Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Mar 11.
Published in final edited form as: Curr Opin Mol Ther. 2009 Dec;11(6):641–651.

Novel functions for small RNA molecules

Chunxiang Zhang 1
PMCID: PMC3593927  NIHMSID: NIHMS203150  PMID: 20072941

Abstract

Small RNAs are short (~ 18 to 30 nucleotides), non-coding RNA molecules that can regulate gene expression in both the cytoplasm and the nucleus via post-transcriptional gene silencing (PTGS), chromatin-dependent gene silencing (CDGS) or RNA activation (RNAa). Three classes of small RNAs have been defined, including microRNAs (miRNAs), siRNAs and Piwi-interacting RNAs (piRNAs). Research has indicated that small RNAs play important roles in cellular processes such as cell differentiation, growth/proliferation, migration, apoptosis/death, metabolism and defense. Accordingly, small RNAs are critical regulators of normal development and physiology. More interestingly, increasing evidence indicates that small RNAs are involved in the pathogenesis of diverse diseases including cancer, cardiovascular disease, stroke, neurodegenerative disease, diabetes, liver disease, kidney disease and infectious disease. More than 20 clinical trials are ongoing to evaluate therapies based on small RNA. Additionally, small RNAs may serve as novel biomarkers and therapeutic targets for the majority of diseases.

Keywords: Biomarker, cell biology, disease, microRNA, small RNA, therapy

Introduction

Small RNAs are defined as short (~ 18 to 30 nucleotides [nt]), non-coding RNA molecules that can inhibit the expression of target genes via post-transcriptional gene silencing (PTGS) and chromatin-dependent gene silencing (CDGS), in both the cytoplasm and the nucleus [13]. In addition, more recent studies revealed that some small RNAs can increase the expression of target genes via an RNA activation (RNAa) mechanism [46]. It is generally agreed that there are three main classes of small RNAs: microRNAs (miRNAs), siRNAs and Piwi-interacting RNAs (piRNAs) [1]. Recently, there has been an increase in the number of studies of small RNAs, reflecting the novel association of these molecules with many critical biological functions [7]. A given small RNA sequence is able to regulate the expression of multiple genes because it can bind to target genes as either an imperfect or perfect complement [8] Thus, small RNAs are functionally as important as transcription factors [9]. As a group, small RNAs may directly regulate more than 30% of the genes in a cell [10]. It is not surprising, therefore, that small RNAs are involved in the regulation of all major cellular functions, including cell differentiation, growth/proliferation, migration, apoptosis/death, metabolism and defense [11, 12]. Given these diverse roles, small RNAs could be pivotal regulators in development, physiology and disease [11]. This review article summarizes the novel functions of these small RNA molecules.

Classification, biogenesis and gene-regulatory mechanism of small RNAs

Although many classes of small RNAs have emerged, various characteristics related to the origins, structures, associated effector proteins and biological roles of small RNAs have resulted in the classification of these molecules into three main categories: miRNAs, siRNAs and piRNAs [1]. miRNAs are ssRNA molecules with a length of approximately 21 or 22 nt. miRNAs are initially transcribed in the nucleus to form large pri-miRNA transcripts, which are subsequently processed in the nucleus into approximately 70-nt pre-miRNAs. These pre-miRNAs are then transported into the cytoplasm to become mature miRNAs [11]. The inhibition of the expression of target genes by miRNAs then occurs via PTGS and CDGS in both the cytoplasm and the nucleus [1, 13]. Currently more than 850 mature human miRNA sequences have been identified [14], many of which are highly conserved among many species. siRNAs were discovered from the phenomenon known as RNAi, in which administrated dsRNAs selectively inhibit the expression of target genes with a homologous nt sequence [1]. Because these products of processed dsRNAs were able to efficiently reconstitute silencing complexes, they were named siRNAs [15]. It is well established that siRNAs can be produced from RNA transcribed in the nucleus (endogenous siRNAs), can be virally derived or can be introduced experimentally as chemically synthesized dsRNAs (exogenous siRNAs) [1]. siRNAs have a distinct size distribution that is able to inhibit the expression of genes by PTGS and CDGS [1]. piRNAs are the most recently discovered class of siRNAs [16] and, as their name suggests, are germ-cell-specific small RNAs that bind to the Piwi clade of Argonaute proteins [17]. Piwi is a critical protein for their target gene regulation (17). In addition to the inhibitory effects of piRNAs on their target genes, recent studies indicate that some piRNAs also are able to increase their target gene expression via an RNAa mechanism [46].

Cellular functions of small RNAs

Small RNAs in cell differentiation

During development, the expression of small RNAs occurs in spatiotemporal, tissue- and cell-specific manners, suggesting the involvement of small RNAs in cell differentiation. The first evidence that miRNAs might play a role in the differentiation of embryonic stem (ES) cells came from the identification of changes in the expression of miRNAs during the differentiation of ES cells, in which multiple miRNAs were either downregulated or upregulated [17,18]. The critical role of miRNAs in the differentiation of ES cells was further verified in mice with the genes encoding Dicer (a key enzyme in small RNA biogenesis) inactivated by gene knockout (KO) [18]. In these mice, the inhibition of the effects of miRNA resulted in abnormal differentiation of ES cells and the premature death of embryos [18,19]. More recently, Xu and colleagues reported that the expression of miRNA-145 in EC cells was high during differentiation but low other times [20]. Studies have been conducted to elucidate the roles of miRNAs in adult cell differentiation [2130]. One example is the study of differentiation in cancer cells. Dedifferentiation is an important feature of cancer cells [21], and miRNAs are dysregulated in many cancers [22,23]. The modulation of miRNAs has strong effects on the differentiation of cancer cells [23]. The role of the miRNA-1 (miR-1) in the differentiation of cardiomyocytes was discovered in 2005 [24]. The miR-1 gene was identified as a direct transcriptional target of several regulators of muscle differentiation [24]. The role of miRNAs in the differentiation of adult vascular smooth muscle cells (VSMCs) was investigated in a study by Zhang and colleagues [25]. miR-145 was identified as a critical controller of VSMC differentiation both in rat VSMCs in vitro and in rat carotid arteries in vivo, via its target genes such as Kruppel-like factor 5 (KLF5) [25]. Further studies revealed that miRNAs also play important roles in the differentiation of other cell types [2630], for example: miR-143 in adipocyte differentiation [26]; miR-181 in myoblast differentiation [27]; miR-142s, miR-181 and miR-223 in hematopoietic cell differentiation [28]; miR-25 in airway smooth muscle cell differentiation [29]; and multiple miRNAs in neural cell differentiation [30].

Small RNAs in cell growth/proliferation

During the past 5 years, the biological roles of miRNAs in the growth and proliferation of cancer have been studied. miRNAs were identified as critical regulators of the proliferation of many cancer cells [22,23].

The role of miRNAs in the growth of cardiomyocytes has been documented in recent studies [3133]. The overexpression of miR-21, miR-23a, miR-23b, miR-24, miR-195 or miR-214 via adenovirus-mediated gene transfer induced the hypertrophic growth of cultured cardiomyocytes; whereas the overexpression of miR-150, miR-181b and miR-1 caused a reduction in the size of cardiomyocytes [3133]. These effects of miRNAs on cardiomyocyte growth were confirmed by further studies both in vitro and in vivo [3436].

In recent studies by Zhang and colleagues, it was demonstrated that miR-221, miR-222 and miR-145 play important roles in the proliferation of VSMCs [31,37]. The overexpression of miR-221 and miR-222 increased the proliferation of VSMCs, whereas knockdown of the two miRNAs decreased proliferation [37]. In contrast, VSMC proliferation was significantly inhibited by the overexpression of miR-145 [31]. The effects of miR-221, miR-222 and miR-145 on VSMC proliferation were also demonstrated by two other independent research groups [38,39]. In addition, miR-143 was identified as a regulator of the proliferation of VSMCs by Cordes et al [39].

Small RNAs in cell migration

miRNAs are critical regulators of the migration of cancer cells; for example, miR-23b [40], miR-146b [41] and miR-34a [42,43] are reported to be involved in this process. The effects of miR-221 and miR-222 on the migration of vascular endothelial cells were initially determined by assays of tube formation and wound healing [44]. The results suggest that the influence of miR-221 and miR-222 on the migration of endothelial cells occurs, at least in part, through c-kit [44]. The effects of other miRNAs, such as let-7 [45,46], miR-27b [45,46], miR-126 [47] and miR-210 [48] on human endothelial cell migration have also been demonstrated. For example, Davis et al reported that miR-221 had a pro-migratory effect on VSMCs [38].

Small RNAs in cell death and apoptosis

The role of miRNAs in the apoptosis of cancer cells was described in a review article by Wang and Lee [49]. Multiple miRNAs that are deregulated in cancer cells were found to regulate apoptotic pathways. For example, miR-29b [50], miR-15-16[49], let-7[49], miR-98[49], miR-21[51] and miR-17-92 [52] were involved in regulating the apoptosis of cancer cells [49]. The biological roles of miRNAs in cell apoptosis/death were also demonstrated in other types of cells. [5355]. In studies by Zhang and colleagues, miR-21 was demonstrated to be an important anti-apoptotic miRNA in cardiac cells and in VSMCs both in vitro and in vivo via the regulation of the target genes of miR-21, such as PDCD4 [5355].

Small RNAs in cell metabolism and cell defense

Small RNAs were reported to have roles in cell metabolism, including lipid metabolism [56] and glucose homeostasis [57]. In addition, miRNAs and siRNAs are also involved in cell defensive responses to diverse injuries such as oxidative stress [58], bacterial infections [59] and viral infections [60].

Small RNAs in developmental biology

The roles of small RNAs in development were first demonstrated in Dicer KO mice. These mice did not survive for more than 7.5 days after gastrulation, suggesting a vital role of miRNAs in early development [61,62].

Small RNAs in early embryonic development

miR-15 and miR-16 inhibited Nodal signaling and dorsal mesoderm patterning in the early embryo [63]. Spemann's organizer and head structures were reduced in size by the overexpression of miR-15 and miR-16, but were increased by the inhibition of these miRNAs. miR-430, a highly abundant miRNA that is required for the clearance of maternal mRNAs, was demonstrated to directly decrease the expression of squint mRNA, a member of the Nodal family [64]. Interestingly, lefty mRNA, an antagonist of Nodal, was also downregulated by miR-430. When miR-430 complementary sites of squint were mutated, early embryonic development was disrupted [64].

Small RNAs in cardiac development

The role of miRNAs in cardiac development was demonstrated by investigating the role of miR-1 in this process [24,65]. Cardiomyocytes in miR-1-2 KO mutant mice failed to exit the cell cycle properly, resulting in hyperplasia [65]. These defects resulted in the prenatal or early postnatal death of approximately half of the mutant mice. In addition to miR-21, miR-133a and miR-206 are also involved in cardiac development [66].

Small RNAs in neuronal development

The role of small RNAs in neuronal development was first demonstrated by the requirement for miR-273 in the establishment of left-right asymmetry in ASE neurons in Caenorhabditis elegans [67]. miR-124a, which is specific to neuronal tissue, helps to acquire and maintain the neuronal cellular identity by directly silencing a large number of target mRNAs, including the mRNAs for polypyrimidine tract-binding protein and the RE1-silencing transcription factor [68].

Small RNAs in germline development

Mutations in piRNA-pathway genes including piwi, zucchini and squash resulted in severe defects in oogenesis, including loss of germline stem cells in Drosophila. The mouse genome encodes three Piwi homologs: Miwi, Miwi2 and Mili [69]. Mutations in each of the three genes led to the degeneration of the male germline, suggesting piRNAs might play an important role in the mouse female germline [69].

Small RNAs in disease

Small RNAs in cancer

Cancer is a complex disease that involves a variety of changes in gene expression that result in abnormal cell growth, migration and apoptosis [69]. As these genes and cellular functions are regulated by miRNAs, cancer became the first field of miRNA research [22].

miRNA expression profiles have been generated by microarray analysis in multiple cancer types, including bladder cancer [70], breast cancer [71], chronic lymphocytic leukemia (CLL) [72], colorectal cancer [73,74], gastric cancer [75], glioblastoma [76], hepatocellular carcinoma [77], lung cancer [78], nasopharyngeal carcinoma [79], oral cancer [80], ovarian cancer [81], pancreatic tumor [82], pituitary tumor [83] and prostate cancer [84]. These studies demonstrated that many miRNAs are aberrantly expressed in diverse cancers. The uniqueness of miRNA profiling in particular cancer types may provide important information for cancer diagnosis; different cancers may have different miRNA signatures [7,12]. Further studies demonstrated that many of these aberrantly expressed miRNAs were either tumor suppressors (TS-miRs) or oncogenes (onco-miRs) [22,85,86]. For example, let-7 was reported to be a TS-miR for lung cancer via its target gene RAS [87], and miR-34a was reported to be a TS-miR for many cancers via its multiple target genes such as BCL2 [88]. In contrast, miR-155 [89] and miR-372 [90] were reported to be onco-miRs.

Studies revealed that the some miRNAs are related to tumor invasion and metastasis [91,92]. For example, miR-31 [93], miR-200 [94], miR-193b [95] and miR-23b [40] were reported to be inhibitors of tumor invasion and metastasis. However, tumor invasion and metastasis were promoted by some miRNAs such as miR-373 [40], miR-520c [96] and miR-10b [97]. In addition, studies demonstrated that expression patterns of miRNAs are able to aid in cancer prognosis [98101]. Thus, small RNAs may play important roles in cancer development and progression, as well as prognosis, diagnosis and the evaluation of treatment response [22,23].

Small RNAs in cardiovascular disease

Cardiac hypertrophy and heart failure are the most common pathological responses to several cardiovascular diseases. Cardiac hypertrophy often leads to heart failure, and is a major determinant of mortality and morbidity in cardiovascular diseases. miRNAs are important regulators for the differentiation and growth of cardiac cells, and it is therefore reasonable to hypothesize that miRNAs play important roles in cardiac hypertrophy and heart failure. Four independent research groups almost simultaneously reported results profiling the miRNA expression signature of mouse hearts in which hypertrophy was induced [3134]. The modulation of some of these dysregulated miRNAs had strong effects on cardiac myocyte hypertrophy in vitro [3133]. In vivo, the overexpression of miR-195, an miRNA that was upregulated in hypertrophic hearts, was sufficient to induce cardiac hypertrophy [33], while a gene mutation or 'decoy' approach confirmed the roles of miR-133 [34] and miR-208 [36] in cardiomyocyte hypertrophy. The roles of miR-23a [102] and miR-1 [103] in cardiac hypertrophy were identified in two studies. Furthermore, the role of miRNAs in human cardiac hypertrophy and heart failure was investigated in several clinical studies [33,104,105]. A critical link between miRNAs and heart failure has also been reported [106108].

Cardiovascular diseases are the leading cause of death in developed countries. Several recent studies have suggested that miRNAs might play critical roles in the pathophysiology of acute myocardial infarction (AMI) [109112]. The potential involvement of miRNAs in AMI was suggested in a study in miR-126-null mice that demonstrated the survival rate of miR-126-deficient mice following AMI was significantly lower than of miR-126-expressing mice [110]. van Rooij et al studies the expression signature in late phase AMI and found that miR-29 was critical for cardiac fibrosis during the repair process after AMI [112]. In addition, a study in an in vitro ischemia/reperfusion (I/R) injury model revealed that myocardial infarct size was reduced in mouse hearts pre-injected with heat-shock-induced miRNAs including miR-21 [112]. Moreover, the miRNA expression signature and the role of miR-21 in the early phase of AMI were identified by Zhang and colleagues [113]. In a mouse model, Ren et al found that miR-320 was involved in the regulation of cardiac I/R injury by targeting Hsp20 [114].

The formation of neointimal lesions is a common pathological feature of diverse cardiovascular diseases [53]. Using microarray analysis and a model of neointimal lesion formation, the miRNA expression profile in the vascular wall during neointimal lesion formation was determined [53]. Compared with healthy, uninjured arteries, microarray analysis demonstrated that aberrant miRNA expression was a characteristic of vascular walls after angioplasty [53]. Modulating an aberrantly overexpressed miRNA, miR-21, via antisense-mediated knockdown had a significantly negative effect on neointimal lesion formation in rat arteries after angioplasty [53]. These results indicated that miRNAs might be important regulators in the development of proliferative vascular diseases.

More recently, a series of studies were conducted by different research groups to determine the roles of miR-221 [37,38], miR-222 [37], miR-143 and miR-145 [25,39,113] in proliferative vascular disease. Zhang and colleagues demonstrated that knocking-down miR-221 and miR-222 inhibited neointimal growth in rat carotid arteries after angioplasty by inhibiting the proliferation of VSMCs [37]. The cellular effects of miR-221 were further demonstrated by another study, in which Davis et al demonstrated that miR-221 increased the proliferation of VSMCs and migration through its target gene, p27Kip1 [38]. miR-145 was also identified as the most abundant miRNA in normal arteries, and the expression of this miRNA was significantly downregulated in injured or atherosclerotic vascular walls with neointimal lesion formation [25,39]. Interestingly, both studies identified miR-145 as a critical regulator for the differentiation and proliferation of VSMCs. The restoration of miR-145 in balloon-injured rat carotid arteries significantly inhibited neointimal growth [25]. Moreover, KO of miR-145 and miR-143 was sufficient to elicit neointimal lesion formation in mouse vessels [113].

Small RNAs in stroke and neurodegenerative disease

In a model of middle cerebral artery occlusion (MCAO), two studies revealed the role of miRNAs in the pathogenesis of stroke [115,116]. In the first study, the expression profiles of miRNAs were first identified by Jeyaseelan et al, in which multiple miRNAs were found to be involved in ischemic injury responses in the brain [115]. In the second study, of the 238 miRNAs evaluated, 8 had increased expression and 12 had decreased expression in at least 4 out of 5 reperfusion time points studied between 3 h and 3 days, compared with controls [116]. Bioinformatic analysis indicated a correlation in expression levels between altered miRNA and several mRNAs known to mediate inflammation, transcription, neuroprotection, receptor function and ionic homeostasis [116].

The roles of miRNAs in neurodegenerative disease were described in a review article by Barbato et al [117]. Alzheimer's disease (AD) is the best studied degenerative disease affecting the CNS. Several studies demonstrated that multiple dysregulated miRNAs could be associated with aging and could contribute to the development of AD, including miR-9, miR-128, miR-29, miR-107, let-7, miR-101, miR-15a and miR-106b [117121]. miR-133b was identified as an miRNA expressed in midbrain dopaminergic neurons, but the expression of this miRNA was deficient in midbrain tissue from patients with Parkinson's disease [122]. miR-133b was suggested to regulate the maturation and function of dopaminergic neurons within a negative feedback circuit through suppression of paired-like homeodomain 3 (Pitx3) [123]. In addition, levels of miR-19, miR-101 and miR-130 were related to the severity of neurodegenerative disease because these miRNAs target ataxin-1, which is increased in diseases such as spinocerebellar ataxia type 1 [117].

Small RNAs in diabetes

miRNAs have emerged as novel regulators in both type I and type II diabetes [122]. The first miRNA that was linked to type I diabetes was miR-375 [124126], which was found to be selectively expressed in pancreatic endocrine cell lines [124]. Overexpression of miR-375 resulted in suppressed glucose-stimulated insulin secretion, whereas inhibition of miR-375 enhanced insulin secretion, both via miR-375-mediated suppression of myotrophin and pyruvate dehydrogenase kinase [125,126]. Mice lacking miR-375 were hyperglycemic [126], and exhibited increased total pancreatic α-cell counts, raised levels of fasting and fed plasma glucagon, and increased gluconeogenesis and hepatic glucose output [126]. Pancreatic β-cell mass was reduced as a result of impaired proliferation [126]. More recently, the role of miRNAs in type II diabetes was also reported. For example, the overexpression of miR-125a in insulin target tissues was related to type II diabetes in rats [127].

Small RNAs in liver disease

The expression of miRNAs is dysregulated in many liver diseases such as viral hepatitis, hepatocellular cancer and polycystic liver diseases (for a review, see reference [128]). For example, the expression of miR-1, miR-30, miR-122, miR-128, miR-196, miR-296, miR-351, miR-431 and miR-448 was demonstrated to be related to viral and virus-infected cell functions during viral hepatitis [129131]. The expression of miR-21, miR-224, miR-34a, miR-221, miR-222, miR-106a, miR-203, miR-122, miR-422b, miR-145 and miR-199a was related to hepatocellular cancer [40]. In addition, decreased expression of miR-15a was associated with polycystic liver disease in model cell lines [132].

Small RNAs in kidney diseases

A role for miRNAs in kidney diseases was recently discovered [133135]. Studies of conditional Dicer KO mice revealed critical roles for miRNAs in kidney development, and the maintenance of the structural and functional integrity of the renal collecting system and glomerular barrier [133]. Kidney podocyte-specific deletion of Dicer resulted in proteinuria and severe renal dysfunction in mice [134]. miRNA expression profiles were characterized in normal and diseased kidneys [133], and miR-15a was implicated in pathways linked to cystic kidney disease [133]. In addition, miR-192 and miR-377 promoted matrix deposition [133], while miR-200 and miR-205 were related to epithelial-to-mesenchymal transition [133]. In contrast, miR-21 might have protective roles in kidney disease [135].

Small RNAs in infectious diseases

Viruses encode miRNAs that target their own mRNA or the mRNA of the cells that they infect [136,137]. For example, miRBART2 is encoded by Epstein-Barr virus (EBV) and inhibits EBV DNA polymerase BALF5 [136]. miR-H2-3p, miR-H6, miR-H3 and miR-H4 provided novel candidate mechanisms for the regulation of viral latency and productive replication [36, 38]. simian virus 40 (SV40) -encoded microRNAs were able to regulate viral gene expression and reduce susceptibility to cytotoxic T cells in cultured TC-7/Db cells [139]. miR-UL112, expressed by human CMV, targeted the MHC class I polypeptide-related sequence B and inhibited NK cell killing of virus-infected cells [140]. In an alternative mechanism, viral miRNA targeted the apoptosis pathway to promote the survival of infected cells, which was supported by miR-BART5, expressed by EBV to inhibit the pro-apoptotic protein PUMA [141]. The detailed roles of individual miRNAs in infective diseases should be investigated in future studies. The role of siRNAs in infective diseases is well characterized and is described in a review article by Ge et al [142].

The use of small RNAs in diagnosis

The expression of miRNAs is tissue-specific. Many studies in diseased tissues revealed that different diseases had different miRNA expression signatures. These findings provided an opportunity to use these signatures in the diagnosis of diverse diseases (for reviews of the diagnostic roles of tissue miRNAs, see references [143,144]). Interestingly, studies revealed that miRNAs exist in circulating blood. In contrast to the original hypothesis that miRNAs are only stable within the cells, miRNAs are relatively stable in the blood because of their ability to bind to other materials such as exosomes [145, 146] . Increasing evidence suggests that miRNAs in diseased tissues could be released into circulating blood, which can then be measured and used as a diagnostic biomarker [145146]. Circulating miRNAs have been proven to be biomarkers in cancer [145, 146], stroke [145] and heart disease [147]. For example, serum levels of miR-141 (an miRNA expressed in prostate cancer) were able to distinguish patients with prostate cancer from healthy controls [146], and plasma miR-208 has been demonstrated as a biomarker for myocardial injury [147].

Small RNAs in therapy

miRNAs are aberrantly expressed in many diseases, some being upregulated and others downregulated. Thus, two major miRNA-based therapeutic strategies are restoring the expression of miRNAs reduced in diseases and, conversely, inhibiting overexpressed miRNAs (for a review of the therapeutic use of siRNAs, see reference [148]).

Artificial antisense oligonucleotides that downregulate complementary miRNA sequences are designated as anti-miRs, also known as miRNA inhibitors [148,149]. Many chemical modifications of miRNA inhibitors have been designed to enhance miRNA inhibition, increase stability and tissue uptake. miR-122 expressed in the liver was implicated in cholesterol and lipid metabolism. Locked nucleic acid (LNA)-anti-miR-122 was used to efficiently silence miR-122 in monkeys, and resulted in a long-lasting and reversible decrease in total plasma cholesterol, without evidence of LNA-associated toxicities or histopathological changes [150].

The upregulation of a specific miRNA in vivo in the clinic is another strategy for the treatment of diverse diseases. Although pre-miRNAs and miRNA mimics are successful in upregulating the expression of miRNAs in vitro, the application of these mimics in vivo remains questionable due to their weak tissue uptake and short period of effect. Virus expressing miRNAs such as adenovirus was successfully used to upregulate the expression of miRNAs in animal studies [25,37,113]. Additional approaches to upregulate miRNAs in patients should be investigated.

Although small RNA-based modalities have not been accepted as formal therapeutic medicines, the promising results from animal studies prompted an increasing number of clinical trials of small RNA-based therapeutics for the treatment of many human diseases. There are more than 20 ongoing clinical trials of miRNAs [151153]. Some of the small RNA-based drugs that are currently undergoing clinical trials are listed in Table 1, together with their therapeutic target(s), possible indications and developing companies [151,154164].

Table 1.

Small RNA-based therapeutics in clinical development.

Small RNAs Molecular target Target diseases Highest development status Reference
bevasiranib (OPKO Health Inc) VEGF Wet AMD and diabetic macular edema Phase III [151,154]
oblimersen sodium (Genasense) (Genta Inc) BCL2 B-cell lymphoma, breast tumor, chronic lymphocytic leukemia, colorectal tumor, leukemia, macroglobulinemia, melanoma, multiple myeloma, myeloid leukemia, nasopharyngeal carcinoma, non-Hodgkin's lymphoma, NSCLC and prostate tumor Phase III [156,157]
AGN-211745 (Sirna-027) (Allergan Inc) VEGFR-1 mRNA Wet AMD Phase II [158]
PF-4523655 (RTP801i-14) (Quark Pharmaceuticals Inc/Pfizer Inc) Hypoxia-inducible gene and RTP801 Wet AMD Phase II [159]
ALN-RSV01 (Alnylam Pharmaceuticals Inc/Cubist Pharmaceuticals Inc/ Kyowa Hakko Kirin Co Ltd) Respiratory syncytial virus N gene Respiratory syncytial virus infection Phase II [160]
imetelstat (GRN-163L) (Geron Corp) Telomerase Breast tumor, chronic lymphocytic leukemia, multiple myeloma, ocular disease, NSCLC and solid tumor Phase II [161]
shRNA therapeutic (BLT-HIV) with a recombinant virus vector (rHIV7-shl-TAR-CCR5RZ) (Benitec Ltd) HIV-1 tat and rev shared exons HIV infection and associated lymphoma Phase I [162]
NUCB-1000 (Nucleonics Inc) HBV genome HBV infection Phase I [164]
CALAA-01 (Calando Pharmaceuticals Inc) Ribonucleotide reductase M2 subunit Solid tumor Phase I [164]
TD-101 (TransDerm Inc) Keratins (K6A) Pachyonychia congenital Phase I (ClinicalTrials.gov identifier: NCT00716014)
SPC-3649 (Santaris Pharma A/S) miR-122 HCV infection and hyperlipidemia Phase I [155]

AMD age-related macular degeneration, shRNA short hairpin RNA

The first siRNA-based clinical trial was initiated in 2004 by Acuity Pharmaceuticals Inc [151,154]. The company tested an siRNA, bevasiranib (now being developed by OPKO Health Inc following its merger with Acuity), for the treatment of wet age-related macular degeneration (Table 1). Bevasiranib targets the expression of VEGF. In this clinical trial, an inhibitory effect on the overgrowth of blood vessels behind the retina (the cause of severe and irreversible loss of vision) occurred in patients receiving the siRNA. Bevasiranib, which is in phase III trials for wet age-related macular degeneration, is also undergoing a phase II clinical trial for the treatment of diabetic macular edema [151,154].

The first clinical trial of an anti-infective miRNA is the miR-122 inhibitor (anti-miR-122) SPC-3649 for the treatment of HCV infection, initiated by Santaris Pharma A/S in 2009 [155]. miR-122 is the therapeutic target of this inhibitor because this molecule facilitates the replication of HCV in the liver.

Recently, Genta Inc completed a clinical trial of the RNA-based therapy oblimersen sodium (Genasense), which targets BCL2, in the treatment of relapsed or refractory CLL [156,157] (Table 1). The company had filed an NDA with the FDA after positive clinical findings. The FDA has recommended conducting a confirmatory clinical trial for this potential novel drug [157].

Some disadvantages were identified in the initial clinical trials of small RNA-based therapies, such as off-target effects. However, the interest in these new therapeutic approaches from researchers, clinical doctors and the drug development industry suggests that rapid advances and new applications for small RNA-based therapies should be expected.

Conclusion

In summary, as shown in Figure 1, recent research progress has identified many novel functions of small RNAs in cell biology. Small RNAs have important roles in development and disease via regulation of cell differentiation, growth/proliferation, migration, apoptosis/death, metabolism and defense. Increasing evidence reveals that small RNAs are involved in the pathogenesis of diverse diseases such as cancer, cardiovascular disease, stroke, neurodegenerative disease, diabetes, liver diseases, kidney diseases and infective diseases. Small RNAs may serve as novel biomarkers and therapeutic targets for the majority of diseases. Detailed understanding of target genes of small RNAs and of potential side effects are needed to improve the safety, efficacy and reliability of small RNA-based therapy. Further advanced therapeutic strategies to characterize and modulate the aberrantly expressed miRNAs under different disease conditions should be developed to realize the potential of miRNA-based technology in clinical diagnosis and therapy.

Acknowledgements

The author's research was supported by a National Institutes of Health Grant (HL080133) and a grant from the American Heart Association (09GRNT2250567).

References

• of special interest

  • 1.Farazi TA, Juranek SA, Tuschl T. The growing catalog of small RNAs and their association with distinct Argonaute/Piwi family members. Development. 2008;135(7):1201–1214. doi: 10.1242/dev.005629. [DOI] [PubMed] [Google Scholar]
  • 2.Carthew RW, Sontheimer EJ. Origins and mechanisms of miRNAs and siRNAs. Cell. 2009;136(4):642–655. doi: 10.1016/j.cell.2009.01.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Kim VN, Han J, Siomi MC. Biogenesis of small RNAs in animals. Nat Rev Mol Cell Biol. 2009;10(2):126–139. doi: 10.1038/nrm2632. [DOI] [PubMed] [Google Scholar]
  • 4. Li LC, Okino ST, Zhao H, Pookot D, Place RF, Urakami S, Enokida H, Dahiya R. Small dsRNAs induce transcriptional activation in human cells. Proc Natl Acad Sci USA. 2006;103(46):17337–17342. doi: 10.1073/pnas.0607015103. #x02022; An early report of the activation function of a small RNA on its target gene. The synthetic 21-nt dsRNAs targeting E-cadherin, p21(WAF1/CIP1) (p21) and VEGF increased the expression of the target genes.
  • 5.Pushparaj PN, Aarthi JJ, Kumar SD, Manikandan J. RNAi and RNAa - The Yin and Yang of RNAome. Bioinformation. 2008;2(6):235–237. doi: 10.6026/97320630002235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Garber K. Genetics. Small RNAs reveal an activating side. Science. 2006;314(5800):741–742. doi: 10.1126/science.314.5800.741a. [DOI] [PubMed] [Google Scholar]
  • 7.Sun BK, Tsao H. Small RNAs in development and disease. J Am Acad Dermatol. 2008;59(5):725–737. doi: 10.1016/j.jaad.2008.08.017. [DOI] [PubMed] [Google Scholar]
  • 8.Ha M, Pang M, Agarwal V, Chen ZJ. Interspecies regulation of microRNAs and their targets. Biochim Biophys Acta. 2008;1779(11):735–742. doi: 10.1016/j.bbagrm.2008.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Chen K, Rajewsky N. The evolution of gene regulation by transcription factors and microRNAs. Nat Rev Genet. 2007;8(2):93–103. doi: 10.1038/nrg1990. [DOI] [PubMed] [Google Scholar]
  • 10.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(1):15–20. doi: 10.1016/j.cell.2004.12.035. [DOI] [PubMed] [Google Scholar]
  • 11.Wang Y, Stricker HM, Gou D, Liu L. MicroRNA: past and present. Front Biosci. 2007;12:2316–2329. doi: 10.2741/2234. [DOI] [PubMed] [Google Scholar]
  • 12.Zhang C. MicroRNomics: A newly emerging approach for disease biology. Physiol Genomics. 2008;33(2):139–147. doi: 10.1152/physiolgenomics.00034.2008. [DOI] [PubMed] [Google Scholar]
  • 13.Moazed D. Small RNAs in transcriptional gene silencing and genome defence. Nature. 2009;457(7228):413–420. doi: 10.1038/nature07756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Friedman JM, Jones PA. MicroRNAs: Critical mediators of differentiation, development and disease. Swiss Med Wkly. 2009;139(33–34):466–472. doi: 10.4414/smw.2009.12794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Hamilton AJ, Baulcombe DC. A species of small antisense RNA in posttranscriptional gene silencing in plants. Science. 1999;286(5441):950–952. doi: 10.1126/science.286.5441.950. [DOI] [PubMed] [Google Scholar]
  • 16.Aravin AA, Naumova NM, Tulin AV, Vagin VV, Rozovsky YM, Gvozdev VA. Double-stranded RNA-mediated silencing of genomic tandem repeats and transposable elements in the D. melanogaster germline. Curr Biol. 2001;11(13):1017–1027. doi: 10.1016/s0960-9822(01)00299-8. [DOI] [PubMed] [Google Scholar]
  • 17.Lin H. piRNAs in the germ line. Science. 2007;316(5823):397. doi: 10.1126/science.1137543. [DOI] [PubMed] [Google Scholar]
  • 18.Houbaviy HB, Murray MF, Sharp PA. Embryonic stem cell-specific microRNAs. Dev Cell. 2003;5(2):351–358. doi: 10.1016/s1534-5807(03)00227-2. [DOI] [PubMed] [Google Scholar]
  • 19.Suh MR, Lee Y, Kim JY, Kim SK, Moon SH, Lee JY, Cha KY, Chung HM, Yoon HS, Moon SY, Kim VN, et al. Human embryonic stem cells express a unique set of microRNAs. Dev Biol. 2004;270(2):488–498. doi: 10.1016/j.ydbio.2004.02.019. [DOI] [PubMed] [Google Scholar]
  • 20.Xu N, Papagiannakopoulos T, Pan G, Thomson JA, Kosik KS. MicroRNA-145 regulates OCT4, SOX2, and KLF4 and represses pluripotency in human embryonic stem cells. Cell. 2009;137(4):647–658. doi: 10.1016/j.cell.2009.02.038. [DOI] [PubMed] [Google Scholar]
  • 21.Fusenig NE, Breitkreutz D, Boukamp P, Tomakidi P, Stark HJ. Differentiation and tumor progression. Recent Results Cancer Res. 1995;139:1–19. doi: 10.1007/978-3-642-78771-3_1. [DOI] [PubMed] [Google Scholar]
  • 22.Zhang B, Pan X, Cobb GP, Anderson TA. microRNAs as oncogenes and tumor suppressors. Dev Biol. 2007;302(1):1–12. doi: 10.1016/j.ydbio.2006.08.028. [DOI] [PubMed] [Google Scholar]
  • 23.Mishra PJ, Merlino G. MicroRNA reexpression as differentiation therapy in cancer. J Clin Invest. 2009;119(8):2119–2123. doi: 10.1172/JCI40107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Zhao Y, Samal E, Srivastava D. Serum response factor regulates a muscle-specific microRNA that targets Hand2 during cardiogenesis. Nature. 2005;436(7048):214–220. doi: 10.1038/nature03817. [DOI] [PubMed] [Google Scholar]
  • 25.Cheng Y, Liu X, Yang J, Lin Y, Xu D, Lu Q, Deitch EA, Huo Y, Delphin E, Zhang C. MicroRNA-145, a novel smooth muscle cell phenotypic marker and modulator, controls vascular neointimal lesion formation. Circ Res. 2009;105(5):158–166. doi: 10.1161/CIRCRESAHA.109.197517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Esau C, Kang X, Peralta E, Hanson E, Marcusson EG, Ravichandran LV, Sun Y, Koo S, Perera RJ, Jain R, Dean NM, et al. MicroRNA-143 regulates adipocyte differentiation. J Biol Chem. 2004;279(50):52361–52365. doi: 10.1074/jbc.C400438200. [DOI] [PubMed] [Google Scholar]
  • 27.Naguibneva I, Ameyar-Zazoua M, Polesskaya A, Ait-Si-Ali S, Groisman R, Souidi M, Cuvellier S, Harel-Bellan A. The microRNA miR-181 targets the homeobox protein Hox-A11 during mammalian myoblast differentiation. Nat Cell Biol. 2006;8(3):278–284. doi: 10.1038/ncb1373. [DOI] [PubMed] [Google Scholar]
  • 28.Chen CZ, Li L, Lodish HF, Bartel DP. MicroRNAs modulate hematopoietic lineage differentiation. Science. 2004;303(5654):83–86. doi: 10.1126/science.1091903. [DOI] [PubMed] [Google Scholar]
  • 29.Kuhn AR, Schlauch K, Lao R, Halayko AJ, Gerthoffer WT, Singer CA. MicroRNA expression in human airway smooth muscle cells: Role of miR-25 in regulation of airway smooth muscle phenotype. Am J Respir Cell Mol Biol. 2009 doi: 10.1165/rcmb.2009-0123OC. doi:10.1165/rcmb.2009-0123OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Le MT, Xie H, Zhou B, Chia PH, Rizk P, Um M, Udolph G, Yang H, Lim B, Lodish HF. MicroRNA-125b promotes neuronal differentiation in human cells by repressing multiple targets. Mol Cell Biol. 2009;29(19):5290–5305. doi: 10.1128/MCB.01694-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Cheng Y, Ji R, Yue J, Yang J, Liu X, Chen H, Dean DB, Zhang C. MicroRNAs are aberrantly expressed in hypertrophic heart: Do they play a role in cardiac hypertrophy? Am J Pathol. 2007;170(6):1831–1840. doi: 10.2353/ajpath.2007.061170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Sayed D, Hong C, Chen IY, Lypowy J, Abdellatif M. MicroRNAs play an essential role in the development of cardiac hypertrophy. Circ Res. 2007;100(3):416–424. doi: 10.1161/01.RES.0000257913.42552.23. [DOI] [PubMed] [Google Scholar]
  • 33. Van Rooij E, Sutherland LB, Liu N, Williams AH, McAnally J, Gerard RD, Richardson JA, Olson EN. A signature pattern of stress-responsive microRNAs that can evoke cardiac hypertrophy and heart failure. Proc Natl Acad Sci USA. 2006;103(48):18255–18260. doi: 10.1073/pnas.0608791103. • The first study to demonstrate that miRNAs are critical regulators in heart disease. Revealed that multiple miRNAs were involved in cardiac hypertrophy and heart failure.
  • 34.Carè A, Catalucci D, Felicetti F, Bonci D, Addario A, Gallo P, Bang ML, Segnalini P, Gu Y, Dalton ND, Elia L, et al. MicroRNA-133 controls cardiac hypertrophy. Nat Med. 2007;13(5):613–618. doi: 10.1038/nm1582. [DOI] [PubMed] [Google Scholar]
  • 35.Tatsuguchi M, Seok HY, Callis TE, Thomson JM, Chen JF, Newman M, Rojas M, Hammond SM, Wang DZ. Expression of microRNAs is dynamically regulated during cardiomyocyte hypertrophy. J Mol Cell Cardiol. 2007;42(6):1137–1141. doi: 10.1016/j.yjmcc.2007.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Van Rooij E, Sutherland LB, Qi X, Richardson JA, Hill J, Olson EN. Control of stress-dependent cardiac growth and gene expression by a microRNA. Science. 2007;316(5824):575–579. doi: 10.1126/science.1139089. [DOI] [PubMed] [Google Scholar]
  • 37.Liu X, Cheng Y, Zhang S, Lin Y, Yang J, Zhang C. A necessary role of miR-222 and miR-221 in vascular smooth muscle cell proliferation and neointimal hyperplasia. Circ Res. 2009;104(4):476–487. doi: 10.1161/CIRCRESAHA.108.185363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Davis BN, Hilyard AC, Nguyen PH, Lagna G, Hata A. Induction of microRNA-221 by platelet-derived growth factor signaling is critical for modulation of vascular smooth muscle phenotype. J Biol Chem. 2009;284(6):3728–3738. doi: 10.1074/jbc.M808788200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Cordes KR, Sheehy NT, White MP, Berry EC, Morton SU, Muth AN, Lee TH, Miano JM, Ivey KN, Srivastava D. miR-145 and miR-143 regulate smooth muscle cell fate and plasticity. Nature. 2009;460(7256):705–710. doi: 10.1038/nature08195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Salvi A, Sabelli C, Moncini S, Venturin M, Arici B, Riva P, Portolani N, Giulini SM, De Petro G, Barlati S. MicroRNA-23b mediates urokinase and c-Met downmodulation and a decreased migration of human hepatocellular carcinoma cells. FEBS J. 2009;276(11):2966–2982. doi: 10.1111/j.1742-4658.2009.07014.x. [DOI] [PubMed] [Google Scholar]
  • 41.Xia H, Qi Y, Ng SS, Chen X, Li D, Chen S, Ge R, Jiang S, Li G, Chen Y, He ML, et al. MicroRNA-146b inhibits glioma cell migration and invasion by targeting MMPs. Brain Res. 2009;1269:158–165. doi: 10.1016/j.brainres.2009.02.037. [DOI] [PubMed] [Google Scholar]
  • 42.Li N, Fu H, Tie Y, Hu Z, Kong W, Wu Y, Zheng X. miR-34a inhibits migration and invasion by down-regulation of c-Met expression in human hepatocellular carcinoma cells. Cancer Lett. 2009;275(1):44–53. doi: 10.1016/j.canlet.2008.09.035. [DOI] [PubMed] [Google Scholar]
  • 43.Yan D, Zhou X, Chen X, Hu DN, Dong XD, Wang J, Lu F, Tu L, Qu J. MicroRNA-34a inhibits uveal melanoma cell proliferation and migration through downregulation of c-Met. Invest Ophthalmol Vis Sci. 2009;50(4):1559–1565. doi: 10.1167/iovs.08-2681. [DOI] [PubMed] [Google Scholar]
  • 44.Poliseno L, Tuccoli A, Mariani L, Evangelista M, Citti L, Woods K, Mercatanti A, Hammond S, Rainaldi G. MicroRNAs modulate the angiogenic properties of HUVECs. Blood. 2006;108(9):3068–3071. doi: 10.1182/blood-2006-01-012369. [DOI] [PubMed] [Google Scholar]
  • 45.Kuehbacher A, Urbich C, Zeiher AM, Dimmeler S. Role of Dicer and Drosha for endothelial microRNA expression and angiogenesis. Circ Res. 2007;101(1):59–68. doi: 10.1161/CIRCRESAHA.107.153916. [DOI] [PubMed] [Google Scholar]
  • 46.Suárez Y, Fernández-Hernando C, Pober JS, Sessa WC. Dicer dependent microRNAs regulate gene expression and functions in human endothelial cells. Circ Res. 2007;100(8):1164–1173. doi: 10.1161/01.RES.0000265065.26744.17. [DOI] [PubMed] [Google Scholar]
  • 47.Wang S, Aurora AB, Johnson BA, Qi X, McAnally J, Hill JA, Richardson JA, Bassel-Duby R, Olson EN. The endothelialspecific microRNA miR-126 governs vascular integrity and angiogenesis. Dev Cell. 2008;15(2):261–271. doi: 10.1016/j.devcel.2008.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Fasanaro P, D'Alessandra Y, Di Stefano V, Melchionna R, Romani S, Pompilio G, Capogrossi MC, Martelli F. MicroRNA-210 modulates endothelial cell response to hypoxia and inhibits the receptor tyrosine kinase ligand Ephrin-A3. J Biol Chem. 2008;283(23):15878–15883. doi: 10.1074/jbc.M800731200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Wang Y, Lee CG. MicroRNA and cancer – Focus on apoptosis. J Cell Mol Med. 2009;13(1):12–23. doi: 10.1111/j.1582-4934.2008.00510.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Mott JL, Kobayashi S, Bronk SF, Gores GJ. mir-29 regulates Mcl-1 protein expression and apoptosis. Oncogene. 2007;26(42):6133–6140. doi: 10.1038/sj.onc.1210436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Chan JA, Krichevsky AM, Kosik KS. MicroRNA-21 is an antiapoptotic factor in human glioblastoma cells. Cancer Res. 2005;65(14):6029–6033. doi: 10.1158/0008-5472.CAN-05-0137. [DOI] [PubMed] [Google Scholar]
  • 52.Nagel S, Venturini L, Przybylski GK, Grabarczyk P, Schmidt CA, Meyer C, Drexler HG, Macleod RA, Scherr M. Activation of miR-17-92 by NK-like homeodomain proteins suppresses apoptosis via reduction of E2F1 in T-cell acute lymphoblastic leukemia. Leuk Lymphoma. 2009;50(1):101–108. doi: 10.1080/10428190802626632. [DOI] [PubMed] [Google Scholar]
  • 53. Ji R, Cheng Y, Yue J, Yang J, Liu X, Chen H, Dean DB, Zhang C. MicroRNA expression signature and antisensemediated depletion reveal an essential role of microRNA in vascular neointimal lesion formation. Circ Res. 2007;100(11):1579–1588. doi: 10.1161/CIRCRESAHA.106.141986. • Determined the potential involvement of miRNAs in vascular diseases. Aberrant miRNA expression was a characteristic of vascular walls with neointimal formation. Inhibition of an aberrantly overexpressed miRNA, miR-21, had a significantly negative effect on neointimal growth.
  • 54.Lin Y, Liu X, Cheng Y, Yang J, Huo Y, Zhang C. Involvement of microRNAs in hydrogen peroxide-mediated gene regulation and cellular injury response in vascular smooth muscle cells. J Biol Chem. 2009;284(12):7903–7913. doi: 10.1074/jbc.M806920200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Cheng Y, Liu X, Zhang S, Lin Y, Yang J, Zhang C. MicroRNA-21 protects against the H2O2-induced injury on cardiac myocytes via its target gene PDCD4. J Mol Cell Cardiol. 2009;47(1):5–14. doi: 10.1016/j.yjmcc.2009.01.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Xie H, Sun L, Lodish HF. Targeting microRNAs in obesity. Expert Opin Ther Targets. 2009;13(10):1227–1238. doi: 10.1517/14728220903190707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Poy MN, Spranger M, Stoffel M. microRNAs and the regulation of glucose and lipid metabolism. Diabetes Obes Metab. 2007;9(Suppl 2):S67–S73. doi: 10.1111/j.1463-1326.2007.00775.x. [DOI] [PubMed] [Google Scholar]
  • 58.Simone NL, Soule BP, Ly D, Saleh AD, Savage JE, Degraff W, Cook J, Harris CC, Gius D, Mitchell JB. Ionizing radiation-induced oxidative stress alters miRNA expression. PLoS One. 2009;4(7):e6377. doi: 10.1371/journal.pone.0006377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Jin H. Endogenous small RNAs and antibacterial immunity in plants. FEBS Lett. 2008;582(18):2679–2684. doi: 10.1016/j.febslet.2008.06.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Pfeffer S. Micro RNA and viral infections in mammals. J Soc Biol. 2007;201(4):377–384. doi: 10.1051/jbio:2007908. [DOI] [PubMed] [Google Scholar]
  • 61. Bernstein E, Kim SY, Carmell MA, Murchison EP, Alcorn H, Li MZ, Mills AA, Elledge SJ, Anderson KV, Hannon GJ. Dicer is essential for mouse development. Nat Genet. 2003;35(3):215–217. doi: 10.1038/ng1253. • The roles of small RNAs in development were first demonstrated in mice that were Dicer-deficient (a key enzyme in small RNA biogenesis). These mice did not survive, suggesting the vital role of miRNAs in early development.
  • 62.Murchison EP, Partridge JF, Tam OH, Cheloufi S, Hannon GJ. Characterization of Dicer-deficient murine embryonic stem cells. Proc Natl Acad Sci USA. 2005;102(34):12135–12140. doi: 10.1073/pnas.0505479102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Martello G, Zacchigna L, Inui M, Montagner M, Adorno M, Mamidi A, Morsut L, Soligo S, Tran U, Dupont S, Cordenonsi M, et al. MicroRNA control of Nodal signalling. Nature. 2007;449(7159):183–188. doi: 10.1038/nature06100. [DOI] [PubMed] [Google Scholar]
  • 64.Choi WY, Giraldez AJ, Schier AF. Target protectors reveal dampening and balancing of nodal agonist and antagonist by miR-430. Science. 2007;318(5848):271–274. doi: 10.1126/science.1147535. [DOI] [PubMed] [Google Scholar]
  • 65.Zhao Y, Ransom JF, Li A, Vedantham V, von Drehle M, Muth AN, Tsuchihashi T, McManus MT, Schwartz RJ, Srivastava D. Dysregulation of cardiogenesis, cardiac conduction, and cell cycle in mice lacking miR-1-2. Cell. 2007;129(2):303–317. doi: 10.1016/j.cell.2007.03.030. [DOI] [PubMed] [Google Scholar]
  • 66.Cordes KR, Srivastava D. MicroRNA regulation of cardiovascular development. Circ Res. 2009;104(6):724–732. doi: 10.1161/CIRCRESAHA.108.192872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Johnston RJ, Hobert O. A microRNA controlling left/right neuronal asymmetry in Caenorhabditis elegans. Nature. 2003;426(6968):845–849. doi: 10.1038/nature02255. [DOI] [PubMed] [Google Scholar]
  • 68.Ko HY, Lee DS, Kim S. Noninvasive imaging of microRNA124a-mediated repression of the chromosome 14 ORF 24 gene during neurogenesis. FEBS J. 2009;276(17):4854–4865. doi: 10.1111/j.1742-4658.2009.07185.x. [DOI] [PubMed] [Google Scholar]
  • 69.Blagosklonny MV. Molecular theory of cancer. Cancer Biol Ther. 2005;4(6):621–627. doi: 10.4161/cbt.4.6.1818. [DOI] [PubMed] [Google Scholar]
  • 70.Gottardo F, Liu CG, Ferracin M, Calin GA, Fassan M, Bassi P, Sevignani C, Byrne D, Negrini M, Pagano F, Gomella LG, et al. Micro-RNA profiling in kidney and bladder cancers. Urol Oncol. 2007;25(5):387–392. doi: 10.1016/j.urolonc.2007.01.019. [DOI] [PubMed] [Google Scholar]
  • 71.Iorio MV, Ferracin M, Liu CG, Veronese A, Spizzo R, Sabbioni S, Magri E, Pedriali M, Fabbri M, Campiglio M, Ménard S, et al. MicroRNA gene expression deregulation in human breast cancer. Cancer Res. 2005;65(16):7065–7070. doi: 10.1158/0008-5472.CAN-05-1783. [DOI] [PubMed] [Google Scholar]
  • 72. Calin GA, Liu CG, Sevignani C, Ferracin M, Felli N, Dumitru CD, Shimizu M, Cimmino A, Zupo S, Dono M, Dell'Aquila ML, et al. MicroRNA profiling reveals distinct signatures in B cell chronic lymphocytic leukemias. Proc Natl Acad Sci USA. 2004;101(32):11755–11760. doi: 10.1073/pnas.0404432101. • An early study demonstrating that miRNAs are involved in human cancer using the genome-wide approach of expression profiling.
  • 73.Schetter AJ, Leung SY, Sohn JJ, Zanetti KA, Bowman ED, Yanaihara N, Yuen ST, Chan TL, Kwong DL, Au GK, Liu CG, et al. MicroRNA expression profiles associated with prognosis and therapeutic outcome in colon adenocarcinoma. JAMA. 2008;299(4):425–436. doi: 10.1001/jama.299.4.425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Motoyama K, Inoue H, Takatsuno Y, Tanaka F, Mimori K, Uetake H, Sugihara K, Mori M. Over- and under-expressed microRNAs in human colorectal cancer. Int J Oncol. 2009;34(4):1069–1075. doi: 10.3892/ijo_00000233. [DOI] [PubMed] [Google Scholar]
  • 75.Xia L, Zhang D, Du R, Pan Y, Zhao L, Sun S, Hong L, Liu J, Fan D. miR-15b and miR-16 modulate multidrug resistance by targeting BCL2 in human gastric cancer cells. Int J Cancer. 2008;123(2):372–379. doi: 10.1002/ijc.23501. [DOI] [PubMed] [Google Scholar]
  • 76.Ciafrè SA, Galardi S, Mangiola A, Ferracin M, Liu CG, Sabatino G, Negrini M, Maira G, Croce CM, Farace MG. Extensive modulation of a set of microRNAs in primary glioblastoma. Biochem Biophys Res Commun. 2005;334(4):1351–1358. doi: 10.1016/j.bbrc.2005.07.030. [DOI] [PubMed] [Google Scholar]
  • 77.Murakami Y, Yasuda T, Saigo K, Urashima T, Toyoda H, Okanoue T, Shimotohno K. Comprehensive analysis of microRNA expression patterns in hepatocellular carcinoma and non-tumorous tissues. Oncogene. 2006;25(17):2537–2545. doi: 10.1038/sj.onc.1209283. [DOI] [PubMed] [Google Scholar]
  • 78.Yanaihara N, Caplen N, Bowman E, Seike M, Kumamoto K, Yi M, Stephens RM, Okamoto A, Yokota J, Tanaka T, Calin GA, et al. Unique microRNA molecular profiles in lung cancer diagnosis and prognosis. Cancer Cell. 2006;9(3):189–198. doi: 10.1016/j.ccr.2006.01.025. [DOI] [PubMed] [Google Scholar]
  • 79.Sengupta S, den Boon JA, Chen IH, Newton MA, Stanhope SA, Cheng YJ, Chen CJ, Hildesheim A, Sugden B, Ahlquist P. MicroRNA 29c is down-regulated in nasopharyngeal carcinomas, up-regulating mRNAs encoding extracellular matrix proteins. Proc Natl Acad Sci USA. 2008;105(15):5874–5878. doi: 10.1073/pnas.0801130105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Yu T, Wang XY, Gong RG, Li A, Yang S, Cao YT, Wen YM, Wang CM, Yi XZ. The expression profile of microRNAs in a model of 7,12-dimethyl-benz[a]anthrance-induced oral carcinogenesis in Syrian hamster. J Exp Clin Cancer Res. 2009;28:64. doi: 10.1186/1756-9966-28-64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Pan Q, Luo X, Chegini N. Differential expression of microRNAs in myometrium and leiomyomas and regulation by ovarian steroids. J Cell Mol Med. 2008;12(1):227–240. doi: 10.1111/j.1582-4934.2007.00207.x. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 82.Roldo C, Missiaglia E, Hagan JP, Falconi M, Capelli P, Bersani S, Calin GA, Volinia S, Liu CG, Scarpa A, Croce CM. MicroRNA expression abnormalities in pancreatic endocrine and acinar tumors are associated with distinctive pathologic features and clinical behavior. J Clin Oncol. 2006;24(29):4677–4684. doi: 10.1200/JCO.2005.05.5194. [DOI] [PubMed] [Google Scholar]
  • 83.Bottoni A, Zatelli MC, Ferracin M, Tagliati F, Piccin D, Vignali C, Calin GA, Negrini M, Croce CM, Degli Uberti EC. Identification of differentially expressed microRNAs by microarray: A possible role for microRNA genes in pituitary adenomas. J Cell Physiol. 2007;210(2):370–377. doi: 10.1002/jcp.20832. [DOI] [PubMed] [Google Scholar]
  • 84.Mattie MD, Benz CC, Bowers J, Sensinger K, Wong L, Scott GK, Fedele V, Ginzinger D, Getts R, Haqq C. Optimized high-throughput microRNA expression profiling provides novel biomarker assessment of clinical prostate and breast cancer biopsies. Mol Cancer. 2006;5:24. doi: 10.1186/1476-4598-5-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Esquela-Kerscher A, Slack FJ. Oncomirs – microRNAs with a role in cancer. Nat Rev Cancer. 2006;6(4):259–269. doi: 10.1038/nrc1840. [DOI] [PubMed] [Google Scholar]
  • 86.Mocellin S, Pasquali S, Pilati P. Oncomirs: From tumor biology to molecularly targeted anticancer strategies. Mini Rev Med Chem. 2009;9(1):70–80. doi: 10.2174/138955709787001802. [DOI] [PubMed] [Google Scholar]
  • 87.Johnson SM, Grosshans H, Shingara J, Byrom M, Jarvis R, Cheng A, Labourier E, Reinert KL, Brown D, Slack FJ. RAS is regulated by the let-7 microRNA family. Cell. 2005;120(5):635–647. doi: 10.1016/j.cell.2005.01.014. [DOI] [PubMed] [Google Scholar]
  • 88.Bommer GT, Gerin I, Feng Y, Kaczorowski AJ, Kuick R, Love RE, Zhai Y, Giordano TJ, Qin ZS, Moore BB, MacDougald OA, et al. p53-mediated activation of miRNA34 candidate tumor-suppressor genes. Curr Biol. 2007;17(15):1298–1307. doi: 10.1016/j.cub.2007.06.068. [DOI] [PubMed] [Google Scholar]
  • 89.Tam W, Dahlberg JE. miR-155/BIC as an oncogenic microRNA. Genes Chromosomes Cancer. 2006;45(2):211–212. doi: 10.1002/gcc.20282. [DOI] [PubMed] [Google Scholar]
  • 90.Voorhoeve PM, le Sage C, Schrier M, Gillis AJ, Stoop H, Nagel R, Liu YP, van Duijse J, Drost J, Griekspoor A, Zlotorynski Ee, et al. A genetic screen implicates miRNA-372 and miRNA-373 as oncogenes in testicular germ cell tumors. Cell. 2006;124(6):1169–1181. doi: 10.1016/j.cell.2006.02.037. [DOI] [PubMed] [Google Scholar]
  • 91.Bracken CP, Gregory PA, Khew-Goodall Y, Goodall GJ. The role of microRNAs in metastasis and epithelial-mesenchymal transition. Cell Mol Life Sci. 2009;66(10):1682–1699. doi: 10.1007/s00018-009-8750-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Edmonds MD, Hurst DR, Vaidya KS, Stafford LJ, Chen D, Welch DR. Breast cancer metastasis suppressor 1 coordinately regulates metastasis-associated microRNA expression. Int J Cancer. 2009;125(8):1778–1785. doi: 10.1002/ijc.24616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Valastyan S, Reinhardt F, Benaich N, Calogrias D, Szász AM, Wang ZC, Brock JE, Richardson AL, Weinberg RA. A pleiotropically acting microRNA, miR-31, inhibits breast cancer metastasis. Cell. 2009;137(6):1032–1046. doi: 10.1016/j.cell.2009.03.047. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 94.Dykxhoorn DM, Wu Y, Xie H, Yu F, Lal A, Petrocca F, Martinvalet D, Song E, Lim B, Lieberman J. miR-200 enhances mouse breast cancer cell colonization to form distant metastases. PLoS One. 2009;4(9):e7181. doi: 10.1371/journal.pone.0007181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Li XF, Yan PJ, Shao ZM. Downregulation of miR-193b contributes to enhance urokinase-type plasminogen activator (uPA) expression and tumor progression and invasion in human breast cancer. Oncogene. 2009;28(44):3937–3948. doi: 10.1038/onc.2009.245. [DOI] [PubMed] [Google Scholar]
  • 96.Huang Q, Gumireddy K, Schrier M, le Sage C, Nagel R, Nair S, Egan DA, Li A, Huang G, Klein-Szanto AJ, Gimotty PA, et al. The microRNAs miR-373 and miR-520c promote tumour invasion and metastasis. Nat Cell Biol. 2008;10(2):202–210. doi: 10.1038/ncb1681. [DOI] [PubMed] [Google Scholar]
  • 97.Ma L, Teruya-Feldstein J, Weinberg RA. Tumour invasion and metastasis initiated by microRNA-10b in breast cancer. Nature. 2007;449(7163):682–688. doi: 10.1038/nature06174. [DOI] [PubMed] [Google Scholar]
  • 98.Mathé EA, Nguyen GH, Bowman ED, Zhao Y, Budhu A, Schetter AJ, Braun R, Reimers M, Kumamoto K, Hughes D, Altorki NK, et al. MicroRNA expression in squamous cell carcinoma and adenocarcinoma of the esophagus: Associations with survival. Clin Cancer Res. 2009;15(19):6192–6200. doi: 10.1158/1078-0432.CCR-09-1467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Schetter AJ, Nguyen GH, Bowman ED, Mathé EA, Yuen ST, Hawkes JE, Croce CM, Leung SY, Harris CC. Association of inflammation-related and microRNA gene expression with cancer-specific mortality of colon adenocarcinoma. Clin Cancer Res. 2009;15(18):5878–5887. doi: 10.1158/1078-0432.CCR-09-0627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Raponi M, Dossey L, Jatkoe T, Wu X, Chen G, Fan H, Beer DG. MicroRNA classifiers for predicting prognosis of squamous cell lung cancer. Cancer Res. 2009;69(14):5776–5783. doi: 10.1158/0008-5472.CAN-09-0587. [DOI] [PubMed] [Google Scholar]
  • 101.Wang QZ, Xu W, Habib N, Xu R. Potential uses of microRNA in lung cancer diagnosis, prognosis, and therapy. Curr Cancer Drug Targets. 2009;9(4):572–594. doi: 10.2174/156800909788486731. [DOI] [PubMed] [Google Scholar]
  • 102.Lin Z, Murtaza I, Wang K, Jiao J, Gao J, Li PF. miR-23a functions downstream of NFATc3 to regulate cardiac hypertrophy. Proc Natl Acad Sci USA. 2009;106(29):12103–12108. doi: 10.1073/pnas.0811371106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Ikeda S, He A, Kong SW, Lu J, Bejar R, Bodyak N, Lee KH, Ma Q, Kang PM, Golub TR, Pu WT. MicroRNA-1 negatively regulates expression of the hypertrophy-associated calmodulin and Mef2a genes. Mol Cell Biol. 2009;29(8):2193–2204. doi: 10.1128/MCB.01222-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Yang B, Lin H, Xiao J, Lu Y, Luo X, Li B, Zhang Y, Xu C, Bai Y, Wang H, Chen G, et al. The muscle-specific microRNA miR-1 regulates cardiac arrhythmogenic potential by targeting GJA1 and KCNJ2. Nat Med. 2007;13(4):486–491. doi: 10.1038/nm1569. [DOI] [PubMed] [Google Scholar]
  • 105.Mann DL. MicroRNAs and the failing heart. N Engl J Med. 2007;356(25):2644–2645. doi: 10.1056/NEJMcibr072068. [DOI] [PubMed] [Google Scholar]
  • 106.Naga Prasad SV, Duan ZH, Gupta MK, Surampudi VS, Volinia S, Calin GA, Liu CG, Kotwal A, Moravec CS, Starling RC, Perez DM, et al. A unique microRNA profile in end-stage heart failure indicates alterations in specific cardiovascular signaling networks. J Biol Chem. 2009;284(40):27487–27499. doi: 10.1074/jbc.M109.036541. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 107.Wang N, Zhou Z, Liao X, Zhang T. Role of microRNAs in cardiac hypertrophy and heart failure. IUBMB Life. 2009;61(6):566–571. doi: 10.1002/iub.204. [DOI] [PubMed] [Google Scholar]
  • 108.Sucharov C, Bristow MR, Port JD. miRNA expression in the failing human heart: Functional correlates. J Mol Cell Cardiol. 2008;45(2):185–192. doi: 10.1016/j.yjmcc.2008.04.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Ikeda S, Kong SW, Lu J, Bisping E, Zhang H, Allen PD, Golub TR, Pieske B, Pu WT. Altered microRNA expression in human heart disease. Physiol Genomics. 2007;31(3):367–373. doi: 10.1152/physiolgenomics.00144.2007. [DOI] [PubMed] [Google Scholar]
  • 110.Wang S, Aurora AB, Johnson BA, Qi X, McAnally J, Hill JA, Richardson JA, Bassel-Duby R, Olson EN. The endothelial-specific microRNA miR-126 governs vascular integrity and angiogenesis. Dev Cell. 2008;15(2):261–271. doi: 10.1016/j.devcel.2008.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.van Rooij E, Sutherland LB, Thatcher JE, DiMaio JM, Naseem RH, Marshall WS, Hill JA, Olson EN. Dysregulation of microRNAs after myocardial infarction reveals a role of miR-29 in cardiac fibrosis. Proc Natl Acad Sci USA. 2008;105(35):13027–13032. doi: 10.1073/pnas.0805038105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Yin C, Wang X, Kukreja RC. Endogenous microRNAs induced by heat-shock reduce myocardial infarction following ischemia-reperfusion in mice. FEBS Lett. 2008;582(30):4137–4142. doi: 10.1016/j.febslet.2008.11.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Dong S, Cheng Y, Yang J, Li J, Liu X, Wang X, Wang D, Krall TJ, Delphin ES, Zhang C. MicroRNA expression signature and the role of microRNA-21 in the early phase of acute myocardial infarction. J Biol Chem. 2009;284(43):29514–29525. doi: 10.1074/jbc.M109.027896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Ren XP, Wu J, Wang X, Sartor MA, Qian J, Jones K, Nicolaou P, Pritchard TJ, Fan GC. MicroRNA-320 is involved in the regulation of cardiac ischemia/reperfusion injury by targeting heat-shock protein 20. Circulation. 2009;119(17):2357–2366. doi: 10.1161/CIRCULATIONAHA.108.814145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Jeyaseelan K, Lim KY, Armugam A. MicroRNA expression in the blood and brain of rats subjected to transient focal ischemia by middle cerebral artery occlusion. Stroke. 2008;39(3):959–966. doi: 10.1161/STROKEAHA.107.500736. [DOI] [PubMed] [Google Scholar]
  • 116.Dharap A, Bowen K, Place R, Li LC, Vemuganti R. Transient focal ischemia induces extensive temporal changes in rat cerebral microRNAome. J Cereb Blood Flow Metab. 2009;29(4):675–687. doi: 10.1038/jcbfm.2008.157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Barbato C, Ruberti F, Cogoni C. Searching for MIND: MicroRNAs in neurodegenerative diseases. J Biomed Biotechnol. 2009;2009:871313. doi: 10.1155/2009/871313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Lukiw WJ. Micro-RNA speciation in fetal, adult and Alzheimer's disease hippocampus. Neuroreport. 2007;18(3):297–300. doi: 10.1097/WNR.0b013e3280148e8b. [DOI] [PubMed] [Google Scholar]
  • 119.Hébert SS, Horré K, Nicolaï L, Papadopoulou AS, Mandemakers W, Silahtaroglu AN, Kauppinen S, Delacourte A, De Strooper B. Loss of microRNA cluster miR-29a/b-1 in sporadic Alzheimer's disease correlates with increased BACE1/β-secretase expression. Proc Natl Acad Sci USA. 2008;105(17):6415–6420. doi: 10.1073/pnas.0710263105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Cogswell JP, Ward J, Taylor IA, Waters M, Shi Y, Cannon B, Kelnar K, Kemppainen J, Brown D, Chen C, Prinjha RK, et al. Identification of miRNA changes in Alzheimer's disease brain and CSF yields putative biomarkers and insights into disease pathways. J Alzheimers Dis. 2008;14(1):27–41. doi: 10.3233/jad-2008-14103. [DOI] [PubMed] [Google Scholar]
  • 121.Wang WX, Rajeev BW, Stromberg AJ, Ren N, Tang G, Huang Q, Rigoutsos I, Nelson PT. The expression of microRNA miR-107 decreases early in Alzheimer's disease and may accelerate disease progression through regulation of β-site amyloid precursor protein-cleaving enzyme 1. J Neurosci. 2008;28(5):1213–1223. doi: 10.1523/JNEUROSCI.5065-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Kim J, Inoue K, Ishii J, Vanti WB, Voronov SV, Murchison E, Hannon G, Abeliovich A. A microRNA feedback circuit in midbrain dopamine neurons. Science. 2007;317(5842):1220–1224. doi: 10.1126/science.1140481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Pandey AK, Agarwal P, Kaur K, Datta M. MicroRNAs in diabetes: Tiny players in big disease. Cell Physiol Biochem. 2009;23(4–6):221–232. doi: 10.1159/000218169. [DOI] [PubMed] [Google Scholar]
  • 124.Poy MN, Eliasson L, Krutzfeldt J, Kuwajima S, Ma X, Macdonald PE, Pfeffer S, Tuschl T, Rajewsky N, Rorsman P, Stoffel M. A pancreatic islet-specific microRNA regulates insulin secretion. Nature. 2004;432(7014):226–230. doi: 10.1038/nature03076. [DOI] [PubMed] [Google Scholar]
  • 125.El Ouaamari A, Baroukh N, Martens GA, Lebrun P, Pipeleers D, van Obberghen E. miR-375 targets 3'-phosphoinositide-dependent protein kinase-1 and regulates glucose-induced biological responses in pancreatic β-cells. Diabetes. 2008;57(10):2708–2717. doi: 10.2337/db07-1614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Poy MN, Hausser J, Trajkovski M, Braun M, Collins S, Rorsman P, Zavolan M, Stoffel M. miR-375 maintains normal pancreatic α- and β-cell mass. Proc Natl Acad Sci USA. 2009;106(14):5813–5818. doi: 10.1073/pnas.0810550106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Herrera BM, Lockstone HE, Taylor JM, Wills QF, Kaisaki PJ, Barrett A, Camps C, Fernandez C, Ragoussis J, Gauguier D, McCarthy MI, et al. MicroRNA-125a is over-expressed in insulin target tissues in a spontaneous rat model of type 2 diabetes. BMC Med Genomics. 2009;2:54. doi: 10.1186/1755-8794-2-54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Chen XM. MicroRNA signatures in liver diseases. World J Gastroenterol. 2009;15(14):1665–1672. doi: 10.3748/wjg.15.1665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Jopling CL, Yi M, Lancaster AM, Lemon SM, Sarnow P. Modulation of hepatitis C virus RNA abundance by a liver-specific microRNA. Science. 2005;309(5740):1577–1581. doi: 10.1126/science.1113329. [DOI] [PubMed] [Google Scholar]
  • 130.Randall G, Panis M, Cooper JD, Tellinghuisen TL, Sukhodolets KE, Pfeffer S, Landthaler M, Landgraf P, Kan S, Lindenbach BD, Chien M, et al. Cellular cofactors affecting hepatitis C virus infection and replication. Proc Natl Acad Sci USA. 2007;104(31):12884–12889. doi: 10.1073/pnas.0704894104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Pedersen IM, Cheng G, Wieland S, Volinia S, Croce CM, Chisari FV, David M. Interferon modulation of cellular microRNAs as an antiviral mechanism. Nature. 2007;449(7164):919–922. doi: 10.1038/nature06205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Lee SO, Masyuk T, Splinter P, Banales JM, Masyuk A, Stroope A, Larusso N. MicroRNA15a modulates expression of the cell-cycle regulator Cdc25A and affects hepatic cystogenesis in a rat model of polycystic kidney disease. J Clin Invest. 2008;118(11):3714–3724. doi: 10.1172/JCI34922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Saal S, Harvey SJ. MicroRNAs and the kidney: Coming of age. Curr Opin Nephrol Hypertens. 2009;18(14):317–323. doi: 10.1097/MNH.0b013e32832c9da2. [DOI] [PubMed] [Google Scholar]
  • 134.Harvey SJ, Jarad G, Cunningham J, Goldberg S, Schermer B, Harfe BD, McManus MT, Benzing T, Miner JH. Podocyte-specific deletion of Dicer alters cytoskeletal dynamics and causes glomerular disease. J Am Soc Nephrol. 2008;19(11):2150–2158. doi: 10.1681/ASN.2008020233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Kato M, Arce L, Natarajan R. MicroRNAs and their role in progressive kidney diseases. Clin J Am Soc Nephrol. 2009;4(7):1255–1266. doi: 10.2215/CJN.00520109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Grassmann R, Jeang KT. The roles of microRNAs in mammalian virus infection. Biochim Biophys Acta. 2008;1779(11):706–711. doi: 10.1016/j.bbagrm.2008.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Lu LF, Liston A. MicroRNA in the immune system, microRNA as an immune system. Immunology. 2009;127(3):291–298. doi: 10.1111/j.1365-2567.2009.03092.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Umbach JL, Kramer MF, Jurak I, Karnowski HW, Coen DM, Cullen BR. MicroRNAs expressed by herpes simplex virus 1 during latent infection regulate viral mRNAs. Nature. 2008;454(7205):780–783. doi: 10.1038/nature07103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Sullivan CS, Grundhoff AT, Tevethia S, Pipas JM, Ganem D. SV40-encoded microRNAs regulate viral gene expression and reduce susceptibility to cytotoxic T cells. Nature. 2005;435(7042):682–686. doi: 10.1038/nature03576. [DOI] [PubMed] [Google Scholar]
  • 140.Stern-Ginossar N, Elefant N, Zimmermann A, Wolf DG, Saleh N, Biton M, Horwitz E, Prokocimer Z, Prichard M, Hahn G, Goldman-Wohl D, Greenfield C, Yagel S, Hengel H, Altuvia Y, Margalit H, Mandelboim O. Host immune system gene targeting by a viral miRNA. Science. 2007;317(5836):376–381. doi: 10.1126/science.1140956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Choy EY, Siu KL, Kok KH, Lung RW, Tsang CM, To KF, Kwong DL, Tsao SW, Jin DY. An Epstein-Barr virus-encoded microRNA targets PUMA to promote host cell survival. J Exp Med. 2008;205(11):2551–2560. doi: 10.1084/jem.20072581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Ge Q, Eisen HN, Chen J. Use of siRNAs to prevent and treat influenza virus infection. Virus Res. 2004;102(1):37–42. doi: 10.1016/j.virusres.2004.01.013. [DOI] [PubMed] [Google Scholar]
  • 143.Bartels CL, Tsongalis GJ. MicroRNAs: Novel biomarkers for human cancer. Clin Chem. 2009;55(4):623–631. doi: 10.1373/clinchem.2008.112805. [DOI] [PubMed] [Google Scholar]
  • 144.Waldman SA, Terzic A. A study of microRNAs in silico and in vivo: Diagnostic and therapeutic applications in cancer. FEBS J. 2009;276(8):2157–2164. doi: 10.1111/j.1742-4658.2009.06934.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Cortez MA, Calin GA. MicroRNA identification in plasma and serum: A new tool to diagnose and monitor diseases. Expert Opin Biol Ther. 2009;9(6):703–711. doi: 10.1517/14712590902932889. • Demonstrated that circulating cell-free miRNAs could be used in the diagnosis of diverse diseases.
  • 146.Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, Peterson A, Noteboom J, O'Briant KC, Allen A, Lin DW, et al. Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci USA. 2008;105(30):10513–10518. doi: 10.1073/pnas.0804549105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Ji X, Takahashi R, Hiura Y, Hirokawa G, Fukushima Y, Iwai N. Plasma miR-208 as a biomarker of myocardial injury. Clin Chem. 2009;55(11):1944–1949. doi: 10.1373/clinchem.2009.125310. [DOI] [PubMed] [Google Scholar]
  • 148.Brown BD, Naldini L. Exploiting and antagonizing microRNA regulation for therapeutic and experimental applications. Nat Rev Genet. 2009;10(8):578–585. doi: 10.1038/nrg2628. [DOI] [PubMed] [Google Scholar]
  • 149.Pushparaj PN, Melendez AJ. Short interfering RNA (siRNA) as a novel therapeutic. Clin Exp Pharmacol Physiol. 2006;33(5–6):504–510. doi: 10.1111/j.1440-1681.2006.04399.x. [DOI] [PubMed] [Google Scholar]
  • 150.Elmén J, Lindow M, Schütz S, Lawrence M, Petri A, Obad S, Lindholm M, Hedtjärn M, Hansen HF, Berger U, Gullans S, et al. LNA-mediated microRNA silencing in non-human primates. Nature. 2008;452(7189):896–899. doi: 10.1038/nature06783. [DOI] [PubMed] [Google Scholar]
  • 151.Castanotto D, Rossi JJ. The promises and pitfalls of RNA-interference-based therapeutics. Nature. 2009;457(7228):426–433. doi: 10.1038/nature07758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.de Fougerolles A, Vornlocher HP, Maraganore J, Lieberman J. Interfering with disease: A progress report on siRNA-based therapeutics. Nat Rev Drug Discov. 2007;6(6):443–453. doi: 10.1038/nrd2310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Austin, TX, USA: Mirna Therapeutics Inc; 2009. Clinical trials featuring oligo-based therapeutics. www.mirnatherapeutics.com/technology/clinical_trials.aspx. [Google Scholar]
  • 154.Singerman L. Combination therapy using the small interfering RNA bevasiranib. Retina. 2009;29(6 Suppl):S49–S50. doi: 10.1097/IAE.0b013e3181ad2341. [DOI] [PubMed] [Google Scholar]
  • 155.Hoersholm, Denmark: Santaris Pharma A/S; 2008. Santaris Pharma begins human clinical testing of world's first microRNA. www.santaris.com/NewsReleases/santaris-pharma-begins-human-clinical-testing-of-world-s-first-microrna/Default.aspx. [Google Scholar]
  • 156.Banerjee D. Genasense (Genta Inc) Curr Opin Investig Drugs. 2001;2(4):574–580. [PubMed] [Google Scholar]
  • 157.Genta Inc. FDA appeal decision indicates that Genasense(R) approval in chronic lymphocytic leukemia will require additional confirmatory trial. Press Release. 2009 Mar 09; [Google Scholar]
  • 158.Shen J, Samul R, Silva RL, Akiyama H, Liu H, Saishin Y, Hackett SF, Zinnen S, Kossen K, Fosnaugh K, Vargeese C, et al. Suppression of ocular neovascularization with siRNA targeting VEGF receptor 1. Gene Ther. 2006;13(3):225–234. doi: 10.1038/sj.gt.3302641. [DOI] [PubMed] [Google Scholar]
  • 159.2009. Fremont, CA, USA: Quark Pharmaceuticals Inc; PF-4523655. www.quarkpharma.com/qbi-en/products/53. [Google Scholar]
  • 160.DeVincenzo J, Cehelsky JE, Alvarez R, Elbashir S, Harborth J, Toudjarska I, Nechev L, Murugaiah V, Van Vliet A, Vaishnaw AK, Meyers R. Evaluation of the safety, tolerability and pharmacokinetics of ALN-RSV01, a novel RNAi antiviral therapeutic directed against respiratory syncytial virus (RSV) Antiviral Res. 2008;77(3):225–231. doi: 10.1016/j.antiviral.2007.11.009. [DOI] [PubMed] [Google Scholar]
  • 161.Harley CB. Telomerase and cancer therapeutics. Nat Rev Cancer. 2008;8(3):167–179. doi: 10.1038/nrc2275. [DOI] [PubMed] [Google Scholar]
  • 162.Zhou J, Li H, Li S, Zaia J, Rossi JJ. Novel dual inhibitory function aptamer-siRNA delivery system for HIV-1 therapy. Mol Ther. 2008;16(8):1481–1489. doi: 10.1038/mt.2008.92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.RNA-interference therapy for HBV infection enters phase I clinical trial. Expert Rev Anti Infect Ther. 2008;6(1):5–8. No authors listed. [Google Scholar]
  • 164.Davis ME. The first targeted delivery of siRNA in humans via a self-assembling, cyclodextrin polymer-based nanoparticle: From concept to clinic. Mol Pharm. 2009;6(3):659–668. doi: 10.1021/mp900015y. [DOI] [PubMed] [Google Scholar]

RESOURCES