Your browser doesn't support javascript.
loading
Conservative iron chelation for neurodegenerative diseases such as Parkinson's disease and amyotrophic lateral sclerosis.
Devos, David; Cabantchik, Z Ioav; Moreau, Caroline; Danel, Véronique; Mahoney-Sanchez, Laura; Bouchaoui, Hind; Gouel, Flore; Rolland, Anne-Sophie; Duce, James A; Devedjian, Jean-Christophe.
Afiliación
  • Devos D; Service de Pharmacologie Clinique et Service de Neurologie NS-Park/FCRIN Network LICEND COEN Center Lille, Université de Lille, CHU de Lille, INSERM, UMRS_1171, Lille, France. david.devos@chru-lille.fr.
  • Cabantchik ZI; Service de Neurologie NS-Park/FCRIN Network LICEND COEN Center Lille, Université de Lille, CHU de Lille, INSERM, UMRS_1171, Lille, France. david.devos@chru-lille.fr.
  • Moreau C; Département de Pharmacologie Médicale, Université Lille INSERM 1171, CHU de Lille, 59037, Lille, France. david.devos@chru-lille.fr.
  • Danel V; Della Pergola Chair, Alexander Silberman Institute of Life Sciences, Hebrew University, 91904, Jerusalem, Israel.
  • Mahoney-Sanchez L; Service de Neurologie NS-Park/FCRIN Network LICEND COEN Center Lille, Université de Lille, CHU de Lille, INSERM, UMRS_1171, Lille, France.
  • Bouchaoui H; Service de Neurologie NS-Park/FCRIN Network LICEND COEN Center Lille, Université de Lille, CHU de Lille, INSERM, UMRS_1171, Lille, France.
  • Gouel F; Service de Pharmacologie Clinique et Service de Neurologie NS-Park/FCRIN Network LICEND COEN Center Lille, Université de Lille, CHU de Lille, INSERM, UMRS_1171, Lille, France.
  • Rolland AS; Service de Pharmacologie Clinique et Service de Neurologie NS-Park/FCRIN Network LICEND COEN Center Lille, Université de Lille, CHU de Lille, INSERM, UMRS_1171, Lille, France.
  • Duce JA; Service de Pharmacologie Clinique et Service de Neurologie NS-Park/FCRIN Network LICEND COEN Center Lille, Université de Lille, CHU de Lille, INSERM, UMRS_1171, Lille, France.
  • Devedjian JC; Service de Pharmacologie Clinique et Service de Neurologie NS-Park/FCRIN Network LICEND COEN Center Lille, Université de Lille, CHU de Lille, INSERM, UMRS_1171, Lille, France.
J Neural Transm (Vienna) ; 127(2): 189-203, 2020 02.
Article en En | MEDLINE | ID: mdl-31912279
ABSTRACT
Focal iron accumulation associated with brain iron dyshomeostasis is a pathological hallmark of various neurodegenerative diseases (NDD). The application of iron-sensitive sequences in magnetic resonance imaging has provided a useful tool to identify the underlying NDD pathology. In the three major NDD, degeneration occurs in central nervous system (CNS) regions associated with memory (Alzheimer's disease, AD), automaticity (Parkinson's disease, PD) and motor function (amyotrophic lateral sclerosis, ALS), all of which require a high oxygen demand for harnessing neuronal energy. In PD, a progressive degeneration of the substantia nigra pars compacta (SNc) is associated with the appearance of siderotic foci, largely caused by increased labile iron levels resulting from an imbalance between cell iron import, storage and export. At a molecular level, α-synuclein regulates dopamine and iron transport with PD-associated mutations in this protein causing functional disruption to these processes. Equally, in ALS, an early iron accumulation is present in neurons of the cortico-spinal motor pathway before neuropathology and secondary iron accumulation in microglia. High serum ferritin is an indicator of poor prognosis in ALS and the application of iron-sensitive sequences in magnetic resonance imaging has become a useful tool in identifying pathology. The molecular pathways that cascade down from such dyshomeostasis still remain to be fully elucidated but strong inroads have been made in recent years. Far from being a simple cause or consequence, it has recently been discovered that these alterations can trigger susceptibility to an iron-dependent cell-death pathway with unique lipoperoxidation signatures called ferroptosis. In turn, this has now provided insight into some key modulators of this cell-death pathway that could be therapeutic targets for the NDD. Interestingly, iron accumulation and ferroptosis are highly sensitive to iron chelation. However, whilst chelators that strongly scavenge intracellular iron protect against oxidative neuronal damage in mammalian models and are proven to be effective in treating systemic siderosis, these compounds are not clinically suitable due to the high risk of developing iatrogenic iron depletion and ensuing anaemia. Instead, a moderate iron chelation modality that conserves systemic iron offers a novel therapeutic strategy for neuroprotection. As demonstrated with the prototype chelator deferiprone, iron can be scavenged from labile iron complexes in the brain and transferred (conservatively) either to higher affinity acceptors in cells or extracellular transferrin. Promising preclinical and clinical proof of concept trials has led to several current large randomized clinical trials that aim to demonstrate the efficacy and safety of conservative iron chelation for NDD, notably in a long-term treatment regimen.
Asunto(s)
Palabras clave

Texto completo: 1 Bases de datos: MEDLINE Asunto principal: Enfermedad de Parkinson / Terapia por Quelación / Quelantes del Hierro / Deferiprona / Esclerosis Amiotrófica Lateral / Hierro Tipo de estudio: Clinical_trials / Prognostic_studies Límite: Animals / Humans Idioma: En Revista: J Neural Transm (Vienna) Año: 2020 Tipo del documento: Article País de afiliación: Francia

Texto completo: 1 Bases de datos: MEDLINE Asunto principal: Enfermedad de Parkinson / Terapia por Quelación / Quelantes del Hierro / Deferiprona / Esclerosis Amiotrófica Lateral / Hierro Tipo de estudio: Clinical_trials / Prognostic_studies Límite: Animals / Humans Idioma: En Revista: J Neural Transm (Vienna) Año: 2020 Tipo del documento: Article País de afiliación: Francia