AU2008361210A1 - An endogenous short hairpin RNA and the use of the same - Google Patents

An endogenous short hairpin RNA and the use of the same Download PDF

Info

Publication number
AU2008361210A1
AU2008361210A1 AU2008361210A AU2008361210A AU2008361210A1 AU 2008361210 A1 AU2008361210 A1 AU 2008361210A1 AU 2008361210 A AU2008361210 A AU 2008361210A AU 2008361210 A AU2008361210 A AU 2008361210A AU 2008361210 A1 AU2008361210 A1 AU 2008361210A1
Authority
AU
Australia
Prior art keywords
seq
sequence
cells
shr
expression construct
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
AU2008361210A
Inventor
Chunjing Bian
James Q. Yin
Robert C. Zhao
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Institute of Basic Medical Sciences of CAMS
Original Assignee
Institute of Basic Medical Sciences of CAMS
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Institute of Basic Medical Sciences of CAMS filed Critical Institute of Basic Medical Sciences of CAMS
Publication of AU2008361210A1 publication Critical patent/AU2008361210A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.

Description

WO 2010/022553 PCT/CN2008/072133 An endogenous short hairpin RNA and the use of the same Field of the Invention The present invention relates to a RNA molecule, particularly to an endogenous short hairpin RNA as well as the application of the invented RNA to induce the formation of hematopoietic cells by repressing ElIA-like inhibitor of differentiation-I (referred to hereinafter as EID 1). This application was supported by the program of Ministry of Science and Technology of P.R. China; National Natural Science Foundation of China; Beijing Ministry of Science and Technology as well as Cheung Kong Scholars programme. Background of the Invention Both bone marrow mesenchymal stem cells (referred to herein as BMSCs) and hematopoietic stem cells (referred to herein as HSCs) are characterized by their capacity to self-renew to supply differentiated cells during the lifetime of the organism. Said BMSCs expanded in culture can be distinguished from HSCs by the lack of expression of CD45 and CD34 (G. Chamberlain, J. Fox, B. Ashton, J. Middleton, Stem Cells. 25, 2739-49 (2007).). Since it is a great challenge to obtain a large number of HSCs for clinical application (B.P. Sorrentino, Nat Rev Immunol. 4(11):878-88 (2004).), BMSCs would serve as an alternate source for HSCs if BMSCs can be induced to differentiate into hematopoietic cells. It is also known that mesenchymal stem cells (BMSCs) are multipotent stem cells that can differentiate into cells of bone, endothelium, adipose tissue, cartilage, muscle, and brain. However, whether or not BMSCs can become hematopoietic stem cells (HSCs) in vitro and in vivo remains controversial. (Y. Jiang et al., Nature 418, 41-49 (2002); M. Serafini et al., J Exp Med. 204,129-39 (2007); M.F. Pittenger, Science. 284, 143-7 (1999); K.W. Liechty et al., Nat Med. 6, 1282-6 (2000), and M. Angelopoulou et al., Exp. Hematol 31, 413-420 (2003)). In the present invention, it has been demonstrated that when BMSCs are cultured in the medium containing a cocktail of cytokines and growth factors, BMSCs expressing Flk-1 (fetal liver kinase-1) but negative for CD34 and CD31 appear and also can be readily expanded. It seems that expression of Flk-1 renders BMSCs greater differentiation potential (L.Liao, L Li, R.C. Zhao, Philos Trans R Soc Lond B Biol Sci. 362:1107-12 (2007)). 1 WO 2010/022553 PCT/CN2008/072133 These Flk-1l cells could even be induced to differentiate into hematopoietic cells although at a very low frequency (H. Guo et al., Exp. Hematol.31: 650-658 (2003)). In addition, it is also very important for the person skilled in the art to understand what molecular mechanisms are controlling the self-renewal and differentiation of BMSCs in order to make BMSCs an alternate source for HSCs. It has been shown by the prior art that miRNAs are differentially expressed in different hematopoietic cell types and play important regulatory roles. For example, miR-181 is associated with B lymphoid development (C.Z. Chen et al., Science 303, 83-86 (2004)), miRNA-142 and -223 with T lymphopoiesis (S.H. Ramkissoon et al., Leukemia Research, 30, 643-647 (2005).), miRNA-221 and -222 with erythropoiesis (N. Felli et al., Proc Natl Acad Sci U S A. 102, 18081-18086 (2005).), miRNA-223 with granulocytic differentiation (F. Fazi et al., Cell, 123, 819-831 (2005).) and miR-10, -126 and -17 with megakaryocytopoiesis (R. Garzon et al., Proc Natl Acad Sci U S A.103, 5078-5083 (2006). ). Moreover, some miRNAs (miR-128 and -181) have been shown to prevent the differentiation of HSCs (III, R.W. Georgantas et al., Proc Natl Acad Sci U S A.104, 2750-2755 (2007).). Although some miRNAs such as miR-130a and miR-10a have been found to target the transcription factor genes HOXA1 and MAFB, which are important for cellular differentiation (R. Garzon et al., Proc Natl Acad Sci U S A.103, 5078-5083 (2006)), it remains unclear whether any miRNAs or other small RNAs are involved in the determination of the self-renewal and differentiation of BMSCs and HSCs at early stages of development. In fact, little is known about intrinsic effectors of BMSC fate decisions. Recently, we have reported that a novel class of endogenous shRNAs is generated from the introns of protein-coding genes in human cells (Yin JQ and Zhao RC. Methods. 43(2):123-30 (2007). T. Gu et al. (Accompanying manuscript) 2008. See an accompanying manuscript). Summary of the Invention The first object of the invention is to provide an endogenous short hairpin RNA or a complement thereof having a sequence of SEQ. ID. NO.: 1 that induces the formation of hematopoietic cells particularly by repressing EID1. The said SEQ. ID. NO.: 1 shows: 5'-CAA AUA CUC ACC AUU GUG UUA CA-3'. 2 WO 2010/022553 PCT/CN2008/072133 The said sequence of SEQ. ID. NO.: 1 essentially includes the nucleic acid of 5'-CAA AUA CUC A-3'. The second object of the invention is to provide an expression construct including the sequence of SEQ. ID. NO.: 1. The third object of the invention is to provide a vector comprising an expression construct having an expression construct including the sequence of SEQ. ID. NO.: 1. All vectors known by the person skilled in the art can be used in the present invention. In an embodiment of the invention, the vector preferably is a retrovirus plasmid (retrovirus vector pMSCV ) that expresses the sequence of SEQ. ID. NO.: 1. The fourth object of the invention is to provide an application of the invented RNA or a complement thereof having a sequence of SEQ. ID. NO.: 1 to induce the formation of hematopoietic cells or a method of treatment for a subject suffering from dysfunction of formation of blood cells or a method of treatment for hematopoiesis in mammalian including human. In other word, the invention also provides a use of an endogenous short hairpin RNA having SEQ. ID. NO.: 1 or SEQ. ID. NO.: 2 in manufacture of medicament for treatment of a subject suffering from dysfunction of hematopoiesis in mammalian including human. The said SEQ. ID. NO.: 2 shows 5'-UGU AAC ACA AUG GUG AGU AUU UG-3'. The % identity of a polynucleotide is determined by GAP (Needleman and Wunsch, 1970) analysis (GCG program) with a gap creation penalty=5, and a gap extension penalty=0.3. Unless stated otherwise, the query sequence is at most 23 nucleotides in length. Preferably, the sequence is at least 10 nucleotides in length, and the GAP analysis aligns the two sequences over a region of at most 23 nucleotides. And the GAP analysis aligns the two sequences over a region of at least 10 nucleotides. With regard to the defined polynucleotides, it will be appreciated that % identity figures higher than those provided above will encompass preferred embodiments. 3 WO 2010/022553 PCT/CN2008/072133 Thus, where applicable, in light of the minimum % identity figures, it is preferred that a polynucleotide of the invention comprises a sequence which is at least 40%, more preferably at least 45%, more preferably at least 50%, more preferably at least 55%, more preferably at least 60%, more preferably at least 65%, more preferably at least 70%, more preferably at least 75%, more preferably at least 80%, more preferably at least 85%, more preferably at least 90%, more preferably at least 91%, more preferably at least 92%, more preferably at least 93%, more preferably at least 94%, more preferably at least 95%, more preferably at least 96%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99%, more preferably at least 99.1%, more preferably at least 99.2%, more preferably at least 99.3%, more preferably at least 99.4%, more preferably at least 99.5%, more preferably at least 99.6%, more preferably at least 99.7%, more preferably at least 99.8%, and even more preferably at least 99.9% identical to the relevant nominated SEQ. ID. NO.: 1. Polynucleotides of the present invention are natural ones, and may possess one or more mutations which are deletions, insertions, or substitutions of nucleotide residues. Mutants can be either naturally occurring (that is to say, isolated from a natural source) or synthetic (for example, by performing site- directed mutagenesis on the nucleic acid). Oligonucleotides and/or polynucleotides of the invention hybridize to a sill- gene of the present invention, or a region flanking said gene, under stringent conditions. The term "stringent hybridization conditions" and the like as used herein refers to parameters with which the art is familiar, including the variation of the hybridization temperature with length of an oligonucleotide. Nucleic acid hybridization parameters may be found in references which compile such methods, Sambrook, et al. (supra), and Ausubel, et al. (supra). For example, stringent hybridization conditions, as used herein, can refer to hybridization at 65'C in hybridization buffer (3.5xSSC, 0.02% Ficoll, 0.02% polyvinyl pyrrolidone, 0.02% Bovine Serum Albumin (BSA), 2.5 mM NaH2PO4 (pH7), 0.5% SDS, 2 niM EDTA), followed by one or more washes in 0.2.xSSC, 0.01% BSA at 500C. Alternatively, the nucleic acid and/or oligonucleotides (which may also be referred to as "primers" or "probes") hybridize to the region of the an insect genome of interest, such as the genome of a honeybee, under conditions used in nucleic acid amplification techniques such as PCR. 4 WO 2010/022553 PCT/CN2008/072133 BMSCs transduced with a newly identified short hairpin RNA (shRNA) of the present invention can differentiate into hematopoietic stem cells (HSCs) and their descendant multipotent progenitor cells that have the capacity of further differentiating into blood cells in vitro, according to the invention, on transplantation into sublethally irradiated NOD/SCID mice, transduced human BMSCs engrafted and differentiated into all hematopoietic lineages including lymphocytes and myelocytes. Furthermore this new shRNA alters BMSC fate by repressing the translation of EID 1. Thus a model has been established that the endogenous shRNA can confer definitive tropism to BMSCs and unveil an intrinsic roadmap for programming hematopoiesis-related genes in HSCs. Like artificial shRNAs, the two strands of their stems are perfectly complementary with a length of at least 21 nucleotides. To identify shRNAs important in regulating the self-renewal and directional differentiation of stem cells, we employed a custom microarray for a high-throughput screen (see. table 1). Table. 1 The different expression patterns of sRNA between BMSC and HSC Name Intensity Intensity Ratio (sRNA) (BMSC) (HSC) (HSC/BMSC) ZK-693 2943 9 0.0031 ZK-871 1853 9 0.0049 ZK-125 1846 12 0.0063 ZK-633 5281 62 0.0117 ZK-313 2360 39 0.0167 ZK-428 2582 48 0.0186 ZK-358 3800 178 0.0468 ZK-381 2505 160 0.0639 ZK-290 2147 138 0.0641 ZK-401 3000 197 0.0658 ZK-574 1598 106 0.0661 ZK-1011 3176 253 0.0797 ZK-266 2641 216 0.0818 ZK-921 2225 248 0.1116 ZK-826 11330 1388 0.1225 5 WO 2010/022553 PCT/CN2008/072133 ZK-960 11452 1643 0.1434 ZK-1040 9095 1454 0.1598 ZK-768 4641 939 0.2023 ZK-296 4722 1011 0.2141 ZK-630 3109 766 0.2463 ZK-588 4896 1264 0.2582 ZK-798 3122 854 0.2735 ZK-548 2367 694 0.2932 ZK-908 1055 5329 5.0533 ZK-249 309 1571 5.0852 ZK-907 298 4168 14. 0090 *ZK-249 is referred as shR-CH in this paper. In these experiments of the present invention, several hundreds of new short hairpin RNAs derived from human introns were identified already. The custom array analysis successfully revealed a differential expression of shRNAs between human BMSCs and HSCs. The majority of shRNAs were more highly expressed in BMSCs than in HSCs. According to the invention, two shRNAs, namely shR-CH having SEQ. ID. NO.:. 1 and a complementary sequence of shR-CH having SEQ. ID. NO.: 2, displayed stronger signals in HSCs than MSCs (Fig. 1A). The shR-CH precursor was expected to have effect as same as the shR-CH. To confirm the differential expression pattern of shR-CH, quantitative real-time PCR was performed. The expression of shR-CH was found to be at least 10 times higher in HSCs than in MSCs. To determine the length of mature shR-CH, this endogenous shRNA has been cloned. As shown in Figure 2B, mature shR-CH was about 21nt in length and its precursor formed a perfectly complementary hairpin structure. The said shR-CH resides within the first intron of SH3PXD2B gene that is located on chromosome 5 and is not phylogenetically conserved as most endogenous shRNAs (17 Accompanying manuscript). To identify potential target genes of shR-CH, similar bioinformatics based approaches as those used in prior art to predict potential targets of miRNAs was used. Among the predicted target genes of shR-CH (see the following Table2), Table.2 Prediction of shR-CH target genes No Gene Function 6 WO 2010/022553 PCT/CN2008/072133 1 NIPAI Related with neural system development 2 FLJ35424 No detailed reports 3 FLJ21625 No detailed reports 4 EIDI CREBBP/EP300 inhibitory protein I:Rb binding protein EIDI (ElA-like inhibitor of differentiation-1)(NM_014335.2) was selected for validation by qRT-PCR and Western blot assay because it contains five naturally-occurring putative shR-CH binding sites at its 3'UTR region (see Fig IC). More importantly, EIDI has been shown to have a close relationship with two essential hematopoiesis-related transcriptional coactivators (S. Miyake et al., Mol Cell Biol. 20, 8889-902 (2000); W. Xu et al., Blood. 107, 4407-16 (2006).). Several lines of studies have been reported that the transcriptional coactivators CBP (Cyclic adenosine monophosphate response element binding protein (CREB-binding protein) and its paralogue p300 interact with over 312 proteins, at least 65 of which are encoded by genes that are essential for hematopoiesis (D. N. Messina, J. Glasscock, W. Gish, M. Lovett. Genome Res. 14:2041-2047 (2004); L.H. Kasper, Nature. 419, 738-43 (2002)). CBP/p300 was thought to provide both an assembly platform as well as protein acetyltransferase functions with many transcription factors and histones that regulate gene expression (V. V. Ogryzko et al., Cell 87, 953-959 (1996); X. Liu et al., Nature. 451, 846-50 (2008)).The differential expression of shR-CH between human BMSCs and HSCs raised the possibility that it might be a key factor for HSCs to maintain their sternness and thus dictate the directional differentiation from BMSCs to HSCs. To investigate this possibility, vectors containing the murine stem-cell retrovirus backbone, a RNA polymerase III (pol III)-specific U6 gene promoter and shR-CH or mock shRNA in BMSCs (Fig. 2A) have been constructed. To examine the biological effect of ectopic expression of shR-CH on BMSC differentiation, mesenchymal stem cells from human bone marrow are infected with those vectors. As shown in Fig. 2B, the levels of shR-CH were significantly elevated by transduction of shR-CH vectors, compared with those in control or mock infected cells. Elevated levels of shR-CH can be sustained for a long time in BMSCs infected with shR-CH (data not shown). Next whether increased levels of shR-CH affected the expression of its target mRNA was determined. Levels of EIDI mRNA were analyzed via application of RT-PCR at day 2 following transduction of shR-CH vectors. As shown in Figure 2C and 2D, whereas there was only a slight change in the level of EID 1 mRNA, a remarkably decreased level of 7 WO 2010/022553 PCT/CN2008/072133 EIDI protein was detected when assayed by Western blotting. Consistent with the potential involvement of EID1 in shR-CH-dependent hematopoietic differentiation, endogenous EIDI protein was down-regulated in HSCs, coupled with a concomitant occurrence in expression of CD45 marker. In BMSCs and HSCs , it was noted that a reciprocal relationship between the expression of EID1 and the expression of the CD45 marker that is characteristic for hematopoietic cells. Moreover, CD45 was upregulated after shR-CH induced repression of EID1, raising the possibility that EID1 is involved in suppressing the hematopoietic program in BMSCs. According to the present invention a single BMSC was cloned by the single cell cloning method to reach this end and rule out the possibility of HSC contamination. The results from a fluorescence activated cell sorter illustrated that the cloned BMSCs were characterized with the presence of specific CD29, CD105 and CD44 surface antigens and Flk-1 nuclear antigen, and without the CD34, CD45 and CD133 specific for HSCs (Suppl. Fig. 1). Then these cloned cells were infected with vectors expressing shR-CH or mock shRNA and seeded onto the 24-well plate coated with fibronectin and collagen, and supplemented with a cocktail of hematopoietic cytokines and growth factors. Cells originating from transduced BMSCs were identified on the basis of the green fluorescent protein (GFP) marker carried by the vector, and differentiation of BMSCs to hematopoietic cells was characterized by expression of specific CD45 surface antigen (Fig. 3A). Ex vivo expansion of infected BMSCs was determined by using a modified CFU assay. 5000 seed cells from each group were selected and cultured for an additional 14 days. shR-CH overexpression rendered a substantial growth advantage during the 14-day culture as determined by in vitro colony-forming capacity (CFU-GM). This was also reflected by the predominance of GFP- and CD45- positive cells in the shR-CH culture (Fig. 3B). Although the same number of cells (~5,000) was initially seeded in three cases, shR-CH-transduced cells proliferated and differentiated significantly (Fig. 3C) whereas very few mock-infected BMSCs could change their phenotypes by day 14, suggesting that non-infected BMSCs and mock-infected BMSCs fail to thrive under these conditions. In contrast, many cells infected with shR-CH vectors could form CD45-positive colonies. This significant increase in hematopoietic cells indicated that increased levels of shR-CH induced the hematopoietic differentiation of BMSCs. The induction of hematopoietic differentiation was shR-CH specific since transduction with mock vectors had no effects on CD45 expression and the formation 8 WO 2010/022553 PCT/CN2008/072133 of CFU. These results strongly suggest that the enforced expression of shR-CH can overcome the blockade of hematopoietic differentiation and set out the program for the directional differentiation of hematopoiesis in BMSCs. If an increase in shR-CH level is required for definitive differentiation of BMSCs, the direct inhibition of its target gene, EID1, should have similar effects on BMSC differentiation. Therefore, we tested whether the EID1 gene was of functional importance in inducing the directional differentiation of BMSCs. Quantitative real time RT-PCR analysis illustrated that EID 1 -siR effectively reduced endogenous EID 1 expression to 40% of that observed with a mock siRNA in the infected BMSCs (Fig. 2C). As expected, BMSCs treated with EID1-siRNA were indeed induced to differentiate into hematopoietic cells, as determined by the specific expression of CD45 antigen (Fig. 3). This si-EID1 is highly specific for its target mRNA since mock siRNAs showed no effects on the differentiation of BMSCs. Therefore, like the ectopic expression of shR-CH, the knockdown of EID1 expression in BMSCs can result in the differentiation of these stem cells toward hematopoietic cells. Together, the findings described above provide the direct evidence that shR-CH dictates the differentiation of MSCs via the repression of EID1. To directly assess whether the ectopic expression of shR-CH induced the directional differentiation of human BMSCs in vivo, the capacity of infected BMSCs for multilineage hematopoietic repopulation in mice was examined. In initial experiments, human BMSCs were infected with either the retrovirus that expressed shR-CH or a control vector that expressed unrelated shRNA and were then transplanted into sublethally irradiated recipient mice. BMSC-derived GFP-expressing hematopoietic cells were detected through 60 days post infected-BMSC transplantation. By using antibodies against the corresponding antigens specific for human cells, immunochemical staining of mouse bone marrow indicated that these BMSC-derived cells could differentiate into multiple hematopoietic lineages as shown by expression of markers for hematopoietic progenitors and then further into myeloid cells marked by CD33 and lymphoid cells expressing the CD3 marker (Fig. 4A). In contrast, very few human CD45-, CD33- or CD3-positive cells were seen in the bone marrow sections of control and mock-transduced groups. The few human CD45-, CD33- or CD3-positive cells seen in the control and mock-transduced groups may reflect the intrinsic capacity of Flk- 1 9 WO 2010/022553 PCT/CN2008/072133 BMSCs to differentiate into HSC. Several organs were also collected and determined the presence of human BMSC-derived cells by GFP illumination. However, no positive results were obtained. These findings strongly suggest that shR-CH induce the directional differentiation of BMSCs into hematopoietic cells but not to other types of cells. To quantify the phenotype of the BMSC-derived cells in bone marrow, the cells were analyzed by flow cytometry by using three monoclonal antibodies against human leukocyte and stem cell markers. Consistent with the observation on the bone marrow sections, FACS profiles of bone marrow cells from recipients also showed robust engraftment of the BMSC-derived cells in medullary canal (Fig. 4B). After 8 weeks, the lineage composition of medullary canal cells descending from infected human BMSC (GFP* cells) was examined. The shR-CH expression in human BMSCs led to a significant increase in lymphoid (CD3) cells in medullary canal (2.8% vs 0.5% in the control). Similarly, there was also a substantial elevation in CD33-positive myeloid lineage cells (Fig. 4B). The presence of human BMSC-derived hematopoietic cells in bone marrow of the mouse recipients indicated that the ectopic expression of shR-CH in human BMSCs initiated a program for the hematopoietic lineage differentiation. The preferential expression of shR-CH in HSCs implicates an important role of shR-CH in hematopoiesis. The findings in the invention show that ectopic expression of shR-CH is able to initiate and direct the human BMSCs to differentiate into hematopoietic cell lines in vitro and vivo (Fig. 3 and 4) substantiate that notion. Through the repression of EID1, shR-CH probably up-regulates the activities of hematopoiesis-related genes such as CBP and p300 (Fig 4C). CBP and p300 knockout or point-mutant mice were reported to have dramatically reduced numbers of definitive erythroid, myeloid, and B-lymphocytic progenitors in bone marrow (L.H. Kasper, Nature. 419, 738-43 (2002); Y. Chen, P. Haviemik, K.D. Bunting, Y.C. Yang, Blood. 110, 2889-98 (2007); A.L. Kung et al., Genes Dev. 14, 272-7 (2000)). Other studies further showed that CBP and p300 were fate decision factors for HSCs, responsible for HSC self-renewal and hematopoietic differentiation, respectively (V.I. Rebel et al., Proc Natl Acad Sci U S A. 99, 14789-94 (2002); M.L. Sandberg et al., Dev Cell. 8, 153-66 (2005)). To test whether the expression of genes downstream of CBP/p300 is affected by the change of shR-CH and EIDI, the expression of RUNX1, 10 WO 2010/022553 PCT/CN2008/072133 an important hematopoiesis-related transcription factor, in BMSCs expressing shR-CH and in MSCs treated with si-EID1 was also measured. As shown in Fig.4D, RUNX1 was up-regulated significantly in both cases, indicating that the transcriptional activity of CBP/p300 was increased. The capacity of BMSCs to contribute to progeny of all three germ layers makes them a potential resource for regenerative medicine (Y. Jiang et al., Nature 418, 41-49 (2002).3, Y.S. Yoon et al., J. Clin. Invest 115, 326-338 (2005); M. T. Reyes et al., Blood 98, 2615-2625 (2001).). However, before the clinical trials, it is important to develop methods that allow reproducible differentiation of BMSCs. According to the study of the present invention, it showed for the first time that transplantation of shR-CH-infected BMSCs can give rise to high levels of BMSC-derived hematopoietic cells in bone marrow of the recipients. These findings also provide another explanation for the enhanced myelopoiesis and megakaryocytopoiesis caused by co-transplantation of human BMSCs and HSCs (K.W. Liechty et al., Nat Med. 6, 1282-6 (2000)). Moreover, it has been shown that BMSCs can prevent from lethal graft-versus-host disease, leading to a speedy recovery of hematopoiesis (X. Chen, H. Xu, C. M. Wan, McCaigue, G. Li, .Stem Cells. 24, 2052-9 (2006); G. Ren et al., Cell Stem Cell 2, 141-150 (2008)). In other words, shR-CH-infected BMSCs could serve as an alternate source of hematopoietic stem cells for transplantation. In summary, these studies characterized a new endogenous shRNA in structure and function, and identified a regulatory way in which endogenous shRNAs participate to control the directional differentiation of mesenchymal stem cells. Brief description of the drawings Figure 1 shows an identification and characterization of a novel shR-CH in human stem cells, wherein Figure 1A indicates the differential expression of newly identified shR-CH. Data are averages of at least three independent determinations. Error bars indicate standard deviations. *P<0.01 and **P<0.001, expression of shR-CH detected by microarrays or qRT-PCR assay in BMSCs compared with HSCs, respectively. Figure 1B indicates the cloning and sequencing of mature shR-CH and the secondary structure of the same. Solid red line stands for the sequence of mature shR-CH. Figure 1C indicates the alignment of the sequences of mature shR-CH and its three putative binding sites at the 3'UTR region of EID 1 mRNA. 11 WO 2010/022553 PCT/CN2008/072133 Figure 2 shows that the ectopic expression of shR-CH can effectively inhibit the expression of endogenous EIDI gene at the translational level. Wherein Fig 2A indicates the construction of the mock-sRNA /GFP and shR-CH/GFP retroviral vectors used in this study. Fig 2B indicates the differential expression of newly identified shR-CH in different cases. Data are averages of at least three independent determinations. Error bars indicate standard deviations. *p < 0.001. And Fig 2C indicates Real Time-PCR analysis on the expression levels of EID1 mRNA under various conditions were carried out and normalized to that of GAPDH, and the resultant expression levels in different cases are normalized to their levels in the control. Data are averages of at least three independent determinations. Error bars indicate standard deviations. Fig 2D indicates the levels of EIDI protein from different cases were analyzed by Western blotting. The Western blot was stripped and re-probed with actin antibody to check for equal loading of total protein. Figure 3 shows that the enforced expression of shR-CH can induce the proliferation and differentiation of human BMSCs in vitro. Cells from a single clone were used. Fig 3A indicates the morphological alterations of normal BMSC, mock-, shR-CH and siElID1-infected BMSCs illustrated by different staining methods. Fig 3B indicates CFC assay of CD45 hematopoietic cells. Cells were cultured in growth medium for 24 hours after transduction and then transferred into differentiation medium for 12 hours before immunostaining for CD45 specific for hematopoietic cells. Observation under inverted microscope showed that both the shR-CH-infected BMSCs and the siElID1-infected BMSCs generated the colonies effectively. Fig 3C indicates quantitative analysis of colonies from three different cases. At Day 14, the colony number from the BMSCs infected by shR-CH, siEIDI or mock vectors gave the result of n>6/case; *p < 0.001. Value inside bars represents fold increase. Figure 4 shows the effects of shR-CH forced expression of on hematopoietic lineage differentiation in vivo, wherein Fig 4A indicates the immunofluorescent and Hochest33342 staining of mouse bone marrow sections at 60 days post-trans. Enumeration of cells were stained with the human-specific antibodies against CD45, CD3 or CD33 in control, mock-shRNA and shR-CH sections of bone marrow obtained at 60 days post-transplantation. Fig 4B indicates hematopoietic reconstitution from human BMSCs grafted in NOD-SCID mice. 10' GFP'CD45' 12 WO 2010/022553 PCT/CN2008/072133 BMSCs were transplanted in sublethally irradiated NOD-SCID mice. Representative flow cytometry profiles of BM of NOD-SCID (human CD45) mice >8wk after transplant demonstrate multilineage (lymphoid and myeloid cells) reconstitution. For each quadrant, the fraction of cells relative to the total number of all the cells in mice bone marrow is given. Fig 4C and Fig 4D indicate that Real time-PCR analysis on the expression levels of RUNX1 mRNA under various conditions were carried out and normalized to that of GAPDH, and the resultant expression levels in different cases are normalized to their levels in the control. Data are averages of at least three independent determinations. Error bars indicate standard deviations. Detailed description of the invention Example 1 Cells and Cell Culture Human bone marrow (BM) was obtained from 30 healthy donors (ages 20 to 50 years) following informed consent according to guidelines from the Committee of Beijing Union Hospital on the Use of Human Subjects in Research. Isolation and culture of BM-derived FlklK CD31-CD34- cells from healthy donors were performed as described previously with some modifications. Briefly, based on a known method in the art, mononuclear cells were separated by a Ficoll-Paque gradient centrifugation (specific gravity 1.077 g/mL; Nycomed Pharma AS, Oslo, Norway) and cultured in DF12 medium containing 5% FCS, 20 ng/mL EGF, 100 U/mL penicillin and 100 g/mL streptomycin (Gibco Life Technologies) at 37'C and a 5%CO 2 humidified atmosphere. The floating cells were discarded at 18 hours. Culture media were changed every 2 days. At day 6, cells were harvested and further depleted of hematopoietic cells with magnetic-activated cell separation (MACS) CD45, GlyA, and CD34 micromagnetic beads (Miltenyi Biotec, Auburn, CA). Cells were then replated and passaged. At passages 4 cells were harvested by trypsinization, transfected with retrovirus vectors and used in the transplantation assay. Cells derived from single clone were used in other assays. To ensure single-cell originality of each cell colony, sorted cells were plated in wells coated with fibronectin (Sigma, St Louis, MO) and collagen (Sigma) for each patient. Culture medium was Dulbecco modified Eagle medium and Ham F12 medium (DF12) containing 40% MCDB-201 medium complete with trace elements (MCDB) (Sigma), 13 WO 2010/022553 PCT/CN2008/072133 2% fetal calf serum (FCS; Gibco Life Technologies, Paisley, United Kingdom), 1x insulin-transferrin-selenium (Gibco Life Technologies), 10- 9 M dexamethasone (Sigma), 10- 4 M ascorbic acid 2-phosphate (Sigma), 20 ng/mL interleukin-6 (Sigma), 10 ng/mL epidermal growth factor (Sigma), 10 ng/mL platelet-derived growth factor BB (Sigma), 50 ng/mL fetal liver tyrosine kinase 3 (Flt-3) ligand (Sigma), 30 ng/mL bone morphogenetic protein-4 (Sigma), and 100 U/mL penicillin and 100 g/mL streptomycin. Culture media were changed every 4 to 6 days. Wells with a single adherent cell were identified during the first 24 hours. The appearance of cell colonies was checked daily. Wells containing more than one colony were discarded. The cells deriving from a single cell colony usually reached 90% confluence after four weeks' culture. Then they were harvested by trypsinization and culture expanded. The phenotype of the cells was evaluated by FACS. CD133 hematopoietic stem cells were isolated from cord blood (presented by Beijing Cord Blood Bank) by using the CD 133 positive immunomagnetic beads (MACS). Example 2 Plasmid construction A retrovirus vector pMSCV encoding shRNAs expressed from the U6 promoter was generated by the method in the art (Michael T Hemann 2003)., the short hairpin DNA sequence (SEQ. ID. NO.: 3 and SEQ. ID. NO.: 4, Table 4) coincident with the pre-shR-CH was chemically synthesized as a construct. The retroviruses containing the shR-CH-expressing cassette were packaged by H293T cells. The retroviral titers of the mock-GFP and shR-CH-GFP producer cells were 3 x 105/mL and 4 x 105/mL respectively, as assessed by transfer of GFP expression to H293T cells.(Fig.2 A) Example 3 RNA Isolation and Purification Total RNA was extracted from different cells by using TRIzol (Invitrogen), as described in the art, and then small RNA was isolated from large RNA by using mirVana miRNA Isolation kit (Ambion, Austin, TX) according to the manufacturer's instructions. The concentration of small RNA was measured by the UV absorbance at 260 nm. Example 4 Cloning and sequencing of shRNA Short hairpin RNAs were polyadenylated at 37 0 C for 30 min in a 50-ul reaction volume including 1.5 ug RNA and 5 U poly(A) polymerase (Takara). Poly(A)-tailed small RNA was recovered by phenol/chloroform extraction and ethanol precipitation. 14 WO 2010/022553 PCT/CN2008/072133 Reverse transcription was performed by applying 1.5 ug RNA and 1 ug of RT primer (SEQ. ID. NO.: 5, Table 4) with 200 U of SuperScript III reverse-transcriptase (Invitrogen). The cDNA amplification was carried out for 40 cycles at a final annealing temperature of 60 0 C by using primers SEQ. ID. NO.: 6 (Table 4) targeting specifically the novel shRNA shR-CH and SEQ. ID. NO.: 7 (Table 4). A 5' adapter (SEQ. ID. NO.: 8, Table 4) was ligated to small RNA by using T4 RNA ligase (TAKARA) and the ligation products were recovered by phenol/chloroform extraction followed by ethanol precipitation. Reverse transcription was performed by means of 1.5 ug RNA and 1 ug specific RT primer, SEQ. ID. NO.: 9 (Table 4). The cDNA amplification was carried out for 40 cycles at a final annealing temperature of 60 0 C via primers, SEQ. ID. NO.: 10 and SEQ. ID. NO.: 11 (Table 4). The PCR products were separated on 2% agarose with Goldview staining. A gel fragment spanning 100 nt internal standards was excised and DNA was eluted into elution buffer (0.5M NH 4 Ac, 10 mM Mg(Ac) 2 , and 1 mM EDTA) at 37 0 C and recovered by phenol/chloroform extraction followed by ethanol precipitation. PCR products spanning the insert were directly submitted for sequencing (Shanghai Sangon Biological Engineering Technology & Services Co., Ltd).(Fig. IB) Example 5 Real-time PCR The expression levels of small RNA and related genes were determined by real-time RT-PCR. SYBR green assays with SYBR Premix Ex TaqTM (TAKARA) were run on the 7500HT real-time PCR instrument (Applied Biosystems). Reverse transcription of small RNA was performed by using 1.5 ug RNA and 1 ug shR-CH specific RT primer . (SEQ. ID. NO.: 9, Table 4). Primers, SEQ. ID. NO.: 6 and SEQ. ID. NO.: 11 (Table 4) were used for the following real-time PCR. Reverse transcription of hematopoiesis-related genes were performed by using 1.5ug RNA and lug universe primer oligo-dT(1 8). The primers used for the cDNA amplication were listed in Table 3. The results refer to Fig.lA, Fig.2B, Fig.2C, Fig.4D. Table 3 Primers used for the cDNA amplication SEQ.ID.NO. Gene Sequence 12 E6 (sense primer) 5'-CTCGCTTCGGCAGCACA-3' 15 WO 2010/022553 PCT/CN2008/072133 13 E6(antisense primer) 5'-AACGCTTCACGAATTTGCGT-3' 14 EID 1 (sense primer) 5'-CAGATGCTCTTGGAAACACA-3' 15 EID 1 (antisense primer) 5'-CTGCCAATACAAAAGCCCAC-3' 16 0-actin(sense primer) 5'-GAGACCTTCAACACCCCAGCC-3' 17 0-actin(antisense primer) 5'-AATGTCACGCACGATTTCCC-3' Table 4 Sequence of nucleotide acids referred in this paper SEQ.ID. NO. Sequence 1 5'-CAA AUA CUC ACC AUU GUG UUA CA-3' 2 5'-UGU AAC ACA AUG GUG AGU AUU UG-3' 3 5'-GAT CCC GTG TAA CAC AAT GGT GAG TAT TTGactagaatataCAA ATA CTC ACC ATT GTG TTA CAT TTT TTG GAA A-3 4 5'-AGC TTT TCC AAA AAA TGT AA CAC AAT GGT GAG TAT TTG tatattetactCAA ATA CTC ACC ATT GTG TTA CAC GG-3' 5 ATT CTA GAG GCC GAG GCG GCC GAC ATG-d (T) 30 6 5'-ACA CTC CAG CTG GGC AA A TAC TCA CC A TTG-3' 7 5'-ATT CTA GAG GCC GAG GCG GCC GAC ATG T-3' 8 5'-CGA CUG GAG CAC GAG GAC ACU GAC AUG GAC UGA AGG AGU AGA AA-3' 9 5'-CTC AAC TGG TGT CGT GGA GTC GGC AAT TCA GTT GAG TGT AACA C-3' 10 5'-GGA CAC TGA CAT GGA CTG AAG GAG TA-3' 11 5'-GGT GTC GTG GAG TCG-3' Example 6 Western blotting Cells were washed twice with cold PBS and then extracted in 20 ul of RIPA lysis buffer (50mM Tris-HCl pH7.5; 1% NP-40; 150mM NaCl; lmg/ml aprotinin; lmg/ml leupeptin; 1mM Na 3
VO
4 ; 1mM NaF; 1mM PMSF) at 4 C for 30 min. Total protein was resolved on 10% SDS-polyacrylamide gel eletrophoresis and bands of protein transferred to a polyvinylidene difluoride (PVDF) membrane (Amersham). The membrane was blocked with 5% nonfat milk TBS buffer overnight at RT, and incubated for 2 hours with primary antibodies. The expression of beta-actin was used as loading control. The antibodies used included beta-actin (Santa Cruz Biotechnology), and EIDI (UPSTATE). The membranes then were incubated for one hour with HRP-conjugated rabbit anti-goat secondary antibody. Immunocomplexes were visualized with a commercial ECL kit.(Fig.2D) 16 WO 2010/022553 PCT/CN2008/072133 Example 7 Bioinformatic Analysis The algorithm, miRanda 3.0 and targetscan 3.1( Miranda KC et al., Cell. 126, 1203-1217 (2006); Grimson A, Farh KK et al., Mol Cel. 27, 91-105 (2007)), were used in the invention for target prediction for shRNA. HSCs-overexpressed human-shR-CH was selected for target verification. Among hundreds of predicted targets, one potential targets for human shR-CH, EID1 was chosen for further experimental validation. The result was shown in Fig. 1C. Eample 8 Small RNA MicroArray and Granulocyte-macrophage colony-forming unit (CFU-GM) assay Small RNA MicroArray 1. The custom sRNA MicroArrays were provided by CapitoBio Corporation. The details were described in the above (Yin JQ and Zhao RC .Methods. 43(2):123-30 (2007)). Gr anulocyte-macr ophage colony-forming unit (CFU-GM) assay The BM-derived mesenchymal stem cells stably expressing shR-CH were analyzed by CFU-GM assay to assess the hematopoietic colony forming ability. Cells from a single clone were used in this assay. The cells suspending in the supernatant were collected and seeded in the CFU mix. The cells were then incubated at 37 C under 5% CO 2 in a humidified atmosphere. At day 14, the numbers of colony-forming units (CFU) (CFU, defined as >50 cells/colony) were scored based on the morphology and size of colonies.(Fig.3B, Fig.3C) Example 9 Immunostaining of cells and tissues Cells from a single clone were fixed with paraformaldehyde 4% for 30 min at room temperature and then washed three times with PBS. The cells were permeabilized with Triton 0.2% in PBS for 5 min. After four washes in PBS, the cells were blocked with a blocking buffer (Dako) for 30 min and then incubated in PBS/BSA 0.1% with an anti-albumin (Dako) or anti-CD45, CD34, CD19, CD71, CD3 and CD33 antibody (PE; Becton Dickinson), or with rabbit IgG (Jackson ImmunoResearch) as a negative control for one hour at room temperature. After staining, the cells were washed three times in PBS and incubated with an antirabbit whole IgG-Cy3 (Jackson ImmunoResearch) or anti-rabbit whole IgG-Cy2 (Jackson 17 WO 2010/022553 PCT/CN2008/072133 ImmunoResearch) in PBS/BSA 0.1% for one hour at room temperature. The cells were washed again (four times in PBS) and the nuclei were stained with Hochest 33342 diluted in PBS for 5 min at room temperature. The stained cells were visualized by using a fluorescence microscope (Leica DMIRB) and images captured using Magnafire software. The results refer to Fig.3A, Fig.4A. Example 10 Transplantation and FACS assay Six to eight week-old male nonobese diabetic ( NOD)/ LtSz-scid/scid (SCID ) mice were bred and maintained under defined flora conditions in individually ventilated (high-efficiency particle-arresting filtered air) sterile micro-isolator cages (Techniplast, Milan, Italy). All animal handle and experiment procedures were approved by the Animal Care and Use Committee of the Chinese Academy of Medical Sciences. Mice were sublethally irradiated (300 cGy) with a cesium source (MDS Nordion; Gammacell, Ottawa, QC, Canada) prior to transplantation. BMSCs (2.5x 10 5 cells/mice) transduced without or with shR-CH vectors or mock vectors in 0.4 ml of physiological saline (PS) were respectively injected via tail vain into the irradiated mice. The peripheral white blood cell count was done once a week. Mice were killed 2 months later by cervical dislocation. The BM from both femora and tibiae were collected. They were air dried and then stained with Jenner-Giemsa (BDH Ltd, Poole, United Kingdom). Conventional 4-um histologic sections of decalcified tibia were cut from formalin-fixed, paraffin-embedded material and stained. The BM of mice that received a transplant was assessed for presence of human cells by FACS. Mononuclear cells were harvested as described in the previous paragraph and red blood cells were lysed by adding 8.3% ammonium chloride. Single-cell suspensions were then prepared. After blocking of Fc receptors with human serum, cells were determined by labeling with anti-CD45, -CD33, or -CD3 phycoerythrin (PE; Becton Dickinson). Cells labeled with anti-immunoglobulin G (anti-IgG) monoclonal antibody (mAb) were used as control. In addition, cells were gated to include both lymphoid and myeloid fractions.(Fig.4B) Statistical analysis Data are described as means+SEM of the indicated number of separate experiments. A one-way analysis of variance was performed for multiple comparisons. If there was significant variation between treatment and control groups, the mean values were compared by using Student's two-tailed t-test. 18 WO 2010/022553 PCT/CN2008/072133 The invention has been described in detail with reference to preferred embodiments thereof However, it will be appreciated that those skilled in the art, upon consideration of this disclosure, may make modification and improvements within the spirit and scope of the invention as set forth in the following claims. 19

Claims (14)

1. An endogenous short hairpin RNA having a sequence of SEQ. ID. NO.: 1 that induces the formation of hematopoietic cells particularly by repressing EID1.
2. A complement of endogenous short hairpin RNA according to claim 1 having a sequence of SEQ. ID. NO.: 2.
3. The endogenous short hairpin RNA according to claim 1, wherein said sequence of SEQ. ID. NO.: 1 essentially includes the nucleic acid of 5'-CAA AUA CUC A-3'.
4. An expression construct including the sequence of SEQ. ID. NO.: 1, wherein the expression construct has sequence of SEQ. ID. NO.: 3.
5. An expression construct including the sequence of SEQ. ID. NO.: 1, wherein the expression construct has a complement having sequence of SEQ. ID. NO.: 4.
6. A vector comprising an expression construct having an expression construct including the sequence of SEQ. ID. NO.: 1.
7. The vector according claim 6, wherein the vector is a retrovirus plasmid.
8. A polynucleotide comprising a sequence which is at least 70% identical to the relevant nominated SEQ. ID. NO.: 1.
9. A polynucleotide comprising a sequence which is at least 80% identical to the relevant nominated SEQ. ID. NO.: 1.
10. A polynucleotide comprising a sequence which is at least 90% identical to the relevant nominated SEQ. ID. NO.: 1.
11. A use of the RNA or a complement thereof having a sequence of SEQ. ID. NO.: 1 according to claim 1 or 2 in manufacture of a medicament for treatment for a subject suffering from dysfunction of formation of blood cells.
12. The use of claim 11, wherein the dysfunction of formation of blood cells includes hematopoiesis.
13. The use of claim 11 or 12, wherein the subject is mammalian animal including human.
14. A method of treatment for a subject suffering from dysfunction of formation of blood cells, wherein effective amount of a RNA or a complement thereof having a sequence of SEQ. ID. NO.: 1 or claim 2 is administrated enough to induce the formation of hematopoietic cells. 20 WO 2010/022553 PCT/CN2008/072133 AMENDED CLAIMS received by the International Bureau on 28 July 2009 (28.07.2009) What is claimed is: 1. An endogenous short hairpin RNA having a sequence of SEQ. ID. NO.: 1 that induces the formation of hematopoietic cells particularly by repressing ElD 1. 2. A complement of endogenous short hairpin RNA according to claim I having a sequence of SEQ. ID. NO.: 2. 3. The endogenous short hairpin RNA according to claim 1, wherein said sequence of SEQ. ID. NO.: 1 essentially includes the nucleic acid of 5'-CAA AUA CUC A-3'. 4. An expression construct including the sequence of SEQ. ID. NO.: 1, wherein the expression construct has sequence of SEQ. ID. NO.: 3. 5. An expression construct including the sequence of SEQ. ID. NO.: 1, wherein the expression construct has a complement having sequence of SEQ. ID. NO.: 4. 6. A vector comprising an expression construct including the sequence of SEQ. ID. NO.: 3 or SEQ ID NO. 4. 7. The vector according claim 6, wherein the vector is a retrovirus plasmid. 8. A polynucleotide comprising a sequence which is at least 70% identical to the relevant nominated SEQ. ID. NO.: 3 or SEQ ID NO. 4. 9. A polynucleotide comprising a sequence which is at least 80% identical to the relevant nominated SEQ. ID. NO.: 3 or SEQ ID NO. 4. 10. A polynucleotide comprising a sequence which is at least 90% identical to the relevant nominated SEQ. ID. NO.: 3 or SEQ ID NO. 4. 11. A use of the RNA or a complement thereof having a sequence of SEQ. ID. NO.: 1 according to claim 1 or 2 in manufacture of a medicament for treatment for a subject suffering from dysfunction of formation of blood cells. 12. The use of claim 11, wherein the dysfunction of formation of blood cells includes hematopoiesis. 13. The use of claim 11 or 12, wherein the subject is mammalian animal including human. 14. A method of treatment for a subject suffering from dysfunction of formation of blood cells, wherein effective amount of a RNA or a complement thereof having a sequence of SEQ. ID. NO.: 1 or claim 2 is administrated enough to induce the formation of hematopoietic cells. 21 AMENDED SHEET (ARTICLE 19)
AU2008361210A 2008-08-25 2008-08-25 An endogenous short hairpin RNA and the use of the same Abandoned AU2008361210A1 (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/CN2008/072133 WO2010022553A1 (en) 2008-08-25 2008-08-25 An endogenous short hairpin rna and the use of the same

Publications (1)

Publication Number Publication Date
AU2008361210A1 true AU2008361210A1 (en) 2010-03-04

Family

ID=41720773

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2008361210A Abandoned AU2008361210A1 (en) 2008-08-25 2008-08-25 An endogenous short hairpin RNA and the use of the same

Country Status (4)

Country Link
EP (1) EP2329024A4 (en)
CN (1) CN102131929A (en)
AU (1) AU2008361210A1 (en)
WO (1) WO2010022553A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103088065B (en) * 2013-01-25 2014-12-10 北京银杏德济生物技术有限公司 Method capable of forming hematopoietic stem cells by quickly inducing reversal decision of mesenchymal stem cells in large scale with high purity
EP2805705B1 (en) * 2013-05-23 2016-11-09 IP Gesellschaft für Management mbH Packaging with one or more administration units comprising a sodium salt of (R)-3-[6-amino-pyridin-3-yl]-2-(1-cyclohexyl-1 H-imidazol-4-yl)-propionic acid
CN116904469B (en) * 2023-09-12 2024-01-23 首都儿科研究所 Inhibitor for p300 protein expression, preparation method and application thereof

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040265849A1 (en) * 2002-11-22 2004-12-30 Applera Corporation Genetic polymorphisms associated with Alzheimer's disease, methods of detection and uses thereof
MXPA06004428A (en) * 2003-10-22 2006-07-03 Aventis Pharma Inc Retroviral vectors for delivery of interfering rna.
CN101024830A (en) * 2007-02-01 2007-08-29 王鸿艳 Clone method of ShRNA
WO2009017670A2 (en) * 2007-07-26 2009-02-05 University Of Massachusetts Ras-mediated epigenetic silencing effectors and uses thereof

Also Published As

Publication number Publication date
WO2010022553A1 (en) 2010-03-04
EP2329024A1 (en) 2011-06-08
EP2329024A4 (en) 2012-02-22
CN102131929A (en) 2011-07-20

Similar Documents

Publication Publication Date Title
Pei et al. AMPK/FIS1-mediated mitophagy is required for self-renewal of human AML stem cells
Herrera-Merchan et al. miR-33-mediated downregulation of p53 controls hematopoietic stem cell self-renewal
Kong et al. MIR-23A microRNA cluster inhibits B-cell development
Lee et al. Cellular and genetic characterization of human adult bone marrow-derived neural stem-like cells: a potential antiglioma cellular vector
Wojtowicz et al. Ectopic miR-125a expression induces long-term repopulating stem cell capacity in mouse and human hematopoietic progenitors
Hu et al. Bmi1 suppresses adipogenesis in the hematopoietic stem cell niche
JP2023171952A (en) Methods and compositions for expansion of cell population
JP2016513974A (en) Compositions and methods for reprogramming hematopoietic stem cell lineage
Sommerkamp et al. Differential alternative polyadenylation landscapes mediate hematopoietic stem cell activation and regulate glutamine metabolism
Santos et al. Leukemia inhibitory factor (LIF) overexpression increases the angiogenic potential of bone marrow mesenchymal stem/stromal cells
US20110280861A1 (en) Method for mir-125a in promoting hematopoietic stem cell self renewal and expansion
Honda et al. Side population cells expressing ABCG2 in human adult dental pulp tissue
Zou et al. Duplexes of 21‐nucleotide RNAs mediate RNA interference in differentiated mouse ES cells
US9163234B2 (en) Culture method
CA2628865A1 (en) Methods and compositions for modulation of stem cell aging
Wang et al. E3-ligase Skp2 regulates β-catenin expression and maintains hematopoietic stem cell homing
US8828965B2 (en) MiR-150 for the treatment of blood disorders
AU2008361210A1 (en) An endogenous short hairpin RNA and the use of the same
Chao et al. CD71high population represents primitive erythroblasts derived from mouse embryonic stem cells
Bai et al. Evidence for the existence of CD34+ angiogenic stem cells in human first‐trimester decidua and their therapeutic for ischaemic heart disease
CN110551688B (en) Composition for inducing reprogramming of somatic cells into hematopoietic stem/progenitor cells and promoting in-vitro expansion of hematopoietic stem/progenitor cells and application thereof
US20080305085A1 (en) Compositions And Methods For Stem Cell Expansion
CN101368180A (en) Small RNA numerator for differentiation of mesenchyma stem cell into hematopoiesis cell and function target point thereof
Kim et al. Generation of CD34+ cells from human embryonic stem cells using a clinically applicable methodology and engraftment in the fetal sheep model
US20150299712A1 (en) Modulation of Hematopoietic Stem Cell Differentiation

Legal Events

Date Code Title Description
MK4 Application lapsed section 142(2)(d) - no continuation fee paid for the application