron microscopy (TEM) analysis showed mitochondrial vacuolization associated with the accumulation of lipofuscin-like aggregates containing undegraded material. Autophagy dysregulation was also associated with iron accumulation and lipid peroxidation. In addition, mutant fibroblasts showed altered mitochondrial bioenergetics. Antioxidants such as pantothenate, vitamin E and α-lipoic prevented lipid peroxidation and iron accumulation. However, antioxidants were not able to correct the expression levels of WDR45, neither the autophagy defect nor cell bioenergetics. Our study demonstrated that WDR45 mutations in BPAN cellular models impaired autophagy, iron metabolism and cell bioenergetics. Antioxidants partially improved cell physiopathology; however, autophagy and cell bioenergetics remained affected.
Collapse
Affiliation(s)
- Alejandra Suárez-Carrillo
- Centro Andaluz de Biología del Desarrollo, ABD-CSIC-Universidad Pablo de Olavide, 41013 Sevilla, Spain; (A.S.-C.); (M.Á.-C.); (A.R.-G.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (R.P.-P.); (D.R.-L.); (D.G.-F.); (J.M.R.-D.); (M.M.-C.)
| | - Mónica Álvarez-Córdoba
- Centro Andaluz de Biología del Desarrollo, ABD-CSIC-Universidad Pablo de Olavide, 41013 Sevilla, Spain; (A.S.-C.); (M.Á.-C.); (A.R.-G.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (R.P.-P.); (D.R.-L.); (D.G.-F.); (J.M.R.-D.); (M.M.-C.)
| | - Ana Romero-González
- Centro Andaluz de Biología del Desarrollo, ABD-CSIC-Universidad Pablo de Olavide, 41013 Sevilla, Spain; (A.S.-C.); (M.Á.-C.); (A.R.-G.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (R.P.-P.); (D.R.-L.); (D.G.-F.); (J.M.R.-D.); (M.M.-C.)
| | - Marta Talaverón-Rey
- Centro Andaluz de Biología del Desarrollo, ABD-CSIC-Universidad Pablo de Olavide, 41013 Sevilla, Spain; (A.S.-C.); (M.Á.-C.); (A.R.-G.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (R.P.-P.); (D.R.-L.); (D.G.-F.); (J.M.R.-D.); (M.M.-C.)
| | - Suleva Povea-Cabello
- Centro Andaluz de Biología del Desarrollo, ABD-CSIC-Universidad Pablo de Olavide, 41013 Sevilla, Spain; (A.S.-C.); (M.Á.-C.); (A.R.-G.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (R.P.-P.); (D.R.-L.); (D.G.-F.); (J.M.R.-D.); (M.M.-C.)
| | - Paula Cilleros-Holgado
- Centro Andaluz de Biología del Desarrollo, ABD-CSIC-Universidad Pablo de Olavide, 41013 Sevilla, Spain; (A.S.-C.); (M.Á.-C.); (A.R.-G.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (R.P.-P.); (D.R.-L.); (D.G.-F.); (J.M.R.-D.); (M.M.-C.)
| | - Rocío Piñero-Pérez
- Centro Andaluz de Biología del Desarrollo, ABD-CSIC-Universidad Pablo de Olavide, 41013 Sevilla, Spain; (A.S.-C.); (M.Á.-C.); (A.R.-G.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (R.P.-P.); (D.R.-L.); (D.G.-F.); (J.M.R.-D.); (M.M.-C.)
| | - Diana Reche-López
- Centro Andaluz de Biología del Desarrollo, ABD-CSIC-Universidad Pablo de Olavide, 41013 Sevilla, Spain; (A.S.-C.); (M.Á.-C.); (A.R.-G.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (R.P.-P.); (D.R.-L.); (D.G.-F.); (J.M.R.-D.); (M.M.-C.)
| | - David Gómez-Fernández
- Centro Andaluz de Biología del Desarrollo, ABD-CSIC-Universidad Pablo de Olavide, 41013 Sevilla, Spain; (A.S.-C.); (M.Á.-C.); (A.R.-G.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (R.P.-P.); (D.R.-L.); (D.G.-F.); (J.M.R.-D.); (M.M.-C.)
| | - José Manuel Romero-Domínguez
- Centro Andaluz de Biología del Desarrollo, ABD-CSIC-Universidad Pablo de Olavide, 41013 Sevilla, Spain; (A.S.-C.); (M.Á.-C.); (A.R.-G.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (R.P.-P.); (D.R.-L.); (D.G.-F.); (J.M.R.-D.); (M.M.-C.)
| | - Manuel Munuera-Cabeza
- Centro Andaluz de Biología del Desarrollo, ABD-CSIC-Universidad Pablo de Olavide, 41013 Sevilla, Spain; (A.S.-C.); (M.Á.-C.); (A.R.-G.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (R.P.-P.); (D.R.-L.); (D.G.-F.); (J.M.R.-D.); (M.M.-C.)
| | - Antonio Díaz
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, NY 10461, USA;
- Institute for Aging Studies, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Susana González-Granero
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia and CIBERNED-ISCIII, 46100 Valencia, Spain; (S.G.-G.); (J.M.G.-V.)
| | - José Manuel García-Verdugo
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia and CIBERNED-ISCIII, 46100 Valencia, Spain; (S.G.-G.); (J.M.G.-V.)
| | - José A. Sánchez-Alcázar
- Centro Andaluz de Biología del Desarrollo, ABD-CSIC-Universidad Pablo de Olavide, 41013 Sevilla, Spain; (A.S.-C.); (M.Á.-C.); (A.R.-G.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (R.P.-P.); (D.R.-L.); (D.G.-F.); (J.M.R.-D.); (M.M.-C.)
| |
Collapse
|
4
|
Álvarez-Córdoba M, Talaverón-Rey M, Povea-Cabello S, Cilleros-Holgado P, Gómez-Fernández D, Piñero-Pérez R, Reche-López D, Munuera-Cabeza M, Suárez-Carrillo A, Romero-González A, Romero-Domínguez JM, López-Cabrera A, Armengol JÁ, Sánchez-Alcázar JA. Patient-Derived Cellular Models for Polytarget Precision Medicine in Pantothenate Kinase-Associated Neurodegeneration. Pharmaceuticals (Basel) 2023; 16:1359. [PMID: 37895830 PMCID: PMC10609847 DOI: 10.3390/ph16101359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 09/21/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023] Open
Abstract
The term neurodegeneration with brain iron accumulation (NBIA) brings together a broad set of progressive and disabling neurological genetic disorders in which iron is deposited preferentially in certain areas of the brain. Among NBIA disorders, the most frequent subtype is pantothenate kinase-associated neurodegeneration (PKAN) caused by pathologic variants in the PANK2 gene codifying the enzyme pantothenate kinase 2 (PANK2). To date, there are no effective treatments to stop the progression of these diseases. This review discusses the utility of patient-derived cell models as a valuable tool for the identification of pharmacological or natural compounds for implementing polytarget precision medicine in PKAN. Recently, several studies have described that PKAN patient-derived fibroblasts present the main pathological features associated with the disease including intracellular iron overload. Interestingly, treatment of mutant cell cultures with various supplements such as pantothenate, pantethine, vitamin E, omega 3, α-lipoic acid L-carnitine or thiamine, improved all pathophysiological alterations in PKAN fibroblasts with residual expression of the PANK2 enzyme. The information provided by pharmacological screenings in patient-derived cellular models can help optimize therapeutic strategies in individual PKAN patients.
Collapse
Affiliation(s)
- Mónica Álvarez-Córdoba
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide University), 41013 Seville, Spain; (M.Á.-C.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (D.G.-F.); (R.P.-P.); (D.R.-L.); (M.M.-C.); (A.S.-C.); (A.R.-G.); (J.M.R.-D.); (A.L.-C.)
| | - Marta Talaverón-Rey
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide University), 41013 Seville, Spain; (M.Á.-C.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (D.G.-F.); (R.P.-P.); (D.R.-L.); (M.M.-C.); (A.S.-C.); (A.R.-G.); (J.M.R.-D.); (A.L.-C.)
| | - Suleva Povea-Cabello
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide University), 41013 Seville, Spain; (M.Á.-C.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (D.G.-F.); (R.P.-P.); (D.R.-L.); (M.M.-C.); (A.S.-C.); (A.R.-G.); (J.M.R.-D.); (A.L.-C.)
| | - Paula Cilleros-Holgado
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide University), 41013 Seville, Spain; (M.Á.-C.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (D.G.-F.); (R.P.-P.); (D.R.-L.); (M.M.-C.); (A.S.-C.); (A.R.-G.); (J.M.R.-D.); (A.L.-C.)
| | - David Gómez-Fernández
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide University), 41013 Seville, Spain; (M.Á.-C.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (D.G.-F.); (R.P.-P.); (D.R.-L.); (M.M.-C.); (A.S.-C.); (A.R.-G.); (J.M.R.-D.); (A.L.-C.)
| | - Rocío Piñero-Pérez
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide University), 41013 Seville, Spain; (M.Á.-C.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (D.G.-F.); (R.P.-P.); (D.R.-L.); (M.M.-C.); (A.S.-C.); (A.R.-G.); (J.M.R.-D.); (A.L.-C.)
| | - Diana Reche-López
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide University), 41013 Seville, Spain; (M.Á.-C.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (D.G.-F.); (R.P.-P.); (D.R.-L.); (M.M.-C.); (A.S.-C.); (A.R.-G.); (J.M.R.-D.); (A.L.-C.)
| | - Manuel Munuera-Cabeza
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide University), 41013 Seville, Spain; (M.Á.-C.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (D.G.-F.); (R.P.-P.); (D.R.-L.); (M.M.-C.); (A.S.-C.); (A.R.-G.); (J.M.R.-D.); (A.L.-C.)
| | - Alejandra Suárez-Carrillo
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide University), 41013 Seville, Spain; (M.Á.-C.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (D.G.-F.); (R.P.-P.); (D.R.-L.); (M.M.-C.); (A.S.-C.); (A.R.-G.); (J.M.R.-D.); (A.L.-C.)
| | - Ana Romero-González
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide University), 41013 Seville, Spain; (M.Á.-C.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (D.G.-F.); (R.P.-P.); (D.R.-L.); (M.M.-C.); (A.S.-C.); (A.R.-G.); (J.M.R.-D.); (A.L.-C.)
| | - Jose Manuel Romero-Domínguez
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide University), 41013 Seville, Spain; (M.Á.-C.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (D.G.-F.); (R.P.-P.); (D.R.-L.); (M.M.-C.); (A.S.-C.); (A.R.-G.); (J.M.R.-D.); (A.L.-C.)
| | - Alejandra López-Cabrera
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide University), 41013 Seville, Spain; (M.Á.-C.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (D.G.-F.); (R.P.-P.); (D.R.-L.); (M.M.-C.); (A.S.-C.); (A.R.-G.); (J.M.R.-D.); (A.L.-C.)
| | - José Ángel Armengol
- Department of Physiology, Anatomy and Cellular Biology, Pablo de Olavide University, 41013 Seville, Spain;
| | - José Antonio Sánchez-Alcázar
- Andalusian Centre for Developmental Biology (CABD-CSIC-Pablo de Olavide University), 41013 Seville, Spain; (M.Á.-C.); (M.T.-R.); (S.P.-C.); (P.C.-H.); (D.G.-F.); (R.P.-P.); (D.R.-L.); (M.M.-C.); (A.S.-C.); (A.R.-G.); (J.M.R.-D.); (A.L.-C.)
| |
Collapse
|
5
|
Talaverón-Rey M, Álvarez-Córdoba M, Villalón-García I, Povea-Cabello S, Suárez-Rivero JM, Gómez-Fernández D, Romero-González A, Suárez-Carrillo A, Munuera-Cabeza M, Cilleros-Holgado P, Reche-López D, Piñero-Pérez R, Sánchez-Alcázar JA. Alpha-lipoic acid supplementation corrects pathological alterations in cellular models of pantothenate kinase-associated neurodegeneration with residual PANK2 expression levels. Orphanet J Rare Dis 2023; 18:80. [PMID: 37046296 PMCID: PMC10091671 DOI: 10.1186/s13023-023-02687-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 04/02/2023] [Indexed: 04/14/2023] Open
Abstract
BACKGROUND Neurodegeneration with brain iron accumulation (NBIA) disorders are a group of neurodegenerative diseases that have in common the accumulation of iron in the basal nuclei of the brain which are essential components of the extrapyramidal system. Frequent symptoms are progressive spasticity, dystonia, muscle rigidity, neuropsychiatric symptoms, and retinal degeneration or optic nerve atrophy. One of the most prevalent subtypes of NBIA is Pantothenate kinase-associated neurodegeneration (PKAN). It is caused by pathogenic variants in the gene of pantothenate kinase 2 (PANK2) which encodes the enzyme responsible for the first reaction on the coenzyme A (CoA) biosynthesis pathway. Thus, deficient PANK2 activity induces CoA deficiency as well as low expression levels of 4'-phosphopantetheinyl proteins which are essential for mitochondrial metabolism. METHODS This study is aimed at evaluating the role of alpha-lipoic acid (α-LA) in reversing the pathological alterations in fibroblasts and induced neurons derived from PKAN patients. Iron accumulation, lipid peroxidation, transcript and protein expression levels of PANK2, mitochondrial ACP (mtACP), 4''-phosphopantetheinyl and lipoylated proteins, as well as pyruvate dehydrogenase (PDH) and Complex I activity were examined. RESULTS Treatment with α-LA was able to correct all pathological alterations in responsive mutant fibroblasts with residual PANK2 enzyme expression. However, α-LA had no effect on mutant fibroblasts with truncated/incomplete protein expression. The positive effect of α-LA in particular pathogenic variants was also confirmed in induced neurons derived from mutant fibroblasts. CONCLUSIONS Our results suggest that α-LA treatment can increase the expression levels of PANK2 and reverse the mutant phenotype in PANK2 responsive pathogenic variants. The existence of residual enzyme expression in some affected individuals raises the possibility of treatment using high dose of α-LA.
Collapse
Affiliation(s)
- Marta Talaverón-Rey
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-UPO), Universidad Pablo de Olavide, 41013, Seville, Spain
| | - Mónica Álvarez-Córdoba
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-UPO), Universidad Pablo de Olavide, 41013, Seville, Spain
| | - Irene Villalón-García
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-UPO), Universidad Pablo de Olavide, 41013, Seville, Spain
| | - Suleva Povea-Cabello
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-UPO), Universidad Pablo de Olavide, 41013, Seville, Spain
| | - Juan M Suárez-Rivero
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-UPO), Universidad Pablo de Olavide, 41013, Seville, Spain
| | - David Gómez-Fernández
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-UPO), Universidad Pablo de Olavide, 41013, Seville, Spain
| | - Ana Romero-González
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-UPO), Universidad Pablo de Olavide, 41013, Seville, Spain
| | - Alejandra Suárez-Carrillo
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-UPO), Universidad Pablo de Olavide, 41013, Seville, Spain
| | - Manuel Munuera-Cabeza
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-UPO), Universidad Pablo de Olavide, 41013, Seville, Spain
| | - Paula Cilleros-Holgado
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-UPO), Universidad Pablo de Olavide, 41013, Seville, Spain
| | - Diana Reche-López
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-UPO), Universidad Pablo de Olavide, 41013, Seville, Spain
| | - Rocío Piñero-Pérez
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-UPO), Universidad Pablo de Olavide, 41013, Seville, Spain
| | - José A Sánchez-Alcázar
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-UPO), Universidad Pablo de Olavide, 41013, Seville, Spain.
| |
Collapse
|
6
|
Cavestro C, Diodato D, Tiranti V, Di Meo I. Inherited Disorders of Coenzyme A Biosynthesis: Models, Mechanisms, and Treatments. Int J Mol Sci 2023; 24:ijms24065951. [PMID: 36983025 PMCID: PMC10054636 DOI: 10.3390/ijms24065951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/09/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Coenzyme A (CoA) is a vital and ubiquitous cofactor required in a vast number of enzymatic reactions and cellular processes. To date, four rare human inborn errors of CoA biosynthesis have been described. These disorders have distinct symptoms, although all stem from variants in genes that encode enzymes involved in the same metabolic process. The first and last enzymes catalyzing the CoA biosynthetic pathway are associated with two neurological conditions, namely pantothenate kinase-associated neurodegeneration (PKAN) and COASY protein-associated neurodegeneration (CoPAN), which belong to the heterogeneous group of neurodegenerations with brain iron accumulation (NBIA), while the second and third enzymes are linked to a rapidly fatal dilated cardiomyopathy. There is still limited information about the pathogenesis of these diseases, and the knowledge gaps need to be resolved in order to develop potential therapeutic approaches. This review aims to provide a summary of CoA metabolism and functions, and a comprehensive overview of what is currently known about disorders associated with its biosynthesis, including available preclinical models, proposed pathomechanisms, and potential therapeutic approaches.
Collapse
Affiliation(s)
- Chiara Cavestro
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20126 Milan, Italy
| | - Daria Diodato
- Unit of Muscular and Neurodegenerative Disorders, Ospedale Pediatrico Bambino Gesù, 00165 Rome, Italy
| | - Valeria Tiranti
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20126 Milan, Italy
| | - Ivano Di Meo
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20126 Milan, Italy
| |
Collapse
|
7
|
Bi-Allelic Mutations in Zebrafish pank2 Gene Lead to Testicular Atrophy and Perturbed Behavior without Signs of Neurodegeneration. Int J Mol Sci 2022; 23:ijms232112914. [DOI: 10.3390/ijms232112914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 10/01/2022] [Accepted: 10/22/2022] [Indexed: 11/17/2022] Open
Abstract
Coenzyme A (CoA) is an essential cofactor in all living organisms, being involved in a large number of chemical reactions. Sequence variations in pantothenate kinase 2 (PANK2), the first enzyme of CoA biosynthesis, are found in patients affected by Pantothenate Kinase Associated Neurodegeneration (PKAN), one of the most common forms of neurodegeneration, with brain iron accumulation. Knowledge about the biochemical and molecular features of this disorder has increased a lot in recent years. Nonetheless, the main culprit of the pathology is not well defined, and no treatment option is available yet. In order to contribute to the understanding of this disease and facilitate the search for therapies, we explored the potential of the zebrafish animal model and generated lines carrying biallelic mutations in the pank2 gene. The phenotypic characterization of pank2-mutant embryos revealed anomalies in the development of venous vascular structures and germ cells. Adult fish showed testicular atrophy and altered behavioral response in an anxiety test but no evident signs of neurodegeneration. The study suggests that selected cell and tissue types show a higher vulnerability to pank2 deficiency in zebrafish. Deciphering the biological basis of this phenomenon could provide relevant clues for better understanding and treating PKAN.
Collapse
|
8
|
Álvarez-Córdoba M, Reche-López D, Cilleros-Holgado P, Talaverón-Rey M, Villalón-García I, Povea-Cabello S, Suárez-Rivero JM, Suárez-Carrillo A, Munuera-Cabeza M, Piñero-Pérez R, Sánchez-Alcázar JA. Therapeutic approach with commercial supplements for pantothenate kinase-associated neurodegeneration with residual PANK2 expression levels. Orphanet J Rare Dis 2022; 17:311. [PMID: 35945593 PMCID: PMC9364590 DOI: 10.1186/s13023-022-02465-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/24/2022] [Indexed: 12/24/2022] Open
Abstract
Background Neurodegeneration with brain iron accumulation (NBIA) is a group of rare neurogenetic disorders frequently associated with iron accumulation in the basal nuclei of the brain characterized by progressive spasticity, dystonia, muscle rigidity, neuropsychiatric symptoms, and retinal degeneration or optic nerve atrophy. Pantothenate kinase-associated neurodegeneration (PKAN) is one of the most widespread NBIA subtypes. It is caused by mutations in the gene of pantothenate kinase 2 (PANK2) that result in dysfunction in PANK2 enzyme activity, with consequent deficiency of coenzyme A (CoA) biosynthesis, as well as low levels of essential metabolic intermediates such as 4′-phosphopantetheine, a necessary cofactor for essential cytosolic and mitochondrial proteins. Methods In this manuscript, we examined the therapeutic effectiveness of pantothenate, panthetine, antioxidants (vitamin E and omega 3) and mitochondrial function boosting supplements (L-carnitine and thiamine) in mutant PANK2 cells with residual expression levels. Results Commercial supplements, pantothenate, pantethine, vitamin E, omega 3, carnitine and thiamine were able to eliminate iron accumulation, increase PANK2, mtACP, and NFS1 expression levels and improve pathological alterations in mutant cells with residual PANK2 expression levels. Conclusion Our results suggest that several commercial compounds are indeed able to significantly correct the mutant phenotype in cellular models of PKAN. These compounds alone or in combinations are of common use in clinical practice and may be useful for the treatment of PKAN patients with residual enzyme expression levels. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-022-02465-9.
Collapse
Affiliation(s)
- Mónica Álvarez-Córdoba
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013, Sevilla, Spain
| | - Diana Reche-López
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013, Sevilla, Spain
| | - Paula Cilleros-Holgado
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013, Sevilla, Spain
| | - Marta Talaverón-Rey
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013, Sevilla, Spain
| | - Irene Villalón-García
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013, Sevilla, Spain
| | - Suleva Povea-Cabello
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013, Sevilla, Spain
| | - Juan M Suárez-Rivero
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013, Sevilla, Spain
| | - Alejandra Suárez-Carrillo
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013, Sevilla, Spain
| | - Manuel Munuera-Cabeza
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013, Sevilla, Spain
| | - Rocío Piñero-Pérez
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013, Sevilla, Spain
| | - José A Sánchez-Alcázar
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013, Sevilla, Spain.
| |
Collapse
|
9
|
Li WB, Shen NX, Zhang C, Xie HC, Li ZY, Cao L, Chen LZ, Zeng YJ, Fan CX, Chen Q, Shi YW, Song XW. Novel PANK2 Mutations in Patients With Pantothenate Kinase-Associated Neurodegeneration and the Genotype–Phenotype Correlation. Front Aging Neurosci 2022; 14:848919. [PMID: 35462688 PMCID: PMC9019683 DOI: 10.3389/fnagi.2022.848919] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
Pantothenate kinase-associated neurodegeneration (PKAN) is a rare genetic disorder caused by mutations in the mitochondrial pantothenate kinase 2 (PANK2) gene and displays an inherited autosomal recessive pattern. In this study, we identified eight PANK2 mutations, including three novel mutations (c.1103A > G/p.D368G, c.1696C > G/p.L566V, and c.1470delC/p.R490fs494X), in seven unrelated families with PKAN. All the patients showed an eye-of-the-tiger sign on the MRI, six of seven patients had dystonia, and two of seven patients had Parkinsonism. Biallelic mutations of PANK2 decreased PANK2 protein expression and reduced mitochondrial membrane potential in human embryonic kidney (HEK) 293T cells. The biallelic mutations from patients with early-onset PKAN, a severity phenotype, showed decreased mitochondrial membrane potential more than that from late-onset patients. We systematically reviewed all the reported patients with PKAN with PANK2 mutations. The results indicated that the early-onset patients carried a significantly higher frequency of biallelic loss-of-function (LoF) mutations compared to late-onset patients. In general, patients with LoF mutations showed more severe phenotypes, including earlier onset age and loss of gait. Although there was no significant difference in the frequency of biallelic missense mutations between the early-onset and late-onset patients, we found that patients with missense mutations in the mitochondrial trafficking domain (transit peptide/mitochondrial domain) of PANK2 exhibited the earliest onset age when compared to patients with mutations in the other two domains. Taken together, this study reports three novel mutations and indicates a correlation between the phenotype and mitochondrial dysfunction. This provides new insight for evaluating the clinical severity of patients based on the degree of mitochondrial dysfunction and suggests genetic counseling not just generalized identification of mutated PANK2 in clinics.
Collapse
Affiliation(s)
- Wen-Bin Li
- Department of Neurology, Institute of Neuroscience, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Nan-Xiang Shen
- Department of Neurology, Institute of Neuroscience, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Chao Zhang
- Suzhou Hospital of Anhui Medical University (Suzhou Municipal Hospital of Anhui Province), Suzhou, China
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Huan-Cheng Xie
- Department of Neurology, Institute of Neuroscience, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Zong-Yan Li
- Department of Neurology, Institute of Neuroscience, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Li Cao
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Li-Zhi Chen
- Department of Neurology, Institute of Neuroscience, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Yuan-jin Zeng
- Department of Neurology, Institute of Neuroscience, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Cui-Xia Fan
- Department of Neurology, Institute of Neuroscience, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Qian Chen
- Department of Neurology, Institute of Neuroscience, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Yi-Wu Shi
- Department of Neurology, Institute of Neuroscience, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
- *Correspondence: Yi-Wu Shi,
| | - Xing-Wang Song
- Department of Neurology, Institute of Neuroscience, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
- Xing-Wang Song,
| |
Collapse
|
10
|
Villalón-García I, Álvarez-Córdoba M, Povea-Cabello S, Talaverón-Rey M, Villanueva-Paz M, Luzón-Hidalgo R, Suárez-Rivero JM, Suárez-Carrillo A, Munuera-Cabeza M, Salas JJ, Falcón-Moya R, Rodríguez-Moreno A, Armengol JA, Sánchez-Alcázar JA. Vitamin E prevents lipid peroxidation and iron accumulation in PLA2G6-Associated Neurodegeneration. Neurobiol Dis 2022; 165:105649. [PMID: 35122944 DOI: 10.1016/j.nbd.2022.105649] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/11/2022] [Accepted: 01/31/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND PLA2G6-Associated Neurodegeneration (PLAN) is a rare neurodegenerative disease with autosomal recessive inheritance, which belongs to the NBIA (Neurodegeneration with Brain Iron Accumulation) group. Although the pathogenesis of the disease remains largely unclear, lipid peroxidation seems to play a central role in the pathogenesis. Currently, there is no cure for the disease. OBJECTIVE In this work, we examined the presence of lipid peroxidation, iron accumulation and mitochondrial dysfunction in two cellular models of PLAN, patients-derived fibroblasts and induced neurons, and assessed the effects of α-tocopherol (vitamin E) in correcting the pathophysiological alterations in PLAN cell cultures. METHODS Pathophysiological alterations were examined in fibroblasts and induced neurons generated by direct reprograming. Iron and lipofuscin accumulation were assessed using light and electron microscopy, as well as biochemical analysis techniques. Reactive Oxygen species production, lipid peroxidation and mitochondrial dysfunction were measured using specific fluorescent probes analysed by fluorescence microscopy and flow cytometry. RESULTS PLAN fibroblasts and induced neurons clearly showed increased lipid peroxidation, iron accumulation and altered mitochondrial membrane potential. All these pathological features were reverted with vitamin E treatment. CONCLUSIONS PLAN fibroblasts and induced neurons reproduce the main pathological alterations of the disease and provide useful tools for disease modelling. The main pathological alterations were corrected by Vitamin E supplementation in both models, suggesting that blocking lipid peroxidation progression is a critical therapeutic target.
Collapse
Affiliation(s)
- Irene Villalón-García
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Spain
| | - Mónica Álvarez-Córdoba
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Spain
| | - Suleva Povea-Cabello
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Spain
| | - Marta Talaverón-Rey
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Spain
| | - Marina Villanueva-Paz
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Spain
| | - Raquel Luzón-Hidalgo
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Spain
| | - Juan M Suárez-Rivero
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Spain
| | - Alejandra Suárez-Carrillo
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Spain
| | - Manuel Munuera-Cabeza
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Spain
| | - Joaquín J Salas
- Departamento de Bioquímica y Biología Molecular de Productos Vegetales, Instituto de la Grasa (CSIC), Sevilla, Spain.
| | - Rafael Falcón-Moya
- Laboratorio de Neurociencia Celular y Plasticidad, Departamento de Fisiología, Anatomía y Biología Celular, Universidad Pablo de Olavide, Sevilla, Spain.
| | - Antonio Rodríguez-Moreno
- Laboratorio de Neurociencia Celular y Plasticidad, Departamento de Fisiología, Anatomía y Biología Celular, Universidad Pablo de Olavide, Sevilla, Spain.
| | - José A Armengol
- Departamento de Fisiología, Anatomía y Biología Celular, Universidad Pablo de Olavide, Sevilla, Spain.
| | - José A Sánchez-Alcázar
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Spain.
| |
Collapse
|
11
|
Coenzyme a Biochemistry: From Neurodevelopment to Neurodegeneration. Brain Sci 2021; 11:brainsci11081031. [PMID: 34439650 PMCID: PMC8392065 DOI: 10.3390/brainsci11081031] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/29/2021] [Accepted: 07/29/2021] [Indexed: 12/21/2022] Open
Abstract
Coenzyme A (CoA) is an essential cofactor in all living organisms. It is involved in a large number of biochemical processes functioning either as an activator of molecules with carbonyl groups or as a carrier of acyl moieties. Together with its thioester derivatives, it plays a central role in cell metabolism, post-translational modification, and gene expression. Furthermore, recent studies revealed a role for CoA in the redox regulation by the S-thiolation of cysteine residues in cellular proteins. The intracellular concentration and distribution in different cellular compartments of CoA and its derivatives are controlled by several extracellular stimuli such as nutrients, hormones, metabolites, and cellular stresses. Perturbations of the biosynthesis and homeostasis of CoA and/or acyl-CoA are connected with several pathological conditions, including cancer, myopathies, and cardiomyopathies. In the most recent years, defects in genes involved in CoA production and distribution have been found in patients affected by rare forms of neurodegenerative and neurodevelopmental disorders. In this review, we will summarize the most relevant aspects of CoA cellular metabolism, their role in the pathogenesis of selected neurodevelopmental and neurodegenerative disorders, and recent advancements in the search for therapeutic approaches for such diseases.
Collapse
|
12
|
Camarena V, Huff TC, Wang G. Epigenomic regulation by labile iron. Free Radic Biol Med 2021; 170:44-49. [PMID: 33493555 PMCID: PMC8217092 DOI: 10.1016/j.freeradbiomed.2021.01.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/17/2020] [Accepted: 01/11/2021] [Indexed: 12/21/2022]
Abstract
Iron is an essential micronutrient metal for cellular functions but can generate highly reactive oxygen species resulting in oxidative damage. For these reasons its uptake and metabolism is highly regulated. A small but dynamic fraction of ferrous iron inside the cell, termed intracellular labile iron, is redox-reactive and ready to participate multiples reactions of intracellular enzymes. Due to its nature its determination and precise quantification has been a roadblock. However, recent progress in the development of intracellular labile iron probes are allowing the reevaluation of our current understanding and unmasking new functions. The role of intracellular labile iron in regulating the epigenome was recently discovered. This chapter examine how intracellular labile iron can modulate histone and DNA demethylation and how its pool can mediate a signaling pathway from cAMP serving as a sensor of the metabolic needs of the cells.
Collapse
Affiliation(s)
- Vladimir Camarena
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Tyler C Huff
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Gaofeng Wang
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
13
|
Álvarez-Córdoba M, Talaverón-Rey M, Villalón-García I, Povea-Cabello S, Suárez-Rivero JM, Suárez-Carrillo A, Munuera-Cabeza M, Salas JJ, Sánchez-Alcázar JA. Down regulation of the expression of mitochondrial phosphopantetheinyl-proteins in pantothenate kinase-associated neurodegeneration: pathophysiological consequences and therapeutic perspectives. Orphanet J Rare Dis 2021; 16:201. [PMID: 33952316 PMCID: PMC8101147 DOI: 10.1186/s13023-021-01823-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 04/20/2021] [Indexed: 02/06/2023] Open
Abstract
Background Neurodegeneration with brain iron accumulation (NBIA) is a group of genetic neurological disorders frequently associated with iron accumulation in the basal nuclei of the brain characterized by progressive spasticity, dystonia, muscle rigidity, neuropsychiatric symptoms, and retinal degeneration or optic nerve atrophy. Pantothenate kinase-associated neurodegeneration (PKAN) is the most widespread NBIA disorder. It is caused by mutations in the gene of pantothenate kinase 2 (PANK2) which catalyzes the first reaction of coenzyme A (CoA) biosynthesis. Thus, altered PANK2 activity is expected to induce CoA deficiency as well as low levels of essential metabolic intermediates such as 4′-phosphopantetheine which is a necessary cofactor for critical proteins involved in cytosolic and mitochondrial pathways such as fatty acid biosynthesis, mitochondrial respiratory complex I assembly and lysine and tetrahydrofolate metabolism, among other metabolic processes. Methods In this manuscript, we examined the effect of PANK2 mutations on the expression levels of proteins with phosphopantetheine cofactors in fibroblast derived from PKAN patients. These proteins include cytosolic acyl carrier protein (ACP), which is integrated within the multifunctional polypeptide chain of the fatty acid synthase involved in cytosolic fatty acid biosynthesis type I (FASI); mitochondrial ACP (mtACP) associated with mitocondrial fatty acid biosynthesis type II (FASII); mitochondrial alpha-aminoadipic semialdehyde synthase (AASS); and 10-formyltetrahydrofolate dehydrogenases (cytosolic, ALD1L1, and mitochondrial, ALD1L2). Results In PKAN fibroblasts the expression levels of cytosolic FAS and ALD1L1 were not affected while the expression levels of mtACP, AASS and ALD1L2 were markedly reduced, suggesting that 4′-phosphopantetheinylation of mitochondrial but no cytosolic proteins were markedly affected in PKAN patients. Furthermore, the correction of PANK2 expression levels by treatment with pantothenate in selected mutations with residual enzyme content was able to correct the expression levels of mitochondrial phosphopantetheinyl-proteins and restore the affected pathways. The positive effects of pantothenate in particular mutations were also corroborated in induced neurons obtained by direct reprograming of mutant PANK2 fibroblasts. Conclusions Our results suggest that the expression levels of mitochondrial phosphopantetheinyl-proteins are severely reduced in PKAN cells and that in selected mutations pantothenate increases the expression levels of both PANK2 and mitochondrial phosphopantetheinyl-proteins associated with remarkable improvement of cell pathophysiology. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-021-01823-3.
Collapse
Affiliation(s)
- Mónica Álvarez-Córdoba
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas, Universidad Pablo de Olavide, Carretera de Utrera Km 1, 41013, Sevilla, Spain.,Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013, Sevilla, Spain
| | - Marta Talaverón-Rey
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas, Universidad Pablo de Olavide, Carretera de Utrera Km 1, 41013, Sevilla, Spain.,Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013, Sevilla, Spain
| | - Irene Villalón-García
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas, Universidad Pablo de Olavide, Carretera de Utrera Km 1, 41013, Sevilla, Spain.,Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013, Sevilla, Spain
| | - Suleva Povea-Cabello
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas, Universidad Pablo de Olavide, Carretera de Utrera Km 1, 41013, Sevilla, Spain.,Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013, Sevilla, Spain
| | - Juan M Suárez-Rivero
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas, Universidad Pablo de Olavide, Carretera de Utrera Km 1, 41013, Sevilla, Spain.,Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013, Sevilla, Spain
| | - Alejandra Suárez-Carrillo
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas, Universidad Pablo de Olavide, Carretera de Utrera Km 1, 41013, Sevilla, Spain.,Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013, Sevilla, Spain
| | - Manuel Munuera-Cabeza
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas, Universidad Pablo de Olavide, Carretera de Utrera Km 1, 41013, Sevilla, Spain.,Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013, Sevilla, Spain
| | - Joaquín J Salas
- Departamento de Bioquímica Y Biología Molecular de Productos Vegetales, Instituto de La Grasa (CSIC), Sevilla, Spain
| | - José A Sánchez-Alcázar
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas, Universidad Pablo de Olavide, Carretera de Utrera Km 1, 41013, Sevilla, Spain. .,Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, 41013, Sevilla, Spain.
| |
Collapse
|
14
|
Exploring Yeast as a Study Model of Pantothenate Kinase-Associated Neurodegeneration and for the Identification of Therapeutic Compounds. Int J Mol Sci 2020; 22:ijms22010293. [PMID: 33396642 PMCID: PMC7795310 DOI: 10.3390/ijms22010293] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/18/2020] [Accepted: 12/23/2020] [Indexed: 12/11/2022] Open
Abstract
Mutations in the pantothenate kinase 2 gene (PANK2) are the cause of pantothenate kinase-associated neurodegeneration (PKAN), the most common form of neurodegeneration with brain iron accumulation. Although different disease models have been created to investigate the pathogenic mechanism of PKAN, the cascade of molecular events resulting from CoA synthesis impairment is not completely understood. Moreover, for PKAN disease, only symptomatic treatments are available. Despite the lack of a neural system, Saccharomyces cerevisiae has been successfully used to decipher molecular mechanisms of many human disorders including neurodegenerative diseases as well as iron-related disorders. To gain insights into the molecular basis of PKAN, a yeast model of this disease was developed: a yeast strain with the unique gene encoding pantothenate kinase CAB1 deleted, and expressing a pathological variant of this enzyme. A detailed functional characterization demonstrated that this model recapitulates the main phenotypes associated with human disease: mitochondrial dysfunction, altered lipid metabolism, iron overload, and oxidative damage suggesting that the yeast model could represent a tool to provide information on pathophysiology of PKAN. Taking advantage of the impaired oxidative growth of this mutant strain, a screening for molecules able to rescue this phenotype was performed. Two molecules in particular were able to restore the multiple defects associated with PKAN deficiency and the rescue was not allele-specific. Furthermore, the construction and characterization of a set of mutant alleles, allowing a quick evaluation of the biochemical consequences of pantothenate kinase (PANK) protein variants could be a tool to predict genotype/phenotype correlation.
Collapse
|
15
|
Oxidative Stress, a Crossroad Between Rare Diseases and Neurodegeneration. Antioxidants (Basel) 2020; 9:antiox9040313. [PMID: 32326494 PMCID: PMC7222183 DOI: 10.3390/antiox9040313] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/06/2020] [Accepted: 04/13/2020] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress is an imbalance between production and accumulation of oxygen reactive species and/or reactive nitrogen species in cells and tissues, and the capacity of detoxifying these products, using enzymatic and non-enzymatic components, such as glutathione. Oxidative stress plays roles in several pathological processes in the nervous system, such as neurotoxicity, neuroinflammation, ischemic stroke, and neurodegeneration. The concepts of oxidative stress and rare diseases were formulated in the eighties, and since then, the link between them has not stopped growing. The present review aims to expand knowledge in the pathological processes associated with oxidative stress underlying some groups of rare diseases: Friedreich’s ataxia, diseases with neurodegeneration with brain iron accumulation, Charcot-Marie-Tooth as an example of rare neuromuscular disorders, inherited retinal dystrophies, progressive myoclonus epilepsies, and pediatric drug-resistant epilepsies. Despite the discrimination between cause and effect may not be easy on many occasions, all these conditions are Mendelian rare diseases that share oxidative stress as a common factor, and this may represent a potential target for therapies.
Collapse
|
16
|
Subramanian C, Yao J, Frank MW, Rock CO, Jackowski S. A pantothenate kinase-deficient mouse model reveals a gene expression program associated with brain coenzyme a reduction. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165663. [PMID: 31918006 DOI: 10.1016/j.bbadis.2020.165663] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/12/2019] [Accepted: 12/29/2019] [Indexed: 12/12/2022]
Abstract
Pantothenate kinase (PanK) is the first enzyme in the coenzyme A (CoA) biosynthetic pathway. The differential expression of the four-active mammalian PanK isoforms regulates CoA levels in different tissues and PANK2 mutations lead to Pantothenate Kinase Associated Neurodegeneration (PKAN). The molecular mechanisms that potentially underlie PKAN pathophysiology are investigated in a mouse model of CoA deficiency in the central nervous system (CNS). Both PanK1 and PanK2 contribute to brain CoA levels in mice and so a mouse model with a systemic deletion of Pank1 together with neuronal deletion of Pank2 was generated. Neuronal Pank2 expression in double knockout mice decreased starting at P9-11 triggering a significant brain CoA deficiency. The depressed brain CoA in the mice correlates with abnormal forelimb flexing and weakness that, in turn, contributes to reduced locomotion and abnormal gait. Biochemical analysis reveals a reduction in short-chain acyl-CoAs, including acetyl-CoA and succinyl-CoA. Comparative gene expression analysis reveals that the CoA deficiency in brain is associated with a large elevation of Hif3a transcript expression and significant reduction of gene transcripts in heme and hemoglobin synthesis. Reduction of brain heme levels is associated with the CoA deficiency. The data suggest a response to oxygen/glucose deprivation and indicate a disruption of oxidative metabolism arising from a CoA deficiency in the CNS.
Collapse
Affiliation(s)
| | - Jiangwei Yao
- St. Jude Children's Research Hospital, Memphis, TN 38105-3678, USA
| | - Matthew W Frank
- St. Jude Children's Research Hospital, Memphis, TN 38105-3678, USA
| | - Charles O Rock
- St. Jude Children's Research Hospital, Memphis, TN 38105-3678, USA
| | | |
Collapse
|
17
|
Alvarez-Cordoba M, Villanueva-Paz M, Villalón-García I, Povea-Cabello S, Suárez-Rivero JM, Talaverón-Rey M, Abril-Jaramillo J, Vintimilla-Tosi AB, Sánchez-Alcázar JA. Precision medicine in pantothenate kinase-associated neurodegeneration. Neural Regen Res 2019; 14:1177-1185. [PMID: 30804242 PMCID: PMC6425824 DOI: 10.4103/1673-5374.251203] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Neurodegeneration with brain iron accumulation is a broad term that describes a heterogeneous group of progressive and invalidating neurologic disorders in which iron deposits in certain brain areas, mainly the basal ganglia. The predominant clinical symptoms include spasticity, progressive dystonia, Parkinson’s disease-like symptoms, neuropsychiatric alterations, and retinal degeneration. Among the neurodegeneration with brain iron accumulation disorders, the most frequent subtype is pantothenate kinase-associated neurodegeneration (PKAN) caused by defects in the gene encoding the enzyme pantothenate kinase 2 (PANK2) which catalyzed the first reaction of the coenzyme A biosynthesis pathway. Currently there is no effective treatment to prevent the inexorable course of these disorders. The aim of this review is to open up a discussion on the utility of using cellular models derived from patients as a valuable tool for the development of precision medicine in PKAN. Recently, we have described that dermal fibroblasts obtained from PKAN patients can manifest the main pathological changes of the disease such as intracellular iron accumulation accompanied by large amounts of lipofuscin granules, mitochondrial dysfunction and a pronounced increase of markers of oxidative stress. In addition, PKAN fibroblasts showed a morphological senescence-like phenotype. Interestingly, pantothenate supplementation, the substrate of the PANK2 enzyme, corrected all pathophysiological alterations in responder PKAN fibroblasts with low/residual PANK2 enzyme expression. However, pantothenate treatment had no favourable effect on PKAN fibroblasts harbouring mutations associated with the expression of a truncated/incomplete protein. The correction of pathological alterations by pantothenate in individual mutations was also verified in induced neurons obtained by direct reprograming of PKAN fibroblasts. Our observations indicate that pantothenate supplementation can increase/stabilize the expression levels of PANK2 in specific mutations. Fibroblasts and induced neurons derived from patients can provide a useful tool for recognizing PKAN patients who can respond to pantothenate treatment. The presence of low but significant PANK2 expression which can be increased in particular mutations gives valuable information which can support the treatment with high dose of pantothenate. The evaluation of personalized treatments in vitro of fibroblasts and neuronal cells derived from PKAN patients with a wide range of pharmacological options currently available, and monitoring its effect on the pathophysiological changes, can help for a better therapeutic strategy. In addition, these cell models will be also useful for testing the efficacy of new therapeutic options developed in the future.
Collapse
Affiliation(s)
- Mónica Alvarez-Cordoba
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - Marina Villanueva-Paz
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - Irene Villalón-García
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - Suleva Povea-Cabello
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - Juan M Suárez-Rivero
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - Marta Talaverón-Rey
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | | | | | - José A Sánchez-Alcázar
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| |
Collapse
|
18
|
Seibler P, Burbulla LF, Dulovic M, Zittel S, Heine J, Schmidt T, Rudolph F, Westenberger A, Rakovic A, Münchau A, Krainc D, Klein C. Iron overload is accompanied by mitochondrial and lysosomal dysfunction in WDR45 mutant cells. Brain 2018; 141:3052-3064. [PMID: 30169597 PMCID: PMC7190033 DOI: 10.1093/brain/awy230] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 07/08/2018] [Accepted: 07/15/2018] [Indexed: 01/10/2023] Open
Abstract
Beta-propeller protein-associated neurodegeneration is a subtype of monogenic neurodegeneration with brain iron accumulation caused by de novo mutations in WDR45. The WDR45 protein functions as a beta-propeller scaffold and plays a putative role in autophagy through its interaction with phospholipids and autophagy-related proteins. Loss of WDR45 function due to disease-causing mutations has been linked to defects in autophagic flux in patient and animal cells. However, the role of WDR45 in iron homeostasis remains elusive. Here we studied patient-specific WDR45 mutant fibroblasts and induced pluripotent stem cell-derived midbrain neurons. Our data demonstrated that loss of WDR45 increased cellular iron levels and oxidative stress, accompanied by mitochondrial abnormalities, autophagic defects, and diminished lysosomal function. Restoring WDR45 levels partially rescued oxidative stress and the susceptibility to iron treatment, and activation of autophagy reduced the observed iron overload in WDR45 mutant cells. Our data suggest that iron-containing macromolecules and organelles cannot effectively be degraded through the lysosomal pathway due to loss of WDR45 function.
Collapse
Affiliation(s)
- Philip Seibler
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Lena F Burbulla
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Marija Dulovic
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Simone Zittel
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Johanne Heine
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Thomas Schmidt
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | | | - Ana Westenberger
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | | | | | - Dimitri Krainc
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| |
Collapse
|
19
|
Pagani F, Trivedi A, Khatri D, Zizioli D, Garrafa E, Mitola S, Finazzi D. Silencing of pantothenate kinase 2 reduces endothelial cell angiogenesis. Mol Med Rep 2018; 18:4739-4746. [PMID: 30221726 DOI: 10.3892/mmr.2018.9480] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 08/23/2018] [Indexed: 11/05/2022] Open
Abstract
Coenzyme A (CoA) is an essential cofactor of cellular metabolism that is involved in ~4% of cellular reactions. Its de novo production relies on five subsequent enzymatic steps, starting with the phosphorylation of vitamin B5. Pantothenate kinase 2 (PANK2) and coenzyme A synthase (COASY) catalyze the first and last steps of this pathway. Mutations in these genes lead to severe and progressive movement disorders, with neurodegeneration and iron accumulation in the basal ganglia, known as PANK2‑ and COASY protein‑associated neurodegeneration, respectively. Given the ubiquitous role of CoA in cellular metabolism, it is still not clear why patients carrying PANK2 and COASY mutations develop almost exclusively neurological symptoms. Important clues are the energetic profile of neural cells as well as the high levels of PANK2 expression in the brain; however, other features may contribute to this selective tissue vulnerability. Notably, when pank2 or coasy expression was suppressed in zebrafish evident perturbation of neuronal development was observed, as well as severe defects in vasculature formation. Supplementation of CoA to fish water prevented the appearance of the phenotype, thereby confirming the specific connection with the availability of the metabolic cofactor. The present study investigated the associations between PANK2 defects and angiogenesis in a mammalian setting, and revealed that PANK2 expression was required for normal angiogenetic properties of human umbilical vein endothelial cells.
Collapse
Affiliation(s)
- Francesca Pagani
- Section of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, I‑25123 Brescia, Italy
| | - Akansha Trivedi
- Section of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, I‑25123 Brescia, Italy
| | - Deepak Khatri
- Section of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, I‑25123 Brescia, Italy
| | - Daniela Zizioli
- Section of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, I‑25123 Brescia, Italy
| | - Emirena Garrafa
- Section of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, I‑25123 Brescia, Italy
| | - Stefania Mitola
- Section of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, I‑25123 Brescia, Italy
| | - Dario Finazzi
- Section of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, I‑25123 Brescia, Italy
| |
Collapse
|
20
|
Pantothenate Rescues Iron Accumulation in Pantothenate Kinase-Associated Neurodegeneration Depending on the Type of Mutation. Mol Neurobiol 2018; 56:3638-3656. [PMID: 30173408 DOI: 10.1007/s12035-018-1333-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 08/24/2018] [Indexed: 12/18/2022]
Abstract
Neurodegeneration with brain iron accumulation (NBIA) is a group of inherited neurologic disorders in which iron accumulates in the basal ganglia resulting in progressive dystonia, spasticity, parkinsonism, neuropsychiatric abnormalities, and optic atrophy or retinal degeneration. The most prevalent form of NBIA is pantothenate kinase-associated neurodegeneration (PKAN) associated with mutations in the gene of pantothenate kinase 2 (PANK2), which is essential for coenzyme A (CoA) synthesis. There is no cure for NBIA nor is there a standard course of treatment. In the current work, we describe that fibroblasts derived from patients harbouring PANK2 mutations can reproduce many of the cellular pathological alterations found in the disease, such as intracellular iron and lipofuscin accumulation, increased oxidative stress, and mitochondrial dysfunction. Furthermore, mutant fibroblasts showed a characteristic senescent morphology. Treatment with pantothenate, the PANK2 enzyme substrate, was able to correct all pathological alterations in responder mutant fibroblasts with residual PANK2 enzyme expression. However, pantothenate had no effect on mutant fibroblasts with truncated/incomplete protein expression. The positive effect of pantothenate in particular mutations was also confirmed in induced neurons obtained by direct reprograming of mutant fibroblasts. Our results suggest that pantothenate treatment can stabilize the expression levels of PANK2 in selected mutations. These results encourage us to propose our screening model as a quick and easy way to detect pantothenate-responder patients with PANK2 mutations. The existence of residual enzyme expression in some affected individuals raises the possibility of treatment using high dose of pantothenate.
Collapse
|
21
|
Drecourt A, Babdor J, Dussiot M, Petit F, Goudin N, Garfa-Traoré M, Habarou F, Bole-Feysot C, Nitschké P, Ottolenghi C, Metodiev MD, Serre V, Desguerre I, Boddaert N, Hermine O, Munnich A, Rötig A. Impaired Transferrin Receptor Palmitoylation and Recycling in Neurodegeneration with Brain Iron Accumulation. Am J Hum Genet 2018; 102:266-277. [PMID: 29395073 DOI: 10.1016/j.ajhg.2018.01.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 01/05/2018] [Indexed: 12/29/2022] Open
Abstract
Neurodegeneration with brain iron accumulation (NBIA) is a genetically heterogeneous condition characterized by progressive dystonia with iron accumulation in the basal ganglia. How NBIA-associated mutations trigger iron overload remains poorly understood. After studying fibroblast cell lines from subjects carrying both known and unreported biallelic mutations in CRAT and REPS1, we ascribe iron overload to the abnormal recycling of transferrin receptor (TfR1) and the reduction of TfR1 palmitoylation in NBIA. Moreover, we describe palmitoylation as a hitherto unreported level of post-translational TfR1 regulation. A widely used antimalarial agent, artesunate, rescued abnormal TfR1 palmitoylation in cultured fibroblasts of NBIA subjects. These observations suggest therapeutic strategies aimed at targeting impaired TfR1 recycling and palmitoylation in NBIA.
Collapse
|
22
|
Tello C, Darling A, Lupo V, Pérez-Dueñas B, Espinós C. On the complexity of clinical and molecular bases of neurodegeneration with brain iron accumulation. Clin Genet 2017; 93:731-740. [PMID: 28542792 DOI: 10.1111/cge.13057] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 05/04/2017] [Accepted: 05/18/2017] [Indexed: 02/06/2023]
Abstract
Neurodegeneration with brain iron accumulation (NBIA) is a group of inherited heterogeneous neurodegenerative rare disorders. These patients present with dystonia, spasticity, parkinsonism and neuropsychiatric disturbances, along with brain magnetic resonance imaging (MRI) evidence of iron accumulation. In sum, they are devastating disorders and to date, there is no specific treatment. Ten NBIA genes are accepted: PANK2, PLA2G6, C19orf12, COASY, FA2H, ATP13A2, WDR45, FTL, CP, and DCAF17; and nonetheless, a relevant percentage of patients remain without genetic diagnosis, suggesting that other novel NBIA genes remain to be discovered. Overlapping complex clinical pictures render an accurate differential diagnosis difficult. Little is known about the pathophysiology of NBIAs. The reported NBIA genes take part in a variety of pathways: CoA synthesis, lipid and iron metabolism, autophagy, and membrane remodeling. The next-generation sequencing revolution has achieved relevant advances in genetics of Mendelian diseases and provide new genes for NBIAs, which are investigated according to 2 main strategies: genes involved in disorders with similar phenotype and genes that play a role in a pathway of interest. To achieve an effective therapy for NBIA patients, a better understanding of the biological process underlying disease is crucial, moving toward a new age of precision medicine.
Collapse
Affiliation(s)
- C Tello
- Unit of Genetics and Genomics of Neuromuscular and Neurodegenerative Disorders, Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain
| | - A Darling
- Department of Neuropediatrics, Hospital Sant Joan de Déu, Barcelona, Spain.,Unit U703, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| | - V Lupo
- Unit of Genetics and Genomics of Neuromuscular and Neurodegenerative Disorders, Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain
| | - B Pérez-Dueñas
- Department of Neuropediatrics, Hospital Sant Joan de Déu, Barcelona, Spain.,Unit U703, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| | - C Espinós
- Unit of Genetics and Genomics of Neuromuscular and Neurodegenerative Disorders, Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain
| |
Collapse
|
23
|
Arber C, Angelova PR, Wiethoff S, Tsuchiya Y, Mazzacuva F, Preza E, Bhatia KP, Mills K, Gout I, Abramov AY, Hardy J, Duce JA, Houlden H, Wray S. iPSC-derived neuronal models of PANK2-associated neurodegeneration reveal mitochondrial dysfunction contributing to early disease. PLoS One 2017; 12:e0184104. [PMID: 28863176 PMCID: PMC5581181 DOI: 10.1371/journal.pone.0184104] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 08/17/2017] [Indexed: 01/22/2023] Open
Abstract
Mutations in PANK2 lead to neurodegeneration with brain iron accumulation. PANK2 has a role in the biosynthesis of coenzyme A (CoA) from dietary vitamin B5, but the neuropathological mechanism and reasons for iron accumulation remain unknown. In this study, atypical patient-derived fibroblasts were reprogrammed into induced pluripotent stem cells (iPSCs) and subsequently differentiated into cortical neuronal cells for studying disease mechanisms in human neurons. We observed no changes in PANK2 expression between control and patient cells, but a reduction in protein levels was apparent in patient cells. CoA homeostasis and cellular iron handling were normal, mitochondrial function was affected; displaying activated NADH-related and inhibited FADH-related respiration, resulting in increased mitochondrial membrane potential. This led to increased reactive oxygen species generation and lipid peroxidation in patient-derived neurons. These data suggest that mitochondrial deficiency is an early feature of the disease process and can be explained by altered NADH/FADH substrate supply to oxidative phosphorylation. Intriguingly, iron chelation appeared to exacerbate the mitochondrial phenotype in both control and patient neuronal cells. This raises caution for the use iron chelation therapy in general when iron accumulation is absent.
Collapse
Affiliation(s)
- Charles Arber
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, United Kingdom
| | - Plamena R. Angelova
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, United Kingdom
| | - Sarah Wiethoff
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, United Kingdom
| | - Yugo Tsuchiya
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom
| | - Francesca Mazzacuva
- Centre for Translational Omics, Genetics and Genomic Medicine Programme, UCL Institute of Child Health, London, United Kingdom
| | - Elisavet Preza
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, United Kingdom
| | - Kailash P. Bhatia
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, United Kingdom
| | - Kevin Mills
- Centre for Translational Omics, Genetics and Genomic Medicine Programme, UCL Institute of Child Health, London, United Kingdom
| | - Ivan Gout
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom
| | - Andrey Y. Abramov
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, United Kingdom
| | - John Hardy
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, United Kingdom
| | - James A. Duce
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Henry Houlden
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, United Kingdom
| | - Selina Wray
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, United Kingdom
| |
Collapse
|
24
|
Aoun M, Corsetto PA, Nugue G, Montorfano G, Ciusani E, Crouzier D, Hogarth P, Gregory A, Hayflick S, Zorzi G, Rizzo AM, Tiranti V. Changes in Red Blood Cell membrane lipid composition: A new perspective into the pathogenesis of PKAN. Mol Genet Metab 2017; 121:180-189. [PMID: 28456385 DOI: 10.1016/j.ymgme.2017.04.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/14/2017] [Accepted: 04/14/2017] [Indexed: 01/12/2023]
Abstract
Pantothenate Kinase-Associated Neurodegeneration (PKAN) is a form of Neurodegeneration with Brain Iron Accumulation (NBIA) associated with mutations in the pantothenate kinase 2 gene (PANK2). The PANK2 catalyzes the first step of coenzyme A (CoA) biosynthesis, a pathway producing an essential cofactor that plays a key role in energy and lipid metabolism. The majority of PANK2 mutations reduces or abolishes the activity of the enzyme. In around 10% of cases with PKAN, the presence of deformed red blood cells with thorny protrusions in the circulation has been detected. Changes in membrane protein expression and assembly during erythropoiesis were previously explored in patients with PKAN. However, data on red blood cell membrane phospholipid organization are still missing in this disease. In this study, we performed lipidomic analysis on red blood cells from Italian patients affected by PKAN with a particular interest in membrane physico-chemical properties. We showed an increased number of small red blood cells together with membrane phospholipid alteration, particularly a significant increase in sphingomyelin (SM)/phosphatidylcholine (PC) and SM/phosphatidylethanolamine (PE) ratios, in subjects with PKAN. The membrane structural abnormalities were associated with membrane fluidity perturbation. These morphological and functional characteristics of red blood cells in patients with PKAN offer new possible tools in order to shed light on the pathogenesis of the disease and to possibly identify further biomarkers for clinical studies.
Collapse
Affiliation(s)
- Manar Aoun
- Unit of Molecular Neurogenetics, Pierfranco and Luisa Mariani Centre for the Study of Mitochondrial Disorders in Children, Foundation IRCCS Neurological Institute C. Besta, Via Temolo 4, 20126 Milan, Italy
| | - Paola Antonia Corsetto
- Department of Pharmacological and Biomolecular Sciences, Laboratory of Membrane Biochemistry and Applied Nutrition, Università degli Studi di Milano, Milan, Italy
| | - Guillaume Nugue
- IRBA, Unité des Risques Technologiques Emergeants BP 73, 91223 Brétigny sur Orge Cedex, France
| | - Gigliola Montorfano
- Department of Pharmacological and Biomolecular Sciences, Laboratory of Membrane Biochemistry and Applied Nutrition, Università degli Studi di Milano, Milan, Italy
| | - Emilio Ciusani
- Unit of Clinical Pathology and Medical Genetics, Foundation IRCCS Neurological Institute C. Besta, Milan, Italy
| | - David Crouzier
- IRBA, Unité des Risques Technologiques Emergeants BP 73, 91223 Brétigny sur Orge Cedex, France
| | - Penelope Hogarth
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR 97239, United States
| | - Allison Gregory
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR 97239, United States
| | - Susan Hayflick
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR 97239, United States
| | - Giovanna Zorzi
- Unit of Child Neurology, Foundation IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Angela Maria Rizzo
- Department of Pharmacological and Biomolecular Sciences, Laboratory of Membrane Biochemistry and Applied Nutrition, Università degli Studi di Milano, Milan, Italy.
| | - Valeria Tiranti
- Unit of Molecular Neurogenetics, Pierfranco and Luisa Mariani Centre for the Study of Mitochondrial Disorders in Children, Foundation IRCCS Neurological Institute C. Besta, Via Temolo 4, 20126 Milan, Italy.
| |
Collapse
|
25
|
Ingrassia R, Memo M, Garavaglia B. Ferrous Iron Up-regulation in Fibroblasts of Patients with Beta Propeller Protein-Associated Neurodegeneration (BPAN). Front Genet 2017; 8:18. [PMID: 28261264 PMCID: PMC5314138 DOI: 10.3389/fgene.2017.00018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 02/06/2017] [Indexed: 11/13/2022] Open
Abstract
Mutations in WDR45 gene, coding for a beta-propeller protein, have been found in patients affected by Neurodegeneration with Brain Iron Accumulation, NBIA5 (also known as BPAN). BPAN is a movement disorder with Non Transferrin Bound Iron (NTBI) accumulation in the basal ganglia as common hallmark between NBIA classes (Hayflick et al., 2013). WDR45 has been predicted to have a role in autophagy, while the impairment of iron metabolism in the different NBIA subclasses has not currently been clarified. We found the up-regulation of the ferrous iron transporter (-)IRE/Divalent Metal Transporter1 and down-regulation of Transferrin receptor in the fibroblasts of two BPAN affected patients with splicing mutations 235+1G>A (BPAN1) and 517_519ΔVal 173 (BPAN2). The BPAN patients showed a concomitant increase of intracellular ferrous iron after starvation. An altered pattern of iron transporters with iron overload is highlighted in BPAN human fibroblasts, supporting for a role of DMT1 in NBIA. We here present a novel element, about iron accumulation, to the existing knowledge in field of NBIA. Attention is focused to a starvation-dependent iron overload, possibly accounting for iron accumulation in the basal ganglia. Further investigation could clarify iron regulation in BPAN.
Collapse
Affiliation(s)
- Rosaria Ingrassia
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia Brescia, Italy
| | - Maurizio Memo
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia Brescia, Italy
| | - Barbara Garavaglia
- Molecular Neurogenetics Unit, Foundation IRCCS Neurological Institute Carlo Besta Milan, Italy
| |
Collapse
|
26
|
Orellana DI, Santambrogio P, Rubio A, Yekhlef L, Cancellieri C, Dusi S, Giannelli SG, Venco P, Mazzara PG, Cozzi A, Ferrari M, Garavaglia B, Taverna S, Tiranti V, Broccoli V, Levi S. Coenzyme A corrects pathological defects in human neurons of PANK2-associated neurodegeneration. EMBO Mol Med 2016; 8:1197-1211. [PMID: 27516453 PMCID: PMC5048368 DOI: 10.15252/emmm.201606391] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Pantothenate kinase‐associated neurodegeneration (PKAN) is an early onset and severely disabling neurodegenerative disease for which no therapy is available. PKAN is caused by mutations in PANK2, which encodes for the mitochondrial enzyme pantothenate kinase 2. Its function is to catalyze the first limiting step of Coenzyme A (CoA) biosynthesis. We generated induced pluripotent stem cells from PKAN patients and showed that their derived neurons exhibited premature death, increased ROS production, mitochondrial dysfunctions—including impairment of mitochondrial iron‐dependent biosynthesis—and major membrane excitability defects. CoA supplementation prevented neuronal death and ROS formation by restoring mitochondrial and neuronal functionality. Our findings provide direct evidence that PANK2 malfunctioning is responsible for abnormal phenotypes in human neuronal cells and indicate CoA treatment as a possible therapeutic intervention.
Collapse
Affiliation(s)
- Daniel I Orellana
- Proteomics of Iron Metabolism Unit, Division of Neuroscience San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Santambrogio
- Proteomics of Iron Metabolism Unit, Division of Neuroscience San Raffaele Scientific Institute, Milan, Italy
| | - Alicia Rubio
- Stem Cells and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Latefa Yekhlef
- Neuroimmunology Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Cinzia Cancellieri
- Stem Cells and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Sabrina Dusi
- Molecular Neurogenetics Unit, Foundation IRCCS-Neurological Institute "Carlo Besta", Milan, Italy
| | - Serena G Giannelli
- Stem Cells and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Paola Venco
- Molecular Neurogenetics Unit, Foundation IRCCS-Neurological Institute "Carlo Besta", Milan, Italy
| | - Pietro G Mazzara
- Stem Cells and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Anna Cozzi
- Proteomics of Iron Metabolism Unit, Division of Neuroscience San Raffaele Scientific Institute, Milan, Italy
| | - Maurizio Ferrari
- Genomic Unit for the Diagnosis of Human Pathologies, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy Vita-Salute San Raffaele University, Milan, Italy
| | - Barbara Garavaglia
- Molecular Neurogenetics Unit, Foundation IRCCS-Neurological Institute "Carlo Besta", Milan, Italy
| | - Stefano Taverna
- Neuroimmunology Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Valeria Tiranti
- Molecular Neurogenetics Unit, Foundation IRCCS-Neurological Institute "Carlo Besta", Milan, Italy
| | - Vania Broccoli
- Stem Cells and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy Institute of Neuroscience, National Research Council, Milan, Italy
| | - Sonia Levi
- Proteomics of Iron Metabolism Unit, Division of Neuroscience San Raffaele Scientific Institute, Milan, Italy Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
27
|
Arber CE, Li A, Houlden H, Wray S. Review: Insights into molecular mechanisms of disease in neurodegeneration with brain iron accumulation: unifying theories. Neuropathol Appl Neurobiol 2016; 42:220-41. [PMID: 25870938 PMCID: PMC4832581 DOI: 10.1111/nan.12242] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 03/18/2015] [Indexed: 12/14/2022]
Abstract
Neurodegeneration with brain iron accumulation (NBIA) is a group of disorders characterized by dystonia, parkinsonism and spasticity. Iron accumulates in the basal ganglia and may be accompanied by Lewy bodies, axonal swellings and hyperphosphorylated tau depending on NBIA subtype. Mutations in 10 genes have been associated with NBIA that include Ceruloplasmin (Cp) and ferritin light chain (FTL), both directly involved in iron homeostasis, as well as Pantothenate Kinase 2 (PANK2), Phospholipase A2 group 6 (PLA2G6), Fatty acid hydroxylase 2 (FA2H), Coenzyme A synthase (COASY), C19orf12, WDR45 and DCAF17 (C2orf37). These genes are involved in seemingly unrelated cellular pathways, such as lipid metabolism, Coenzyme A synthesis and autophagy. A greater understanding of the cellular pathways that link these genes and the disease mechanisms leading to iron dyshomeostasis is needed. Additionally, the major overlap seen between NBIA and more common neurodegenerative diseases may highlight conserved disease processes. In this review, we will discuss clinical and pathological findings for each NBIA-related gene, discuss proposed disease mechanisms such as mitochondrial health, oxidative damage, autophagy/mitophagy and iron homeostasis, and speculate the potential overlap between NBIA subtypes.
Collapse
Affiliation(s)
- C E Arber
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK
| | - A Li
- Reta Lila Weston Institute, Institute of Neurology, University College London, London, UK
| | - H Houlden
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK
| | - S Wray
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK
| |
Collapse
|
28
|
Kinghorn KJ, Castillo-Quan JI. Mitochondrial dysfunction and defects in lipid homeostasis as therapeutic targets in neurodegeneration with brain iron accumulation. Rare Dis 2016; 4:e1128616. [PMID: 27141409 PMCID: PMC4838319 DOI: 10.1080/21675511.2015.1128616] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 11/11/2015] [Accepted: 12/01/2015] [Indexed: 10/26/2022] Open
Abstract
The PLA2G6 gene encodes a group VIA calcium independent phospholipase A2 (iPLA2β), which hydrolyses glycerophospholipids to release fatty acids and lysophospholipids. Mutations in PLA2G6 are associated with a number of neurodegenerative disorders including neurodegeneration with brain iron accumulation (NBIA), infantile neuroaxonal dystrophy (INAD), and dystonia parkinsonism, collectively known as PLA2G6-associated neurodegeneration (PLAN). Recently Kinghorn et al. demonstrated in Drosophila and PLA2G6 mutant fibroblasts that loss of normal PLA2G6 activity is associated with mitochondrial dysfunction and mitochondrial lipid peroxidation. Furthermore, they were able to show the beneficial effects of deuterated polyunsaturated fatty acids (D-PUFAs), which reduce lipid peroxidation. D-PUFAs were able to rescue the locomotor deficits of flies lacking the fly ortholog of PLA2G6 (iPLA2-VIA), as well as the mitochondrial abnormalities in PLA2G6 mutant fibroblasts. This work demonstrated that the iPLA2-VIA knockout fly is a useful organism to dissect the mechanisms of pathogenesis of PLAN, and that further investigation is required to determine the therapeutic potential of D-PUFAs in patients with PLA2G6 mutations. The fruit fly has also been used to study some of the other genetic causes of NBIA, and here we also describe what is known about the mechanisms of pathogenesis of these NBIA variants. Mitochondrial dysfunction, defects in lipid metabolism, as well as defective Coenzyme A (CoA) biosynthesis, have all been implicated in some genetic forms of NBIA, including PANK2, CoASY, C12orf19 and FA2H.
Collapse
Affiliation(s)
- Kerri J Kinghorn
- Institute of Healthy Ageing and Department of Genetics, Environment and Evolution, University College London, London, UK; Institute of Neurology, University College London, Queen Square, London, UK
| | - Jorge Iván Castillo-Quan
- Institute of Healthy Ageing and Department of Genetics, Environment and Evolution, University College London, London, UK; Institute of Neurology, University College London, Queen Square, London, UK
| |
Collapse
|
29
|
Zizioli D, Tiso N, Guglielmi A, Saraceno C, Busolin G, Giuliani R, Khatri D, Monti E, Borsani G, Argenton F, Finazzi D. Knock-down of pantothenate kinase 2 severely affects the development of the nervous and vascular system in zebrafish, providing new insights into PKAN disease. Neurobiol Dis 2015; 85:35-48. [PMID: 26476142 PMCID: PMC4684146 DOI: 10.1016/j.nbd.2015.10.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Revised: 08/31/2015] [Accepted: 10/11/2015] [Indexed: 01/31/2023] Open
Abstract
Pantothenate Kinase Associated Neurodegeneration (PKAN) is an autosomal recessive disorder with mutations in the pantothenate kinase 2 gene (PANK2), encoding an essential enzyme for Coenzyme A (CoA) biosynthesis. The molecular connection between defects in this enzyme and the neurodegenerative phenotype observed in PKAN patients is still poorly understood. We exploited the zebrafish model to study the role played by the pank2 gene during embryonic development and get new insight into PKAN pathogenesis. The zebrafish orthologue of hPANK2 lies on chromosome 13, is a maternal gene expressed in all development stages and, in adult animals, is highly abundant in CNS, dorsal aorta and caudal vein. The injection of a splice-inhibiting morpholino induced a clear phenotype with perturbed brain morphology and hydrocephalus; edema was present in the heart region and caudal plexus, where hemorrhages with reduction of blood circulation velocity were detected. We characterized the CNS phenotype by studying the expression pattern of wnt1 and neurog1 neural markers and by use of the Tg(neurod:EGFP/sox10:dsRed) transgenic line. The results evidenced that downregulation of pank2 severely impairs neuronal development, particularly in the anterior part of CNS (telencephalon). Whole-mount in situ hybridization analysis of the endothelial markers cadherin-5 and fli1a, and use of Tg(fli1a:EGFP/gata1a:dsRed) transgenic line, confirmed the essential role of pank2 in the formation of the vascular system. The specificity of the morpholino-induced phenotype was proved by the restoration of a normal development in a high percentage of embryos co-injected with pank2 mRNA. Also, addition of pantethine or CoA, but not of vitamin B5, to pank2 morpholino-injected embryos rescued the phenotype with high efficiency. The zebrafish model indicates the relevance of pank2 activity and CoA homeostasis for normal neuronal development and functioning and provides evidence of an unsuspected role for this enzyme and its product in vascular development. Zebrafish pank2 gene is highly expressed in the CNS and the main vascular structures. Pank2 down-regulation severely affects the development of the forebrain. Pank2 down-regulation affects the dorsal aorta, caudal vein and inter-somitic vessels. Pantethine and Coenzyme A restore the normal development in the absence of pank2 expression.
Collapse
Affiliation(s)
- Daniela Zizioli
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123 Brescia, Italy.
| | - Natascia Tiso
- Department of Biology, University of Padova, via U. Bassi 58/B, 35121 Padova, Italy
| | - Adele Guglielmi
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123 Brescia, Italy
| | - Claudia Saraceno
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123 Brescia, Italy
| | - Giorgia Busolin
- Department of Biology, University of Padova, via U. Bassi 58/B, 35121 Padova, Italy
| | - Roberta Giuliani
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123 Brescia, Italy
| | - Deepak Khatri
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123 Brescia, Italy
| | - Eugenio Monti
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123 Brescia, Italy
| | - Giuseppe Borsani
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123 Brescia, Italy
| | - Francesco Argenton
- Department of Biology, University of Padova, via U. Bassi 58/B, 35121 Padova, Italy
| | - Dario Finazzi
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123 Brescia, Italy; Clinical Chemistry Laboratory, Spedali Civili Hospital, 25123 Brescia, Italy.
| |
Collapse
|
30
|
Mora M, Angelini C, Bignami F, Bodin AM, Crimi M, Di Donato JH, Felice A, Jaeger C, Karcagi V, LeCam Y, Lynn S, Meznaric M, Moggio M, Monaco L, Politano L, de la Paz MP, Saker S, Schneiderat P, Ensini M, Garavaglia B, Gurwitz D, Johnson D, Muntoni F, Puymirat J, Reza M, Voit T, Baldo C, Bricarelli FD, Goldwurm S, Merla G, Pegoraro E, Renieri A, Zatloukal K, Filocamo M, Lochmüller H. The EuroBioBank Network: 10 years of hands-on experience of collaborative, transnational biobanking for rare diseases. Eur J Hum Genet 2015; 23:1116-23. [PMID: 25537360 PMCID: PMC4538193 DOI: 10.1038/ejhg.2014.272] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 10/15/2014] [Accepted: 11/10/2014] [Indexed: 11/09/2022] Open
Abstract
The EuroBioBank (EBB) network (www.eurobiobank.org) is the first operating network of biobanks in Europe to provide human DNA, cell and tissue samples as a service to the scientific community conducting research on rare diseases (RDs). The EBB was established in 2001 to facilitate access to RD biospecimens and associated data; it obtained funding from the European Commission in 2002 (5th framework programme) and started operation in 2003. The set-up phase, during the EC funding period 2003-2006, established the basis for running the network; the following consolidation phase has seen the growth of the network through the joining of new partners, better network cohesion, improved coordination of activities, and the development of a quality-control system. During this phase the network participated in the EC-funded TREAT-NMD programme and was involved in planning of the European Biobanking and Biomolecular Resources Research Infrastructure. Recently, EBB became a partner of RD-Connect, an FP7 EU programme aimed at linking RD biobanks, registries, and bioinformatics data. Within RD-Connect, EBB contributes expertise, promotes high professional standards, and best practices in RD biobanking, is implementing integration with RD patient registries and 'omics' data, thus challenging the fragmentation of international cooperation on the field.
Collapse
Affiliation(s)
- Marina Mora
- Muscle Cell Biology Lab, Neuromuscular Diseases and Neuroimmunolgy Unit, Fondazione Istituto Neurologico C. Besta, Milano, Italy
| | - Corrado Angelini
- IRCCS Fondazione San Camillo Hospital, Lido Venice, Italy
- Department of Neurosciences, NPSRR University of Padova, Padova, Italy
| | | | - Anne-Mary Bodin
- EURORDIS, European Organisation for Rare Diseases, Paris, France
| | | | | | - Alex Felice
- Laboratory of Molecular Genetics and Malta BioBank, University of Malta, and Thalassaemia Clinic, Mater Dei Hospital, Msida, Malta
| | | | - Veronika Karcagi
- Department of Molecular Genetics and Diagnostics, National Institute of Environmental Health, Budapest, Hungary
| | - Yann LeCam
- EURORDIS, European Organisation for Rare Diseases, Paris, France
| | - Stephen Lynn
- MRC Centre for Neuromuscular Diseases, Institute of Genetic Medicine, Newcastle University, International Centre for Life, Newcastle upon Tyne, UK
| | - Marija Meznaric
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Maurizio Moggio
- Neuromuscular Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Dino Ferrari Centre, University of Milan, Milan, Italy
| | | | - Luisa Politano
- Division of Cardiomyology and Medical Genetics, Department of Experimental Medicine, Second University of Naples, Naples, Italy
| | - Manuel Posada de la Paz
- Manuel Posada de la Paz, Institute of Rare Diseases Research, IIER, ISCIII and Spain RDR & CIBERER, Madrid, Spain
| | | | - Peter Schneiderat
- Muscle Tissue Culture Collection, Friedrich-Baur-Institute, Neurological Department, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Monica Ensini
- MRC Centre for Neuromuscular Diseases, Institute of Genetic Medicine, Newcastle University, International Centre for Life, Newcastle upon Tyne, UK
| | - Barbara Garavaglia
- Molecular Neurogenetics Unit, Fondazione Istituto Neurologico C. Besta, Milano, Italy
| | - David Gurwitz
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Diana Johnson
- Dubowitz Neuromuscular Centre, MRC Neuromuscular Centre at UCL Institute of Child Health, London, UK
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, MRC Neuromuscular Centre at UCL Institute of Child Health, London, UK
| | - Jack Puymirat
- Department of Human Genetics, Centre Hospitalier Universitaire de Quebec, Quebec City, Quebec, Canada
| | - Mojgan Reza
- MRC Centre for Neuromuscular Diseases, Institute of Genetic Medicine, Newcastle University, International Centre for Life, Newcastle upon Tyne, UK
| | - Thomas Voit
- Inserm U974—Institute of Myology, University Pierre and Marie Curie Paris 6, Paris, France
| | - Chiara Baldo
- Laboratorio di Genetica Umana, E.O. Ospedali Galliera, Genova, Italy
| | | | - Stefano Goldwurm
- Parkinson Institute, Istituti Clinici di Perfezionamento, Milano, Italy
| | - Giuseppe Merla
- Medical Genetics Unit, IRCCS Casa Sollievo della Sofferenza Hospital, San Giovanni Rotondo, Italy
| | - Elena Pegoraro
- Department of Neurosciences, NPSRR University of Padova, Padova, Italy
| | - Alessandra Renieri
- Division of Medical Genetics, University of Siena, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Kurt Zatloukal
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Mirella Filocamo
- Centro di Diagnostica Genetica e Biochimica delle Malattie Metaboliche, Istituto G. Gaslini, Genova, Italy
| | - Hanns Lochmüller
- MRC Centre for Neuromuscular Diseases, Institute of Genetic Medicine, Newcastle University, International Centre for Life, Newcastle upon Tyne, UK
| |
Collapse
|
31
|
Aoun M, Tiranti V. Mitochondria: A crossroads for lipid metabolism defect in neurodegeneration with brain iron accumulation diseases. Int J Biochem Cell Biol 2015; 63:25-31. [DOI: 10.1016/j.biocel.2015.01.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 01/15/2015] [Accepted: 01/29/2015] [Indexed: 11/16/2022]
|
32
|
Meyer E, Kurian MA, Hayflick SJ. Neurodegeneration with Brain Iron Accumulation: Genetic Diversity and Pathophysiological Mechanisms. Annu Rev Genomics Hum Genet 2015; 16:257-79. [PMID: 25973518 DOI: 10.1146/annurev-genom-090314-025011] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Neurodegeneration with brain iron accumulation (NBIA) comprises a heterogeneous group of progressive disorders with the common feature of excessive iron deposition in the brain. Over the last decade, advances in sequencing technologies have greatly facilitated rapid gene discovery, and several single-gene disorders are now included in this group. Identification of the genetic bases of the NBIA disorders has advanced our understanding of the disease processes caused by reduced coenzyme A synthesis, impaired lipid metabolism, mitochondrial dysfunction, and defective autophagy. The contribution of iron to disease pathophysiology remains uncertain, as does the identity of a putative final common pathway by which the iron accumulates. Ongoing elucidation of the pathogenesis of each NBIA disorder will have significant implications for the identification and design of novel therapies to treat patients with these disorders.
Collapse
Affiliation(s)
- Esther Meyer
- Molecular Neurosciences, Developmental Neurosciences Programme, Institute of Child Health, University College London, London WC1N 1EH, United Kingdom; ,
| | | | | |
Collapse
|
33
|
Ceccatelli Berti C, Dallabona C, Lazzaretti M, Dusi S, Tosi E, Tiranti V, Goffrini P. Modeling human Coenzyme A synthase mutation in yeast reveals altered mitochondrial function, lipid content and iron metabolism. MICROBIAL CELL 2015; 2:126-135. [PMID: 28357284 PMCID: PMC5348974 DOI: 10.15698/mic2015.04.196] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Mutations in nuclear genes associated with defective coenzyme A biosynthesis have been identified as responsible for some forms of neurodegeneration with brain iron accumulation (NBIA), namely PKAN and CoPAN. PKAN are defined by mutations in PANK2, encoding the pantothenate kinase 2 enzyme, that account for about 50% of cases of NBIA, whereas mutations in CoA synthase COASY have been recently reported as the second inborn error of CoA synthesis leading to CoPAN. As reported previously, yeast cells expressing the pathogenic mutation exhibited a temperature-sensitive growth defect in the absence of pantothenate and a reduced CoA content. Additional characterization revealed decreased oxygen consumption, reduced activities of mitochondrial respiratory complexes, higher iron content, increased sensitivity to oxidative stress and reduced amount of lipid droplets, thus partially recapitulating the phenotypes found in patients and establishing yeast as a potential model to clarify the pathogenesis underlying PKAN and CoPAN diseases.
Collapse
Affiliation(s)
| | | | | | - Sabrina Dusi
- Unit of Molecular Neurogenetics - Pierfranco and Luisa Mariani Center for the study of Mitochondrial Disorders in Children, IRCCS Foundation Neurological Institute "C. Besta", Milan, Italy
| | - Elena Tosi
- Department of Life Sciences, University of Parma, Parma, Italy
| | - Valeria Tiranti
- Unit of Molecular Neurogenetics - Pierfranco and Luisa Mariani Center for the study of Mitochondrial Disorders in Children, IRCCS Foundation Neurological Institute "C. Besta", Milan, Italy
| | - Paola Goffrini
- Department of Life Sciences, University of Parma, Parma, Italy
| |
Collapse
|
34
|
Schneider SA, Kurian MA. What the future holds for the genetic diagnosis for neurodegeneration with brain iron accumulation syndromes? Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.1016910] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
35
|
Santambrogio P, Dusi S, Guaraldo M, Rotundo LI, Broccoli V, Garavaglia B, Tiranti V, Levi S. Mitochondrial iron and energetic dysfunction distinguish fibroblasts and induced neurons from pantothenate kinase-associated neurodegeneration patients. Neurobiol Dis 2015; 81:144-53. [PMID: 25836419 PMCID: PMC4642744 DOI: 10.1016/j.nbd.2015.02.030] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 02/11/2015] [Accepted: 02/23/2015] [Indexed: 12/31/2022] Open
Abstract
Pantothenate kinase-associated neurodegeneration is an early onset autosomal recessive movement disorder caused by mutation of the pantothenate kinase-2 gene, which encodes a mitochondrial enzyme involved in coenzyme A synthesis. The disorder is characterised by high iron levels in the brain, although the pathological mechanism leading to this accumulation is unknown. To address this question, we tested primary skin fibroblasts from three patients and three healthy subjects, as well as neurons induced by direct fibroblast reprogramming, for oxidative status, mitochondrial functionality and iron parameters. The patients' fibroblasts showed altered oxidative status, reduced antioxidant defence, and impaired cytosolic and mitochondrial aconitase activities compared to control cells. Mitochondrial iron homeostasis and functionality analysis of patient fibroblasts indicated increased labile iron pool content and reactive oxygen species development, altered mitochondrial shape, decreased membrane potential and reduced ATP levels. Furthermore, analysis of induced neurons, performed at a single cell level, confirmed some of the results obtained in fibroblasts, indicating an altered oxidative status and signs of mitochondrial dysfunction, possibly due to iron mishandling. Thus, for the first time, altered biological processes have been identified in vitro in live diseased neurons. Moreover, the obtained induced neurons can be considered a suitable human neuronal model for the identification of candidate therapeutic compounds for this disease.
Collapse
Affiliation(s)
- Paolo Santambrogio
- San Raffaele Scientific Institute, Division of Neuroscience, 20132 Milano, Italy
| | - Sabrina Dusi
- Molecular Neurogenetics Unit, Foundation IRCCS-Neurological Institute "Carlo Besta", 20126 Milano, Italy
| | - Michela Guaraldo
- San Raffaele Scientific Institute, Division of Neuroscience, 20132 Milano, Italy; University Vita-Salute San Raffaele, 20132 Milano, Italy
| | - Luisa Ida Rotundo
- San Raffaele Scientific Institute, Division of Neuroscience, 20132 Milano, Italy
| | - Vania Broccoli
- San Raffaele Scientific Institute, Division of Neuroscience, 20132 Milano, Italy
| | - Barbara Garavaglia
- Molecular Neurogenetics Unit, Foundation IRCCS-Neurological Institute "Carlo Besta", 20126 Milano, Italy
| | - Valeria Tiranti
- Molecular Neurogenetics Unit, Foundation IRCCS-Neurological Institute "Carlo Besta", 20126 Milano, Italy
| | - Sonia Levi
- San Raffaele Scientific Institute, Division of Neuroscience, 20132 Milano, Italy; University Vita-Salute San Raffaele, 20132 Milano, Italy.
| |
Collapse
|
36
|
Levi S, Finazzi D. Neurodegeneration with brain iron accumulation: update on pathogenic mechanisms. Front Pharmacol 2014; 5:99. [PMID: 24847269 PMCID: PMC4019866 DOI: 10.3389/fphar.2014.00099] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 04/17/2014] [Indexed: 12/21/2022] Open
Abstract
Perturbation of iron distribution is observed in many neurodegenerative disorders, including Alzheimer’s and Parkinson’s disease, but the comprehension of the metal role in the development and progression of such disorders is still very limited. The combination of more powerful brain imaging techniques and faster genomic DNA sequencing procedures has allowed the description of a set of genetic disorders characterized by a constant and often early accumulation of iron in specific brain regions and the identification of the associated genes; these disorders are now collectively included in the category of neurodegeneration with brain iron accumulation (NBIA). So far 10 different genetic forms have been described but this number is likely to increase in short time. Two forms are linked to mutations in genes directly involved in iron metabolism: neuroferritinopathy, associated to mutations in the FTL gene and aceruloplasminemia, where the ceruloplasmin gene product is defective. In the other forms the connection with iron metabolism is not evident at all and the genetic data let infer the involvement of other pathways: Pank2, Pla2G6, C19orf12, COASY, and FA2H genes seem to be related to lipid metabolism and to mitochondria functioning, WDR45 and ATP13A2 genes are implicated in lysosomal and autophagosome activity, while the C2orf37 gene encodes a nucleolar protein of unknown function. There is much hope in the scientific community that the study of the NBIA forms may provide important insight as to the link between brain iron metabolism and neurodegenerative mechanisms and eventually pave the way for new therapeutic avenues also for the more common neurodegenerative disorders. In this work, we will review the most recent findings in the molecular mechanisms underlining the most common forms of NBIA and analyze their possible link with brain iron metabolism.
Collapse
Affiliation(s)
- Sonia Levi
- Proteomic of Iron Metabolism, Vita-Salute San Raffaele University Milano, Italy ; San Raffaele Scientific Institute Milano, Italy
| | - Dario Finazzi
- Department of Molecular and Translational Medicine, University of Brescia Brescia, Italy ; Spedali Civili di Brescia Brescia, Italy
| |
Collapse
|
37
|
Williams S, Gregory A, Hogarth P, Hayflick SJ, Gillingham MB. Metabolism and energy requirements in pantothenate kinase-associated neurodegeneration. Mol Genet Metab 2013; 110:336-41. [PMID: 23891537 PMCID: PMC6059611 DOI: 10.1016/j.ymgme.2013.06.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 06/20/2013] [Accepted: 06/21/2013] [Indexed: 01/04/2023]
Abstract
Pantothenate kinase-associated neurodegeneration (PKAN) is an autosomal recessive disorder of coenzyme A homeostasis caused by defects in the mitochondrial pantothenate kinase 2. Patients with PKAN present with a progressive neurological decline and brain iron accumulation, but general energy balance and nutrition status among these patients has not been reported. To determine if defects in PANK2 change basic energy metabolism in humans, we measured body composition, resting energy expenditure, dietary intake, and blood metabolites among 16 subjects with PKAN. Subjects had a broad range of disease severity but, despite the essential role of coenzyme A in energy metabolism, the subjects had remarkably normal body composition, dietary intake and energy metabolism compared to population normal values. We did observe increased resting energy expenditure associated with disease severity, suggesting increased energy needs later in the disease process, and elevated urinary mevalonate levels.
Collapse
Affiliation(s)
- Sarah Williams
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, USA
| | - Allison Gregory
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, USA
| | - Penelope Hogarth
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, USA
- Department of Neurology, Oregon Health & Science University, Portland, USA
| | - Susan J. Hayflick
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, USA
- Department of Neurology, Oregon Health & Science University, Portland, USA
- Department of Pediatrics, Oregon Health & Science University, Portland, USA
- Corresponding author at: Department of Molecular and Medical Genetics, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA. Fax: +1 503 494 6886. (S.J. Hayflick)
| | - Melanie B. Gillingham
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, USA
| |
Collapse
|
38
|
Pandey V, Varun P, Turm H, Hagit T, Bekenstein U, Uriya B, Shifman S, Sagiv S, Kadener S, Sebastian K. A new in vivo model of pantothenate kinase-associated neurodegeneration reveals a surprising role for transcriptional regulation in pathogenesis. Front Cell Neurosci 2013; 7:146. [PMID: 24058333 PMCID: PMC3766815 DOI: 10.3389/fncel.2013.00146] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2013] [Accepted: 08/21/2013] [Indexed: 01/24/2023] Open
Abstract
Pantothenate Kinase-Associated Neurodegeneration (PKAN) is a neurodegenerative disorder with a poorly understood molecular mechanism. It is caused by mutations in Pantothenate Kinase, the first enzyme in the Coenzyme A (CoA) biosynthetic pathway. Here, we developed a Drosophila model of PKAN (tim-fbl flies) that allows us to continuously monitor the modeled disease in the brain. In tim-fbl flies, downregulation of fumble, the Drosophila PanK homologue in the cells containing a circadian clock results in characteristic features of PKAN such as developmental lethality, hypersensitivity to oxidative stress, and diminished life span. Despite quasi-normal circadian transcriptional rhythms, tim-fbl flies display brain-specific aberrant circadian locomotor rhythms, and a unique transcriptional signature. Comparison with expression data from flies exposed to paraquat demonstrates that, as previously suggested, pathways others than oxidative stress are affected by PANK downregulation. Surprisingly we found a significant decrease in the expression of key components of the photoreceptor recycling pathways, which could lead to retinal degeneration, a hallmark of PKAN. Importantly, these defects are not accompanied by changes in structural components in eye genes suggesting that changes in gene expression in the eye precede and may cause the retinal degeneration. Indeed tim-fbl flies have diminished response to light transitions, and their altered day/night patterns of activity demonstrates defects in light perception. This suggest that retinal lesions are not solely due to oxidative stress and demonstrates a role for the transcriptional response to CoA deficiency underlying the defects observed in dPanK deficient flies. Moreover, in the present study we developed a new fly model that can be applied to other diseases and that allows the assessment of neurodegeneration in the brains of living flies.
Collapse
Affiliation(s)
- Varun Pandey
- Biological Chemistry Department, Silberman Institute of Life Sciences, The Hebrew University of Jerusalem Jerusalem, Israel
| | - Pandey Varun
- Biological Chemistry Department, Silberman Institute of Life Sciences, The Hebrew University of Jerusalem Jerusalem, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Schneider SA, Bhatia KP. Excess iron harms the brain: the syndromes of neurodegeneration with brain iron accumulation (NBIA). J Neural Transm (Vienna) 2012; 120:695-703. [PMID: 23212724 DOI: 10.1007/s00702-012-0922-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 11/11/2012] [Indexed: 12/14/2022]
Abstract
Regulation of iron metabolism is crucial: both iron deficiency and iron overload can cause disease. In recent years, our understanding of the syndromes of Neurodegeneration with Brain Iron Accumulation (NBIA) continues to grow considerably. These are characterized by excessive iron deposition in the brain, mainly the basal ganglia. Pantothenate kinase-associated neurodegeneration (PKAN, NBIA1) and PLA2G6-associated neurodegeneration (PLAN, NBIA2) are the core syndromes, but several other genetic causes have been identified (including FA2H, C19orf12, ATP13A2, CP and FTL). These conditions show a wide clinical and pathological spectrum, with clinical overlap between the different NBIA disorders and other diseases including spastic paraplegias, leukodystrophies, and neuronal ceroid lipofuscinosis. Lewy body pathology was confirmed in some clinical subtypes (C19orf12-associated neurodegeneration and PLAN). Research aims at disentangling the various NBIA genes and their related pathways to move towards pathogenesis-targeted therapies. Until then treatment remains symptomatic. Here we will introduce the group of NBIA syndromes and review the main clinical features and investigational findings.
Collapse
Affiliation(s)
- Susanne A Schneider
- Department of Neurology, University Kiel, Arnold Heller Str. 3, 24105, Kiel, Germany.
| | | |
Collapse
|