Structure and expression of ferritin genes in a human promyelocytic cell line that differentiates in vitro

1986 ◽  
Vol 6 (2) ◽  
pp. 566-573
Author(s):  
C C Chou ◽  
R A Gatti ◽  
M L Fuller ◽  
P Concannon ◽  
A Wong ◽  
...  

HL-60 is a human promyelocytic cell line with the capability of differentiating in vitro to give neutrophils, macrophages, or eosinophils. We screened libraries of HL-60 cDNA clones representing different time points during these differentiation processes to isolate clones corresponding to mRNAs whose expression is regulated during terminal differentiation. Upon sequencing this group of regulated clones, one clone encoding the heavy subunit and two clones encoding the light subunit of human ferritin were identified by reference to published amino acid sequences. Southern blot analyses showed that these clones are encoded by distinct multigene families. These clones identify two mRNAs whose ratios vary in a complex manner during both neutrophil and macrophage differentiation.

1986 ◽  
Vol 6 (2) ◽  
pp. 566-573 ◽  
Author(s):  
C C Chou ◽  
R A Gatti ◽  
M L Fuller ◽  
P Concannon ◽  
A Wong ◽  
...  

HL-60 is a human promyelocytic cell line with the capability of differentiating in vitro to give neutrophils, macrophages, or eosinophils. We screened libraries of HL-60 cDNA clones representing different time points during these differentiation processes to isolate clones corresponding to mRNAs whose expression is regulated during terminal differentiation. Upon sequencing this group of regulated clones, one clone encoding the heavy subunit and two clones encoding the light subunit of human ferritin were identified by reference to published amino acid sequences. Southern blot analyses showed that these clones are encoded by distinct multigene families. These clones identify two mRNAs whose ratios vary in a complex manner during both neutrophil and macrophage differentiation.


1997 ◽  
Vol 139 (1) ◽  
pp. 193-204 ◽  
Author(s):  
Peter Mundel ◽  
Hans W. Heid ◽  
Thomas M. Mundel ◽  
Meike Krüger ◽  
Jochen Reiser ◽  
...  

Synaptopodin is an actin-associated protein of differentiated podocytes that also occurs as part of the actin cytoskeleton of postsynaptic densities (PSD) and associated dendritic spines in a subpopulation of exclusively telencephalic synapses. Amino acid sequences determined in purified rat kidney and forebrain synaptopodin and derived from human and mouse brain cDNA clones show no significant homology to any known protein. In particular, synaptopodin does not contain functional domains found in receptor-clustering PSD proteins. The open reading frame of synaptopodin encodes a polypeptide with a calculated Mr of 73.7 kD (human)/74.0 kD (mouse) and an isoelectric point of 9.38 (human)/9.27 (mouse). Synaptopodin contains a high amount of proline (∼20%) equally distributed along the protein, thus virtually excluding the formation of any globular domain. Sequence comparison between human and mouse synaptopodin revealed 84% identity at the protein level. In both brain and kidney, in vivo and in vitro, synaptopodin gene expression is differentiation dependent. During postnatal maturation of rat brain, synaptopodin is first detected by Western blot analysis at day 15 and reaches maximum expression in the adult animal. The exclusive synaptopodin synthesis in the telencephalon has been confirmed by in situ hybridization, where synaptopodin mRNA is only found in perikarya of the olfactory bulb, cerebral cortex, striatum, and hippocampus, i.e., the expression is restricted to areas of high synaptic plasticity. From these results and experiments with cultured cells we conclude that synaptopodin represents a novel kind of proline-rich, actin-associated protein that may play a role in modulating actin-based shape and motility of dendritic spines and podocyte foot processes.


2004 ◽  
Vol 85 (7) ◽  
pp. 2111-2121 ◽  
Author(s):  
Daohong Jiang ◽  
Said A. Ghabrial

Molecular cloning and complete nucleotide sequencing of Penicillium chrysogenum virus (PcV) dsRNAs indicated that PcV virions contained four dsRNA segments with sizes of 3562, 3200, 2976 and 2902 bp. Each dsRNA segment had unique sequences and contained a single large open reading frame (ORF). In vitro translation of transcripts derived from full-length cDNA clones of PcV dsRNAs yielded single products of sizes similar to those predicted from the deduced amino acid sequences of the individual ORFs. Sequence similarity searches revealed that dsRNA1 encodes a putative RNA-dependent RNA polymerase. In this study, it was determined that dsRNA2 encodes the major capsid protein and that p4, encoded by dsRNA4, is virion-associated as a minor component. All four dsRNAs of PcV, like the genomic segments of viruses with multipartite genomes, were found to have extended regions of highly conserved terminal sequences at both ends. In addition to the strictly conserved 5′-terminal 10 nt, a second region consisting of reiteration of the sequence CAA was found immediately upstream of the AUG initiator codon. These (CAA) n repeats are reminiscent of the translational enhancer elements of tobamoviruses. The 3′-terminal 14 nt were also strictly conserved. As PcV and related viruses with four dsRNA segments (genus Chrysovirus) have not been previously characterized at the molecular level, they were provisionally classified in the family Partitiviridae, comprising viruses with bipartite genomes. This study represents the first report on molecular characterization of a chrysovirus and the results suggest the creation of a new family of mycoviruses with multipartite dsRNA genomes to accommodate PcV and related viruses.


Blood ◽  
1994 ◽  
Vol 83 (11) ◽  
pp. 3188-3198 ◽  
Author(s):  
EZ Tchilian ◽  
PC Beverley ◽  
BD Young ◽  
SM Watt

Abstract CD33 monoclonal antibodies recognize a 67-kD glycoprotein of unknown function that is expressed by early myeloid progenitors and their leukemic counterparts. We report here the cloning of the murine homolog of the human CD33 antigen. Two cDNA clones, differing by an 83- nucleotide insertion in the cytoplasmic region, were isolated. The insertion generated a shift in the reading frame within the cytoplasmic tail, resulting in two mouse CD33 isoforms, m33-A and m33-B, with distinct cytoplasmic domains and with predicted protein core molecular weights of 37 kD and 45 kD, respectively. The cDNAs and deduced amino acid sequences show extensive similarity with the human CD33 sequence with the highest homology occurring in the first and second lg-like domains (61% amino acid identity). The most significant divergence between the human and murine proteins occurs in their cytoplasmic portions. The murine CD33 mRNAs were detected in bone marrow, spleen, thymus, brain, liver, the multipotential progenitor cell line, A4, the myelomonocytic cell line, WEHI3B, the myeloid cell line, M1, and the macrophage cell line, P388, by Northern blot analysis. The expression pattern of the murine CD33 homolog suggests that the function of CD33 antigen in hematopoiesis may be conserved between humans and mice.


1987 ◽  
Vol 7 (11) ◽  
pp. 4065-4074
Author(s):  
B E Rich ◽  
J A Steitz

cDNA clones encoding three antigenically related human ribosomal phosphoproteins (P-proteins) P0, P1, and P2 were isolated and sequenced. P1 and P2 are analogous to Escherichia coli ribosomal protein L7/L12, and P0 is likely to be an analog of L10. The three proteins have a nearly identical carboxy-terminal 17-amino-acid sequence (KEESEESD(D/E)DMGFGLFD-COOH) that is the basis of their immunological cross-reactivity. The identities of the P1 and P2 cDNAs were confirmed by the strong similarities of their encoded amino acid sequences to published primary structures of the homologous rat, brine shrimp, and Saccharomyces cerevisiae proteins. The P0 cDNA was initially identified by translation of hybrid-selected mRNA and immunoprecipitation of the products. To demonstrate that the coding sequences are full length, the P0, P1, and P2 cDNAs were transcribed in vitro by bacteriophage T7 RNA polymerase and the resulting mRNAs were translated in vitro. The synthetic P0, P1, and P2 proteins were serologically and electrophoretically identical to P-proteins extracted from HeLa cells. These synthetic P-proteins were incorporated into 60S but not 40S ribosomes and also assembled into a complex similar to that described for E. coli L7/L12 and L10.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3843-3843
Author(s):  
Phil Paul ◽  
Margaret Forster ◽  
Graham Chapman ◽  
Mathew Lesniewski ◽  
Mary J. Laughlin

Abstract Abstract 3843 Background. The defining characteristic of stem cells is their ability for asymmetric division to provide progenitors for specific tissue generation, while maintaining renewal of the stem cell population. LIF protein has been shown to be critical for maintaining embryonic stem cells in an undifferentiated state. UCB HSC in vitro expansion using cytokines has been pursued to augment hematopoietic recovery following UCB transplantation. Our previous studies have shown that a feeder layer of huMSC inhibits UCB HSC proliferation and differentiation during short-term cytokine-driven expansion. We sought to determine whether LIF is secreted by huMSC and if so, at what concentrations, and to elicit its role, if any, in inhibiting cytokine-driven UCB HSC proliferation. Methods. Third passage huMSC were cultured at density 2×10e6/ml in DMEM supplemented with 10% FBS. Supernatant was collected at 24, 36, 48, and 72h and analyzed for LIF secretion by ELISA. UCB was obtained, and MNC were separated on a Histopaque-1077 density gradient. UCB CD133+ cells were isolated using AutoMACS magnetic cell sorter (Miltenyi Biotec) and surface stained for LIF receptor (LIF-R) using anti hLIF-Rα antibody (R&D Systems). LIF-R expression by isolated UCB CD133+ HSC was confirmed by Western blot (n=3). Isolated UCB CD133+ HSCs were plated in 24 well plates at density 3.3×10e3/ml and cultured in StemPro™ media supplemented with 10% FBS, L-glutamine, penicillin, streptomycin and amphotericin B. UCB CD133+ HSCs were culture-expanded for 96h with or without recombinant human LIF (10ng/ml) in a combination of cytokines including: IL-3, IL-6, Flt-3L, SCF, G-CSF, and EPO. At 0, 48, and 96h cell counts were obtained. Given variability of LIF & LIF-R expression by primary human CD133 HSC, we compared the expression of key factors in the LIF signaling pathway between the TF-1 cell line (CD34+/CD38+) and the tumorigenic sub-clone, TF-1a cell line which displays a more primitive HSC phenotype (CD34+/CD38−), using quantitative PCR (qPCR). TF-1 and TF-1a cells (ATCC) were propagated as defined by ATCC. TF-1 cultures were supplemented with human GM-CSF (required for growth of these factor-dependent cells). Cell cultures were maintained at density range 0.3–5×10e5/ml for TF-1 cells and 0.3–3×10e6/ml for TF-1a per ATCC guidelines. Culture aliquots containing equivalent numbers of suspended cells from TF-1 and TF-1a cultures were collected by centrifugation and total RNA prepared (Trizol). RNA was quantified by spectrophotometry and equivalent amounts were used to prepare cDNA by reverse transcription. qPCR was performed in 96-well plates using commercially available Taq-Man primers, with GAPDH used as endogenous control. The assays were run on an ABI 7500 Fast system and SDS software. Comparisons to the control transcript were performed using the 2-(ddCt) method, after verifying baseline, threshold, and completion of reactions as indicated by the plateau phase of the amplification curve. Results. huMSC secreted LIF at all 4 time points, with peak secretion at 48h (mean 52.1(±3.3) pg/ml) (n=3). Surface expression of LIF-R on gated CD133+ cells was 2.61%. At 48h in vitro expansion, higher UCB CD133+ cell counts in cultures without LIF were noted [6.3×10e4 (±0.9)/ml], versus cultures with LIF [4.4×10e4 (±0.8)/ml] suggesting LIF inhibits UCB HSC proliferation. We observed little difference in LIF transcript secreted by TF-1 and TF-1a cells, but a significant down-regulation of LIF-R in TF-1a (RQ=-6.37) compared to TF-1 cells. Similarly, there was a reduction of SOCS3 transcript in TF-1a cells (RQ=-5.23) compared to TF-1 cells. However, expression of Myc, a primary downstream target of LIF-JAK/STAT signaling, did not differ between TF-1 and TF-1a. Conclusions. LIF secretion by huMSC peaks at 48h at a concentration 3 logs lower than that previously used to inhibit embryonic stem cell differentiation (10ng/ml). LIF exerted inhibitory effects on UCB HSC proliferation at early time points (48h) in cytokine-driven in vitro expansion studies. We observed a down-regulation of the LIF-R and the SOCS3 transcript as might be expected. These data are the first to demonstrate and characterize LIF secretion by huMSC. Further studies are ongoing to further clarify cellular pathways involved in the regulation of LIF signaling on UCB CD133+ HSC differentiation during short-term in vitro cytokine expansion. Disclosures: No relevant conflicts of interest to declare.


1987 ◽  
Vol 115 (2) ◽  
pp. 341-346 ◽  
Author(s):  
F. Stewart ◽  
J. A. Thomson ◽  
S. E. A. Leigh ◽  
J. M. Warwick

ABSTRACT Several cDNA clones corresponding to mRNA for the α-subunit of the horse (Equus caballus) pituitary and placental (chorionic) gonadotrophic hormones have been isolated and sequenced. Polyadenylated mRNA was purified from horse pituitary glands (the source of FSH and LH) and horse placental tissues (the source of chorionic gonadotrophin; CG). The mRNA preparations were characterized by in-vitro translation and Northern hybridization techniques using human and ovine gonadotrophin cDNA clones as probes. Complementary DNA libraries were created from the pituitary and placental mRNAs and a human CG α-subunit probe was used to isolate several horse α-subunit cDNA clones. The α-subunit nucleotide sequence from both sources of tissue was identical, thereby indicating that in the horse (as in man) the same gonadotrophin α-subunit gene is expressed in the pituitary and placenta. Our results are consistent with transcription of a single α-subunit gene for all the glycoprotein hormones in the horse, and we suggest that the reported differences between the horse CG and FSH α-subunit amino acid sequences determined by conventional peptide sequencing methods arose due to errors in the FSH α-subunit sequence. Comparison of the deduced amino acid sequence of the horse α-subunit with that of other α-subunit sequences indicated a number of significant differences which may be related to the unusual receptor-binding properties of the equine gonadotrophins. J. Endocr. (1987) 115, 341–346


1987 ◽  
Vol 7 (11) ◽  
pp. 4065-4074 ◽  
Author(s):  
B E Rich ◽  
J A Steitz

cDNA clones encoding three antigenically related human ribosomal phosphoproteins (P-proteins) P0, P1, and P2 were isolated and sequenced. P1 and P2 are analogous to Escherichia coli ribosomal protein L7/L12, and P0 is likely to be an analog of L10. The three proteins have a nearly identical carboxy-terminal 17-amino-acid sequence (KEESEESD(D/E)DMGFGLFD-COOH) that is the basis of their immunological cross-reactivity. The identities of the P1 and P2 cDNAs were confirmed by the strong similarities of their encoded amino acid sequences to published primary structures of the homologous rat, brine shrimp, and Saccharomyces cerevisiae proteins. The P0 cDNA was initially identified by translation of hybrid-selected mRNA and immunoprecipitation of the products. To demonstrate that the coding sequences are full length, the P0, P1, and P2 cDNAs were transcribed in vitro by bacteriophage T7 RNA polymerase and the resulting mRNAs were translated in vitro. The synthetic P0, P1, and P2 proteins were serologically and electrophoretically identical to P-proteins extracted from HeLa cells. These synthetic P-proteins were incorporated into 60S but not 40S ribosomes and also assembled into a complex similar to that described for E. coli L7/L12 and L10.


2004 ◽  
Vol 31 (4) ◽  
pp. 349 ◽  
Author(s):  
Francesca Bagnoli ◽  
Susanna Danti ◽  
Valentina Magherini ◽  
Radiana Cozza ◽  
Anna M. Innocenti ◽  
...  

Two cDNA clones encoding catalase (Cat1 and Cat2) from peach [Prunus persica (L.) Batsch] were identified, that show homologies to other plant catalases. The nucleotide sequences of the two coding regions showed 88% identity to each other. The amino acid sequences predicted from the two full-length clones showed the highest homology to a catalase from cotton and Nicotiana plumbaginifolia L. and included C-terminal tri-peptides typical of those used to target proteins to peroxisomes. Southern hybridisation analysis suggested the existence of two catalase genes in peach. The expression of Cat1 and Cat2 was determined in seeds, vegetative tissue, leaves during the seasonal cycle and in leaves in response to light / dark treatments. Cat1 had high levels of expression only in leaf tissue and was responsive to light and seasonal changes. Cat2 had high levels of expression in in vitro shoots and was also responsive to seasonal changes, but not to light. In situ hybridisations to leaf tissue indicated that the expression of Cat1 was localised mainly in palisade cells, while Cat2 mRNA was present in the vascular tissue. The results of the expression analysis and in situ hybridisation suggest a role for Cat1 in photorespiration and for Cat2 in stress responses.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 943-943
Author(s):  
Amy Sexauer ◽  
Xiaochuan Yang ◽  
Alexander E Perl ◽  
Christian Thiede ◽  
Michael J. Borowitz ◽  
...  

Abstract Abstract 943 FLT3/ITD mutations are present in approximately 23% of adult AML cases. FLT3/ITD therefore presents an attractive therapeutic target for tyrosine kinase inhibitors. To date, the clinical responses to FLT3 inhibitors have been primarily limited to clearance of peripheral blood blasts, while bone marrow blasts remain largely unaffected. AC220 (quizartinib), a novel FLT3 inhibitor with 10–50 times more potency in vivo than other FLT3 inhibitors, has had a high response rate in an exploratory subset of patients in an ongoing clinical trial for relapsed and refractory FLT3/ITD AML. A high proportion of FLT3/ITD AML patients treated with AC220 displayed the typical rapid clearance of peripheral blasts seen with other FLT3 inhibitors. Strikingly, however, (as reported at last year's ASH meeting), the bone marrow blasts underwent terminal differentiation in response to AC220 treatment over the course of 4–8 weeks, as established by the presence of the same FLT3/ITD mutation in mature circulating neutrophils isolated after treatment with AC220. This differentiation was frequently accompanied by a syndrome of fever, skin nodules resembling Sweet's syndrome, and pulmonary nodules. To study the mechanisms associated with this phenomenon, we developed an in vitro model of FLT3 inhibitor-induced differentiation. In order to more closely reproduce the bone marrow microenvironment in vitro, we prepared human stromal cell layers derived from healthy bone marrow donors and co-cultured them with Molm14 cells (FLT3/ITD cell line), as well as with primary AML blasts from FLT3/ITD AML patients in the presence and absence of AC220. In the absence of co-culture with stroma, both the Molm14 cell line and the primary samples underwent rapid apoptosis in the presence of increasing concentrations of AC220, similar to the response of peripheral blood blasts in AC220 -treated patients. However, when grown in co-culture with stroma, Molm14 cells underwent relatively rapid (24–48 hours) morphologic differentiation, as evidenced by the development of characteristic multi-lobed nuclei, and by reduction of nitroblue tetrazolium (NBT) after stimulation with endotoxin (respiratory burst activity). Similar data were observed in Molm14 cells treated with the FLT3 inhibitor sorafenib, indicating that this differentiation is likely a class effect of FLT3 inhibitors. We have also used our model system to study differentiation in primary AML patient blasts. Pre-treatment blasts were obtained from a patient enrolled on the AC220 trial (who exhibited terminal differentiation of marrow blasts in response to AC220), and differentiation was induced in vitro by co-culturing the blasts with stroma in the presence of AC220. After five days of drug exposure during stromal co-culture, the blasts remained viable (assessed by trypan blue exclusion), but morphologically began to resemble myelocytes, and demonstrated reduction of NBT in response to endotoxin. Flow cytometry analysis of these blasts after eight days of AC220 exposure demonstrated increased side scatter, expression of CD15, and loss of CD117 and CD34, all indicative of maturation. Blasts cultured in the absence of stroma displayed decreased P-FLT3, P-STAT5, and P-ERK in response to AC220. In contrast, blasts exposed to AC220 on stroma demonstrated continued activity of P-ERK despite loss of P-FLT3 and P-STAT5. CEBPalpha protein levels were significantly higher in blasts on stroma, but decreased rapidly over the first 48 hours of culture. These findings confirm that a FLT3/ITD mutation can be sufficient to induce a differentiation block, and, in the bone marrow microenvironment, FLT3 inhibition does not induce immediate apotosis, but rather differentiation. The continued activity of P-ERK (presumably by parallel signaling pathways activated by stromal cells) may be responsible for the survival of the blasts in the setting of FLT3 inhibition, with CEBPalpha inducing a differentiation program in this context. We conclude, therefore, that the response of a FLT3/ITD AML patient to potent FLT3 inhibition consists of rapid apoptosis of blasts in the peripheral blood, and, in most cases a slower process of terminal differentiation in bone marrow blasts. Furthermore, our in vitro model will allow us to explore the precise mechanisms whereby differentiation is blocked in FLT3/ITD AML, and how this can be overcome with FLT3 inhibition and other targeted therapies. Disclosures: Borowitz: BD Biosciences: Research Funding. Levis:Ambit Biosciences, Inc: Consultancy.


Sign in / Sign up

Export Citation Format

Share Document