Your browser doesn't support javascript.
loading
Novel aspects of iron homeostasis in pathogenic bloodstream form Trypanosoma brucei.
Gilabert Carbajo, Carla; Cornell, Lucy J; Madbouly, Youssef; Lai, Zhihao; Yates, Phillip A; Tinti, Michele; Tiengwe, Calvin.
Afiliação
  • Gilabert Carbajo C; Department of Life Sciences, Sir Ernst Chain Building, Imperial College London, London, United Kingdom.
  • Cornell LJ; Department of Life Sciences, Sir Ernst Chain Building, Imperial College London, London, United Kingdom.
  • Madbouly Y; Department of Life Sciences, Sir Ernst Chain Building, Imperial College London, London, United Kingdom.
  • Lai Z; Department of Life Sciences, Sir Ernst Chain Building, Imperial College London, London, United Kingdom.
  • Yates PA; Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, United States of America.
  • Tinti M; Wellcome Trust Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom.
  • Tiengwe C; Department of Life Sciences, Sir Ernst Chain Building, Imperial College London, London, United Kingdom.
PLoS Pathog ; 17(6): e1009696, 2021 06.
Article em En | MEDLINE | ID: mdl-34161395
ABSTRACT
Iron is an essential regulatory signal for virulence factors in many pathogens. Mammals and bloodstream form (BSF) Trypanosoma brucei obtain iron by receptor-mediated endocytosis of transferrin bound to receptors (TfR) but the mechanisms by which T. brucei subsequently handles iron remains enigmatic. Here, we analyse the transcriptome of T. brucei cultured in iron-rich and iron-poor conditions. We show that adaptation to iron-deprivation induces upregulation of TfR, a cohort of parasite-specific genes (ESAG3, PAGS), genes involved in glucose uptake and glycolysis (THT1 and hexokinase), endocytosis (Phosphatidic Acid Phosphatase, PAP2), and most notably a divergent RNA binding protein RBP5, indicative of a non-canonical mechanism for regulating intracellular iron levels. We show that cells depleted of TfR by RNA silencing import free iron as a compensatory survival strategy. The TfR and RBP5 iron response are reversible by genetic complementation, the response kinetics are similar, but the regulatory mechanisms are distinct. Increased TfR protein is due to increased mRNA. Increased RBP5 expression, however, occurs by a post-transcriptional feedback mechanism whereby RBP5 interacts with its own, and with PAP2 mRNAs. Further observations suggest that increased RBP5 expression in iron-deprived cells has a maximum threshold as ectopic overexpression above this threshold disrupts normal cell cycle progression resulting in an accumulation of anucleate cells and cells in G2/M phase. This phenotype is not observed with overexpression of RPB5 containing a point mutation (F61A) in its single RNA Recognition Motif. Our experiments shed new light on how T. brucei BSFs reorganise their transcriptome to deal with iron stress revealing the first iron responsive RNA binding protein that is co-regulated with TfR, is important for cell viability and iron homeostasis; two essential processes for successful proliferation.
Assuntos

Texto completo: 1 Coleções: 01-internacional Contexto em Saúde: 3_ND Base de dados: MEDLINE Assunto principal: Trypanosoma brucei brucei / Adaptação Fisiológica / Proteínas de Protozoários / Ferro Idioma: En Revista: PLoS Pathog Ano de publicação: 2021 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Contexto em Saúde: 3_ND Base de dados: MEDLINE Assunto principal: Trypanosoma brucei brucei / Adaptação Fisiológica / Proteínas de Protozoários / Ferro Idioma: En Revista: PLoS Pathog Ano de publicação: 2021 Tipo de documento: Article