scholarly journals The KH-Domain Protein αCP Has a Direct Role in mRNA Stabilization Independent of Its Cognate Binding Site

2003 ◽  
Vol 23 (4) ◽  
pp. 1125-1134 ◽  
Author(s):  
Jian Kong ◽  
Xinjun Ji ◽  
Stephen A. Liebhaber

ABSTRACT Previous studies suggest that high-level stability of a subset of mammalian mRNAs is linked to a C-rich motif in the 3′ untranslated region (3′UTR). High-level expression of human α-globin mRNA (hα-globin mRNA) in erythroid cells has been specifically attributed to formation of an RNA-protein complex comprised of a 3′UTR C-rich motif and an associated 39-kDa poly(C) binding protein, αCP. Documentation of this RNA-protein α-complex has been limited to in vitro binding studies, and its impact has been monitored by alterations in steady-state mRNA. Here we demonstrate that αCP is stably bound to hα-globin mRNA in vivo, that α-complex assembly on the hα-globin mRNA is restricted to the 3′UTR C-rich motif, and that α-complex assembly extends the physical half-life of hα-globin mRNA selectively in erythroid cells. Significantly, these studies also reveal that an artificially tethered αCP has the same mRNA-stabilizing activity as the native α-complex. These data demonstrate a unique contribution of the α-complex to hα-globin mRNA stability and support a model in which the sole function of the C-rich motif is to selectively tether αCP to a subset of mRNAs. Once bound, αCP appears to be fully sufficient to trigger downstream events in the stabilization pathway.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1775-1775
Author(s):  
Maria Elena Fabucci ◽  
Katija Jelicic ◽  
Elena Alfani ◽  
Anna Rita F. Migliaccio

Abstract Alpha haemoglobin stabilizing protein (AHSP) is encoded by a gene abundantly expressed in erythroid cells whose function is to chaperone α-chains in the process of haemoglobin assembly (Yu et al, JCI2007; 117:1856). The central role of the excess of α-chains in the pathogenesis of β-thalassemia and the AHSP ability to limit the toxicity of excessive α-globin suggest that increases of AHSP expression might ameliorate the clinical phenotype of β-thalassemia. To clarify the relationship between AHSP and globin gene expression, we measured the levels of mRNA for these genes in erythroblasts generated in vitro from the blood of 30 normal donors and 8 β-thalassemic patients. Normal erythroblasts presented a marked donor-to-donor variability in the expression levels of all the genes analysed. Inter-quartile range (IQR) analyses indicates that the gene whose expression has the highest variability is α-globin (IQR=31.5), followed by β-globin (IQR=8.74), AHSP (IQR=2.82) and γ-globin (IQR=0.86). The IQR value for the α/non-α globin ratio (1.91) is higher than that of the γ/γ+β ratio (0.11), an indication of the existence of donor variegation in the levels of unbalance between expression of α- and non-α globin genes in cells from different donors. The extent of this variegation is even more apparent by the high IQR level of the α-(non-α) expression difference (IQR=38.6). β-thalassemic erythroblasts expressed normal levels of α- and γ-globin, significantly (P<.05) lower levels of β-globin mRNA and, surprisingly, high levels (by 10-fold) of AHSP mRNA. Subject-variability in gene expression was also observed for β-thalassemic erythroblasts. In this case, the gene whose expression had the highest variability is AHSP (IQR=42.8), followed by α-globin (IQR=11.75), β-globin (IQR=3.32), and γ-globin (IQR=1.74). The IQR for the α/non-α globin ratio (7.2) is higher than that of the γ/γ+β ratio (0.67) also for β-thalassemic erythroblasts. The difference between the variances of the excess of α-expression [α-(non-α)] in β-thalassemic and normal erythroblasts is significant by F test (P=.0023). Statistical analyses of these results indicates that, as expected, the levels of α-globin mRNA are positively correlated to those of the non-α globin genes in normal erythroblasts (R2=.93, P<.001) but not in β-thalassemic cells (R2=.22, P<.24). In contrast, the levels of α-globin mRNA are positively correlated with those of AHSP both in normal (R2=.86, P<.0001) and β-thalassemic (R2=.66, P<.05) erythroblasts. Moreover, in spite of the fact that expression of α-globin is correlated, at least in normal erythroblasts, with that of γ+β mRNA, no correlation is found between levels of AHSP mRNA and those of γ+β mRNA. No correlation is also observed between levels of AHSP mRNA and the α/non-α ratio. In contrast, the levels of AHSP mRNA are correlated with the levels of excess of α-globin mRNA in normal erythroblasts (R2=0.86, P<.0001) and the fact that are not correlated in β-thalassemic cells (R2=.45, P=.066) might be due to the limited experimental points available for analyses. In conclusion, this statistical analyses provides evidence for the existence of a regulatory mechanism that balances expression of AHSP with that of excess of α-globin mRNA in erythroid cells. It is suggested that this regulatory mechanism may represent a target for eventual gene modifiers of the β-thalassemic trait. MEF is the recipient of a Marie Curie training Network Fellowship from EU.


Blood ◽  
1984 ◽  
Vol 64 (4) ◽  
pp. 930-936 ◽  
Author(s):  
I Max-Audit ◽  
U Testa ◽  
D Kechemir ◽  
M Titeux ◽  
W Vainchenker ◽  
...  

To further investigate the erythroid nature of the two human erythroleukemia cell lines, K562 and HEL-60, and to define the ontogeny of pyruvate kinase (PK) isozymes (R, M2) in developing human erythroid cells, we have studied the isozymic alterations, if any, during differentiation of these cell lines in vitro and normoblasts isolated from fetal liver in vivo. PK activity of erythroleukemic cell lines was intermediate between that observed in leukocytes and in fetal liver erythroblasts. These cell lines contained a high level of M2-PK, but R- PK was always present, albeit at low concentrations, in all the clones or subclones we studied. Erythroblasts from fetal liver were separated according to density on a Stractan gradient. R-PK levels were nearly constant in the different fractions, whereas M2-PK levels markedly decreased as the erythroblasts became mature and almost completely disappeared in late erythroid cells. Thus, these results clearly demonstrate the erythroid origin of these cell lines.


Blood ◽  
2012 ◽  
Vol 119 (4) ◽  
pp. 1045-1053 ◽  
Author(s):  
Sebastiaan van Zalen ◽  
Grace R. Jeschke ◽  
Elizabeth O. Hexner ◽  
J. Eric Russell

Abstract The normal accumulation of β-globin protein in terminally differentiating erythroid cells is critically dependent on the high stability of its encoding mRNA. The molecular basis for this property, though, is incompletely understood. Factors that regulate β-globin mRNA within the nucleus of early erythroid progenitors are unlikely to account for the constitutively high half-life of β-globin mRNA in the cytoplasm of their anucleate erythroid progeny. We conducted in vitro protein-RNA binding analyses that identified a cytoplasm-restricted β-globin messenger ribonucleoprotein (mRNP) complex in both cultured K562 cells and erythroid-differentiated human CD34+ cells. This novel mRNP targets a specific guanine-rich pentanucleotide in a region of the β-globin 3′untranslated region that has recently been implicated as a determinant of β-globin mRNA stability. Subsequent affinity-enrichment analyses identified AUF-1 and YB-1, 2 cytoplasmic proteins with well-established roles in RNA biology, as trans-acting components of the mRNP. Factor-depletion studies conducted in vivo demonstrated the importance of the mRNP to normal steady-state levels of β-globin mRNA in erythroid precursors. These data define a previously unrecognized mechanism for the posttranscriptional regulation of β-globin mRNA during normal erythropoiesis, providing new therapeutic targets for disorders of β-globin gene expression.


1992 ◽  
Vol 20 (5) ◽  
pp. 997-1003 ◽  
Author(s):  
M. Needham ◽  
C. Gooding ◽  
K. Hudson ◽  
M. Antoniou ◽  
F. Grosveld ◽  
...  

Blood ◽  
1984 ◽  
Vol 64 (4) ◽  
pp. 930-936 ◽  
Author(s):  
I Max-Audit ◽  
U Testa ◽  
D Kechemir ◽  
M Titeux ◽  
W Vainchenker ◽  
...  

Abstract To further investigate the erythroid nature of the two human erythroleukemia cell lines, K562 and HEL-60, and to define the ontogeny of pyruvate kinase (PK) isozymes (R, M2) in developing human erythroid cells, we have studied the isozymic alterations, if any, during differentiation of these cell lines in vitro and normoblasts isolated from fetal liver in vivo. PK activity of erythroleukemic cell lines was intermediate between that observed in leukocytes and in fetal liver erythroblasts. These cell lines contained a high level of M2-PK, but R- PK was always present, albeit at low concentrations, in all the clones or subclones we studied. Erythroblasts from fetal liver were separated according to density on a Stractan gradient. R-PK levels were nearly constant in the different fractions, whereas M2-PK levels markedly decreased as the erythroblasts became mature and almost completely disappeared in late erythroid cells. Thus, these results clearly demonstrate the erythroid origin of these cell lines.


Blood ◽  
1998 ◽  
Vol 92 (11) ◽  
pp. 4108-4118 ◽  
Author(s):  
Naruyoshi Suwabe ◽  
Satoru Takahashi ◽  
Toru Nakano ◽  
Masayuki Yamamoto

Abstract Although the importance of GATA-1 in both primitive and definitive hematopoietic lineages has been shown in vivo, the precise roles played by GATA-1 during definitive hematopoiesis have not yet been clarified. In vitro differentiation of embryonic stem (ES) cells using OP9 stroma cells can generate primitive and definitive hematopoietic cells separately, and we have introduced a method that separates hematopoietic progenitors and differentiated cells produced in this system. Closer examination showed that the expression of erythroid transcription factors in this system is regulated in a differentiation stage-specific manner. Therefore, we examined differentiation of GATA-1 promoter-disrupted (GATA-1.05) ES cells using this system. Because the GATA-1.05 mice die by 12.5 embryonic days due to the lack of primitive hematopoiesis, the in vitro analysis is an important approach to elucidate the roles of GATA-1 in definitive hematopoiesis. Consistent with the in vivo observation, differentiation of GATA-1.05 mutant ES cells along both primitive and definitive lineages was arrested in this ES cell culture system. Although the maturation-arrested primitive lineage cells did not express detectable amounts of ɛy-globin mRNA, the blastlike cells accumulated in the definitive stage showed β-globin mRNA expression at approximately 70% of the wild type. Importantly, the TER119 antigen was expressed and porphyrin was accumulated in the definitive cells, although the levels of both were reduced to approximately 10%, indicating that maturation of definitive erythroid cells is arrested by the lack of GATA-1 with different timing from that of the primitive erythroid cells. We also found that the hematopoietic progenitor fraction of GATA-1.05 cells contains more colony-forming activity, termed CFU-OP9. These results suggest that theGATA-1.05 mutation resulted in proliferation of proerythroblasts in the definitive lineage.


1998 ◽  
Vol 18 (11) ◽  
pp. 6634-6640 ◽  
Author(s):  
Denise E. Sabatino ◽  
Amanda P. Cline ◽  
Patrick G. Gallagher ◽  
Lisa J. Garrett ◽  
George Stamatoyannopoulos ◽  
...  

ABSTRACT During development, changes occur in both the sites of erythropoiesis and the globin genes expressed at each developmental stage. Previous work has shown that high-level expression of human β-like globin genes in transgenic mice requires the presence of the locus control region (LCR). Models of hemoglobin switching propose that the LCR and/or stage-specific elements interact with globin gene sequences to activate specific genes in erythroid cells. To test these models, we generated transgenic mice which contain the human Aγ-globin gene linked to a 576-bp fragment containing the human β-spectrin promoter. In these mice, the β-spectrin Aγ-globin (βsp/Aγ) transgene was expressed at high levels in erythroid cells throughout development. Transgenic mice containing a 40-kb cosmid construct with the micro-LCR, βsp/Aγ-, ψβ-, δ-, and β-globin genes showed no developmental switching and expressed both human γ- and β-globin mRNAs in erythroid cells throughout development. Mice containing control cosmids with the Aγ-globin gene promoter showed developmental switching and expressed Aγ-globin mRNA in yolk sac and fetal liver erythroid cells and β-globin mRNA in fetal liver and adult erythroid cells. Our results suggest that replacement of the γ-globin promoter with the β-spectrin promoter allows the expression of the β-globin gene. We conclude that the γ-globin promoter is necessary and sufficient to suppress the expression of the β-globin gene in yolk sac erythroid cells.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1195-1195
Author(s):  
Heather M. Rogers ◽  
Xiaobing Yu ◽  
Constance Tom Noguchi

Abstract The basic-helix-loop-helix transcription factor SCL/TAL1, is required for erythropoiesis during development, and conditional deletion in adult hematopoiesis results in hematopoietic stem cells with a competitive repopulation disadvantage and defective erythropoiesis in vitro. However, adult mice with a conditional SCL/TAL1 deletion survive with mild anemia, suggesting defective erythroid proliferation and indicating that SCL/TAL1 is important, but not essential in mature red blood cell production. We find that during erythroid differentiation of primary human hematopoietic CD34+ cells, SCL/TAL1 expression peaks at day 8–10 following erythropoietin (EPO) stimulation, concomitant with peak expression of GATA-1 and EKLF. Treatment with SCL/TAL1 antisense oligonucleotides during erythroid differentiation markedly decreases erythroid differentiation as indicated by decreased expression of GATA-1 and both b- and g-globin expression, along with the absence of the characteristic decrease in GATA-2. Microarray analysis of erythroid cells overexpressing SCL/TAL1 indicate increased gene expression for b- and g-globin, and other genes related to erythropoiesis including EPO receptor (EPO-R), and these results are confirmed in stable cell lines with increasing SCL/TAL1 expression. Examination of EPO-R transcription regulation indicates that E-boxes in the 5′ UTR can bind SCL/TAL1 in vitro and, in addition to the GATA-1 binding motif, provide transcription activity in reporter gene assays. These data indicate that in addition to the importance of SCL/TAL1 DNA binding for proliferation of BFU-E and expression of glycophorin A and protein 4.2, SCL/TAL1 is also necessary for high level expression of EPO-R. Reduction in EPO-R expression likely contributes to the anemia associated with the conditional adult deletion of SCL/TAL1 and to the proliferative defect of erythroid cells observed in vitro. Early expression of SCL/TAL1 in hematopoietic cells may activate expression of EPO-R prior to EPO stimulation of erythropoiesis and induction of GATA-1.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 356-356
Author(s):  
John Strouboulis ◽  
Patrick Rodriguez ◽  
Edgar Bonte ◽  
Jeroen Krijgsveld ◽  
Katarzyna Kolodziej ◽  
...  

Abstract GATA-1 is a key transcription factor essential for the differentiation of the erythroid, megakaryocytic and eosinophilic lineages. GATA-1 functions in erythropoiesis involve lineage-specific gene activation and repression of early hematopoietic transcription programs. GATA-1 is known to interact with other transcription factors, such as FOG-1, TAL-1 and Sp1 and also with CBP/p300 and the SWI/SNF chromatin remodeling complex in vitro. Despite this information the molecular basis of its essential functions in erythropoiesis remains unclear. We show here that GATA-1 is mostly present in a high (> 670kDa) molecular weight complex that appears to be dynamic during erythroid differentiation. In order to characterize the GATA-1 complex(es) from erythroid cells, we employed an in vivo biotinylation tagging approach in mouse erythroleukemic (MEL) cells1. Briefly, this involved the fusion of a small (23aa) peptide tag to GATA-1 and its specific, efficient biotinylation by the bacterial BirA biotin ligase which is co-expressed with tagged GATA-1 in MEL cells. Nuclear extracts expressing biotinylated tagged GATA-1 were bound directly to streptavidin beads and co-purifying proteins were identified by mass spectrometry. In addition to the known GATA-1-interacting transcription factors FOG-1, TAL-1 and Ldb-1, we describe novel interactions with the essential hematopoietic transcription factor Gfi-1b and the chromatin remodeling complexes MeCP1 and ACF/WCRF. Significantly, GATA-1 interaction with the repressive MeCP1 complex requires FOG-1. We also show in erythroid cells that GATA-1, FOG-1 and MeCP1 are stably bound to repressed genes representing early hematopoietic (e.g. GATA-2) or alternative lineage-specific (e.g. eosinophilic) transcription programs, whereas the GATA-1/Gfi1b complex is bound to repressed genes involved in cell proliferation. In contrast, GATA-1 and TAL-1 are bound to the active erythroid-specific EKLF gene. Our findings on GATA-1 complexes provide novel insight as to the critical roles that GATA-1 plays in many aspects of erythropoiesis by revealing the GATA-1 partners in the execution of specific functions.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 5216-5216
Author(s):  
Mario Falchi ◽  
Lilian Varricchio ◽  
Fabrizio Martelli ◽  
Francesca Masiello ◽  
Orietta Piccioni ◽  
...  

Abstract Calreticulin (CALR) mutations disrupting domains of the protein that regulate its intracellular trafficking were discovered in patients with myeloproliferative neoplasms (MPN) without JAK2 mutations and are associated with JAK2 activation (Klampt et al, NEJM 2013;369:2379; Nangalia et al, NEJM 2013;369:2391). Whether JAK2 mutations are associated with altered CALR functions is unknown. We previously reported that erythroid cells (Erys) generated in-vitro from JAK2+-polycythemia vera (PV) patients do not respond to dexamethasone (Dex) because in these cells the glucocorticoid receptor (GRα) is constitutively retained in the nucleus (Varricchio et al, Blood 2011;118:425). This was explained by the high frequency found in PV patients of the A3669G GR polymorphism that increases expression of GRβ, the isoform responsible for nuclear retention of GRα. The A3669G frequency is also increased in primary myelofibrosis (PMF) and, when associated with JAK2 mutations, predicts poor survival (Poletto et al, Blood 2012;120:3112). Although the relationship between GR and CALR in Erys has not been defined as yet, studies in animal models indicate that in non-erythroid cells CALR regulates GRα nuclear export. This suggested that CALR may favor nuclear export of GRα, antagonizing GRβ, also, in Erys, and that this function may be impaired both by JAK2 and CALR mutations found in MPN. To test this hypothesis, we compared content, localization and association of CALR and GR in Erys expanded in-vitro from normal sources (NS) or from JAK2 + and CALR + MPNs. By Western Blot (WB), NS and MPN Erys contained comparable levels of CALR and GR. By FACS, in NS Erys cell-surface CALR expression increased from the proerythroblast (proEry) to the polychromatic (polyEry) stage [mean fluorescent intensity (MFI)=215±51 vs 460±125, p<0.05]. Conversely, confocal microscopy of permeabilized cells (to remove cell-surface proteins) detected greater levels of CALR in the cytoplasm of proErys (Fig 1) than in that of polyErys (not shown). CALR was never detected in the nuclei of NS Erys. Thus, maturation of normal Erys is associated with cytoplasm to cell surface trafficking of CALR. By contrast, JAK2+ -PV proErys expressed greater cell-surface levels of CALR than NS proErys (MFI=304±69, p<0.05) and barely detectable levels of CALR in the cytoplasm (Fig 1). Cytoplasmic levels of CALR were also barely detectable in JAK2 + and CALR + PMF proErys (not shown). These results indicate that CALR trafficking is altered in JAK2 + and CALR + proErys alike. By WB and confocal microscopy in NS proErys GRα was prevalently localized in the nuclei or cytoplasm, depending on whether cells had been exposed to Dex (which provides nuclear export signals) or erythropoietin/stem cell factor (that by activating Ca2+ signaling presumably stimulates CALR). In the cytoplasm, GR was co-localized with CALR in proximity of the nuclear membrane. Multi-regression analyses of 410 Pro-Erys compared single CALR and total and cytoplasmic GR signals as independent parameters against the merged signals (co-localization) as dependent parameter. This showed that co-localization correlated significantly with cytoplasmic levels of both CALR and GR but that the greatest p values were observed for CALR, suggesting that CALR is the driving force in determining the interaction between the two proteins. By contrast, in MPN proErys GRα was localized prevalently in nuclei and CALR remained barely detectable in the cytoplasm and their localization was unaffected by treatments with Dex or erythropoietin/stem cell factor. Treatments of JAK2+ Erys with ruxolitinib increased total CALR levels (by 30%, p<0.05) and reduced its cell-surface expression (MFI=304±69 vs 220±48, p<0.05), increasing that in the cytoplasm of proErys (Fig 1). Although this treatment did not alter expression of GRβ, which remained high, it restored the presence of GRα in the cytoplasm where it was associated with CALR in the perinuclear region (Fig. 1) making JAK2+ Erys responsive to Dex (MTT incorporation 0.334±0.081 vs 0.224±0.045 with and without Dex, p<0.05), suggesting that impairment of the nuclear export signal provided by CALR cooperates with GRβ in retaining GRα in the nucleus of proErys from MPNs. These results provide the first indication that cellular distribution, and possibly GRα nuclear export functions, of CALR are altered in erythroid cells from MPN patients irrespective of their mutation status. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document