Abstract
Multipotent mesenchymal stromal cells (MSC) isolated from various adult tissue sources have the capacity to self-renew and to differentiate into multiple lineages. Both of these processes are tightly regulated by genetic and epigenetic mechanisms. Emerging evidence indicates that the class of single-stranded non-coding RNAs known as “microRNAs” also plays a critical role in this process. First described in nematodes and plants, microRNAs have been shown to modulate major regulatory mechanisms in eukaryotic cells involved in a broad array of cellular functions. Studies with various types of embryonic as well as adult stem cells indicate an intricate network of microRNAs regulating key transcription factors and other genes which in turn determine cell fate. In addition, expression of unique microRNAs in specific cell types serves as a useful diagnostic marker to define a particular cell type. MicroRNAs are also found to be regulated by extracellular signaling pathways that are important for differentiation into specific tissues, suggesting that they play a role in specifying tissue identity. In this review we describe the importance of microRNAs in stem cells focusing on our current understanding of microRNAs in MSC and their derivatives.
Keywords: miRNA, gene regulation, epigenetics, human mesenchymal stem cells, Review
Introduction
Global gene expression analysis of stem cells is one method to produce a clear understanding of the variations in biological conditions across different types of stem cells. A similar approach has begun to help us identify and understand the epigenetic signatures of stem cells. Several microarray-based techniques for the systematic analysis of epigenetic changes have become available allowing broad-scale analysis on a new level of transcriptional and translational regulation, particularly for studying the regulation of microRNAs and their predicted mRNA targets. Understanding the importance of microRNAs in MSC differentiation will require knowledge of the mechanisms explaining mRNA targeting, the mechanisms underlying microRNA regulation, and an accurate assessment of microRNA-mRNA targeting.
The discoveries of artificial silencer RNAs and genomic-encoded microRNAs have led to an explosion in the study of small non-coding RNAs1-7. Various types of small, non-coding RNAs exist as modulators of gene expression, affecting transcription rate, mRNA stability, and mRNA translation into functional proteins. What was once believed to be a specialized function occurring only in plants8, 9 or worms1, 10 has now been shown to control regulatory pathways in several mammalian biological process such as cell growth and apoptosis11-14, viral infection15-19, neural function20-23 and stem cell function21, 24-37. We will outline the issues related to microRNA expression profiling in mesenchymal stem cells as an example of how these studies are useful for classifying differentiating cells and, potentially, understanding molecular networks active during differentiation.
MSC
The plastic-adherent, fibroblast-like cells isolated from bone marrow with osteogenic potential was first described by Friedenstein38 and subsequently described by several studies38-44. Since then the ability of these cells to differentiate into multiple connective tissue cell types has been reported43-47 with the term mesenchymal stem cell first proposed by Caplan48. MSC has been isolated from multiple adult tissue sources such as cord blood, placenta, adipose and dermal tissues, synovial fluid, deciduous teeth, and amniotic fluid49-54. This broad distribution of sources combined with their ability to differentiate into multiple mesenchymal phenotypes such as bone, cartilage, tendon, and adipose tissue has lead them to be evaluated as potential therapeutic candidates for several disease and degenerative applications55-61. The varied source and methodology used for MSC isolation has also largely led to ambiguities resulting in the lack of an universally accepted criteria to define these cells. To address this issue, the International Society of Cellular Therapy has recently designated these cells as “Multipotent mesenchymal stromal cells” (MSC)62 based on the adherence to plastic, expression of specific surface antigen and their differentiation potential.
Despite this level of interest, a clear understanding of the factors involved in their growth regulation remains rudimentary. Several reports have described that only one third of the expanded MSC colonies truly retain tri-lineage differentiation potential, that is, the ability to differentiate into adipocytes, osteocytes and chondrocytes, with the remaining cells in MSC cultures thought to be either having only bi- or mono-lineage or to be committed progenitor cells46, 63. This could largely be due to clonal expansion which in general could alter the phenotype of most stem cell types. Global gene expression analysis has revealed that MSC differentiation into specific mature cell types is a temporally controlled and regulated process involving the activities of various transcription factors, growth facts and signaling pathways64, 65. However, microRNAs would be expected to regulate mRNA translation and/or stability, so perhaps the regulation of microRNA expression patterns represents a novel regulatory network in MSC.
What are microRNAs?
MicroRNAs are single-stranded RNAs of 19−23 nucleotides that derive from a ∼70 nucleotide precursor and are found in a wide variety of organisms, from plants to insects to humans. Estimates suggest there are about 120 microRNA genes in each invertebrate species, and at least 250 genes in mammals, with some groups predicting from 1,000 per genome66-68 to as many as 10,00069. After transcription, microRNA precursors are cleaved in the nucleus by Drosha70, exported to cytoplasm by exportin71, 72, and inserted into a RISC (RNA induced silencing complex) complex after additional cleavage by Dicer73. Evidence for the requirement of the processing of microRNA in stem cell function and differentiation comes from studies of the Drosha complex partners Loquacious (homolog of human TRBP), which is required for germ-line stem cell maintenance74, and DGCR875, which is required for embryonic stem cell self-renewal 37. Similarly, Dicer knockouts exhibit defects in stem cell differentiation25. Clearly, microRNAs underlie key differentiation mechanisms.
Relatively little is known of the regulation of microRNA expression, although a few microRNA transcriptional control sequences have been described14, 76-78. Interestingly, microRNAs are often clustered in the genome and two or more closely-related microRNA precursors have been detected in polycistronic precursors35, 79-81. Other microRNAs are encoded within introns of other genes82, 83 and some microRNAs are edited84, 85, producing a dizzying mix of co-expression, predicted transcriptional control and post-transcriptional modifications. There is much to learn about the regulatory mechanisms controlling microRNA expression.
microRNA regulation mechanisms & target prediction
MicroRNAs are known to negatively regulate gene expression. This could be achieved by direct mRNA cleavage86-90; mRNA decay by deadenylation91, 92 or via translational repression93. Complexes containing microRNAs and those of the RISC complex involved in RNAi are similar. RNA silencing, using a perfectly complementary siRNA, normally cleaves an mRNA target. In animals, microRNAs usually have imperfect complementarity to a 3’ UTR element in their mRNA targets and thus are primarily believed to attenuate translation of the target mRNA. Many studies suggest a continuum of RISC behaviors using similar components, since endogenous microRNAs with perfect complementarity can cleave mRNAs94 and exogenously introduced siRNAs can attenuate translation of mRNAs having imperfect complementarity95, 96.
Few target genes that are regulated by microRNAs are currently documented, but the ones identified have important roles in apoptosis, homeobox regulation, development30, 97-101, cell growth and apoptosis13, 14, viral infection15, 19 and human cancer81, 102-104. This type of gene control represents a new regulatory mechanism, and is predicted to affect many crucial cellular processes including developmental programs.
An important challenge for incorporating microRNA regulation into gene expression mechanisms is the difficulty in predicting or verifying mRNA targets of specific microRNAs. Efforts to map microRNA binding sites specifically in the transcriptome in animal cells have been challenging, relying primarily on computational predictions. However, it has been suggested that microRNAs regulate gene expression of more than 30% of protein coding genes in humans105. One clue to decoding this mystery is the conservation of microRNAs and mRNA target sequences across species. For example, more than a third of the C. elegans microRNAs have easily recognizable homologs in humans106. Sequence conservation argues for conserved function throughout evolution. A number of prominent prediction algorithms have been developed66, 69, 107-110, in most cases based on this observed conservation of complementary “seed” regions conserved across species. A recent review considers these methods and proposes an approach for applying these algorithms111. A few reports extend these predictions to collect supporting evidence for specific biochemical targeting interactions112, microRNA-mRNA presence in polysomes113, or RNA-protein complex composition114, 115. One direct method for validating targets uses a luciferase reporter plasmid with a replaceable 3'UTR13. By cloning a 3'UTR from the suspected target mRNA into this reporter and co-transfecting it with microRNA precursors, inhibition of luciferase production can be detected. In general, many of the computationally-predicted targeting interactions are not consistent between algorithms and relatively little direct experimental evidence substantiates these predictions. In our experience, computed target lists have a high rate of false positives (unpublished results).
microRNA analysis methods
Detection of microRNAs in tissues is possible with a variety of techniques. Several microarrays have been described102, 103, 116-127 and many commercial sources of microRNA arrays can be found (Table 1) Most of these are based on the publicly-available list of microRNAs found in the miRBase database maintained at the Sanger Institute (http://microrna.sanger.ac.uk)128. With most of the array-based methods, it is difficult to claim resolution of specific microRNAs within 1 nt of the probe sequence since the melting temperatures are quite low compared with the longer probes often used for mRNA detection. However, higher specificity can be achieved using direct labeling of microRNAs to obtain RNA:DNA hybridization (Ncode™) or by locked nucleic acid (LNA) oligo probes (mirCURY™). Validation of microarray results originally depended on Northern blots but can also use quantitative real-time PCR (qPCR). In general, qPCR techniques yield the greatest dynamic range, improved specificity, and high sensitivity. In one case, qPCR allowed detection of microRNAs from single cultured neurons or even laser captured somatodendritic compartments129. Other detection techniques include ELISA-like or bead-anchored hybridizations using probes and labeling similar to array methods to perform high-throughput analyses130. A promising technology for detection and quantification of both known and novel microRNAs is MPSS or massively parallel signature sequencing. For example, Lu and colleagues131 sequenced 721,044 17-nt cDNAs prepared from Arabidopsis inflorescence, of which 67,528 were unique. Seventy-seven percent of these matched genome sequences, exceeding by more than 10-fold the previously identified Arabidopsis microRNA library. However, later studies described the importance of filtering these results for genomic hairpin structures and other types of small RNA, reducing the number of predicted or novel structures132, 133. MPSS revealed novel microRNAs in Marek's disease virus that contribute to pathogenesis134. MPSS also revealed 447 new microRNAs in chimpanzee and human brains135. Furthermore, MPSS of mRNAs found ∼25% uncharacterized or novel genes expressed in hESC42. As MPSS becomes more widely available and the costs per sample come down it is possible that direct tag sequencing may become an important technique in the study of microRNAs.
Table 1.
Commercial sources of microRNA microarrays
| Product Name | Source | References |
|---|---|---|
| Human miRNA Microarray | Agilent | 155 |
| mirVana™ | Ambion/Applied Biosystems | 93, 156 |
| Species Specific MicroRNA Arrays | CombiMatrix | |
| MirCURY™ | Exiqon, Inc. | 157, 158 |
| GenoExplorer™ | GenoSensor Corp. | 159-161 |
| NCode™ | Invitrogen, Inc. | 117, 144 |
| Human microRNA Microarray | LC Sciences | 30, 162 |
microRNAs in embryonic and adult stem cells
Several studies have identified populations of microRNAs that are associated with specific stem cell types or those that are regulated during stem cell differentiation (Table 2). MicroRNA has been shown to mediate regulation of stem cell division 27, as well as differentiation in adipocyte116, cardiac136, neural24, 137 and hematopoietic lineages88, 138. For example, the involvement of miR181 in differentiation of the hematopoietic lineage is well established88. miR181a is expressed at relatively low levels in Lin− bone marrow progenitor cells compared to B-lymphocytes and is expressed in high levels in Thymus and primary lymphoid organs. Ectopic expression of miR181a in hematopoietic stem cells leads to increased fraction of B-cell lineage both in vitro and in vivo suggesting it to be a positive regulator for B-cell differentiation88. microRNAs are also found to be highly expressed in the adult brain suggesting their involvement in neuronal function and plasticity139. miR124 and miR128, both highly expressed in adult brain, are also expressed in neurons, while miR23 is restricted to astrocytes. The induction of microRNA expression during neural differentiation of embryonic stem cells further suggest their importance in neural development29.
Table 2.
Studies of MicroRNA in Stem Cells
| Cell type |
Process |
Reference |
|
|---|---|---|---|
| Embryonic Stem Cells |
All |
Review: miRNA in Stem cells |
163 164 |
| Murine |
Dicer in miRNA biogenesis in conditional dicer-deficient ESC | 26 | |
| DGCR8 in miRNA biogenesis and stem cell self-renewal | 37 | ||
| ESC | 35 | ||
| ESC and during differentiation | 32 | ||
| ESC in response to DNA damage | 165 | ||
| ESC-derived neurogenesis |
166 |
||
| Human |
ESC | 141, 143 | |
| expression pattern to characterize hESCs | 144, 146 | ||
| ESC and during Differentiation | 145 | ||
| regulation during Stem Cell Division | 167 | ||
| miR125b in Differentiation |
168 |
||
| Neural cells |
Review-miRNA in Neural cells | ||
| expression during neural differentiation | 22 | ||
| regulation of Lin-28 during neural differentiation | 137 | ||
| miR124 in developing neural tube | 20 | ||
| Neural cell specification |
29; 169 |
||
| Hematopoeitic cells | Review: miRNA in Hematology | 170 | |
| Murine and human erythroid differentiation | 171 | ||
| in CD34+ cells | 33 | ||
| in hematopoeitic lineage differentiation | 88: 172 | ||
| in cord blood CD34+ erythorpoiesis | 173 | ||
| miRs 17−5p, 20a and 106a in monocytopoiesis | 174 | ||
| miR221 and 222 inhibit normal erythropoeisis | 175 | ||
| miR181a in T cell selection | 176 | ||
| Timing of miR-150 in mature B and T cells | 177 | ||
The miR-15a-miR16 cluster resides in a region of human chromosome 13 thought to harbor a tumor suppressor gene102, 140 and also to target Bcl2, potentially modulating apoptosis98. MicroRNAs have also been found to control G1/S checkpoint bypass in Drosophila stem cells27, demonstrating their intimate role in stem cell division.
A unique set of microRNAs is reported to be expressed in mouse embryonic stem cells (ESC) that changes with differentiation during differentiation via embryoid body (EB) formation25, 26, 35, 141, 142. Northern blot and cloning analysis identified several microRNAs in human ESC, several of which were identical to microRNAs previously reported in mouse ESCs143. We recently reported that several microRNAs are highly expressed across multiple hESC lines144. Furthermore, as these cells differentiate, the microRNA profiles change significantly. Combining the profiling of microRNAs and mRNAs, correlation of gene and microRNA expression predicts regulatory interaction of microRNA and mRNAs involved in both maintenance of pluripotency as well as differentiation145. Based on this, microRNA expression pattern in human embryonic stem cells is included in the panel of tests suggested for qualification of human embryonic stem cells146.
Consistent with the observation for individual microRNAs, mouse ES knockout cells lacking Dicer25, required for cleavage of microRNA precursors, or DGCR837, an RNA-binding protein essential for processing microRNAs, exhibits a failure to undergo differentiation and completely prevented mouse ESC from differentiating into embryoid bodies, suggesting that microRNAs are required to inhibit ES self-renewal. Furthermore, knockout mice lacking Argonaute2, the catalytic component of the RISC complex, exhibited severe defects in neural development, including the failure to close neural tube147. Collectively, these studies implicate the importance of microRNAs as key regulators during the process of stem cell maintenance and differentiation.
A unique set of microRNAs are expressed in embryonic stem cells (Table 3). Using Northern blot analysis, cloning, or array methods, a set of microRNAs specific to pluripotent cell types such as mouse embryonic stem (ES) cells35, mouse or human embryonic carcinoma cells22, or human ESC32, 143-145, has been identified. While these studies could not yet ascribe regulatory functions to these microRNA populations, their unique presence in stem cells and their disappearance during differentiation suggest roles in maintaining “stemness” by suppressing pluripotency and/or restricting cell differentiation. In one example, miR-134 was found to be enriched after retinoic acid treatment of mouse ES cells, and miR-134 reduces expression of Nanog and LRH130, enhancing differentiation to specific lineages.
Table 3.
MicroRNAs associated with embryonic stem cells
| miRNA |
Species |
Function |
Reference |
|---|---|---|---|
| miR291,292, 293,294,295 | Mouse | Present in ESC, low in EB, absent in MEF/NIH3T3 | 35 |
| miR29a, 29b, 193, 199 | Present in MEF/NIH3T3, absent in ESC,EB | ||
| miR296 | ESC specific | ||
| miR21, miR22 | Spontaneously diff cells | ||
| miR15a, 16, 19b, 92, 93, 96, 130, 130b |
|
General physiological aspects |
|
| miR106a, 127, 130b, 15b, 16, 18, 19b | Human | Present in single hESC cells | 141 |
| miR20, 21, 290, 291, 292, 293, 294, 295, | |||
| miR92, 93 |
|
|
|
| miR302b, 302c, 302a, 302a, 367 | Human | Present in Both hESC and EC | 143 |
| miR200c, 368, 154, 371, 372, 373 |
|
ESC only |
|
| miR200c, 371, 372, 302a, 302d | Human | Present in hES, low in EB, absent in adult brain | 145 |
| miR373, 302c, 21, 222, 296, 494, 367 | Higher in ESC compared to EB or adult brain | ||
| miR17M, 92, 93 | Higher in EB comapred to ESC or adult brain | ||
| Let7a, miR29a, 143 | Higher in adult tissue followed by ESC and EB | ||
| miR134, 18a, 18b, 16 |
|
Unchanged between hESC & EB |
|
| miR183,290,291−3p, 291−5p, 292−5p, 293, 294, 295, 302a, 302b, 302c, 302d, 367, 292−3p | Mouse | Stem cell specific | 32 |
microRNA expression in MSC and derivatives
Compared with these examples examining microRNAs in ESC, relatively few studies have identified microRNA expression patterns in differentiating MSC or specific pathways that could be targeted (Table 4). In one case, miR-103 and miR-107 were found to be encoded within an intron of the PANK gene in a pattern that is conserved among a large array of organisms148. Both microRNAs (the high degree of sequence homology makes them virtually indistinguishable) are predicted to target a large number of genes involved in acetyl-CoA and lipid metabolism. Another study found that the modulation of a single microRNA could promote the formation of adipocytes from precursor cells116. This differential targeting was shown to be mediated by the translational regulation of ERK5, an intermediate in the LIF signaling pathway116. ERK5 is also known to mediate a neuronal survival response following retrograde signaling of neurotrophins149, 150. A third example found that miR-140 was expressed during cartilage development and that miR-140 may act by inhibiting HDAC4, a likely co-repressor of Runx2151. These initial examples show that, similar to studies in ESC, microRNAs are likely to be mediators of key pathways during MSC differentiation.
Table 4.
microRNAs associated with MSCs
We have recently begun mapping microRNA changes during MSC differentiation using microarrays and qPCR validation152. In general, we observe a great deal of variability between individual donors, making generalizations difficult. For example, MSC prepared from two different donors had a correlation coefficient of 0.912 over all detected mRNAs, and MSC from the same donor prepared from two subsequent passages had a correlation of 0.804. However, using pooled samples of MSC cultures, we identified 27 human microRNAs as regulated during MSC differentiation into adipocytes, osteocytes or chondrocytes. Expression patterns could be clustered to highlight specific microRNAs with restricted expression patterns, confirming previously-described results (e.g., miR-143 and miR-145 enriched in adipocytes) as well as identifying novel markers (e.g. miR-638 and miR-663 in chondrocytes). We found that several of the regulated microRNAs were attributable to changes upon osteocyte differentiation. Standard gene expression analysis found that osteocyte differentiation included pathway signatures for PDGF stimulation, as was first recognized by Ng et al.153. To determine whether the class of osteocyte-specific microRNAs could by regulated by PDGF-stimulated transcriptional mechanisms, we bioinformatically associated PDGF-responsive transcription factor position weight matrices with genomic sequences upstream of regulated microRNAs. Our results predict a significant number of PDGF-responsive transcription factor sites are found upstream of regulated microRNAs, compared with expressed but unregulated microRNAs (Fig. 1). This prediction was confirmed by treating differentiating MSC with AG-370, a tyrphostin inhibitor of the PDGF signaling pathway. MicroRNAs ranking highly on a confidence list of predicted PDGF regulation targets were generally also inhibited by the addition of AG-370 during osteogenic differentiation (examples listed in Table 4). Taken together, these results identify an osteogenic class of microRNAs that are regulated by growth factor signaling during differentiation, demonstrating the regulation of specific microRNAs during MSC differentiation.
Figure 1.

Model of PDGF-regulated microRNAs modulating translation of targeted mRNAs.
Predicted microRNA-target interactions
The importance of microRNA regulation in MSC differentiation is demonstrated in the increasing evidence for specific microRNAs participating in known pathways through mRNA target interactions. There are fewer known examples for bone and connective tissue differentiation but we should expect to find target pathways as these microRNAs are described.
Recently, using a concept similar to the “biclustering” of gene expression data154, we carried out correlation of an mRNA to each microRNA expression level in multiple hESC lines and their differentiated samples organized as separate groups145. A similar approach was also employed to study the changes in miRNA expression during murine embryonic stem cell differentiation32. Such a cross-correlation clustering provides interpretable lists of microRNAs and associated mRNAs that may be hypothesized to interact through specific mechanisms providing impetus towards recognizing microRNAs involved in controlling stem cell fate.
Future directions
As the complexity of the regulatory network in differentiating stem cells begins to become known, identification of regulated microRNAs and their interactions with transcriptional networks and, ultimately, cellular regulatory systems, becomes valuable. The organization of the hierarchical order of all types of stem cells based on their pluripotency and linkage of their functional characteristics to genetic and epigenetic regulatory elements provides a novel means to understand and, potentially, manipulate cell fate. With the ultimate goal of using microRNAs to modulate differentiation of stem cells as potential therapeutic agents, we expect to produce a clearer understanding of microRNAs in specific cellular pathways such as development, proliferation, stem cell renewal, differentiation and in diseases such as cancer.
Acknowledgements
RPH is supported by NIH, the New Jersey Commission on Spinal Cord Research, the New Jersey Commission on Science & Technology, and Invitrogen, Inc.
REFERENCES
- 1.Bartel DP. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 2004;116:281. doi: 10.1016/s0092-8674(04)00045-5. [DOI] [PubMed] [Google Scholar]
- 2.Lai EC. microRNAs: runts of the genome assert themselves. Curr Biol. 2003;13:R925–R936. doi: 10.1016/j.cub.2003.11.017. [DOI] [PubMed] [Google Scholar]
- 3.Kosik KS, Krichevsky AM. The Elegance of the MicroRNAs: A Neuronal Perspective. Neuron. 2005;47:779–782. doi: 10.1016/j.neuron.2005.08.019. [DOI] [PubMed] [Google Scholar]
- 4.Wienholds E, Plasterk RH. MicroRNA function in animal development. FEBS Lett. 2005 doi: 10.1016/j.febslet.2005.07.070. [DOI] [PubMed] [Google Scholar]
- 5.Croce CM, Calin GA. miRNAs, cancer, and stem cell division. Cell. 2005;122:6–7. doi: 10.1016/j.cell.2005.06.036. [DOI] [PubMed] [Google Scholar]
- 6.Harfe BD. MicroRNAs in vertebrate development. Curr Opin Genet Dev. 2005;15:410–415. doi: 10.1016/j.gde.2005.06.012. [DOI] [PubMed] [Google Scholar]
- 7.Pasquinelli AE, Hunter S, Bracht J. MicroRNAs: a developing story. Curr Opin Genet Dev. 2005;15:200–205. doi: 10.1016/j.gde.2005.01.002. [DOI] [PubMed] [Google Scholar]
- 8.Carrington JC, Ambros V. Role of microRNAs in plant and animal development. Science. 2003;301:336–338. doi: 10.1126/science.1085242. [DOI] [PubMed] [Google Scholar]
- 9.Hunter C, Poethig RS. miSSING LINKS: miRNAs and plant development. Curr Opin Genet Dev. 2003;13:372–378. doi: 10.1016/s0959-437x(03)00081-9. [DOI] [PubMed] [Google Scholar]
- 10.Ambros V. MicroRNA pathways in flies and worms: growth, death, fat, stress, and timing. Cell. 2003;113:673–676. doi: 10.1016/s0092-8674(03)00428-8. [DOI] [PubMed] [Google Scholar]
- 11.Bommer GT, Gerin I, Feng Y, et al. p53-mediated activation of miRNA34 candidate tumor-suppressor genes. Curr Biol. 2007;17:1298–1307. doi: 10.1016/j.cub.2007.06.068. [DOI] [PubMed] [Google Scholar]
- 12.Chan JA, Krichevsky AM, Kosik KS. MicroRNA-21 is an antiapoptotic factor in human glioblastoma cells. Cancer Res. 2005;65:6029–6033. doi: 10.1158/0008-5472.CAN-05-0137. [DOI] [PubMed] [Google Scholar]
- 13.Cheng AM, Byrom MW, Shelton J, et al. Antisense inhibition of human miRNAs and indications for an involvement of miRNA in cell growth and apoptosis. Nucleic Acids Res. 2005;33:1290–1297. doi: 10.1093/nar/gki200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Raver-Shapira N, Marciano E, Meiri E, et al. Transcriptional activation of miR-34a contributes to p53-mediated apoptosis. Mol Cell. 2007;26:731–743. doi: 10.1016/j.molcel.2007.05.017. [DOI] [PubMed] [Google Scholar]
- 15.Sullivan CS, Ganem D. MicroRNAs and viral infection. Mol Cell. 2005;20:3–7. doi: 10.1016/j.molcel.2005.09.012. [DOI] [PubMed] [Google Scholar]
- 16.Bennasser Y, Le SY, Yeung ML, et al. HIV-1 encoded candidate micro-RNAs and their cellular targets. Retrovirology. 2004;1:43. doi: 10.1186/1742-4690-1-43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Couturier JP, Root-Bernstein RS. HIV may produce inhibitory microRNAs (miRNAs) that block production of CD28, CD4 and some interleukins. J Theor Biol. 2005;235:169–184. doi: 10.1016/j.jtbi.2005.01.001. [DOI] [PubMed] [Google Scholar]
- 18.Grey F, Antoniewicz A, Allen E, et al. Identification and characterization of human cytomegalovirus-encoded microRNAs. J Virol. 2005;79:12095–12099. doi: 10.1128/JVI.79.18.12095-12099.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Lo HL, Chang T, Yam P, et al. Inhibition of HIV-1 replication with designed miRNAs expressed from RNA polymerase II promoters. Gene Ther. 2007 doi: 10.1038/sj.gt.3303011. [DOI] [PubMed] [Google Scholar]
- 20.Cao X, Pfaff SL, Gage FH. A functional study of miR-124 in the developing neural tube. Genes Dev. 2007;21:531–536. doi: 10.1101/gad.1519207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Greco SJ, Rameshwar P. MicroRNAs regulate synthesis of the neurotransmitter substance P in human mesenchymal stem cell-derived neuronal cells. Proc Natl Acad Sci U S A. 2007;104:15484–15489. doi: 10.1073/pnas.0703037104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Sempere LF, Freemantle S, Pitha-Rowe I, et al. Expression profiling of mammalian microRNAs uncovers a subset of brain-expressed microRNAs with possible roles in murine and human neuronal differentiation. Genome Biol. 2004;5:R13. doi: 10.1186/gb-2004-5-3-r13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Wayman GA, Soderling TR, Goodman RH, et al. Neuron-enriched microRNA 132 regulates activity-dependent dendritic growth and synaptogenesis. Society for Neuroscience Abstracts. 2006;36:321.321. [Google Scholar]
- 24.Kuwabara T, Hsieh J, Nakashima K, et al. A small modulatory dsRNA specifies the fate of adult neural stem cells. Cell. 2004;116:779–793. doi: 10.1016/s0092-8674(04)00248-x. [DOI] [PubMed] [Google Scholar]
- 25.Kanellopoulou C, Muljo SA, Kung AL, et al. Dicer-deficient mouse embryonic stem cells are defective in differentiation and centromeric silencing. Genes Dev. 2005;19:489–501. doi: 10.1101/gad.1248505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Murchison EP, Partridge JF, Tam OH, et al. Characterization of Dicer-deficient murine embryonic stem cells. Proc Natl Acad Sci U S A. 2005;102:12135–12140. doi: 10.1073/pnas.0505479102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Hatfield SD, Shcherbata HR, Fischer KA, et al. Stem cell division is regulated by the microRNA pathway. Nature. 2005;435:974–978. doi: 10.1038/nature03816. [DOI] [PubMed] [Google Scholar]
- 28.Kidner CA, Martienssen RA. The role of ARGONAUTE1 (AGO1) in meristem formation and identity. Dev Biol. 2005;280:504–517. doi: 10.1016/j.ydbio.2005.01.031. [DOI] [PubMed] [Google Scholar]
- 29.Smirnova L, Grafe A, Seiler A, et al. Regulation of miRNA expression during neural cell specification. Eur J Neurosci. 2005;21:1469–1477. doi: 10.1111/j.1460-9568.2005.03978.x. [DOI] [PubMed] [Google Scholar]
- 30.Tay YM, Tam WL, Ang YS, et al. MicroRNA-134 Modulates the Differentiation of Mouse Embryonic Stem Cells where it Causes Post-transcriptional Attenuation of Nanog and LRH1. Stem Cells. 2007 doi: 10.1634/stemcells.2007-0295. [DOI] [PubMed] [Google Scholar]
- 31.Abrahante JE, Daul AL, Li M, et al. The Caenorhabditis elegans hunchback-like gene lin-57 / hbl-1 controls developmental time and is regulated by microRNAs. Dev Cell. 2003;4:625. doi: 10.1016/s1534-5807(03)00127-8. [DOI] [PubMed] [Google Scholar]
- 32.Chen C, Ridzon D, Lee CT, et al. Defining embryonic stem cell identity using differentiation-related microRNAs and their potential targets. Mamm Genome. 2007;18:316–327. doi: 10.1007/s00335-007-9032-6. [DOI] [PubMed] [Google Scholar]
- 33.Georgantas RW, 3rd, Hildreth R, Morisot S, et al. CD34+ hematopoietic stem-progenitor cell microRNA expression and function: a circuit diagram of differentiation control. Proc Natl Acad Sci U S A. 2007;104:2750–2755. doi: 10.1073/pnas.0610983104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Gillis A, Stoop H, Hersmus R, et al. High-throughput microRNAome analysis in human germ cell tumours. J Pathol. 2007 doi: 10.1002/path.2230. [DOI] [PubMed] [Google Scholar]
- 35.Houbaviy HB, Murray MF, Sharp PA. Embryonic stem cell-specific MicroRNAs. Dev Cell. 2003;5:351. doi: 10.1016/s1534-5807(03)00227-2. [DOI] [PubMed] [Google Scholar]
- 36.Ren G, Li T, Lan JQ, et al. Lentiviral RNAi-induced downregulation of adenosine kinase in human mesenchymal stem cell grafts: A novel perspective for seizure control. Exp Neurol. 2007 doi: 10.1016/j.expneurol.2007.07.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Wang Y, Medvid R, Melton C, et al. DGCR8 is essential for microRNA biogenesis and silencing of embryonic stem cell self-renewal. Nat Genet. 2007 doi: 10.1038/ng1969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Friedenstein AJ. Marrow stromal fibroblasts. Calcif Tissue Int. 1995;56(Suppl 1):S17. [Google Scholar]
- 39.Prockop DJ. Marrow stromal cells as stem cells for nonhematopoietic tissues. Science. 1997;276:71–74. doi: 10.1126/science.276.5309.71. [DOI] [PubMed] [Google Scholar]
- 40.Bianco P, Riminucci M, Gronthos S, et al. Bone marrow stromal stem cells: nature, biology, and potential applications. Stem Cells. 2001;19:180–192. doi: 10.1634/stemcells.19-3-180. [DOI] [PubMed] [Google Scholar]
- 41.Phinney DG. Building a consensus regarding the nature and origin of mesenchymal stem cells. J Cell Biochem Suppl. 2002;38:7–12. doi: 10.1002/jcb.10084. [DOI] [PubMed] [Google Scholar]
- 42.Brandenberger R, Khrebtukova I, Thies RS, et al. MPSS profiling of human embryonic stem cells. BMC Dev Biol. 2004;4:10. doi: 10.1186/1471-213X-4-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Bruder SP, Jaiswal N, Haynesworth SE. Growth kinetics, self-renewal, and the osteogenic potential of purified human mesenchymal stem cells during extensive subcultivation and following cryopreservation. J Cell Biochem. 1997;64:278–294. doi: 10.1002/(sici)1097-4644(199702)64:2<278::aid-jcb11>3.0.co;2-f. [DOI] [PubMed] [Google Scholar]
- 44.Bruder SP, Kurth AA, Shea M, et al. Bone regeneration by implantation of purified, culture-expanded human mesenchymal stem cells. J Orthop Res. 1998;16:155–162. doi: 10.1002/jor.1100160202. [DOI] [PubMed] [Google Scholar]
- 45.Dennis JE, Merriam A, Awadallah A, et al. A quadripotential mesenchymal progenitor cell isolated from the marrow of an adult mouse. J Bone Miner Res. 1999;14:700–709. doi: 10.1359/jbmr.1999.14.5.700. [DOI] [PubMed] [Google Scholar]
- 46.Pittenger MF, Mackay AM, Beck SC, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284:143–147. doi: 10.1126/science.284.5411.143. [DOI] [PubMed] [Google Scholar]
- 47.Haynesworth SE, Baber MA, Caplan AI. Cell surface antigens on human marrow-derived mesenchymal cells are detected by monoclonal antibodies. Bone. 1992;13:69–80. doi: 10.1016/8756-3282(92)90363-2. [DOI] [PubMed] [Google Scholar]
- 48.Caplan AI. Mesenchymal stem cells. J Orthop Res. 1991;9:641–650. doi: 10.1002/jor.1100090504. [DOI] [PubMed] [Google Scholar]
- 49.Chunmeng S, Tianmin C. Skin: a promising reservoir for adult stem cell populations. Med Hypotheses. 2004;62:683–688. doi: 10.1016/j.mehy.2003.12.022. [DOI] [PubMed] [Google Scholar]
- 50.De Bari C, Dell'Accio F, Tylzanowski P, et al. Multipotent mesenchymal stem cells from adult human synovial membrane. Arthritis Rheum. 2001;44:1928–1942. doi: 10.1002/1529-0131(200108)44:8<1928::AID-ART331>3.0.CO;2-P. [DOI] [PubMed] [Google Scholar]
- 51.Shi S, Gronthos S. Perivascular niche of postnatal mesenchymal stem cells in human bone marrow and dental pulp. J Bone Miner Res. 2003;18:696–704. doi: 10.1359/jbmr.2003.18.4.696. [DOI] [PubMed] [Google Scholar]
- 52.Bieback K, Kern S, Kluter H, et al. Critical parameters for the isolation of mesenchymal stem cells from umbilical cord blood. Stem Cells. 2004;22:625–634. doi: 10.1634/stemcells.22-4-625. [DOI] [PubMed] [Google Scholar]
- 53.In 't Anker PS, Scherjon SA, Kleijburg-van der Keur C, et al. Amniotic fluid as a novel source of mesenchymal stem cells for therapeutic transplantation. Blood. 2003;102:1548–1549. doi: 10.1182/blood-2003-04-1291. [DOI] [PubMed] [Google Scholar]
- 54.Li CD, Zhang WY, Li HL, et al. Isolation and Identification of a Multilineage Potential Mesenchymal Cell from Human Placenta. Placenta. 2005 doi: 10.1016/j.placenta.2005.08.003. [DOI] [PubMed] [Google Scholar]
- 55.Pittenger MF, Martin BJ. Mesenchymal stem cells and their potential as cardiac therapeutics. Circ Res. 2004;95:9–20. doi: 10.1161/01.RES.0000135902.99383.6f. [DOI] [PubMed] [Google Scholar]
- 56.Fibbe WE, Noort WA. Mesenchymal stem cells and hematopoietic stem cell transplantation. Ann N Y Acad Sci. 2003;996:235–244. doi: 10.1111/j.1749-6632.2003.tb03252.x. [DOI] [PubMed] [Google Scholar]
- 57.Le Blanc K, Rasmusson I, Sundberg B, et al. Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cells. Lancet. 2004;363:1439–1441. doi: 10.1016/S0140-6736(04)16104-7. [DOI] [PubMed] [Google Scholar]
- 58.Marion NW, Mao JJ. Mesenchymal stem cells and tissue engineering. Methods Enzymol. 2006;420:339–361. doi: 10.1016/S0076-6879(06)20016-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Shi S, Bartold PM, Miura M, et al. The efficacy of mesenchymal stem cells to regenerate and repair dental structures. Orthod Craniofac Res. 2005;8:191–199. doi: 10.1111/j.1601-6343.2005.00331.x. [DOI] [PubMed] [Google Scholar]
- 60.Gao J, Caplan AI. Mesenchymal stem cells and tissue engineering for orthopaedic surgery. Chir Organi Mov. 2003;88:305–316. [PubMed] [Google Scholar]
- 61.Tuan RS, Boland G, Tuli R. Adult mesenchymal stem cells and cell-based tissue engineering. Arthritis Res Ther. 2003;5:32–45. doi: 10.1186/ar614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Dominici M, Le Blanc K, Mueller I, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8:315–317. doi: 10.1080/14653240600855905. [DOI] [PubMed] [Google Scholar]
- 63.Muraglia A, Cancedda R, Quarto R. Clonal mesenchymal progenitors from human bone marrow differentiate in vitro according to a hierarchical model. J Cell Sci. 2000;113(Pt 7):1161–1166. doi: 10.1242/jcs.113.7.1161. [DOI] [PubMed] [Google Scholar]
- 64.Doi M, Nagano A, Nakamura Y. Molecular cloning and characterization of a novel gene, EMILIN-5, and its possible involvement in skeletal development. Biochem Biophys Res Commun. 2004;313:888–893. doi: 10.1016/j.bbrc.2003.11.181. [DOI] [PubMed] [Google Scholar]
- 65.Qi H, Aguiar DJ, Williams SM, et al. Identification of genes responsible for osteoblast differentiation from human mesodermal progenitor cells. Proc Natl Acad Sci U S A. 2003;100:3305–3310. doi: 10.1073/pnas.0532693100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Lewis BP, Burge CB, Bartel DP. Conserved Seed Pairing, Often Flanked by Adenosines, Indicates that Thousands of Human Genes are MicroRNA Targets. Cell. 2005;120:15. doi: 10.1016/j.cell.2004.12.035. [DOI] [PubMed] [Google Scholar]
- 67.Xie X, Lu J, Kulbokas EJ, et al. Systematic discovery of regulatory motifs in human promoters and 3' UTRs by comparison of several mammals. Nature. 2005;434:338–345. doi: 10.1038/nature03441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Bentwich I, Avniel A, Karov Y, et al. Identification of hundreds of conserved and nonconserved human microRNAs. Nat Genet. 2005;37:766–770. doi: 10.1038/ng1590. [DOI] [PubMed] [Google Scholar]
- 69.Miranda KC, Huynh T, Tay Y, et al. A pattern-based method for the identification of MicroRNA binding sites and their corresponding heteroduplexes. Cell. 2006;126:1203–1217. doi: 10.1016/j.cell.2006.07.031. [DOI] [PubMed] [Google Scholar]
- 70.Lee Y, Ahn C, Han J, et al. The nuclear RNase III Drosha initiates microRNA processing. Nature. 2003;425:415. doi: 10.1038/nature01957. [DOI] [PubMed] [Google Scholar]
- 71.Yi R, Qin Y, Macara IG, et al. Exportin-5 mediates the nuclear export of pre-microRNAs and short hairpin RNAs. Genes Dev. 2003;17:3011. doi: 10.1101/gad.1158803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Lund E, Guttinger S, Calado A, et al. Nuclear export of microRNA precursors. Science. 2004;303:95. doi: 10.1126/science.1090599. [DOI] [PubMed] [Google Scholar]
- 73.Hutvagner G, McLachlan J, Pasquinelli AE, et al. A cellular function for the RNA-interference enzyme Dicer in the maturation of the let-7 small temporal RNA. Science. 2001;293:834. doi: 10.1126/science.1062961. [DOI] [PubMed] [Google Scholar]
- 74.Forstemann K, Tomari Y, Du T, et al. Normal microRNA maturation and germ-line stem cell maintenance requires Loquacious, a double-stranded RNA-binding domain protein. PLoS Biol. 2005;3:e236. doi: 10.1371/journal.pbio.0030236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Han J, Lee Y, Yeom KH, et al. The Drosha-DGCR8 complex in primary microRNA processing. Genes Dev. 2004;18:3016. doi: 10.1101/gad.1262504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Lee Y, Kim M, Han J, et al. MicroRNA genes are transcribed by RNA polymerase II. Embo J. 2004;23:4051. doi: 10.1038/sj.emboj.7600385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Cai X, Hagedorn CH, Cullen BR. Human microRNAs are processed from capped, polyadenylated transcripts that can also function as mRNAs. RNA. 2004;10:1957. doi: 10.1261/rna.7135204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Rodriguez A, Griffiths-Jones S, Ashurst JL, et al. Identification of mammalian microRNA host genes and transcription units. Genome Res. 2004;14:1902. doi: 10.1101/gr.2722704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Lagos-Quintana M, Rauhut R, Yalcin A, et al. Identification of tissue-specific microRNAs from mouse. Curr Biol. 2002;12:735. doi: 10.1016/s0960-9822(02)00809-6. [DOI] [PubMed] [Google Scholar]
- 80.Seitz H, Royo H, Bortolin ML, et al. A large imprinted microRNA gene cluster at the mouse Dlk1-Gtl2 domain. Genome Res. 2004;14:1741. doi: 10.1101/gr.2743304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.He L, Thomson JM, Hemann MT, et al. A microRNA polycistron as a potential human oncogene. Nature. 2005;435:828–833. doi: 10.1038/nature03552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Ying SY, Lin SL. Intronic microRNAs. Biochem.Biophys.Res Commun. 2005;326:515. doi: 10.1016/j.bbrc.2004.10.215. [DOI] [PubMed] [Google Scholar]
- 83.Ruby JG, Jan CH, Bartel DP. Intronic microRNA precursors that bypass Drosha processing. Nature. 2007;448:83–86. doi: 10.1038/nature05983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Luciano DJ, Mirsky H, Vendetti NJ, et al. RNA editing of a miRNA precursor. RNA. 2004;10:1174. doi: 10.1261/rna.7350304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Kawahara Y, Zinshteyn B, Sethupathy P, et al. Redirection of silencing targets by adenosine-to-inosine editing of miRNAs. Science. 2007;315:1137–1140. doi: 10.1126/science.1138050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Aukerman MJ, Sakai H. Regulation of flowering time and floral organ identity by a MicroRNA and its APETALA2-like target genes. Plant Cell. 2003;15:2730–2741. doi: 10.1105/tpc.016238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Bagga S, Bracht J, Hunter S, et al. Regulation by let-7 and lin-4 miRNAs results in target mRNA degradation. Cell. 2005;122:553–563. doi: 10.1016/j.cell.2005.07.031. [DOI] [PubMed] [Google Scholar]
- 88.Chen CZ, Li L, Lodish HF, et al. MicroRNAs modulate hematopoietic lineage differentiation. Science. 2004;303:83–86. doi: 10.1126/science.1091903. [DOI] [PubMed] [Google Scholar]
- 89.Yekta S, Shih IH, Bartel DP. MicroRNA-directed cleavage of HOXB8 mRNA. Science. 2004;304:594–596. doi: 10.1126/science.1097434. [DOI] [PubMed] [Google Scholar]
- 90.Yu Z, Raabe T, Hecht NB. MicroRNA Mirn122a reduces expression of the posttranscriptionally regulated germ cell transition protein 2 (Tnp2) messenger RNA (mRNA) by mRNA cleavage. Biol Reprod. 2005;73:427–433. doi: 10.1095/biolreprod.105.040998. [DOI] [PubMed] [Google Scholar]
- 91.Giraldez AJ, Mishima Y, Rihel J, et al. Zebrafish MiR-430 promotes deadenylation and clearance of maternal mRNAs. Science. 2006;312:75–79. doi: 10.1126/science.1122689. [DOI] [PubMed] [Google Scholar]
- 92.Wu L, Fan J, Belasco JG. MicroRNAs direct rapid deadenylation of mRNA. Proc Natl Acad Sci U S A. 2006;103:4034–4039. doi: 10.1073/pnas.0510928103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Zhang L, Huang J, Yang N, et al. microRNAs exhibit high frequency genomic alterations in human cancer. Proc Natl Acad Sci U S A. 2006;103:9136–9141. doi: 10.1073/pnas.0508889103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Hutvagner G, Zamore PD. A microRNA in a multiple-turnover RNAi enzyme complex. Science. 2002;297:2056–2060. doi: 10.1126/science.1073827. [DOI] [PubMed] [Google Scholar]
- 95.Saxena S, Jonsson ZO, Dutta A. Small RNAs with imperfect match to endogenous mRNA repress translation. Implications for off-target activity of small inhibitory RNA in mammalian cells. J Biol Chem. 2003;278:44312. doi: 10.1074/jbc.M307089200. [DOI] [PubMed] [Google Scholar]
- 96.Zeng Y, Cullen BR. Sequence requirements for micro RNA processing and function in human cells. Rna. 2003;9:112–123. doi: 10.1261/rna.2780503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Mansfield JH, Harfe BD, Nissen R, et al. MicroRNA-responsive ‘sensor’ transgenes uncover Hox-like and other developmentally regulated patterns of vertebrate microRNA expression. Nat.Genet. 2004;36:1079. doi: 10.1038/ng1421. [DOI] [PubMed] [Google Scholar]
- 98.Cimmino A, Calin GA, Fabbri M, et al. miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc Natl Acad Sci U S A. 2005 doi: 10.1073/pnas.0506654102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Nakahara K, Kim K, Sciulli C, et al. Targets of microRNA regulation in the Drosophila oocyte proteome. Proc Natl Acad Sci U S A. 2005;102:12023–12028. doi: 10.1073/pnas.0500053102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.O'Donnell KA, Wentzel EA, Zeller KI, et al. c-Myc-regulated microRNAs modulate E2F1 expression. Nature. 2005;435:839–843. doi: 10.1038/nature03677. [DOI] [PubMed] [Google Scholar]
- 101.Tanno B, Cesi V, Vitali R, et al. Silencing of endogenous IGFBP-5 by micro RNA interference affects proliferation, apoptosis and differentiation of neuroblastoma cells. Cell Death Differ. 2005;12:213–223. doi: 10.1038/sj.cdd.4401546. [DOI] [PubMed] [Google Scholar]
- 102.Calin GA, Dumitru CD, Shimizu M, et al. Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci U S A. 2002;99:15524–15529. doi: 10.1073/pnas.242606799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Calin GA, Liu CG, Sevignani C, et al. MicroRNA profiling reveals distinct signatures in B cell chronic lymphocytic leukemias. Proc.Natl.Acad.Sci.U.S.A. 2004;101:11755. doi: 10.1073/pnas.0404432101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Zhu S, Si ML, Wu H, et al. MicroRNA-21 targets the tumor suppressor gene tropomyosin 1 (TPM1). J Biol Chem. 2007 doi: 10.1074/jbc.M611393200. [DOI] [PubMed] [Google Scholar]
- l05.Berezikov E, Guryev V, van de BJ, et al. Phylogenetic Shadowing and Computational Identification of Human microRNA Genes. Cell. 2005;120:21. doi: 10.1016/j.cell.2004.12.031. [DOI] [PubMed] [Google Scholar]
- 106.Lim LP, Lau NC, Weinstein EG, et al. The microRNAs of Caenorhabditis elegans. Genes Dev. 2003;17:991. doi: 10.1101/gad.1074403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Grun D, Wang YL, Langenberger D, et al. microRNA Target Predictions across Seven Drosophila Species and Comparison to Mammalian Targets. PLoS Comput Biol. 2005;1:e13. doi: 10.1371/journal.pcbi.0010013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.John B, Enright AJ, Aravin A, et al. Human MicroRNA Targets. PLoS.Biol. 2004;2:e363. doi: 10.1371/journal.pbio.0020363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Lewis BP, Shih IH, Jones-Rhoades MW, et al. Prediction of mammalian microRNA targets. Cell. 2003;115:787–798. doi: 10.1016/s0092-8674(03)01018-3. [DOI] [PubMed] [Google Scholar]
- 110.Enright AJ, John B, Gaul U, et al. MicroRNA targets in Drosophila. Genome Biol. 2003;5:R1. doi: 10.1186/gb-2003-5-1-r1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Sethupathy P, Megraw M, Hatzigeorgiou AG. A guide through present computational approaches for the identification of mammalian microRNA targets. Nat Methods. 2006;3:881–886. doi: 10.1038/nmeth954. [DOI] [PubMed] [Google Scholar]
- 112.Lall S, Grun D, Krek A, et al. A genome-wide map of conserved microRNA targets in C. elegans. Curr Biol. 2006;16:460–471. doi: 10.1016/j.cub.2006.01.050. [DOI] [PubMed] [Google Scholar]
- 113.Nakamoto M, Jin P, O'Donnell WT, et al. Physiological identification of human transcripts translationally regulated by a specific microRNA. Hum Mol Genet. 2005;14:3813–3821. doi: 10.1093/hmg/ddi397. [DOI] [PubMed] [Google Scholar]
- 114.Easow G, Teleman AA, Cohen SM. Isolation of microRNA targets by miRNP immunopurification. Rna. 2007;13:1198–1204. doi: 10.1261/rna.563707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Beitzinger M, Peters L, Zhu JY, et al. Identification of Human microRNA Targets From Isolated Argonaute Protein Complexes. RNA Biol. 2007:4. doi: 10.4161/rna.4.2.4640. [DOI] [PubMed] [Google Scholar]
- 116.Esau C, Kang X, Peralta E, et al. MicroRNA-143 regulates adipocyte differentiation. J Biol Chem. 2004;279:52361–52365. doi: 10.1074/jbc.C400438200. [DOI] [PubMed] [Google Scholar]
- 117.Goff LA, Yang M, Bowers J, et al. Rational probe optimization and enhanced detection strategy for microRNAs using microarrays. RNA Biology. 2005;2:e9–e16. doi: 10.4161/rna.2.3.2059. [DOI] [PubMed] [Google Scholar]
- 118.Krichevsky AM, King KS, Donahue CP, et al. A microRNA array reveals extensive regulation of microRNAs during brain development. Rna. 2003;9:1274. doi: 10.1261/rna.5980303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Babak T, Zhang W, Morris Q, et al. Probing microRNAs with microarrays: Tissue specificity and functional inference. RNA. 2004;10:1813. doi: 10.1261/rna.7119904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Barad O, Meiri E, Avniel A, et al. MicroRNA expression detected by oligonucleotide microarrays: system establishment and expression profiling in human tissues. Genome Res. 2004;14:2486–2494. doi: 10.1101/gr.2845604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Liu CG, Calin GA, Meloon B, et al. An oligonucleotide microchip for genome-wide microRNA profiling in human and mouse tissues. Proc.Natl.Acad.Sci.U.S.A. 2004;101:9740. doi: 10.1073/pnas.0403293101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Miska EA, Alvarez-Saavedra E, Townsend M, et al. Microarray analysis of microRNA expression in the developing mammalian brain. Genome Biol. 2004;5:R68. doi: 10.1186/gb-2004-5-9-r68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Sun Y, Koo S, White N, et al. Development of a micro-array to detect human and mouse microRNAs and characterization of expression in human organs. Nucleic Acids Res. 2004;32:e188. doi: 10.1093/nar/gnh186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Baskerville S, Bartel DP. Microarray profiling of microRNAs reveals frequent coexpression with neighboring miRNAs and host genes. Rna. 2005;11:241–247. doi: 10.1261/rna.7240905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Liang RQ, Li W, Li Y, et al. An oligonucleotide microarray for microRNA expression analysis based on labeling RNA with quantum dot and nanogold probe. Nucleic Acids Res. 2005;33:e17. doi: 10.1093/nar/gni019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Lim LP, Lau NC, Garrett-Engele P, et al. Microarray analysis shows that some microRNAs downregulate large numbers of target mRNAs. Nature. 2005;433:769–773. doi: 10.1038/nature03315. [DOI] [PubMed] [Google Scholar]
- 127.Beuvink I, Kolb FA, Budach W, et al. A novel microarray approach reveals new tissue-specific signatures of known and predicted mammalian microRNAs. Nucleic Acids Res. 2007;35:e52. doi: 10.1093/nar/gkl1118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Griffiths-Jones S, Grocock RJ, van Dongen S, et al. miRBase: microRNA sequences, targets and gene nomenclature. Nucleic Acids Res. 2006;34:D140–144. doi: 10.1093/nar/gkj112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Kye MJ, Liu T, Levy SF, et al. Somatodendritic microRNAs identified by laser capture and multiplex RT-PCR. Rna. 2007 doi: 10.1261/rna.480407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Mora JR, Getts RC. Enzymatic microRNA detection in microtiter plates with DNA dendrimers. Biotechniques. 2006;41:420, 422, 424. doi: 10.2144/000112270. [DOI] [PubMed] [Google Scholar]
- 131.Lu C, Tej SS, Luo S, et al. Elucidation of the small RNA component of the transcriptome. Science. 2005;309:1567–1569. doi: 10.1126/science.1114112. [DOI] [PubMed] [Google Scholar]
- 132.Jones-Rhoades MW, Bartel DP. Computational Identification of Plant MicroRNAs and Their Targets, Including a Stress-Induced miRNA. Mol.Cell. 2004;14:787. doi: 10.1016/j.molcel.2004.05.027. [DOI] [PubMed] [Google Scholar]
- 133.Lu C, Kulkarni K, Souret FF, et al. MicroRNAs and other small RNAs enriched in the Arabidopsis RNA-dependent RNA polymerase-2 mutant. Genome Res. 2006;16:1276–1288. doi: 10.1101/gr.5530106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Burnside J, Bernberg E, Anderson A, et al. Marek's disease virus encodes MicroRNAs that map to meq and the latency-associated transcript. J Virol. 2006;80:8778–8786. doi: 10.1128/JVI.00831-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Berezikov E, Thuemmler F, van Laake LW, et al. Diversity of microRNAs in human and chimpanzee brain. Nat Genet. 2006;38:1375–1377. doi: 10.1038/ng1914. [DOI] [PubMed] [Google Scholar]
- 136.Zhao Y, Samal E, Srivastava D. Serum response factor regulates a muscle-specific microRNA that targets Hand2 during cardiogenesis. Nature. 2005;436:214–220. doi: 10.1038/nature03817. [DOI] [PubMed] [Google Scholar]
- 137.Wu L, Belasco JG. Micro-RNA regulation of the mammalian lin-28 gene during neuronal differentiation of embryonal carcinoma cells. Mol Cell Biol. 2005;25:9198–9208. doi: 10.1128/MCB.25.21.9198-9208.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Chen CZ, Lodish HF. MicroRNAs as regulators of mammalian hematopoiesis. Semin Immunol. 2005;17:155–165. doi: 10.1016/j.smim.2005.01.001. [DOI] [PubMed] [Google Scholar]
- 139.He L, Hannon GJ. MicroRNAs: small RNAs with a big role in gene regulation. Nat Rev Genet. 2004;5:522–531. doi: 10.1038/nrg1379. [DOI] [PubMed] [Google Scholar]
- 140.Lagos-Quintana M, Rauhut R, Lendeckel W, et al. Identification of novel genes coding for small expressed RNAs. Science. 2001;294:853. doi: 10.1126/science.1064921. [DOI] [PubMed] [Google Scholar]
- 141.Tang F, Hajkova P, Barton SC, et al. MicroRNA expression profiling of single whole embryonic stem cells. Nucleic Acids Res. 2006;34:e9. doi: 10.1093/nar/gnj009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Yang S, Tutton S, Pierce E, et al. Specific double-stranded RNA interference in undifferentiated mouse embryonic stem cells. Mol Cell Biol. 2001;21:7807–7816. doi: 10.1128/MCB.21.22.7807-7816.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Suh MR, Lee Y, Kim JY, et al. Human embryonic stem cells express a unique set of microRNAs. Dev Biol. 2004;270:488–498. doi: 10.1016/j.ydbio.2004.02.019. [DOI] [PubMed] [Google Scholar]
- 144.Josephson R, Ording CJ, Liu Y, et al. Qualification of embryonal carcinoma 2102Ep as a reference for human embryonic stem cell research. Stem Cells. 2007;25:437–446. doi: 10.1634/stemcells.2006-0236. [DOI] [PubMed] [Google Scholar]
- 145.Lakshmipathy U, Love B, Goff L, et al. Micro RNA expression pattern of undifferentiated and differentiated human embryonic stem cells. Stem Cells and Regeneration. 2007 doi: 10.1089/scd.2007.0026. In Press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Pal R, Mandal A, Rao HS, et al. A panel of tests to standardize the characterization of human embryonic stem cells. Regen Med. 2007;2:179–192. doi: 10.2217/17460751.2.2.179. [DOI] [PubMed] [Google Scholar]
- 147.Liu J, Carmell MA, Rivas FV, et al. Argonaute2 is the catalytic engine of mammalian RNAi. Science. 2004;305:1437. doi: 10.1126/science.1102513. [DOI] [PubMed] [Google Scholar]
- 148.Wilfred BR, Wang WX, Nelson PT. Energizing miRNA research: A review of the role of miRNAs in lipid metabolism, with a prediction that miR-103/107 regulates human metabolic pathways. Mol Genet Metab. 2007;91:209–217. doi: 10.1016/j.ymgme.2007.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Watson FL, Heerssen HM, Bhattacharyya A, et al. Neurotrophins use the Erk5 pathway to mediate a retrograde survival response. Nat.Neurosci. 2001;4:981. doi: 10.1038/nn720. [DOI] [PubMed] [Google Scholar]
- 150.Cavanaugh JE. Role of extracellular signal regulated kinase 5 in neuronal survival. Eur.J Biochem. 2004;271:2056. doi: 10.1111/j.1432-1033.2004.04131.x. [DOI] [PubMed] [Google Scholar]
- 151.Tuddenham L, Wheeler G, Ntounia-Fousara S, et al. The cartilage specific microRNA-140 targets histone deacetylase 4 in mouse cells. FEBS Lett. 2006;580:4214–4217. doi: 10.1016/j.febslet.2006.06.080. [DOI] [PubMed] [Google Scholar]
- 152.Goff LA, Ricupero C, Fenstermacher S, et al. Regulation of microRNA Expression in Human Mesenchymal Stem Cells and their Differentiated Progeny. 2007.
- 153.Ng F, Boucher S, Koh S, et al. Signaling pathways active in mesenchymal stem cells differentiation. Communicated [Google Scholar]
- 154.Cheng Y, Church GM. Biclustering of expression data. Proc Int Conf Intell Syst Mol Biol. 2000;8:93–103. [PubMed] [Google Scholar]
- 155.Wang H, Ach RA, Curry B. Direct and sensitive miRNA profiling from low-input total RNA. Rna. 2007;13:151–159. doi: 10.1261/rna.234507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Davison TS, Johnson CD, Andruss BF. Analyzing micro-RNA expression using microarrays. Methods Enzymol. 2006;411:14–34. doi: 10.1016/S0076-6879(06)11002-2. [DOI] [PubMed] [Google Scholar]
- 157.Castoldi M, Schmidt S, Benes V, et al. A sensitive array for microRNA expression profiling (miChip) based on locked nucleic acids (LNA). Rna. 2006;12:913–920. doi: 10.1261/rna.2332406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Moffat ID, Boutros PC, Celius T, et al. microRNAs in Adult Rodent Liver Are Refractory to Dioxin Treatment. Toxicol Sci. 2007;99:470–487. doi: 10.1093/toxsci/kfm189. [DOI] [PubMed] [Google Scholar]
- 159.Weston MD, Pierce ML, Rocha-Sanchez S, et al. MicroRNA gene expression in the mouse inner ear. Brain research. 2006;1111:95–104. doi: 10.1016/j.brainres.2006.07.006. [DOI] [PubMed] [Google Scholar]
- 160.Baroukh N, Ravier MA, Loder MK, et al. MicroRNA-124a regulates Foxa2 expression and intracellular signaling in pancreatic beta-cell lines. J Biol Chem. 2007;282:19575–19588. doi: 10.1074/jbc.M611841200. [DOI] [PubMed] [Google Scholar]
- 161.Chen XM, Splinter PL, O'Hara SP, et al. A Cellular Micro-RNA, let-7i, Regulates Toll-like Receptor 4 Expression and Contributes to Cholangiocyte Immune Responses against Cryptosporidium parvum Infection. J Biol Chem. 2007;282:28929–28938. doi: 10.1074/jbc.M702633200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Huang J, Wang F, Argyris E, et al. Cellular microRNAs contribute to HIV-1 latency in resting primary CD4(+) T lymphocytes. Nat Med. 2007;13:1241–1247. doi: 10.1038/nm1639. [DOI] [PubMed] [Google Scholar]
- 163.Zhang B, Pan X, Anderson TA. MicroRNA: A new player in stem cells. J Cell Physiol. 2006;209:266–269. doi: 10.1002/jcp.20713. [DOI] [PubMed] [Google Scholar]
- 164.Muljo SA, Kanellopoulou C. Mouse embryonic stem cells as a model genetic system to dissect and exploit the RNA interference machinery. Methods Mol Biol. 2006;342:57–72. doi: 10.1385/1-59745-123-1:57. [DOI] [PubMed] [Google Scholar]
- 165.Ishii H, Saito T. Radiation-induced response of micro RNA expression in murine embryonic stem cells. Med Chem. 2006;2:555–563. doi: 10.2174/1573406410602060555. [DOI] [PubMed] [Google Scholar]
- 166.Krichevsky AM, Sonntag KC, Isacson O, et al. Specific microRNAs modulate embryonic stem cell-derived neurogenesis. Stem Cells. 2006;24:857–864. doi: 10.1634/stemcells.2005-0441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Shcherbata HR, Hatfield S, Ward EJ, et al. The MicroRNA pathway plays a regulatory role in stem cell division. Cell Cycle. 2006;5:172–175. doi: 10.4161/cc.5.2.2343. [DOI] [PubMed] [Google Scholar]
- 168.Lee YS, Kim HK, Chung S, et al. Depletion of human micro-RNA miR-125b reveals that it is critical for the proliferation of differentiated cells but not for the down-regulation of putative targets during differentiation. J Biol Chem. 2005;280:16635–16641. doi: 10.1074/jbc.M412247200. [DOI] [PubMed] [Google Scholar]
- 169.Wulczyn FG, Smirnova L, Rybak A, et al. Post-transcriptional regulation of the let-7 microRNA during neural cell specification. Faseb J. 2007;21:415–426. doi: 10.1096/fj.06-6130com. [DOI] [PubMed] [Google Scholar]
- 170.Lawrie CH. MicroRNAs and haematology: small molecules, big function. Br J Haematol. 2007;137:503–512. doi: 10.1111/j.1365-2141.2007.06611.x. [DOI] [PubMed] [Google Scholar]
- 171.Zhan M, Miller CP, Papayannopoulou T, et al. MicroRNA expression dynamics during murine and human erythroid differentiation. Exp Hematol. 2007;35:1015–1025. doi: 10.1016/j.exphem.2007.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Fatica A, Rosa A, Fazi F, et al. MicroRNAs and hematopoietic differentiation. Cold Spring Harb Symp Quant Biol. 2006;71:205–210. doi: 10.1101/sqb.2006.71.014. [DOI] [PubMed] [Google Scholar]
- 173.Choong ML, Yang HH, McNiece I. MicroRNA expression profiling during human cord blood-derived CD34 cell erythropoiesis. Exp Hematol. 2007;35:551–564. doi: 10.1016/j.exphem.2006.12.002. [DOI] [PubMed] [Google Scholar]
- 174.Fontana L, Pelosi E, Greco P, et al. MicroRNAs 17−5p-20a-106a control monocytopoiesis through AML1 targeting and M-CSF receptor upregulation. Nat Cell Biol. 2007;9:775–787. doi: 10.1038/ncb1613. [DOI] [PubMed] [Google Scholar]
- 175.Felli N, Fontana L, Pelosi E, et al. MicroRNAs 221 and 222 inhibit normal erythropoiesis and erythroleukemic cell growth via kit receptor down-modulation. Proc Natl Acad Sci U S A. 2005;102:18081–18086. doi: 10.1073/pnas.0506216102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Li QJ, Chau J, Ebert PJ, et al. miR-181a is an intrinsic modulator of T cell sensitivity and selection. Cell. 2007;129:147–161. doi: 10.1016/j.cell.2007.03.008. [DOI] [PubMed] [Google Scholar]
- 177.Zhou B, Wang S, Mayr C, et al. miR-150, a microRNA expressed in mature B and T cells, blocks early B cell development when expressed prematurely. Proc Natl Acad Sci U S A. 2007;104:7080–7085. doi: 10.1073/pnas.0702409104. [DOI] [PMC free article] [PubMed] [Google Scholar]
