Skip to main content
PLOS ONE logoLink to PLOS ONE
. 2013 Apr 2;8(4):e60300. doi: 10.1371/journal.pone.0060300

Protein Distribution during Human Erythroblast Enucleation In Vitro

Amanda J Bell 1, Timothy J Satchwell 1, Kate J Heesom 1, Bethan R Hawley 1, Sabine Kupzig 2, Matthew Hazell 2, Rosey Mushens 2, Andrew Herman 1, Ashley M Toye 1,2,*
Editor: Andrew C Wilber3
PMCID: PMC3614867  PMID: 23565219

Abstract

Enucleation is the step in erythroid terminal differentiation when the nucleus is expelled from developing erythroblasts creating reticulocytes and free nuclei surrounded by plasma membrane. We have studied protein sorting during human erythroblast enucleation using fluorescence activated cell sorting (FACS) to obtain pure populations of reticulocytes and nuclei produced by in vitro culture. Nano LC mass spectrometry was first used to determine the protein distribution profile obtained from the purified reticulocyte and extruded nuclei populations. In general cytoskeletal proteins and erythroid membrane proteins were preferentially restricted to the reticulocyte alongside key endocytic machinery and cytosolic proteins. The bulk of nuclear and ER proteins were lost with the nucleus. In contrast to the localization reported in mice, several key erythroid membrane proteins were detected in the membrane surrounding extruded nuclei, including band 3 and GPC. This distribution of key erythroid membrane and cytoskeletal proteins was confirmed using western blotting. Protein partitioning during enucleation was investigated by confocal microscopy with partitioning of cytoskeletal and membrane proteins to the reticulocyte observed to occur at a late stage of this process when the nucleus is under greatest constriction and almost completely extruded. Importantly, band 3 and CD44 were shown not to restrict specifically to the reticulocyte plasma membrane. This highlights enucleation as a stage at which excess erythroid membrane proteins are discarded in human erythroblast differentiation. Given the striking restriction of cytoskeleton proteins and the fact that membrane proteins located in macromolecular membrane complexes (e.g. GPA, Rh and RhAG) are segregated to the reticulocyte, we propose that the membrane proteins lost with the nucleus represent an excess mobile population of either individual proteins or protein complexes.

Introduction

During the final stages of erythroid terminal differentiation, the orthochromatic erythroblast enucleates to form the reticulocyte. Whilst undergoing this dramatic process, erythroid membrane proteins, cytoskeletal proteins and other cellular machinery required by the nascent reticulocyte must be selectively retained or will be lost with the extruded nucleus [1]. Studies using mouse erythroblasts have shown that the spectrin cytoskeleton, along with microtubules, myosin and actin partitions to the reticulocyte as the nucleus is removed [2], [3], [4]. Key erythroid membrane surface proteins were observed to be segregated to the nascent reticulocyte following enucleation including band 3 [5], [6], GPA [5], GPC [5] and RhAG [5] in murine cells. Several membrane proteins are selectively lost, such as the Beta 1 integrin [7], the vitamin C transporter SVCT2 [8] and erythroblast macrophage protein (EMP) [7], [9]. A mechanism has been proposed whereby retention of erythrocyte membrane proteins occurs by attachment to the cytoskeletal network via associated adaptor proteins or indirectly via multiprotein membrane protein complexes comprising band 3 or GPC [10]. Supporting this hypothesis, GPA cytoskeletal attachment is greater in erythroblasts than in reticulocytes [7], and the disruption of cytoskeletal attachment in ankyrin and protein 4.1R knockout mice resulted in the mislocalisation of specific membrane proteins (band 3 and RhAG for ankyrin disruption and GPC for protein 4.1) to the plasma membrane surrounding the nucleus [5].

It is currently unknown whether the protein sorting mechanism during enucleation is similar in humans. Griffiths et al recently presented confocal images of a selected number of membrane proteins, including GPA, GPC and Rh. Some immunofluorescence surrounding the extruding nucleus was perceivable in the images presented, and both basigin and beta 1 integrin were lost along with the nucleus [11]. However, partitioning of the majority of key erythrocyte membrane proteins (e.g. band 3, RhAG, Glut1, CD44) and many cytoskeletal proteins (alpha and beta spectrin, ankyrin or protein 4.2) was not investigated. We hypothesized that since differences in membrane protein multiprotein complex composition are known to exist between humans and mice [10], subtle differences may exist in the sorting process that occurs during enucleation. Identifying potential disparities is important to fully understand how specific protein deficiencies occur in human red blood cell diseases such as Hereditary Spherocytosis.

This study has adopted a global proteomic approach in combination with biochemical and detailed immunofluorescence analysis to explore the protein distribution and partitioning that occurs during human erythroblast enucleation. In general we find that there is a preferential restriction of erythroid membrane proteins to the reticulocyte and that this partitioning occurs at a very late stage during enucleation. Importantly, a substantial proportion of some membrane proteins, in particular band 3, CD44, GPC, Glut1 and stomatin are lost in the plasma membrane surrounding the nucleus in humans.

Methods

Antibodies

Monoclonal mouse antibodies used were BRIC256 (GPA), BRIC170 (band 3), LA1818 (RhAG), BRIC69 (Rh), BRIC4 (GPC), BRIC272/BRIC274 (ankyrin), BRIC273 (protein 4.2), BRAC65 (beta spectrin), BRIC172/BRIC276 (alpha spectrin), BRIC32 (CD47) (IBGRL, Filton, Bristol, UK), beta actin (Sigma), PDI (Assay Designs) and calnexin (RDI). BRIC272, BRIC273, BRIC276 and BRAC65 are all novel unpublished monoclonal antibodies. The novel antibodies were characterised using GFP-tagged cDNA expression, shRNA knockdown in K562 cells, and by using mature erythrocytes with a known protein deficiency. Rabbit monoclonal antibody used was beta 1 integrin (Novus). Rabbit polyclonal antibodies used were band 3, RhAG, GPC, Rh, Glut1, protein 4.1, p55, stomatin and CD44 (all available in house), flotillin-2 (Cell Signalling), alpha adducin (Santa Cruz). A goat polyclonal to lamin B was purchased from Santa Cruz. Secondary antibodies used were goat anti–mouse-Alexa 488 and goat anti-rabbit-Alexa 594 (Invitrogen), rabbit anti-mouse RPE, HRP-conjugated swine anti-rabbit and rabbit anti-mouse (Dako) and HRP conjugated donkey anti-goat (Jackson ImmunoResearch).

Erythroblast Cell Culture

Peripheral blood mononuclear cells were isolated from platelet apheresis waste blood (NHSBT, Bristol) from healthy donors with written informed consent for research use in accordance with the Declaration of Helsinki and approved by local Research Ethics Committee (Southmead Research Ethics Committee reference 08/H0102/26 and Bristol Research Ethics Committee Centre reference 12/SW/0199). Erythroblasts were expanded and differentiated using either the whole population of Peripheral Blood Mononuclear cells or from CD34+ as described previously [11], [12], [13]. The culture method for the PBMC population was modified as follows; a lineage depletion step (Lineage Cell Depletion Kit, Miltenyi Biotec, UK) was performed following Percoll on day 5 to ensure complete removal of lineage positive cells at this stage. IMDM (Source Biosciences) supplemented with 2% (v/v) fetal bovine serum (Hyclone, Fisher Scientific UK Ltd), 10 µg/ml insulin (Sigma), 200 µg/ml holotransferrin (Sigma), 3% (v/v) AB serum (Sigma) and 3 U/ml heparin (Sigma) replaced StemSpan SFEM during the expansion (phase 2) and differentiation phases (phase 3). Therefore, during Phase 2 of the culture IMDM base medium was supplemented with 2 U/ml Epo (Bristol Royal Infirmary, Bristol, UK), 1 µM dexamethasone (Sigma), 40 ng/ml IGF-1 (R&D systems), 40 µg/ml cholesterol-rich lipids (Sigma) and SCF (100 ng/ml). For Phase 3 of the culture, IMDM base medium was supplemented with 10 U/ml Epo, 1 mg/ml holotransferrin (Sigma), 3% human AB plasma (Sigma), 10 µg/ml insulin (Sigma), 1 µM thyroid hormone (Sigma), 40 ng/ml IGF-1, and 40 µg/ml cholesterol-rich lipids.

FACS Sorting

5×107 batches of enucleating erythroblasts were washed with PBS, then dual labelled with Hoechst 33342 (5 µg/ml) (Sigma) and BRIC256 (GPA) (detected with PE conjugated secondary). The reticulocyte and nuclei populations were then sorted using a BD Influx Cell Sorter. 1×105 cells from each population were cytospun as previously described [13]. The reticulocyte or nuclei populations were pelleted and stored at −80°C.

Proteomics

1×106 reticulocytes or nuclei were fractionated by 1D SDS-PAGE, gel lanes were cut into 4 equal portions and in-gel digested with trypsin. Extracted peptides were subjected to Nano LC mass spectrometry as described [14] but with modifications. The raw data files were processed using Proteome Discoverer software v1.2 (Thermo Scientific) and searched against the UniProt/SwissProt Human database release version 57.3 (20326 entries) using the SEQUEST (Ver. 28 Rev. 13) algorithm. Peptide precursor mass tolerance was set at 10 ppm, and MS/MS tolerance was set at 0.8 Da. Search criteria included carbamidomethylation of cysteine (+57.0214) as a fixed modification and oxidation of methionine (+15.9949) as a variable modification. Searches were performed with full tryptic digestion and a maximum of 1 missed cleavage was allowed. The reverse database search option was enabled and all peptide data was filtered to satisfy false discovery rate (FDR) of 5%. The Proteome Discoverer software generates a reverse “decoy” database from the same protein database and any peptides passing the initial filtering parameters that were derived from this decoy database are defined as false positive identifications. The minimum cross-correlation factor (Xcorr) filter was readjusted for each individual charge state separately to optimally meet the predetermined target FDR of 5% based on the number of random false positive matches from the reverse decoy database. Thus each data set has its own passing parameters.

Immunofluorescence

Immunostaining of enucleating erythroblasts were conducted as described previously [13]. Briefly, 6 × 105 cells were fixed in suspension in 0.5% acrolein in PBS (Sigma-Aldrich), washed 3 times in PBS-0.1M glycine before being cytospun onto coverslips coated with Cell-Tak (BD Biosciences). Cells were then permeabilized with 0.05% Triton X-100 for 5 minutes at room temperature and then blocked in PBS-4% BSA for 45 minutes, incubated with primary antibodies in PBS-4% BSA for 1 hour, washed with PBS, and incubated for 1 hour with goat anti–mouse Alexa 488–conjugated (Invitrogen) secondary antibodies and 4′,6-diamidino-2-phenylindole (Invitrogen). Coverslips were washed and mounted on microscope slides using Mowiol (Calbiochem) containing 2.5% (w/v) Dabco antifade reagent (Sigma-Aldrich). Confocal images were taken using a Leica AOBS SP2 confocal microscope (63×/1.4 NA oil-immersion lens and processed using Adobe Photoshop 9.0).

SDS-PAGE and Western Blotting

0.5−1×106 cells were lysed for 10 min on ice in lysis buffer (20 mM Tris-HCl, pH 8.0, 137 mM NaCl, 10 mM EDTA, 100 mM NaF, 1% (v/v) Nonidet P-40, 10% (v/v) glycerol, 10 mM Na3VO4, 2 mM PMSF and protease inhibitors, Calbiochem). Omnicleave (10 U/µl, Epicentre) was added to lysis buffer supplemented with 10 mM MgCl2 to digest the DNA present in the nuclei pellets. Equal numbers of lysed reticulocytes and nuclei were loaded and separated by SDS-PAGE and then immunoblotted.

Results

Protein Distribution in Reticulocyte and Nuclei Populations by Proteomics

GPA expression combined with Hoechst staining was exploited to separate reticulocytes and extruded nuclei [15] produced by in vitro erythroblast culture [11], [12], [13]. Three discrete populations were identified by flow cytometry; the reticulocyte population (GPAhigh:Hoechstnegative), extruded nuclei (GPAlow:Hoechstpositive) and nucleated erythroblasts (GPAhigh, Hoechstpositive) (Figure 1A). These populations were isolated using FACS sorting, and cytospins (Figure 1B) confirmed the purity of the reticulocyte (95.3+/−0.65% (n = 3, +/− SEM)) and nuclei (96.9+/−0.56% (n = 3,+/− SEM)) populations.

Figure 1. FACs sorting of reticulocytes and extruded nuclei.

Figure 1

A) Extruded nuclei and reticulocytes were separated by fluorescence activated cell sorting based on fluorescence intensity of DNA (Hoechst) and GPA (BRIC256) staining as outlined in the Materials and Methods. B) Representative cytospins from the sorted reticulocyte (upper panel) and extruded nuclei (lower panel) populations are shown.

To determine the protein distribution during enucleation, a proteomic comparison of the reticulocyte and extruded nuclei populations was undertaken. Tables 14 show a summarised list of peptides detected in the reticulocyte and nuclei populations. Table 1 shows membrane protein peptides detected, Table 2 shows examples of cytoskeletal or cytoskeletal interacting proteins detected, Table 3 nuclear and ER proteins and Table 4 cytosolic proteins and endocytic machinery. As expected, reticulocytes were enriched for peptides of cytoskeletal and erythrocyte membrane proteins. In addition a host of peptides derived from proteins from other cellular compartments such as cytosolic enzymes and endocytic proteins (e.g. Lamp1, clathrin, adaptor proteins, dynamin, sorting nexins,) were enriched in reticulocytes.

Table 1. Proteomic profile of membrane protein distribution in sorted populations of reticulocytes and extruded nuclei.

Nuclei Reticulocytes
Accession Description Total peptides Unique peptides Total peptides Unique peptides
Q9HDC9 Adipocyte plasma membrane-associated protein 22 12 6 4
Q02094 Ammonium transporter Rh type A 3 2 6 2
B4DNW4 Aquaporin 1 9 3 12 4
Q5T5M0 Aquaporin 7 4 1 3 1
Q9NP58 ATP-binding cassette sub-family B member 6, mitochondrial 4 4 40 18
P02730 Band 3 anion transport protein 195 28 477 36
Q54A51 Basigin 36 8 32 8
B6EAT9 CD44 2 2 4 2
E9PB22 CD47 3 1
Q99808 Equilibrative nucleoside transporter 1 11 5 14 5
Q96PL5 Erythroid membrane-associated protein 14 7
O75955 Flotillin-1 9 8 46 20
Q14254 Flotillin-2 6 5 42 20
P11166 Glucose transporter, type 1 51 10 78 12
P04921 Glycophorin-C 13 2 29 3
Q86SU0 Immunoglobulin-like domain-containing receptor 1 9 1 1 1
P20702 Integrin alpha-X 6 2 1 1
P05556 Integrin beta-1 11 7 2 2
P23276 Kell blood group glycoprotein 6 4
O75387 Large neutral amino acids transporter small subunit 3 2 1 4 2
P51811 Membrane transport protein XK 4 3
O15173 Membrane-associated progesterone receptor component 2 25 6 4 2
P53985 Monocarboxylate transporter 1 7 4 6 4
O15439 Multidrug resistance-associated protein 4 2 2 12 10
Q6PIU2 Neutral cholesterol ester hydrolase 1 52 16 12 7
P20020 Plasma membrane calcium-transporting ATPase 1 7 5 14 11
Q16720 Plasma membrane calcium-transporting ATPase 3 6 4 9 7
P23634 Plasma membrane calcium-transporting ATPase 4 12 10 20 16
Q9Y4D8 Probable E3 ubiquitin-protein ligase C12orf51 1 1 44 33
Q5VSJ9 Rh blood group, CcEe antigens 3 2 8 4
E9PS74 SLC43A3 3 2 4 3
Q96QG1 Sodium/calcium exchanger SCL8A3 2 2
B7Z3U6 Sodium/potassium-transporting ATPase subunit alpha-1 14 9 14 12
P54709 Sodium/potassium-transporting ATPase subunit beta-3 4 3 2 2
P27105 Stomatin 118 16 126 16
Q9UJZ1 Stomatin-like protein 2 5 4 13 11
Q9H1E5 Thioredoxin-related transmembrane protein 4 3 2
A6NJC0 TMCC2 65 13 18 6
P02786 Transferrin receptor protein 1 81 26 75 25
B7Z1P7 Transmembrane and coiled-coil domain family 2 151 25 36 15
Q13336 Urea transporter 1 2 2
Q9Y6M5 Zinc transporter 1 2 1 4 4

Sorted populations of reticulocytes and extruded nuclei were fractionated by 1D SDS-PAGE and subjected to Nano LC mass spectrometry. An abridged list containing key erythroid membrane proteins of interest is shown. Total peptide column is the total number of peptides (and therefore an indication of a particular protein’s abundance) detected in the population, whilst the unique peptide column indicates the number of unique peptides detected. To assess differences between nuclei and reticulocyte populations the total peptide number should be used.

Table 4. Proteomic profile of cytosolic and endocytic protein distribution in sorted populations of reticulocytes and extruded nuclei.

Nuclei Reticulocytes
Accession Description Total peptides Unique peptides Total peptides Unique peptides
P62258 14-3-3 protein epsilon 43 16 85 20
P61981 14-3-3 protein gamma 26 12 34 11
P63104 14-3-3 protein zeta/delta 35 12 50 11
Q01813 6-phosphofructokinase type C 1 1 47 21
B4DQJ8 6-phosphogluconate dehydrogenase, decarboxylating 24 12 84 24
P49588 Alanyl-tRNA synthetase, cytoplasmic 13 10 93 37
Q10567 AP-1 complex subunit beta-1 3 3 28 21
O95782 AP-2 complex subunit alpha-1 6 6 69 32
P63010 AP-2 complex subunit beta 6 5 46 27
Q2M2I8 AP2-associated protein kinase 1 5 5
C9JPM4 ARF4 23 6 7 4
P53396 ATP-citrate synthase 26 16 166 49
P07738 Bisphosphoglycerate mutase 34 13 78 17
P11586 C-1-tetrahydrofolate synthase, cytoplasmic 26 17 141 48
P07384 Calpain-1 catalytic subunit 7 6 78 36
P00915 Carbonic anhydrase 1 32 11 64 14
P00918 Carbonic anhydrase 2 49 15 92 21
P04040 Catalase 95 32 376 47
Q00610 Clathrin heavy chain 1 49 35 243 80
P53675 Clathrin heavy chain 2 6 6 40 17
P30046 D-dopachrome decarboxylase 1 1 7 5
Q16531 DNA damage-binding protein 1 41 22 89 48
P46734 Dual specificity mitogen-activated protein kinase kinase 3 33 15 57 20
P50570 Dynamin-2 3 3 23 16
E9PD66 E3 ubiquitin-protein ligase HUWE1 12 7 118 84
Q15075 Early endosome antigen 1 2 2 1 1
P13639 Elongation factor 2 23 11 84 32
P60842 Eukaryotic initiation factor 4A-I 52 18 90 23
P49327 Fatty acid synthase 19 15 135 76
P30043 Flavin reductase 123 16 253 19
P04075 Fructose-bisphosphate aldolase A 55 20 181 31
P11413 Glucose-6-phosphate 1-dehydrogenase 13 9 84 31
P48506 Glutamate–cysteine ligase catalytic subunit 4 4 81 31
E7EU54 Glyceraldehyde-3-phosphate dehydrogenase 42 11 104 14
P49840 Glycogen synthase kinase-3 alpha 3 3 7 4
P08107 Heat shock 70 kDa protein 1A/1B 77 23 96 32
P34932 Heat shock 70 kDa protein 4 10 7 77 35
P17066 Heat shock 70 kDa protein 6 39 9 35 8
P07900 Heat shock protein HSP 90-alpha 114 35 177 45
P08238 Heat shock protein HSP 90-beta 77 31 94 33
P54652 Heat shock-related 70 kDa protein 2 61 11 54 12
P69905 Hemoglobin subunit alpha 532 14 713 17
P68871 Hemoglobin subunit beta 802 20 1199 21
P07195 L-lactate dehydrogenase B chain 35 14 118 23
P11279 Lysosome-associated membrane glycoprotein 1 3 2 10 5
P32119 Peroxiredoxin-2 107 18 282 18
P30041 Peroxiredoxin-6 41 13 95 18
Q13492 Phosphatidylinositol-binding clathrin assembly protein 1 1 18 13
P00558 Phosphoglycerate kinase 1 46 20 102 28
F2Z2J9 Phosphoglycerate mutase 1 1 55 15
P08397 Porphobilinogen deaminase 55 18 111 22
Q9UKV8 Protein argonaute-2 6 4 43 22
P00491 Purine nucleoside phosphorylase 67 16 141 21
P30613 Pyruvate kinase isozymes R/L 17 13 97 32
P50395 Rab GDP dissociation inhibitor beta 54 28 132 40
Q96NA2 Rab-interacting lysosomal protein 2 2 27 14
Q99986 Serine/threonine-protein kinase VRK1 41 20
F5GWT4 Serine/threonine-protein kinase WNK1 1 1 24 20
A6NKH4 Sorting nexin 1 8 7
B4DEK4 Sorting nexin 2 1 1 16 13
Q9NRS6 Sorting nexin-15 3 3
Q9Y5X3 Sorting nexin-5 1 1 4 4
Q9UNH7 Sorting nexin-6 2 1 10 9
Q9Y5X1 Sorting nexin-9 4 3
Q9H2G2 STE20-like serine/threonine-protein kinase 1 1 22 16
P31948 Stress-induced-phosphoprotein 1 28 17 101 39
P17987 T-complex protein 1 subunit alpha 43 22 122 31
P37837 Transaldolase 44 20 74 26
P29401 Transketolase 42 14 117 33
P60174 Triosephosphate isomerase 44 15 104 23
P29144 Tripeptidyl-peptidase 2 1 1 78 45
P54578 Ubiquitin carboxyl-terminal hydrolase 14 18 9 87 27
Q9C0C9 Ubiquitin-conjugating enzyme E2 O 5 3 77 37
Q96RL7 Vacuolar protein sorting-associated protein 13A 11 9
F5GYF5 Vacuolar protein sorting-associated protein 35 1 1 14 11

Sorted populations of reticulocytes and extruded nuclei were fractionated by 1D SDS-PAGE and subjected to Nano LC mass spectrometry. An abridged list containing key cytosolic and endocytic proteins of interest is shown. Total peptide column is the total number of peptides (and therefore an indication of a particular protein’s abundance) detected in the population, whilst the unique peptide column indicates the number of unique peptides detected. To assess differences between nuclei and reticulocyte populations the total peptide number should be used.

Table 2. Proteomic profile of erythroid cytoskeletal protein distribution in sorted populations of reticulocytes and extruded nuclei.

Nuclei Reticulocytes
Accession Description Total peptides Unique peptides Total peptides Unique peptides
Q00013 55 kDa erythrocyte membrane protein 27 15 93 27
P68032 Actin, alpha cardiac muscle 116 16 127 15
P60709 Actin, cytoplasmic 223 25 228 24
P61160 Actin-related protein 2 6 5 22 11
O15143 Actin-related protein 2/3 complex subunit 1B 2 2 14 7
O15144 Actin-related protein 2/3 complex subunit 2 4 3 25 11
P61158 Actin-related protein 3 12 9 44 15
O43707 Alpha-actinin-4 4 4 12 7
P35611 Alpha-adducin 9 5 78 20
P16157 Ankyrin-1 123 65 476 104
E9PE32 Ankyrin-3 8 5 31 9
Q562R1 Beta-actin-like protein 2 41 6 46 6
P35612 Beta-adducin 72 25
B1AK87 Capping protein (Actin filament) muscle Z-line, beta 10 4 43 12
Q96H99 Cortactin 1 1 20 13
Q08495 Dematin 16 10 83 21
A8K8J9 Dynactin 2 (P50), isoform CRA_b 6 5 32 16
Q4KKX0 Erythrocyte membrane protein band 4.2 30 13 164 40
P21333 Filamin-A 27 23 94 66
Q9UEY8 Gamma-adducin 14 7
A2A418 Gelsolin 23 9 15 7
P33176 Kinesin-1 heavy chain 17 12
Q15691 Microtubule-associated protein RP/EB family member 1 5 3 12 8
P12829 Myosin light chain 4 10 5 24 8
P60660 Myosin light polypeptide 6 3 1 4 3
Q3MIV8 Myosin, heavy chain 11, smooth muscle 12 9 43 19
P35580 Myosin-10 48 38 210 106
Q7Z406 Myosin-14 10 8 25 12
P35579 Myosin-9 75 54 373 124
P11171 Protein 4.1 38 17 159 33
P02549 Spectrin alpha chain, erythrocyte 138 87 735 176
P11277 Spectrin beta chain, erythrocyte 105 70 637 165
Q9Y490 Talin-1 66 48 224 102
Q9Y4G6 Talin-2 6 6 24 11
P28289 Tropomodulin-1 4 4 32 16
D9YZV5 Tropomyosin 1 (Alpha) isoform 4 9 5 10 4
Q5VU58 Tropomyosin 3 19 9 37 12
P06753 Tropomyosin alpha-3 chain 11 6 18 6
Q71U36 Tubulin alpha-1 chain 61 13 141 21
P07437 Tubulin beta chain 89 23 249 28
A8MUB1 Tubulin, alpha 1 (Testis specific) 49 9 119 20
B3KPW9 Tubulin, alpha 8 33 7 80 13
B3KS31 Tubulin, beta 6 23 7 61 9
P18206 Vinculin 9 6 57 35

Sorted populations of reticulocytes and extruded nuclei were fractionated by 1D SDS-PAGE and subjected to Nano LC mass spectrometry. An abridged list containing key cytoskeletal proteins of interest is shown. Total peptide column is the total number of peptides (and therefore an indication of a particular protein’s abundance) detected in the population, whilst the unique peptide column indicates the number of unique peptides detected. To assess differences between nuclei and reticulocyte populations the total peptide number should be used.

Table 3. Proteomic profile of nuclear and ER protein distribution in sorted populations of reticulocytes and extruded nuclei.

Nuclei Reticulocytes
Accession Description Total peptides Unique peptides Total peptides Unique peptides
P11021 78 kDa glucose-regulated protein 96 30 48 24
P46013 Antigen KI-67 262 132 14 12
O00148 ATP-dependent RNA helicase DDX39A 57 19 18 9
Q8IWX8 Calcium homeostasis endoplasmic reticulum protein 3 3
P27824 Calnexin 45 15 8 5
P27797 Calreticulin 95 16 27 13
P11387 DNA topoisomerase 1 129 33 2 2
P78527 DNA-dependent protein kinase catalytic subunit 194 113 55 47
O60762 Dolichol-phosphate mannosyltransferase 21 15
P39656 Dolichyl-diphosphooligosaccharide–protein glycosyltransferase 48 kDa subunit 20 11 4 4
P04843 Dolichyl-diphosphooligosaccharide–protein glycosyltransferase subunit 1 56 26 9 7
P49792 E3 SUMO-protein ligase RanBP2 61 47
Q9NZ08 Endoplasmic reticulum aminopeptidase 1 4 4
P30040 Endoplasmic reticulum resident protein 29 20 8 3 1
Q9BS26 Endoplasmic reticulum resident protein 44 16 9 6 3
Q969X5 Endoplasmic reticulum-Golgi intermediate compartment protein 1 3 3
P14625 Endoplasmin 33 22 12 9
Q9P0I2 ER membrane protein complex subunit 3 3 3
O75396 ER-Golgi SNARE of 24 kDa 40 11 8 5
Q9Y5B9 FACT complex subunit SPT16 62 30 4 4
A8K318 Glucosidase 2 subunit beta 23 13 6 6
P09601 Heme oxygenase 1 16 9
Q9BXL5 Hemogen 119 21 42 16
Q5SSJ5 Heterochromatin protein 1-binding protein 3 40 18 1 1
P09429 High mobility group protein B1 160 16 14 5
P26583 High mobility group protein B2 188 17 23 10
Q02539 Histone H1.1 144 10 18 4
P04908 Histone H2A type 1-B/E 182 6 14 4
P68431 Histone H3.1 117 12 22 5
P62805 Histone H4 266 14 33 10
Q5TCI8 Lamin A/C 212 43 40 19
P42166 Lamina-associated polypeptide 2, isoform alpha 148 34 11 5
P42167 Lamina-associated polypeptide 2, isoforms beta/gamma 137 22 13 7
Q14739 Lamin-B receptor 53 17 2 2
P20700 Lamin-B1 162 43 22 13
Q03252 Lamin-B2 103 35 5 4
P43243 Matrin-3 28 13 5 2
Q8N4V1 Membrane magnesium transporter 1 2 1
Q9UNW1 Multiple inositol polyphosphate phosphatase 1 25 14 5 5
Q8NFW8 N-acylneuraminate cytidylyltransferase 111 24 8 4
Q14697 Neutral alpha-glucosidase AB 88 32 22 14
Q8N1F7 Nuclear pore complex protein Nup93 33 18 6 4
Q8TEM1 Nuclear pore membrane glycoprotein 210 60 33 2 2
Q9NR30 Nucleolar RNA helicase 2 39 20
P19338 Nucleolin 99 32 18 15
Q5SRE5 Nucleoporin NUP188 homolog 15 13 1 1
P12270 Nucleoprotein TPR 105 68 17 12
P02545 Prelamin-A/C 284 56 54 26
P07237 Protein disulfide-isomerase 74 22 27 16
P13667 Protein disulfide-isomerase A4 7 7
B7Z254 Protein disulfide-isomerase A6 24 12 7 5
P49257 Protein ERGIC-53 3 3 3 2
Q5JYR6 Ribophorin II 31 10 5 3
P55072 Transitional endoplasmic reticulum ATPase 57 25 133 50
Q9NYU2 UDP-glucose:glycoprotein glucosyltransferase 1 75 45 28 19
O95292 Vesicle-associated membrane protein-associated protein B/C 11 3 2 1

Sorted populations of reticulocytes and extruded nuclei were fractionated by 1D SDS-PAGE and subjected to Nano LC mass spectrometry. An abridged list containing key nuclear proteins and ER proteins of interest is shown. Total peptide column is the total number of peptides (and therefore an indication of a particular protein’s abundance) detected in the population, whilst the unique peptide column indicates the number of unique peptides detected. To assess differences between nuclei and reticulocyte populations the total peptide number should be used.

The extruded nuclei population was enriched for peptides from nuclear proteins (e.g. histones, lamins, DNA topoisomerase, nuclear pore proteins), ER proteins (e.g. PDI, calnexin, calreticulin), and a number of membrane proteins (e.g. integrins). Generally, low numbers of erythrocyte membrane protein peptides were detected in the nuclei but equal numbers of peptides for several membrane proteins including stomatin, transferrin receptor, Na+K+ ATPase and basigin were detected in both the nuclei and reticulocyte samples. Interestingly, peptides from actin and actin binding proteins (e.g cortactin, actinin, ARP 2/3 components) were also detected in the nuclei population, suggesting that some actin and associated proteins are lost with the nucleus at this stage, reflecting their additional role in nuclear processes [16]. It is notable that although higher numbers of peptides for band 3, CD44, GPC, Glut1 and Aquaporin1 were detected in the reticulocytes, considerable numbers of peptides for these proteins were also detected in the nuclei sample. Overall this proteomic dataset confirms the enrichment of erythroid membrane proteins to the reticulocyte and also reflects the fact that the extruded nucleus contains ER proteins, a proportion of cytosol and is surrounded by plasma membrane.

Distribution of Membrane Proteins in Reticulocyte and Nuclei Populations by Western Blotting

The partitioning observed for key membrane and cytoskeletal proteins using proteomics was confirmed by western blotting (Figure 2A–B). Importantly, cytoskeletal proteins (alpha and beta spectrin) or cytoskeletal adaptor proteins (ankyrin, 4.1, adducin, protein 4.2) were clearly restricted to the reticulocyte. Interestingly two components of the erythroid cytoskeleton, p55 and actin, were not totally restricted (Figure 2B). Lamin B, a protein of the nuclear lamina, was found only in the nuclei illustrating the purity of the reticulocyte and nuclei populations (Figure 2C). Nuclei contained high levels of the ER protein calnexin consistent with the loss of the majority of the ER with the nucleus (Figure 2D). Some membrane proteins (e.g. Rh and RhAG) were barely detectable in the nuclei population by western blot, highlighting the sensitivity of the mass spectrometry approach and the heightened retention of these proteins in reticulocytes. Importantly, we consistently detected significant amounts of band 3, GPC, CD44 and Glut1 in both reticulocyte and nuclei samples highlighting differential retention of specific membrane proteins to the reticulocyte during the enucleation process. This work also highlights enucleation as a significant stage of stomatin loss, since stomatin partitioned equally between reticulocyte and nuclei populations whereas another lipid microdomain protein, flotillin-2, was restricted to reticulocytes (Figure 2A).

Figure 2. Erythroid protein distribution in sorted populations of reticulocytes and extruded nuclei.

Figure 2

Sorted populations of extruded nuclei and reticulocytes were lysed and either 5×105 or 1×106 reticulocytes and nuclei were loaded depending on the protein expression levels or antibody sensitivity. Western blotting was conducted on A) membrane proteins using a mouse monoclonal antibody to Band 3, rabbit polyclonals to RhAG, Rh, Flotillin-2, Glut1, GPC, CD44 and stomatin and a rabbit monoclonal to beta 1 integrin B) cytoskeletal proteins using mouse monoclonal antibodies to alpha spectrin, beta spectrin, ankyrin, protein 4.2 and actin and rabbit polyclonals to alpha adducin, protein 4.1 and p55. C) nuclear protein Lamin B using a goat polyclonal. D) ER protein calnexin using a monoclonal antibody. Blots for RhAG, Rh, band 3, GPC, CD44, alpha spectrin, beta spectrin, ankyrin, protein 4.2 and lamin B are representative of 3–4 repeats from 3–4 independent cultures and sorting experiments. Blots for flotillin-2, Glut1, stomatin, beta 1 integrin, alpha adducin, protein 4.1, p55, actin and calnexin are representative of 2 repeats from 2 independent cultures and sorting experiments. All western blots shown were conducted on material isolated from the same reticulocyte and nuclei sorting experiment.

Imaging Protein Distribution during Early and Late Stages of Enucleation

To investigate the localisation and distribution of cytoskeletal and membrane proteins during enucleation, confocal imaging of acrolein fixed erythroblasts was undertaken (Figure 3). No obvious change in membrane protein distribution was observed during the early stages of nuclear extrusion (top row, Figure 3) where the nucleus has polarised and begins to deform the membrane. However, in cells where the nucleus is being deformed as it is squeezed out of the cell (bottom row, Figure 3), complete or partial partitioning of certain erythroid membrane proteins (GPA, GPC, Rh, RhAG and CD47) and cytoskeletal proteins/cytoskeleton associated proteins (alpha spectrin, beta spectrin and ankyrin) to the reticulocyte was observed. We conclude that remodelling of the cytoskeleton and of the majority of membrane components occurs during the late stages of enucleation. Confocal imaging of the ER protein Protein Disulphide Isomerase (PDI) confirmed that ER membrane surrounding the nucleus partitions with the nuclei (Figure 4A). Although the majority of the PDI staining localised as a ring around the nucleus, some remnants of PDI were observed in nascent reticulocytes (results not shown) further supporting the distribution of calnexin shown by Western blot in Figure 2D.

Figure 3. Immunofluorescence of membrane and cytoskeletal protein localisation during human erythroblast enucleation.

Figure 3

Human orthochromatic erythroblasts undergoing enucleation after 144 h of differentiation were removed from culture, fixed in 0.5% acrolein and permeabilised using 0.05% Triton X-100. Images shown are slices through cells in early (upper row) and late stages (lower row) of the enucleation process and detected with monoclonal antibodies against alpha spectrin, beta spectrin, ankyrin, band 3, GPC, GPA, RhAG, Rh, CD47 and a rabbit polyclonal antibody against CD44 and a suitable species specific fluorescent secondary as described in materials and methods. N = 5 for each antibody (although generally between 5–20) except for beta spectrin due to problems with high background fluorescence in the nucleus. Scale bar = 5 µm.

Figure 4. Immunofluorescence microscopy confirms that band 3 and ER are lost during nuclear extrusion.

Figure 4

Human orthochromatic erythroblasts undergoing enucleation after 144 h of differentiation were removed from culture, fixed in 0.5% acrolein and permeabilised using 0.05% Triton X-100. A) Confocal section of enucleating cells labelled with PDI and band 3 antibodies. B) Confocal section showing extruded nuclei (marked with arrows) labelled with band 3 (BRIC170). C) Confocal section of an erythroblast in the late stage of enucleation co-labelled with ankyrin and band 3 antibodies. The non-association of band 3 with ankyrin was observed in every cell identified at the late stage of enucleation (n = 10). Scale bar represents 5 µm.

Interestingly, Figure 3 shows by immunofluorescence that band 3 and CD44 were distributed evenly around the plasma membrane surrounding both the reticulocyte and nucleus throughout enucleation. Band 3 was also detected on isolated extruded nuclei (Figure 4B). However, proteins which connect band 3 to the spectrin cytoskeleton (protein 4.2 and ankyrin) together with other membrane proteins located within band 3 multiprotein complexes (Rh, RhAG, CD47, GPA) were largely excluded from the extruding nucleus as illustrated in Figure 3 and by the co-labelling of band 3 and ankyrin (Figure 4C).

Discussion

We have provided the most detailed study to date of the protein distribution between reticulocytes and the extruded nuclei. This has confirmed that many erythroid membrane and cytoskeletal proteins partition predominantly or exclusively to the reticulocyte during this process. In contrast, nuclear proteins, ER proteins, and a contingent of cytosolic and plasma membrane proteins distribute with the extruded nucleus. This is consistent with observations using electron microscopy where the extruded nucleus is described as being accompanied by a thin rim of cytoplasm, surrounded by plasma membrane [17], [18]. Furthermore, we have demonstrated here that the majority of the ER is lost with the extruded nucleus, building on the observation by imaging that the ER protein calreticulin is lost with the nucleus [11]. ER remnants are still detectable by western blotting (see Figure 2D) and by confocal imaging (results not shown) in the reticulocyte, which we presume are lost upon further reticulocyte maturation.

This work highlights the enucleation step as a significant point of membrane remodelling in human erythropoiesis where excess erythroid membrane proteins are discarded. Unlike in mouse erythroblasts [5], [6], a significant population of human band 3 and to a lesser extent GPC is lost during enucleation. The apparent disparity in distribution, particularly for band 3, between species during enucleation may be due to intrinsic differences in the membrane protein complex composition known to exist between mice and humans [10] or due to mechanistic differences in the process of erythroblast protein sorting. Other membrane proteins were lost during enucleation including CD44, Glut1 and stomatin. For CD44, this further compounds the loss observed in human in vitro cultures during terminal differentiation [19] and since CD44 can bind ankyrin, this additional loss may result from continued competition for ankyrin binding sites with the band 3 population.

It is interesting that several of the proteins lost with the nucleus during enucleation are located in membrane protein complexes. Glut1 and band 3 interact in vitro [20] and stomatin interacts with the C-terminus of Glut1 [21]. Similarly an association may also exist between the GPC and p55 [22] observed in the nuclei population. The loss of these proteins with the nucleus, taken in conjunction with the restriction of the majority of membrane proteins to the reticulocyte, suggests that these represent proteins/complexes that are most likely synthesized in excess which are not attached to the cytoskeleton (e.g. by incorporation into ankyrin or junctional complexes) leaving them vulnerable to loss during enucleation. In addition, low numbers of peptides were detected in the nuclei relative to the reticulocytes for several cytosolic enzymes (e.g. 6-phosphofructokinase and calpain; see Table 4), therefore a mechanism may also exist for segregation of certain key cytosolic proteins in the reticulocyte, perhaps by incorporation into membrane/cytoskeletal complexes.

In summary, isolated pure populations of human reticulocytes and nuclei have been used to study protein partitioning during human erythroblast enucleation. This work is the first reported proteomic dataset for reticulocytes and extruded nuclei and provides the foundations for investigating reticulocyte maturation, sorting defects in human erythrocyte membrane disorders, and for comparison of protein sorting using erythroblasts produced using other cell sources (e.g. iPS or embryonic stem cells). Our observations here during human enucleation are generally supportive of the hypothesis that the cytoskeleton plays an important part in the segregation of membrane proteins to the reticulocyte during enucleation. Nevertheless in humans the partitioning and retention of specific proteins including the abundantly expressed band 3 to the reticulocyte, occurs in a less definitive manner than observed in mice. Further studies are needed to establish whether the loss of proteins during enucleation in human erythroblasts is an active or passive process and to ascertain whether disruption of the cytoskeleton, mimicking that of hereditary anaemias, leads to additional loss of proteins in humans in the same manner as has been reported in mice.

Acknowledgments

The authors thank Professor Jon Morrow (Yale University) for the gift of GFP tagged alpha and beta spectrin. Dr Lesley Bruce (NHSBT Filton) for providing the rabbit polyclonal anti-stomatin antibody and Dr Steve Parsons (NHSBT Filton) for providing the rabbit polyclonal anti-CD44.

Funding Statement

AB was funded by a Wellcome Trust PhD studentship and TJS by a Wellcome Trust project grant (094277). This work was funded in part by National Institute for Health Research programme grant to NHSBT (RP-PG-0310-1004 -AMT) and by NHSBT R&D. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  • 1. Keerthivasan G, Wickrema A, Crispino JD (2011) Erythroblast enucleation. Stem Cells Int 2011: 139851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Geiduschek JB, Singer SJ (1979) Molecular changes in the membranes of mouse erythroid cells accompanying differentiation. Cell 16: 149–163. [DOI] [PubMed] [Google Scholar]
  • 3. Koury ST, Koury MJ, Bondurant MC (1989) Cytoskeletal distribution and function during the maturation and enucleation of mammalian erythroblasts. J Cell Biol 109: 3005–3013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Wang J, Ramirez T, Ji P, Jayapal SR, Lodish HF, et al. (2012) Mammalian erythroblast enucleation requires PI3K-dependent cell polarization. J Cell Sci 125: 340–349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Salomao M, Chen K, Villalobos J, Mohandas N, An X, et al. (2010) Hereditary spherocytosis and hereditary elliptocytosis: aberrant protein sorting during erythroblast enucleation. Blood 116: 267–269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Patel VP, Lodish HF (1987) A fibronectin matrix is required for differentiation of murine erythroleukemia cells into reticulocytes. J Cell Biol 105: 3105–3118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Lee JC, Gimm JA, Lo AJ, Koury MJ, Krauss SW, et al. (2004) Mechanism of protein sorting during erythroblast enucleation: role of cytoskeletal connectivity. Blood 103: 1912–1919. [DOI] [PubMed] [Google Scholar]
  • 8. May JM, Qu ZC, Qiao H, Koury MJ (2007) Maturational loss of the vitamin C transporter in erythrocytes. Biochemical and Biophysical Research Communications 360: 295–298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Soni S, Bala S, Gwynn B, Sahr KE, Peters LL, et al. (2006) Absence of erythroblast macrophage protein (Emp) leads to failure of erythroblast nuclear extrusion. J Biol Chem 281: 20181–20189. [DOI] [PubMed] [Google Scholar]
  • 10. van den Akker E, Satchwell TJ, Williamson RC, Toye AM (2010) Band 3 multiprotein complexes in the red cell membrane; of mice and men. Blood Cells Mol Dis 45: 1–8. [DOI] [PubMed] [Google Scholar]
  • 11.Griffiths RE, Kupzig S, Cogan N, Mankelow TJ, Betin VM, et al. (2012) Maturing reticulocytes internalise plasma membrane in glycophorin A-containing vesicles which fuse with autophagosomes prior to exocytosis. Blood. [DOI] [PMC free article] [PubMed]
  • 12. van den Akker E, Satchwell TJ, Pellegrin S, Daniels G, Toye AM (2010) The majority of the in vitro erythroid expansion potential resides in CD34(-) cells, outweighing the contribution of CD34(+) cells and significantly increasing the erythroblast yield from peripheral blood samples. Haematologica 95: 1594–1598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Satchwell TJ, Bell AJ, Pellegrin S, Kupzig S, Ridgwell K, et al. (2011) Critical band 3 multiprotein complex interactions establish early during human erythropoiesis. Blood 118: 182–191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Pellegrin S, Heesom KJ, Satchwell TJ, Hawley BR, Daniels G, et al. (2012) Differential proteomic analysis of human erythroblasts undergoing apoptosis induced by epo-withdrawal. Plos One 7: e38356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Ji P, Jayapal SR, Lodish HF (2008) Enucleation of cultured mouse fetal erythroblasts requires Rac GTPases and mDia2. Nat Cell Biol 10: 314–321. [DOI] [PubMed] [Google Scholar]
  • 16. Visa N, Percipalle P (2010) Nuclear functions of actin. Cold Spring Harb Perspect Biol 2: a000620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Skutelsky E, Danon D (1967) An electron microscopic study of nuclear elimination from the late erythroblast. J Cell Biol 33: 625–635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Simpson CF, Kling JM (1967) The mechanism of denucleation in circulating erythroblasts. J Cell Biol 35: 237–245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. van den Akker E, Satchwell TJ, Pellegrin S, Flatt JF, Maigre M, et al. (2010) Investigating the key membrane protein changes during in vitro erythropoiesis of protein 4.2 (-) cells (mutations Chartres 1 and 2). Haematologica 95: 1278–1286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Jiang W, Ding Y, Su Y, Jiang M, Hu X, et al. (2006) Interaction of glucose transporter 1 with anion exchanger 1 in vitro. Biochem Biophys Res Commun 339: 1255–1261. [DOI] [PubMed] [Google Scholar]
  • 21. Zhang JZ, Abbud W, Prohaska R, Ismail-Beigi F (2001) Overexpression of stomatin depresses GLUT-1 glucose transporter activity. Am J Physiol Cell Physiol 280: C1277–1283. [DOI] [PubMed] [Google Scholar]
  • 22. Marfatia SM, Morais-Cabral JH, Kim AC, Byron O, Chishti AH (1997) The PDZ domain of human erythrocyte p55 mediates its binding to the cytoplasmic carboxyl terminus of glycophorin C. Analysis of the binding interface by in vitro mutagenesis. J Biol Chem 272: 24191–24197. [DOI] [PubMed] [Google Scholar]

Articles from PLoS ONE are provided here courtesy of PLOS

RESOURCES