-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.

3334 The Human Kell Blood Group Binds the Erythroid 4.1R Protein: New Insights into the 4.1R-Dependent Red Cell Membrane Complex

Red Cells and Erythropoiesis, Structure and Function, Metabolism, and Survival, Excluding Iron
Program: Oral and Poster Abstracts
Session: 101. Red Cells and Erythropoiesis, Structure and Function, Metabolism, and Survival, Excluding Iron: Poster III
Monday, December 7, 2015, 6:00 PM-8:00 PM
Hall A, Level 2 (Orange County Convention Center)

Slim Azouzi1*, Emmanuel Collec1*, Narla Mohandas, DSc2, Xiuli An, MD, PhD3, Yves Colin Aronovicz1 and Caroline Le Van Kim1*

1UMR_S1134, University Paris Diderot/Inserm/INTS, Paris, France
2New York Blood Center, New York, NY
3Laboratory of Membrane Biology, New York Blood Center, New York, NY

Introduction: Protein 4.1R is a cytoskeletal adaptor protein that is responsible for the control of the mechanical stability of erythrocyte membranes, and for the proper anchoring of transmembrane proteins to the membrane skeletal network. Analysis of 4.1R-deficient human and murine erythrocytes revealed the complex array of membrane proteins that bind 4.1R and link these proteins to the spectrin-based skeletal network. 4.1R is composed of four functional domains: the N-terminal 30 kDa domain referred to as the FERM domain, the 16 kDa domain, the 10 kDa spectrin-actin binding domain, and the C-terminal 24 kDa domain. The Kell glycoprotein (93 kDa) is a type II single-span membrane protein which carry the Kell blood group system including the K1 (Kell) and K2 (cellano) antigens. Kell protein has endothelin-3 converting enzyme activity of type II membrane glycoproteins.

In this study we have analyzed the expression of Kell blood group protein in erythrocytes from a patient with hereditary elliptocytosis associated with complete 4.1R deficiency (4.1(-) HE) and performed detailed characterization of the interaction between 4.1R and Kell glycoprotein. Furthermore we also investigated the expression of membrane proteins exhibiting blood group antigens and the functional activities of AQP1, Band 3 and RhAG in the 4.1(-) HE erythrocyte membrane.

Results: Flow cytometry and western blot analyses revealed a severe reduction of Kell in the absence of 4.1R. In vitro pull down and co-immunoprecipitation experiments from erythrocyte membranes showed a direct interaction between Kell and 4.1R. Using different recombinant domains of 4.1R and the cytoplasmic domain of Kell, we demonstrated that the R46R motif in the juxta-membrane region of Kell binds to lobe B of the 4.1R FERM domain. We also observed that 4.1R deficiency is associated with a reduction of XK and DARC proteins, the absence of the glycosylated form of the urea transporter B and a slight decrease of band 3. The functional alteration of the 4.1(-) HE erythrocyte membranes was also determined by measuring various transport activities. We documented a slower rate of HCO3-/Cl- exchange (band 3-dependent), but a normal water (AQP1-dependent) and ammonia  (RhAG-dependent) transport in the absence of 4.1R.

Discussion: In this study, we provide evidence for a direct interaction between Kell and 4.1R and we propose an updated model for the 4.1R- multiprotein complex in human erythrocyte (Fig 1). The lobe A in the 4.1R FERM domain binds to protein transporters such as band 3, NHE1 and UT-B. Functional and structural experiments are required to confirm the presence of UT-B in this complex. The transmembrane proteins GPC, XK, Duffy and Kell bind to the lobe B and the binding site of p55 is located in lobe C.

The deficiency of blood group antigens carrying proteins in HS and HE erythrocytes can be explained by various molecular mechanisms including perturbed trafficking to the erythroblast membrane, aberrant protein sorting during erythroblast enucleation, and selective loss during reticulocyte membrane remodelling. Establishing when and where these proteins associate during erythroid differentiation should provide mechanistic insights into membrane multi-protein complex formation in both normal and abnormal erythropoiesis.

Conclusion: The findings from the present study using 4.1(-) HE human erythrocytes have enabled us to obtain novel insights into the 4.1R complex organization.

Figure 1. Proposed model of the 4.1R-multiprotein complex in human erythrocyte.


Proteins

Normal

4.1(-) HE

GPC

54075 (± 1075)

4000 (± 500)

Band 3

415000(±7000)

317000(±6000)

Kell

4150 (±66)

1220 (±35)

CD47

21300 (±1556)

21000 (± 2350)

Rh

135200 (±283)

121000 (±113)

RhAG

90450 (±2192)

71950 (±13150)

GPA

357500 (±7072)

310500(±12500)

DARC

2500 (±283)

1850 (±250)

CD44

4150 (±71)

4100 (±200)

UT-B*

13125 (± 1968)

2702 (±142)

Lu/BCAM

1300 (±141)

1624 (±153)

AQP1*

256 (±15)

202 (± 22)

GLUT-1

10961 (± 239)

11542 (± 806)

Table 1. Antigen and protein expression of human erythrocytes. Specific antibody binding capacity, as determined by indirect immunofluorescence using QIFIKIT calibrated beads. *Mean of fluorescence intensity given in arbitrary units.

Disclosures: No relevant conflicts of interest to declare.

*signifies non-member of ASH