-Author name in bold denotes the presenting author
-Asterisk * with author name denotes a Non-ASH member
Clinically Relevant Abstract denotes an abstract that is clinically relevant.

PhD Trainee denotes that this is a recommended PHD Trainee Session.

Ticketed Session denotes that this is a ticketed session.

2368 XPO1 (Exportin-1) Is a Major Regulator of Human Erythroid Differentiation. Potential Clinical Applications to Decrease Ineffective Erythropoiesis of Beta-Thalassemia

Hematopoietic Stem and Progenitor Biology
Program: Oral and Poster Abstracts
Session: 501. Hematopoietic Stem and Progenitor Biology: Poster II
Sunday, December 6, 2015, 6:00 PM-8:00 PM
Hall A, Level 2 (Orange County Convention Center)

Flavia Guillem, PhD1,2,3*, Michael Dussiot, PhD4*, Sebastien Causse, PhD5,6*, Guillaume Marcion7*, Emilie-Fleur Gautier, PhD4,8*, Julien Rossignol, MD9,10,11,12*, Jean benoit Arlet, MD, PhD13,14*, Mathilde Lamarque, MD3,15,16*, Patrick Mayeux, PhD8,17,18, Frederique Verdier, PhD4,8*, Xiuli An, MD, PhD19, Jean-Antoine Ribeil, MD, PhD20,21,22,23*, Carmen Garrido, PhD5,6,24*, Genevieve Courtois, PhD25*, Narla Mohandas, D.Sc.26 and Olivier Hermine9,10,12,27*

1Laboratoire INSERM, unité mixte de recherche 1163, centre national de la recherche scientifique (CNRS) e´quipe de recherche labellisée 8254, 24 Boulevard de Montparnasse, 75015 Paris, France, Paris, France
2Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Assistance publique – Hôpitaux de Paris, Hôpital Necker, 24 Boulevard de Montparnasse, 75015 Paris, France, Paris, France
3Laboratory of Excellence GR-Ex, 75015 Paris, France, Paris, France
4Laboratoire d’Excellence GR-Ex, PARIS, France
5INSERM, unité mixte de recherche 866, Equipe labellisée Ligue contre le Cancer and Association pour la Recherche contre le Cancer, and Laboratoire d’Excellence Lipoprotéines et santé (LipSTIC), 21033 Dijon, France, Dijon, France
6University of Burgundy, Faculty of Medicine and Pharmacy, 7 boulevard Jeanne d’Arc, 21033 Dijon, France, Dijon, France
7INSERM, unité mixte de recherche 866, Equipe labellisée Ligue contre le Cancer and Association pour la Recherche contre le Cancer, and Laboratoire d’Excellence Lipoprotéines et santé (LipSTIC), 21033 Dijon, France., Dijon, France
8INSTITUT COCHIN, INSERM U1016, PARIS, France
9Imagine Institute, Sorbonne Paris Cité, Paris Descartes University, Paris, France
10Department of Hematology, Necker Children's hospital, APHP, Paris, France
11Imagine Institute, INSERM UMR 1163, CNRS ERL 8654, Paris, France
12Centre de référence des déficits immunitaires héréditaires (CEREDIH), Necker Children's hospital, APHP, Paris, France
13Service de médecine interne, faculté de médecine Paris Descartes, Sorbonne Paris-Cité et Assistance publique-hôpitaux de Paris, hôpital européen Georges Pompidou, 20, rue Leblanc, 75015 Paris, France, Paris, France
14Centre national de référence des syndromes drépanocytaires majeurs, Paris, France
15Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Assistance publique – Hoˆpitaux de Paris, Hoˆ pital Necker, 24 Boulevard de Montparnasse, 75015 Paris, France., Paris, France
16Laboratoire INSERM, unité mixte de recherche 1163, centre national de la recherche scientifique (CNRS) équipe de recherche labellisée 8254, 24 Boulevard de Montparnasse, 75015 Paris, France., Paris, France
17Laboratoire d'excellence Gr-Ex, Paris, France
18Plateforme de Protéomique 3p5, Université Paris Descartes, PARIS, France
19New York Blood Center, New York, NY
20Hôpital Universitaire Necker -Enfants Malades, Paris, France
21Biotherapy Department, Hopital Universitaire Necker-Enfants Malades, Paris, France
22Paris Descartes Sorbonne Paris Cite University, Paris, France
23Laboratory of Excellence, GR-ex, Paris, France
24Centre anticancéreux George François Leclerc, 1 rue professeur Marion, 21079 Dijon, France, Dijon, France
25Laboratoire Inserm UMR 1163, CNRS ERL 8254, institut Imagine, hôpital Necker, laboratoire d’excellence sur le globule rouge (GR-Ex), Paris, France
26Red Cell Physiology Laboratory, New York Blood Center, New York, NY
27Institut Imagine, INSERM 1163/CNRS ERL 8254, Paris, France

Background
We and others have shown that normal human erythroid cell maturation requires a transient activation of caspase-3 at late stages of maturation (Zermati et al, J Exp Med 2001). We further documented that, in human erythroblasts, the chaperone HSP70 is constitutively expressed and, at late stages of maturation, translocates into the nucleus and protects GATA-1, the master transcriptional factor critical for erythropoiesis, from caspase-3 cleavage (Ribeil et al, Nature 2007). During the maturation of human β-TM erythroblasts, HSP70 is sequestrated by excess of α-globin chains in the cytoplasm and as a consequence, GATA-1 is no longer protected from caspase-3 cleavage resulting in end-stage maturation arrest and apoptosis (Arlet et al, Nature 2013). Understanding the molecular mechanisms that regulate the localization of HSP70 during erythroid differentiation may help to find new therapeutic targets to reduce ineffective erythropoiesis in beta-thalassemia.
Methods
CD34 positive cells from normal and thalassemic peripheral blood were cultured in IMDM/BIT media in the presence of SCF, IL3, IL6 for seven days and subsequently cultured for additional 7 to 9 days in media containing SCF, IL3 and Epo. Erythroblasts differentiation, HSP70 localization were analysed by FACS, AMNIS stream, confocal microscopy and western blot analysis. RNAseq and proteomic analysis of highly purified erythroid cells at all distinct stages of differentiation were used to assess expression levels of various exportins. Duolink and Octet analyses were used to assess protein proximity and affinity of interactions, respectively.
Results
During erythroid differentiation, Hikeshi, the cognate nuclear importin of HSP70, is constitutively expressed and enables HSP70 nucleus entry as assessed by siRNA experiments. However, its expression was not regulated during erythroid differentiation. In contrast, exportin expression analysis showed marked differences in expression levels of XPO1 and XPO7 during erythroid differentiation. XPO1 expression being reduced at the time of c-kit down-regulation and caspase 3 activation while there was a marked increase in XPO7 expression at the late stages of terminal erythroid differentiation. XPO1 interacted in vivo (Duolink analysis) and in vitro with HSP70 (Octet analysis). Likewise, the previously described HSP70 S400A mutant (in the Leucine-rich Nuclear Export Sequence), which is constitutively located in the nucleus interacted with XPO1 with lower affinity compared to HSP70 WT. Stem Cell Factor (SCF) starvation and Pi3k inhibition led to decreased in vivo HSP70/XPO1 interactions. However, neither phosphorylation of HSP70 nor XPO1 were detected by Nanopro and proteomic analysis, and XPO1 expression was not regulated by Pi3K pathway. Expression of RanGTP Activating Protein (RanGAP), a protein critical for XPO1/cargo interaction, was down-regulated at the moment of caspase 3 activation during erythroid maturation, which may explain the decrease in HSP70/XPO-1 interactions. Inhibitors of XPO1 (leptomycin B and KPT 251) were able to induce HSP70 nuclear localization at early stages of differentiation (proE).
In erythroid progenitors from β-TM patients, treatment with the Selective Inhibitor of Nuclear Export compound KPT-251 rescued nuclear HSP70 localization and GATA1 expression, and resulted in improved of erythroid terminal differentiation, without cytotoxicity, of thalassemic erythroid progenitors.
Conclusion

XPO1 is a major regulator of erythropoiesis through the regulation of HSP70 nuclear localization and is a potential new target to decrease ineffective erythropoiesis of thalassemia. Specific XPO-1 inhibitors currently in clinical development are being tested for potential therapy in thalassemic erythroid progenitors.

Disclosures: No relevant conflicts of interest to declare.

*signifies non-member of ASH