Abstract
MicroRNAs are small noncoding RNAs that can bind to the target sites in the 3’‐untranslated region of messenger RNA to regulate posttranscriptional gene expression. Increasing evidence has identified the miR‐29 family, consisting of miR‐29a, miR‐29b‐1, miR‐29b‐2, and miR‐29c, as key regulators of a number of biological processes. Moreover, their abnormal expression contributes to the etiology of numerous diseases. In the current review, we aimed to summarize the differential expression patterns and functional roles of the miR‐29 family in the etiology of diseases including osteoarthritis, osteoporosis, cardiorenal, and immune disease. Furthermore, we highlight the therapeutic potential of targeting members of miR‐29 family in these diseases. We present miR‐29s as promoters of osteoblast differentiation and apoptosis but suppressors of chondrogenic and osteoclast differentiation, fibrosis, and T cell differentiation, with clear avenues for therapeutic manipulation. Further research will be crucial to identify the precise mechanism of miR‐29 family in these diseases and their full potential in therapeutics.
Keywords: cardiorenal disease, immune disease, microRNA, miRNA‐29, osteoarthritis, osteoporosis
In the current review, we aimed to summarize the differential expression patterns and functional roles of the miR‐29 family in the etiology of diseases including osteoarthritis, osteoporosis, cardiorenal and immune disease. We present miR‐29s as promoters of osteoblast differentiation and apoptosis but suppressors of chondrogenic and osteoclast differentiation, fibrosis, and T cell differentiation.

1. INTRODUCTION
MicroRNAs (miRNAs) are short, endogenous, single‐stranded noncoding RNAs of approximately 22 nucleotides in length, initially discovered in Lin‐4 in the nematode Caenorhabditis elegans. 1 , 2 Since this initial discovery, miRNAs have been identified in plants, viruses, and animals including humans. 3 , 4 , 5
Transcription of miRNA generates a primary miRNA (pri‐miRNA), which is cleaved by the RNase III enzyme Drosha and the DGCR8 microprocessor complex subunit (known as Pasha in the model organisms Drosophila melanogaster and Caenorhabditis elegans). 6 , 7 This precursor (pre‐miRNA) is exported to the cytoplasm 8 before cleavage by the RNase III enzyme Dicer, generating a miRNA duplex containing mature miRNA. 9 The duplex unwinds and the mature miRNA assembles into RNA‐induced silencing complex (RISC). 10 , 11 , 12 One strand of the mature miRNA (the “guide” strand) is loaded into Argonaute 2 (AGO2), whilst the “passenger” strand is degraded. 13 , 14 Mature miRNA guides AGO2 from the RISC complex to target sites in the 3ʹ‐untranslated region (3ʹ‐UTR) of messenger RNA (mRNA), inducing gene silencing (Figure 1A). 15 , 16 , 17
Figure 1.

Maturation and function of microRNA (miRNA) and mature sequences of miR‐29s. (A) miRNA is transcribed by RNA polymerase II to generate a primary miRNA (pri‐miRNA), which is cleaved in Dorsha and DGCR8 to generate the precursor miRNA (pre‐miRNA). Pre‐miRNA is exported to the cytoplasm and then cleaved by Dicer to generate a miRNA duplex containing mature miRNA. The duplex unwinds and the mature miRNA assembles into RISC. Mature miRNA mediates gene silencing by guiding AGO2 proteins to target sites in the 3ʹ‐UTR of mRNA. (B) miR‐29 family members have identical seeding regions (blue box and underlined). Tri‐uracil nucleotide at positions 9‐11 exist in miR‐29b and miR‐29c (red box). Nuclear localization sequence at positions 18‐23 is unique to miR‐29b (green box). 3ʹ‐UTR, 3ʹ‐untranslated region; mRNA, messenger RNA
miRNAs influence essentially all developmental processes and disease because miRNAs have conserved interactions with most human mRNAs. 18 Aberrant levels of miRNA expression are found in many diseases where they adversely regulate posttranscriptional gene expression through transcriptional repression and/or degradation of target mRNAs, altering many cellular processes from proliferation and differentiation to apoptosis. 3 , 19 , 20 , 21 , 22 , 23 Therefore, understanding miRNA expression profiles in diseased tissues can guide diagnosis, prognosis, and prediction of therapeutic response.
The miR‐29 family are among the more commonly implicated miRNAs in disease. The miR‐29 family consist of miR‐29a, miR‐29b‐1, miR‐29b‐2, and miR‐29c, generated from two primary transcripts: pri‐miR‐29a/b1 cluster and pri‐miR‐29b2/c cluster, located on chromosomes 7q32.3 and 1q32.2, respectively, in humans. Although the sequences of pre‐miR‐29b1 and ‐2 are different, mature miR‐29b generated from both are identical. 24 , 25 Whilst miR‐29s are broadly conserved within mammals, miR‐29a is the most abundantly expressed family member. 26 , 27 The miR‐29s are identical at nucleotide position 2‐8, the seeding region that plays a role in recognizing the target mRNA (Figure 1B). 28 Despite similar sequences, the miR‐29s have different subcellular localization with miR‐29a mainly located in the cytoplasm whereas miR‐29b and miR‐29c are concentrated in the nucleus. 28 , 29 miR‐29b has a unique six nucleotide segment, which leads to its nuclear localization. 28 This nuclear localization contributes to chromosomal segregation and nuclear morphology through unconventional mechanisms not traditional to mRNA targeting. 30 Both miR‐29b and miR‐29c have a tri‐uracil residue at positions 9–11, leading to rapid decay or turnover, whereas the cytosine residue at nucleotide position 10 of miR‐29a contributes to its stability. 31
Many miRNAs are abnormally expressed in disorders as diverse as osteoarthritis (OA), osteoporosis, cardiorenal disease, and immune disease. The miR‐29 family however is central to the etiology and pathogenesis of these diseases. 32 , 33 , 34 , 35 , 36 In this review, we will explore the expression, regulation, and function of miR‐29 family members, providing fundamental insight into their critical role in the pathogenesis of several debilitating diseases that are of great public health concern.
2. ROLE OF MIR‐29 FAMILY IN OSTEOARTHRITIS
OA is a degenerative joint disease, mostly of the elderly, involving degradation of articular cartilage, subchondral bone sclerosis, chondro‐osteophyte formation, and inflammation of the joint. 37 The etiology of OA is not fully recognized and there are currently no effective treatments for OA aside form pain control, physiotherapy and finally, prosthetic joint replacement in the most severe cases. 38
Articular chondrocytes are generated through the process of chondrogenesis, which begins with the condensation of mesenchymal stem cells (MSCs). 39 MSC‐directed chondrogenic differentiation is an attractive OA treatment target for cartilage repair and regeneration, and Dicer, essential in generation of mature miRNAs, has a critical role in controlling chondrocyte cell proliferation and differentiation. 40 , 41 , 42 miR‐29s are critical for controlling chondrogenic differentiation of MSCs, the downregulation of miR‐29a/b expression in mature chondrocytes compared to MSCs, suggests that high levels of expression may impair development of the mature chondrocyte phenotype. 32 , 43 , 44 , 45 Indeed, miR‐29a overexpression inhibits the expression of chondrocyte‐specific markers, such as Type II collagen and aggrecan, during chondrogenic differentiation, whereas decreased expression of miR‐29a/b, which directly targets the 3ʹ‐UTR of Col2a1, is necessary for human chondrogenic differentiation. 44 , 45 Downregulation of miR‐29a/b is controlled by the transcription factor, Sry‐related box 9 (Sox9) in both human and murine cells. Furthermore, increased Sox9 expression is essential for the formation of MSC condensations. 32 , 44 , 46 , 47 , 48
Transforming growth factor‐β (TGF‐β) signaling is also critical for the initiation of chondrogenesis. 49 , 50 Phosphorylation of Smad2 and Smad3 by TGF‐β leads them to form a heteromeric complex in association with Smad4. 51 , 52 These Smad complexes move into the nucleus and inhibit hypertrophic chondrocyte differentiation. 53 Human OA chondrocytes have been used to show that TGF‐β1 ligand, as well as phosphorylated Smad2/3, downregulate miR‐29a/b/c expression, suggesting miR‐29s in collaboration with the TGF‐β1signalling pathway, may play a role in the etiology of OA. 32 Furthermore, miR‐29b overexpression inhibits TGF‐β1 induced Smad2/3/4 signaling, whilst inhibition of miR‐29b augments the TGF‐β1 response. 32 As well as controlling TGF‐β1 ability to modulate Smad2/3/4 signaling, the downregulation of miR‐29a causes synovial fibroblasts to increase TGF‐β1 expression, whereas miR‐29a overexpression results in decreased TGF‐β1 expression. 54 In summary, the impact of miR‐29s on TGF‐β1‐induced Smad signaling in the etiology of OA warrants further study, which could lead to an attractive treatment strategy for cartilage repair and regeneration in OA.
Many reports have described the expression profiles of miR‐29s in preclinical OA animal models and human OA tissues; miR‐29a is downregulated and miR‐29b is upregulated in OA cartilage compared with healthy cartilage samples. 32 , 55 , 56 OA patients have higher expression of miR‐29c in their plasma and similarly miR‐29c levels in synovial fluid are positively associated with the severity of knee OA. 57 , 58 In mouse models, miR‐29b expression peaks in the days following destabilization of the medial meniscus surgery, but decreases gradually with advancing OA to reach equivalent levels to controls within 6 weeks. 32 , 55 How miR‐29s contribute to cartilage destruction in OA is unclear but their involvement may be linked to the actions of proinflammatory cytokines, such as interleukin‐1 β (IL‐1β) and tumor necrosis factor alpha (TNF‐α), that are released from the synovium and chondrocytes during OA pathogenesis. 59 , 60 IL‐1β has been reported to increase or decrease the expression of miR‐29b in human OA chondrocytes in culture whereas IL‐1β stimulation of the chondrosarcoma SW1353 cell line has no effect on miR‐29b expression. 32 , 61 , 62 There are fewer studies reporting the effects of TNF‐α on miR‐29s in OA but in one study, SW1353 chondrocytes were stimulated with TNF‐α to mimic OA transcriptional reprogramming and miR‐29b expression was down regulated after 24 h exposure. 61 In OA, increased levels of proinflammatory cytokines released by chondrocytes and synovium leads to increased levels of matrix metalloproteinase (MMPs) and the degradation of ECM proteins, such as aggrecan and collagen Type II. 63 , 64 The ability of miR‐29s to mediate the pro‐inflammatory response and alter MMP expression has been the focus of various studies but to date there is little consensus on whether miR‐29s can promote or inhibit MMP expression. miR‐29b repression of MMP‐3 expression has been noted in IL‐1β challenged human and murine chondrocytes as well as miR‐29a restoration of cartilage deteriorationin a in vivo collagenase induced OA model. 32 , 54 In the latter study, undamaged (smooth) articular cartilage was observed, and MMP‐13 expression was reduced in OA joints of miR‐29a transgenic mice compared to wild type mice. 54 Whilst these data suggest that miR‐29a can protect articular cartilage by down regulating MMP‐13, conflicting studies indicate that miR‐29b overexpression can lead to increased expression of MMP1, 13, and Type X collagen and decreased expression of Type II collagen in both rat primary chondrocytes and SW1353 cells. 54 , 55 Similarly, in a rat OA model the injection of a miR‐29b antagomir inhibited the production of MMP1, 13, and type X collagen, whilst protecting Type II collagen and aggrecan from degradation. 55 The proinflammatory cytokine, IL‐1β can also activate nuclear factor‐kappa B (NF‐κB) signaling during the progression of OA, interestingly miR‐29b has been reported to repress NF‐κB signaling and reduce articular cartilage catabolic effects. The effects of miR‐29b on NF‐κB signaling have also been shown in SW1353 chondrocytes where a miR‐29b inhibitor upregulated IL‐1β‐induced activation of NF‐κB signaling. 32 , 65
Although numerous other cytokines, such as IL‐6, IL‐15, IL‐17, IL‐18, and IL‐21 are associated with OA pathogenesis, their effect, if any, on miR‐29 expression in healthy human chondrocytes and synovial tissues remain unclear. 59 For example, IL‐4 is widely recognized as an anabolic cytokine with an ability to inhibit IL‐1β‐induced release of MMP‐13 in OA chondrocytes. 66 As IL‐4 inhibits miR‐29a expression in systemic sclerosis fibroblasts it is possible that a complex interplay between different cytokines and the expression of miR‐29s exist to control MMP production and cartilage degradation in OA. 67 Nevertheless, the precise mechanisms remain unclear and further in vivo and in vitro studies with primary cells are required.
Other signaling pathways associated with OA pathogenesis, such as Wnt/β‐catenin signaling may also involve miR‐29s. Wnt/β‐catenin signaling induces hypertrophic differentiation of chondrocytes which leads to the degeneration of articular cartilage and progression of OA. 68 , 69 Whilst only a few studies have been reported, the miR‐29s can negatively regulate Wnt/β‐catenin signaling through the inhibition of Frizzled receptors FZD3, FZD5, and Disheveled 3, which are required for Wnt activation. 32 , 70 , 71 Wnt/β‐catenin signaling is regulated via extracellular inhibitors, such as Dickkopf‐1 (DKK‐1) and sclerostin. 72 , 73 DKK‐1 prevents the destruction of articular cartilage, subchondral bone sclerosis and chondro‐osteophyte formation, whereas sclerostin deficiency promotes OA in mice via subchondral bone sclerosis. 74 , 75 Whilst this is robust evidence that inhibitors of Wnt/β‐catenin signaling can prevent OA, the precise role of miR‐29s in controlling DKK‐1 and SOST is unknown but further investigations could identify a potential therapeutic target for OA.
3. ROLE OF MIR‐29 FAMILY IN OSTEOPOROSIS
Osteoporosis is an asymptomatic, chronic, and degenerative bone disease resulting in an increased risk of bone fracture and reduced life quality. 76 , 77 , 78 , 79 In osteoporosis a dysregulation of osteoblast and osteoclast differentiation and activity results in an imbalance between bone formation and resorption during bone remodeling. This results in structural changes to both trabecular and cortical bone and a reduced bone mineral density (BMD). 80 , 81 , 82 Osteoblasts and osteoclasts are derived from different embryonic cell lineages; the former differentiate from MSCs whereas the latter derive from hematopoietic progenitors in the monocyte or macrophage lineage. 83 , 84 , 85 Mature osteoblasts synthesize bone matrix and terminally differentiate into osteocytes when embedded in mineralizing osteoid. Osteocytes maintain structural bone integrity and allow bone to adapt to mechanical stimuli. 80 , 86 Osteoporosis is most common in post‐menopausal women where reduced circulating estrogen levels lead to an increase in both bone resorption and bone formation. However, the increase in bone resorption predominates leading to a net bone loss. 87 , 88 , 89
To date, therapeutic agents, such as anti‐resorptive and anabolic drugs are the preferred treatment for osteoporosis. Anti‐resorptive drugs, such as bisphosphonates and anti‐RANKL treatment, for example, denosumab function by inhibiting the recruitment and activity of osteoclasts, leading to a decreased bone remodeling rate. Anabolic drugs, such as 1‐34 parathyroid hormone treatment, (e.g., teriparatide), and anti‐sclerostin, (e.g., romosozumab) by contrast, lead to increased bone formation via a positive remodeling balance. 90 Regardless of the ability of treatment options, the therapeutic options for osteoporosis aren't sufficient for many patients with a high risk of fractures and romosozumab has recently been associated with adverse cardiovascularevents. 90 , 91 It is therefore appropriate for other pathways involved in bone formation and resorption to be considered as therapeutic targets in the quest to maintain or improve bone mass in osteoporosis patients.
Possible new diagnostic biomarkers or targets for pharmaceutical intervention include miRNAs and specifically miR‐29s. In preclinical studies, miR‐29a expression in bone of ovariectomised mice are decreased and in human studies serum levels of miR‐29a levels are lower in postmenopausal women with low BMD. 33 , 92 Serum levels of miR‐29b are also lower in patients with bone fragility fractures and are positively correlated with both histomorphometric parameters of bone formation and circulating levels of procollagen Type 1 N‐terminal propeptide; a recognized biomarker of bone formation. 93 , 94 , 95 Whilst there is a paucity of data reporting the circulating levels of miR‐29s in osteoporosis, there are numerous studies reporting the essential role of miR‐29s for osteoblast and osteoclast differentiation and function during the bone remodeling process. 80
Osteoblast differentiation is a coordinated process that is tightly regulated by growth factors, transcription factors and intracellular signaling pathways. 96 , 97 , 98 , 99 , 100 , 101 , 102 In particular the Wnt/β‐catenin and TGF‐β/bone morphogenetic protein (TGF‐β/BMP) signaling pathways, which have fundamental roles in skeletal development and bone homeostasis. Canonical Wnt signaling requires the phosphorylation and stabilization of cytoplasmic β‐catenin to upregulate the expression of runt‐related transcription factor 2 (Runx2) and the promotion of osteoblast differentiation. 99 Alternative signaling cascades are initiated by TGF‐β/BMP signaling via Smad proteins or noncanonical, non‐Smad pathways. These include various branches of mitogen‐activated protein kinase (MAPK) pathways, such as extracellular signal–regulated kinase (ERK), c‐Jun N‐terminal kinase (JNK), and p38 MAPK (p38). 96 , 97 , 98 , 100 , 101 , 102 In contrast to Wnt/β‐catenin signaling, TGF‐β signaling suppresses the later phase of osteoblast differentiation and matrix mineralization through a reduction of Runx2 expression. 103 Similarly, the class IIa histone deacetylases, HDAC4 and ‐5 are widely expressed in MSCs 104 and can lead to the degradation of Runx2 and impairment of osteoblast differentiation. 102 , 105 They also act as corepressors for TGFβ/Smad3‐mediated transcriptional repression of Runx2 function in osteoblast differentiation. 106 Intriguingly, the expression of miR‐29s increase during osteoblast differentiation, a role consistent with the observation that miR‐29a/b can reduce HDAC4 and induce Runx2 and Wnt/β‐catenin signaling during osteoblast differentiation 33 , 107 , 108 , 109 , 110 , 111 miR‐29a can promote osteoblast differentiation by other routes including the negative regulation of CTNNBIP1 (inhibitor of WNT/β‐catenin signaling), DUSP2 (inhibitor of JNK‐MAPK pathway) and CDK6 (BMP2 antagonist) and ACVR2A, a recognized inhibitor of osteoblast differentiation. 109 , 111 , 112 Furthermore, miR‐29a can also promote osteoblast differentiation by activating Wnt/β‐catenin signaling through the ERK‐MAPK pathway and via a positive feed forward loop in which canonical Wnt signaling induces miR‐29a transcription resulting in the down regulation of antagonists of β‐catenin‐dependent Wnt signaling, DKK1, Kremen2, and secreted fizzled related protein 2. This cascade of events will potentiate Wnt signaling, driving a gene expression program essential for osteoblast differentiation. 107 , 113
The principal function of the fully differentiated osteoblast is to synthesize and excrete a collagen rich ECM in bone (osteoid) which is mineralized through actions of the osteoblast over time. miR‐29a/c negatively regulates the expression of a number of ECM genes and accordingly the mineralization stage of osteoblast differentiation is characterized by decreased ECM gene expression and increased expression of miR‐29a/c. 108 , 114 , 115 For example, miR‐29a/c inhibits the expression of osteonectin or secreted protein acidic and rich in cysteine, which is a collagen‐binding matricellular protein and critical for ECM assembly and deposition. 108 miR‐29b also suppresses the production of collagens Type I, IV, V by the differentiated osteoblast and during ECM mineralization. 111 , 116 The overexpression of miR‐29a in fish bone‐derived cells can lead to increased expression levels of BMP2, osteocalcin, and osteopontin to accelerate differentiation and induce ECM mineralization, but the impact of miR‐29a on ECM mineralization is attenuated at the end stage of the differentiation. 117 These results suggest that miR‐29s are positive regulators for osteoblast differentiation and ECM mineralization, whilst miR‐29s could function as negative regulators to avoid excessive ECM accumulation during skeletal mineralization.
Osteoclast differentiation from hematopoietic precursors is driven by macrophage colony‐stimulating factor and receptor activator for nuclear factor κB ligand (RANKL), which are expressed by osteoblasts, osteocytes and activated T cells. 118 , 119 RANKL activates its receptor, RANK on osteoclast precursors to promote a signaling cascade involving many transcriptional factors, including c‐fos, NF‐κB (p50 and p52), JNK, p38 MAPK, TRAF6, and NFATc1, resulting in osteoclast differentiation and bone resorption. 120 , 121 , 122 , 123 , 124 , 125 The RANK/RANKL interaction together with the soluble decoy receptor osteoprotegerin (OPG) is widely recognized to control osteoclast differentiation and bone resorption. 126 , 127 Osteoclast differentiation is enhanced by proinflammatory cytokines, such as TNF‐α, IL‐1β, IL‐6 and IL‐17, whereas anti‐inflammatory cytokines, such as IL‐4, IL‐12, IL‐33 and interferons inhibit their differentiation. 118 , 119 , 128
Dicer, DGCR8, and AGO2, essential components in the generation of mature miRNAs, have critical roles in osteoclast differentiation. 129 Specifically, the expression of miR‐29a by osteoclast precursors is stimulated by TNF‐α; an observation consistent with other studies reporting that in murine osteoclast precursors and the mouse monocyte cell line, RAW264.7, the expression of miR‐29s are increased during RANKL‐induced osteoclast differentiation, in concert with osteoclast markers Trap and cathepsin K. 130 , 131 In addition, miR‐29 knockdown led to a delay in osteoclast differentiation and the migration of osteoclast precursors but did not affect actin ring formation by mature osteoclasts. 131 Identification of target genes for miR‐29s during osteoclast formation specified that miR‐29s promoted osteoclast formation by targeting RNAs important for cytoskeletal organization, commitment, and osteoclast function. 131 Moreover, miR‐29b can increase osteoclast survival rate by repressing the proapoptotic gene, BCL‐2‐modifying factor. 132 Whilst these data imply that miR‐29s are positive regulators of osteoclast differentiation a conflicting study reported that miR‐29b functions as a negative regulator of human osteoclast differentiation and bone resorption by downregulating the expression of c‐fos and MMP2. 133 This negative regulation of osteoclast formation and function by miR‐29b was recently corroborated in an in vitro study that disclosed upregulation of osteoclast differentiation and pit formation when miR‐29a expression was silenced. 33 Similarly, mice overexpressing miR‐29a in osteoblasts had increased bone mass and estrogen deficiency‐induced bone loss was mitigated in miR‐29a overexpressing mice. Mechanistically, it was found that miR‐29a signaling in osteoblasts is bone protective through repression of osteoclast regulators, RANKL and CXCL12, to reduce osteoclastogenic differentiation. 33 Different culturing conditions may explain the reported discrepancies in the ability of miR‐29s to modify osteoclast formation and activity, but the in vivo data from Lian and colleagues provides compelling insight into the complex interplay between osteoclasts and osteoblasts, revealing the remedial potential of miR‐29a for improving osteoporotic disorders.
Glucocorticoids (GCs) are prescribed for the treatment of many chronic conditions, but their chronic use is associated with frequent and wide‐ranging adverse effects including osteoporosis and bone fractures. 134 These adverse bone effects are also observed in Cushing's disease which is caused by increased secretion of adrenocorticotropic hormone from the pituitary gland that stimulates the synthesis of cortisol by the adrenal glands leading to accelerated bone loss in both men and women. 135 The precise mechanisms that underlie the undesirable effects of GCs on skeletal development are unclear but are likely to involve impairment of both osteoblast and osteoclast differentiation and function. However, the ability of GCs to promote osteoclast formation and activity by increasing RANKL production by both osteoblasts and osteocytes and downregulating its soluble decoy receptor OPG is the prevailing mechanism. This skews the RANKL: OPG ratio towards osteoclastogenesis. 136 Interestingly, miR‐29s have also been reported to mediate the catabolic effects of GCs on the skeleton. GC treatment reduces miR‐29a but not miR‐29b or miR‐29c expression in rat bone and miR‐29a overexpression promotes Runx2 expression and reduces the ability of GCs to inhibit osteoblast differentiation, BMD, and trabecular bone volume. 113 miR‐29a overexpression in mice weakened GCs ability to promote RANKL expression, osteoclast differentiation, and bone erosion, independent of altered OPG expression. 110 , 137 The mechanisms by which miR‐29a protects the skeleton from GCs may involve miR‐29a inhibiting GC‐induced DKK‐1 expression, which would result in increased Wnt/β‐catenin signaling and osteoblast differentiation. 113 Alternatively, miR‐29a may delay or inhibit GC induced bone resorption by repressing tumor necrosis factor superfamily 13b expression, which supports osteoclast differentiation and maturation. 137 In conclusion, the consensus across a variety of published studies is that miR‐29s inhibit osteoclast differentiation; this may make it an attractive target to reduce bone resorption in osteoporosis and other skeletal disorders with excessive osteoclastic bone resorption.
4. ROLE OF MIR‐29 FAMILY IN CARDIORENAL SYNDROME
Cardiorenal syndrome is a complex and severe clinical condition defined as a pathophysiological disorder of the heart and kidneys whereby acute or chronic dysfunction in one organ induces the same in the other. This ultimately leads to both chronic heart failure (CHF) and chronic kidney disease (CKD). 138 , 139 A common pathological feature of cardiorenal syndrome is fibrosis and in particular an excessive accumulation of collagen and fibronectin within the ECM. 140 , 141 Fibrosis is a common consequence of inflammation‐ and oxidative stress‐ related endothelial dysfunction in aging, hypertension, diabetes mellitus (DM), obesity, ischemia, and organ injury. 142 However, the cellular mechanisms leading to fibrosis are unclear and therefore current management of patients with CRS are mainly via supportive therapies to relieve the progression of the diseases. 142 , 143 Cardiac myofibroblasts, the main cell type in the heart, are derived from multiple cell lineages, including resident fibroblasts, smooth muscle cells, epithelial and endothelial cells via epithelial or endothelial‐mesenchymal transition (EMT/EndMT), and fibrocytes. 144 When cardiac myofibroblasts synthesize excess collagen and other ECM proteins, the resultant fibrosis can lead to myocardial infarction, cardiomyopathy, and heart failure. 145 , 146 , 147 , 148
Dicer has a critical role in the development of the ventricular myocardium and the preservation of glomerular and podocyte function and its deletion, unsurprisingly leads to cardiac hypertrophy, ventricular fibrosis, glomerulosclerosis, tubulointerstitial fibrosis, podocyte foot process effacement, and proteinuria. 149 , 150 , 151 , 152 , 153 The heart and kidney have high expression levels of miR‐29s which are lower in animal models and human samples of CHD and CKD possibly implicating miR‐29s as protective agents in these two organs. 154 , 155 , 156 , 157 , 158 Similarly, the expression of miR‐29s in cardiac muscle of mice and humans are also downregulated in the region of a myocardial infarction in mice and humans, which may account for the increased expression of collagen Types I, II, III, and fibrillin 1. 156 In support of this protective role, miR‐29a expression is upregulated in the heart and cardiac fibroblasts of aged zebrafish which may prevent collagen deposition and DNA methylation through the inhibition of DNA methyltransferase, such as DNMT1 and DNMT3a to avoid cardiac damage. 159 Indeed, fibrosis and DNA methylation in human cardiac fibroblasts is a consequence of hypoxia downregulating miR‐29a/b expression. 159
In atrial fibrosis, miR‐29s expression is lower in a canine model of congestive heart failure‐related atrial fibrillation (AF). Similarly, the expression of miR‐29b in both serum and atrial tissues are decreased in humans with congestive heart failure and/or AF whereas overexpression of miR‐29b in canine or murine fibroblasts results in decreased expression of collagen Types I, lll, V, and fibrillin 1. 160 , 161 miR‐29a can also inhibit cardiomyocyte hypertrophy by the inhibition of nuclear factor of activated T cells c4 (NFATc4)and/or the suppression of peroxisome proliferator‐activated receptor δ. 162 , 163 These results together strongly support a protective role for tissue miR‐29s against the development of cardiac hypertrophy and fibrosis. In contrast, the role of circulating miR‐29s is less certain as levels are upregulated in patients with essential hypertension and hypertrophic cardiomyopathy and these levels are positively correlated with left ventricular hypertrophy and myocardial fibrosis. 164 , 165 , 166 Various rodent models have also indicated that the tissue levels of miR‐29s are not protective against transverse aortic constriction (TAC)‐induced cardiac hypertrophy. Increased expression of miR‐29a in cardiac tissue is associated with TAC‐induced cardiac hypertrophy whereas miR‐29 deficient mice or those treated with miR‐29a antagomirs had reduced levels of TAC‐induced cardiac hypertrophy and myocardial fibrosis. 164 , 166 , 168 miR‐29a expression is also upregulated in murine heart tissues of myocardial ischemia‐reperfusion injury and overexpression of miR‐29a promotes cell apoptosis through suppression of insulin‐like growth factor I in a rat myoblast cell line 169 Moreover, suppression of miR‐29a/c can reduce myocardial infarct size and IR injury‐induced cell apoptosis via the upregulation of myeloid cell leukemia 1 (MCL‐1), which is a target of miR‐29s. 170 In summary, the precise roles of miR‐29s on cardiac disease are still unclear as the miR‐29s levels of the tissues appear to be protective for myocardial infarction, congestive heart failure, and AF in some reports whilst conflicting studies indicate that miR‐29s could function as a progressive factor for TAC‐induced cardiac hypertrophy and myocardial IR injury.
Renal myofibroblasts originate from various origins, including bone marrow‐derived fibroblasts, tubular epithelial cells, endothelial cells, pericytes and interstitial fibroblasts and are responsible for any excess matrix production in renal fibrosis. 171 , 172 TGF‐β signaling is a recognized central mediator of renal fibrosis, possibly through its ability to inhibit miR‐29s capacity to suppress the deposition of collagen Types I, III, and IV by mesangial cells, tubular cells and podocytes in both humans and rodent models. 155 , 173 , 174 , 175 A disintegrin and metalloproteinases (ADAMs) are involved in renal fibrosis and TGF‐β/Smad2/3 signaling upregulates Adam 10, 12, 17, 19 expression in renal cells and in unilateral ureteral obstruction models of renal fibrosis. 35 The increase in Adams12 and 19 expression correlated strongly with a decrease in miR‐29s expression and the overexpression of miR‐29s blocked TGF‐β‐mediated upregulation of Adam12 and Adam19 gene expression and improve renal fibrosis. 35 , 36 , 176 These studies strongly suggest ADAMs are involved in renal fibrosis and are regulated by both miR‐29s and TGF‐β making thempotential therapeutic targets for the prevention of renal fibrosis.
Patients with CKD present with cardiac fibrosis, hypertrophy, and dysfunction. 177 , 178 In a rat CKD model, increased levels of circulating cardiotonic steroids (CTS) and activation of Na/K‐ATPase induce cardiac fibrosis and hypertrophy. 179 This in turn negatively regulates miR‐29b resulting in increased collagen Type I synthesis and fibrosis. 178 , 180 This key role for miR‐29b was confirmed in miR‐29b overexpression studies where CTS‐induced collagen Type I synthesis in rat cardiac fibroblasts was inhibited. 178
Diabetic cardiomyopathy (DC) is characterized by myocardial fibrosis, the major cardiovascular complication in patients with DM. 181 The serum levels of IL‐6 are elevated in patients with DM and are positively associated with one‐year mortality outcomes in patients with CHF. 182 , 183 Elevated serum IL‐6 levels in experimental diabetic mice promotes TGF‐β1 expression and the downregulation of miR‐29a. This results in the increased synthesis of collagen Types I and III, an effect that is inhibited by the overexpression of miR‐29a by cardiac fibroblasts. 184 In contrast, the promotion of DM can induce miR‐29s expressions and inhibit MCL1 expression, which results in apoptosis of mouse cardiomyocytes. 185 Insulin, which is used for the treatment of DM, inhibits miR‐29s via increased mammalian target of rapamycin complex 1 (mTORC1) signaling. Conversely, the suppression of mTORC1 signal pathway initiates upregulation of miR‐29s and downregulation of MCL‐1, resulting in the loss of myofibril bundle organization in rats with DM. 185 These results suggest that the impact of DM on the expression and function of miR‐29s in DC may be different depending on the type of cells and experimental conditions studied.
In addition to the effects of DM on cardiac disease, diabetic nephropathy (DN), which is characterized by glomerulosclerosis and tubulointerstitial fibrosis, is the most common cause of CKD in patients with DM. 186 Pathogenesis of DN is attributable to hyperglycemia‐induced TGF‐β/Smad signaling which leads to the fibrotic changes typical of DN. 187 , 188 , 189 Serum and renal levels of miR‐29b are downregulated in patients with Type 2 DM and this leads to the promotion of collagen Types I, III, and IV expression and renal fibrosis in the diabetic mouse. 175 , 190 , 191 Whilst miR‐29s are suppressed in kidneys of diabetic mice, dipeptidyl peptidase‐4 (DPP‐4) inhibitor; a therapeutic drug for Type 2 DM, can restore miR‐29s and potentially limit fibrosis. 159 In addition, Wnt/β‐catenin signaling is impaired in glomeruli of DM‐induced mice whereas DKK‐1 and fibronectin expression are upregulated. 192 This increased expression of DKK‐1 and fibronectin is a likely consequence of reduced levels of miR‐29a as both are normalized in glomeruli of miR‐29a transgenic DM mice. 192 Moreover, overexpression of miR‐29a can rescue the high glucose‐induced cell apoptosis and upregulation of fibronectin in mouse mesangial cells maintained in vitro. 192 Similarly, in murine podocytes and glomeruli of miR‐29a transgenic mice, high glucose‐induced deacetylation and ubiquitination of nephrin, which promotes podocyte apoptosis and renal fibrosis, are also inhibited via the suppression of HDAC4. 193 In contrast to miR‐29a, miR‐29c is higher in high glucose‐treated murine podocytes and glomeruli of diabetic mice which leads to cell apoptosis and increases fibronectin synthesis through a coordinated coupling of Sprouty homolog 1 and Rho kinase. 194 , 195
Angiotensin II (Ang II) activates several intracellular signaling pathways, such as TGF‐β/Smads, NF‐κB, and IL‐6 to promote both cardiac and renal fibrosis and inflammation. 196 , 197 , 198 , 199 Smad7 is induced by activation of Smad2/3 and can suppress TGF‐β‐induced renal and cardiac fibrosis by blocking Smad2/3 phosphorylation via a negative feedback mechanism. 200 Smad7 also plays a key role in suppressing renal inflammation by downregulating NF‐kB signaling. 187 , 201 The central role for Smad7 in CRS was demonstrated in a Smad7 deficiency model, which resulted in enhanced ANG II‐induced loss of miR‐29b expression and the promotion of murine cardiac and renal fibrosis through activation of TGF‐β/Smad3 and NF‐kB signaling. 202 , 203 Angiotensin‐converting enzyme inhibitors can effectively protect against renal fibrosis and reduce the incidence of CKD by inhibiting DPP‐4, phosphorylation of Smad3 and increasing miR‐29s expression in the kidneys of DM‐induced mice and in human endothelial cells. 204 , 205 Indeed, overexpression of miR‐29b can repress Ang II‐induced EMT through the inhibition of phosphoinositide 3‐kinase/AKT signaling, resulting in less renal interstitial fibrosis. 206 Moreover, miR‐29b can also play a protective role in cardiac fibrosis by inhibiting Ang II‐mediated TGF‐β/Smad3 signaling. 207 Whilst the available studies suggest that miR‐29s are essential negative regulators for cardiac and renal fibrosis the precise mechanistic roles of miR‐29s on both diseases remain to be clarified.
5. ROLE OF MIR‐29 FAMILY IN IMMUNE DISEASE
miRNAs are well known to play an important role in maintenance of the immune system. 34 , 208 This was first noted in Dicer‐deficient T cells, which exhibit a preference for Th1 polarization, but has since been shown at many stages of immunological function and development. 208 It is worth noting that miR‐29s are expressed in both T cells and B cells, meaning they have the potential to influence a huge range of processes. 34 Furthermore, miR‐29s are crucial for regulating an immune response to several viruses. 209 , 210 , 211
Adaptive immunity relies on the production of T cells in the thymus, where, in response to antigen stimulation, the thymic epithelium induces naïve T cell production. 212 , 213 T cells will be directed down a T cell type fate, the most commonly studied being Th1 and Th2, important in regulating the response to either intracellular (Th1) or extracellular (Th2) assault. 213 The miR‐29s are vital in both of these steps. Targeted deletion of miR‐29a/b, in vivo, phenocopies Dicer deficient cells, by reducing the threshold for thymic involution, subsequently suppressing T cell production. 34 , 208 , 214 Chandiran and colleagues show that the initial direction of naïve cells down the Th1 cell fate is only possible through noncanonical Notch1‐mediated repression of miR‐29s. 214 , 215 Notch1 repression is attenuated later in Th1 differentiation, by the effects of IFNƳ; this releases the repression of miR‐29s, therefore repressing further production of Th1 cells. 214 The pri‐miR‐29a/b1 cluster is in fact central to a negative regulatory feedback loop, required to maintain T cell balance. 216 Acting through the IFNƳ pathway, miR‐29 suppresses the Th1 cell‐fate, required for cell‐mediated immunity. 216 , 217 miR‐29a/b directly inhibits the expression of the crucial T cell markers IFNƳ (inducer of miR‐29 expression), and T‐box binding transcription factor (T‐bet [inducer of IFNƳ]), through miR‐29 seed sequences in these genes. Treatment of miRNA‐deficient cells with either miR‐29a/b is sufficient to restore wild type levels of T‐bet and IFNƳ, while loss of miR‐29a/b results in unregulated production of Th1 cells as well as IFNƳ and T‐bet. 216 , 217
The importance of miR‐29a in immunological function and disease is in part due to the enrichment of miR‐29s and their targets in both T and B cells. 34 Mice null for miR‐29c in particular exhibit reduced B cell response, and are protected against collagen‐induced arthritis. 218 Repression of B cell miR‐29s expression via AKT and MYC pathways is associated with loss of apoptosis and several B cell malignancies, particularly lymphomas. 218 , 219 , 220 , 221 Likewise, the downregulation of miR‐29s in human immunodeficiency virus (HIV) patients is associated with higher incidence of B cell tumors via the MYC pathway. 223 The role of miR‐29s in these major pathways means that miR‐29s directed therapeutics are being considered for non‐Hodgkin's lymphoma, myeloid leukemia, and aggressive B cell lymphoma as well as the autoimmune disease, Crohn's disease. 222 , 223 , 224 , 225 , 226
The autoimmune disease, multiple sclerosis (MS) is associated with a significant increase in pro‐inflammatory Th1 cells, which have a de‐myelinating effect in the central nervous system. 227 miR‐29ab1 deficiency is noted in both MS and the classic mouse model, experimental autoimmune encephalomyelitis. 216 Ma and colleagues suggest that an increase in Th1 cells, as seen in MS patients, is linked to an increase in miR‐29b specifically, although Smith and colleagues posit that these differences are likely to be cell type and context‐specific, noting a specific increase in miR‐29b within memory cells of MS patients. 216 , 228 One of the first line treatments for MS is Interferon‐β (IFN‐β) treatment, as well as improving clinical symptoms of the disease; IFN‐β reduces inflammatory response and reduces expression of miR‐29s. 229 , 230 While many current treatments look to realigning the Th1/2 balance in MS patients, it is hoped that miR‐29s treatments may provide future therapeutic avenues.
Type 1 DM is an autoimmune disease, caused by a loss of function in the insulin‐secreting β cells of the pancreas. This loss of function follows an influx of proinflammatory cytokines into the pancreatic islets of Langerhans, destroying large numbers of β cells and reducing capacity to secrete insulin in response to rises in blood sugar. 231 Interestingly miR‐29 is among the most highly expressed microRNA in healthy β cells. 232 Experiments in both pre‐diabetic NOD mice and insulin‐impaired MIN6 cells suggest miR‐29s are highly upregulated in prediabetic β cells in direct response to cytokine action. In fact, over expression of miR‐29s caused a diabetes‐like reduction in insulin secretion in both human and mouse islet cells and directly suppresses antiapoptotic gene, MCL1. 233 miR‐29s are considered key markers of diabetes and prediabetes and certainly warrants further study to fully understand its potential in furthering our understanding of the disease and potential therapies. 233 , 234 , 235 , 236
The role of the miR‐29s in HIV infections is an emerging but exciting field. Since miRNAs were first implicated in immunological pathogenesis, several have been implicated in modulating HIV infectivity and miR‐29s seem particularly interesting. 237 , 238 , 239 Silencing Dicer, and therefore miRNAs, results in an enhancement of HIV‐1 replication, suggesting a role in modulating the immune response to HIV infection. 237 It is proposed that expression of miR‐29s is directly repressed by interactions with HIV‐1. In fact, miR‐29s are downregulated in the peripheral blood mononuclear cells (PBMC) of HIV‐1 infected patients, 239 , 240 but not in “elite suppressors” (patients infected with HIV who maintain low viral load without retroviral treatment), who exhibit comparable PBMC miR‐29s levels to control blood. 240 , 241 Furthermore, it has been shown that miR‐29a, induced by IL‐21, directly suppresses replication of the virus via direct interaction with HIV‐1 mRNA, mediating interactions with the RISC complex. 211 , 237 , 242 , 243 , 244 miR‐29s are clearly key to understanding immune response to HIV infection.
6. CONCLUSION AND PROSPECTIVE
In the current review, the miR‐29 family has been shown to promote osteoblast differentiation and apoptosis whilst suppressing chondrogenic differentiation, osteoclast differentiation, fibrosis, and T cell differentiation via the inhibition of target genes (Figure 2). It is clear that miR‐29s could offer therapeutic targets for pathologies involving OA, osteoporosis, cardiorenal disease, and immune disease. However, differing expression levels of miR‐29s in the tissues, cells and serum may need to be taken into consideration (Table 1). In addition to these diseases, further studies have reported that miR‐29s could also provide a promising novel therapeutic approach for reducing excessive collagen deposition in both liver fibrosis and tendinopathy. 245 , 246 , 247 Intravenous administration of miR‐29a has been reported to improve liver fibrosis in mouse models via the downregulation of Col1a1 expression. Watts et al., Similarly, in tendinopathy, intra‐lesional injection of miR‐29a improved the tendon lesion cross‐sectional area in an equine model of collagenase‐induced superficial digital flexor tendon injury via the downregulation of Col3a1 expression. 247 Millar et al., furthermore, the direct injection of a miR‐29a mimic downregulated the expression of Col3a1 in the tendon of a mouse model of patellar tendon injury. 246 These preclinical studies suggest that miR‐29a is also an essential negative regulator for liver fibrosis and tendinopathy. Thus, the role of miR‐29s in the etiology of chronic diseases is extensive and a comprehensive understanding of miR‐29s functions may provide a rational for an attractive treatment strategy for disease prevention and/or cure. Further studies are therefore crucial to clarify the precise mechanism of miR‐29s and their potential therapeutic applications.
Figure 2.

Target genes of miR‐29s in the mechanisms of cell differentiation, fibrosis, and apoptosis. The illustration describes the reported target genes of miR‐29s involved in chondrogenic differentiation, osteoblast differentiation, osteoclast differentiation, fibrosis, apoptosis, and T cell differentiation
Table 1.
Expression of miR‐29s in tissues and serum of humans and animal models of disease
| Osteoarthritis | Osteoporosis | Cardiac disease | Renal disease | Immune disease | |||||||||||
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| References | Species | References | Species | References | Species | References | Species | References | Species | ||||||
| miR‐29a | ↑ | 32 | Human | ー | ↑ |
164 165 166 168 169 184 185 |
Human Human Human Rat Mouse Mouse Rat |
ー | ー | ||||||
| ↓ | 56 | Human | ↓ |
33 92 110 113 |
Mouse Human Mouse Rat |
↓ |
156 157 160 161 167 207 |
Mouse Mouse Mouse Dog Human Mouse |
↓ |
35 36 154 158 175 192 193 205 |
Rat, Mouse Mouse Mouse Mouse Rat, Mouse Mouse Mouse Mouse |
↓ | 240 | Human | |
| miR‐29b | ↑ |
32 55 |
Human Human |
ー | ↑ |
165 185 |
Human Rat |
ー | ↑ | 209 | Human | ||||
| ー | ↓ | 93 | Human | ↓ |
156 157 161 167 207 |
Human, Mouse Mouse Human, Dog Human Mouse |
↓ |
35 36 154 158 175 190 205 |
Rat, Mouse Mouse Mouse Mouse Mouse Mouse Mouse |
↓ | 240 | Human | |||
| miR‐29c | ↑ |
32 57 |
Human Human |
ー | ↑ |
165 185 |
Human Rat |
↑ |
194 195 |
Mouse Human |
ー | ||||
| ー | ー | ↓ |
155 156 157 160 161 207 |
Mouse Mouse Mouse Mouse Dog Mouse |
↓ |
35 36 154 158 174 175 205 |
Rat, Mouse Mouse Mouse Mouse Human, Rat Rat, Mouse Mouse |
↓ | 240 | Human | |||||
CONFLICT OF INTERESTS
The authors declare that there are no conflict of interests.
ACKNOWLEDGMENTS
Masahiro Horita was supported by the Teraoka Scholarship Foundation. Colin Farquharson and Louise A Stephen were supported by the Biotechnology and Biological Sciences Research Council (BBSRC) through an Institute Strategic Program Grant Funding (BB/J004316/1).
Horita M, Farquharson C, Stephen LA. The role of miR‐29 family in disease. J Cell Biochem. 2021;122:696‐715. 10.1002/jcb.29896
REFERENCES
- 1. Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin‐4 encodes small RNAs with antisense complementarity to lin‐14. Cell. 1993;75(5):843‐854. 10.1016/0092-8674(93)90529-y [DOI] [PubMed] [Google Scholar]
- 2. Wightman B, Ha I, Ruvkun G. Posttranscriptional regulation of the heterochronic gene lin‐14 by lin‐4 mediates temporal pattern formation in C. elegans. Cell. 1993;75(5):855‐862. 10.1016/0092-8674(93)90530-4 [DOI] [PubMed] [Google Scholar]
- 3. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116(2):281‐297. 10.1016/s0092-8674(04)00045-5 [DOI] [PubMed] [Google Scholar]
- 4. Perron MP, Provost P. Protein interactions and complexes in human microRNA biogenesis and function. Front Biosci. 2008;13:2537‐2547. 10.2741/2865 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Griffiths‐Jones S, Saini HK, van Dongen S, Enright AJ. miRBase: tools for microRNA genomics. Nucleic Acids Res. 2008;36(Database issue):D154‐D158. 10.1093/nar/gkm952 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Zeng Y, Yi R, Cullen BR. Recognition and cleavage of primary microRNA precursors by the nuclear processing enzyme Drosha. EMBO J. 2005;24(1):138‐148. 10.1038/sj.emboj.7600491 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Cullen BR. Transcription and processing of human microRNA precursors. Mol Cell. 2004;16(6):861‐865. 10.1016/j.molcel.2004.12.002 [DOI] [PubMed] [Google Scholar]
- 8. Yi R, Qin Y, Macara IG, Cullen BR. Exportin‐5 mediates the nuclear export of pre‐microRNAs and short hairpin RNAs. Genes Dev. 2003;17(24):3011‐3016. 10.1101/gad.1158803 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Hutvagner G, McLachlan J, Pasquinelli AE, Balint E, Tuschl T, Zamore PD. A cellular function for the RNA‐interference enzyme Dicer in the maturation of the let‐7 small temporal RNA. Science. 2001;293(5531):834‐838. 10.1126/science.1062961 [DOI] [PubMed] [Google Scholar]
- 10. Chendrimada TP, Gregory RI, Kumaraswamy E, et al. TRBP recruits the Dicer complex to Ago2 for microRNA processing and gene silencing. Nature. 2005;436(7051):740‐744. 10.1038/nature03868 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Diederichs S, Haber DA. Dual role for argonautes in microRNA processing and posttranscriptional regulation of microRNA expression. Cell. 2007;131(6):1097‐1108. 10.1016/j.cell.2007.10.032 [DOI] [PubMed] [Google Scholar]
- 12. Haase AD, Jaskiewicz L, Zhang H, et al. TRBP, a regulator of cellular PKR and HIV‐1 virus expression, interacts with Dicer and functions in RNA silencing. EMBO Rep. 2005;6(10):961‐967. 10.1038/sj.embor.7400509 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Matranga C, Tomari Y, Shin C, Bartel DP, Zamore PD. Passenger‐strand cleavage facilitates assembly of siRNA into Ago2‐containing RNAi enzyme complexes. Cell. 2005;123(4):607‐620. 10.1016/j.cell.2005.08.044 [DOI] [PubMed] [Google Scholar]
- 14. Rand TA, Petersen S, Du F, Wang X. Argonaute2 cleaves the anti‐guide strand of siRNA during RISC activation. Cell. 2005;123(4):621‐629. 10.1016/j.cell.2005.10.020 [DOI] [PubMed] [Google Scholar]
- 15. Liu J. Argonaute2 is the catalytic engine of mammalian RNAi. Science. 2004;305(5689):1437‐1441. 10.1126/science.1102513 [DOI] [PubMed] [Google Scholar]
- 16. Meister G, Landthaler M, Patkaniowska A, Dorsett Y, Teng G, Tuschl T. Human Argonaute2 mediates RNA cleavage targeted by miRNAs and siRNAs. Mol Cell. 2004;15(2):185‐197. 10.1016/j.molcel.2004.07.007 [DOI] [PubMed] [Google Scholar]
- 17. Pillai RS, Artus CG, Filipowicz W. Tethering of human Ago proteins to mRNA mimics the miRNA‐mediated repression of protein synthesis. RNA. 2004;10(10):1518‐1525. 10.1261/rna.7131604 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Friedman RC, Farh KK, Burge CB, Bartel DP. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res. 2009;19(1):92‐105. 10.1101/gr.082701.108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Barbarotto E, Schmittgen TD, Calin GA. MicroRNAs and cancer: profile, profile, profile. Int J Cancer. 2008;122(5):969‐977. 10.1002/ijc.23343 [DOI] [PubMed] [Google Scholar]
- 20. 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. 10.1016/j.cell.2004.12.035 [DOI] [PubMed] [Google Scholar]
- 21. Shirasaki T, Honda M, Shimakami T, et al. MicroRNA‐27a regulates lipid metabolism and inhibits hepatitis C virus replication in human hepatoma cells. J Virol. 2013;87(9):5270‐5286. 10.1128/JVI.03022-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Cullen BR. MicroRNAs as mediators of viral evasion of the immune system. Nat Immunol. 2013;14(3):205‐210. 10.1038/ni.2537 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Inukai S, Slack F. MicroRNAs and the genetic network in aging. J Mol Biol. 2013;425(19):3601‐3608. 10.1016/j.jmb.2013.01.023 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Chang TC, Yu D, Lee YS, et al. Widespread microRNA repression by Myc contributes to tumorigenesis. Nat Genet. 2008;40(1):43‐50. 10.1038/ng.2007.30 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Eyholzer M, Schmid S, Wilkens L, Mueller BU, Pabst T. The tumour‐suppressive miR‐29a/b1 cluster is regulated by CEBPA and blocked in human AML. Br J Cancer. 2010;103(2):275‐284. 10.1038/sj.bjc.6605751 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Kwon, JJ , Nabinger SC, Vega Z, et al. Pathophysiological role of microRNA‐29 in pancreatic cancer stroma. Sci Rep. 2015;5:11450. 10.1038/srep11450 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Tumaneng K, Schlegelmilch K, Russell RC, et al. YAP mediates crosstalk between the Hippo and PI(3)K‐TOR pathways by suppressing PTEN via miR‐29. Nat Cell Biol. 2012;14(12):1322‐1329. 10.1038/ncb2615 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Hwang HW, Wentzel EA, Mendell JT. A hexanucleotide element directs microRNA nuclear import. Science. 2007;315(5808):97‐100. 10.1126/science.1136235 [DOI] [PubMed] [Google Scholar]
- 29. Liao JY, Ma LM, Guo YH, et al. Deep sequencing of human nuclear and cytoplasmic small RNAs reveals an unexpectedly complex subcellular distribution of miRNAs and tRNA 3' trailers. PLoS One. 2010;5(5):e10563. 10.1371/journal.pone.0010563 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Kriegel AJ, Terhune SS, Greene AS, Noon KR, Pereckas MS, Liang M. Isomer‐specific effect of microRNA miR‐29b on nuclear morphology. J Biol Chem. 2018;293(36):14080‐14088. 10.1074/jbc.RA117.001705 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Zhang Z, Zou J, Wang GK, et al. Uracils at nucleotide position 9‐11 are required for the rapid turnover of miR‐29 family. Nucleic Acids Res. 2011;39(10):4387‐4395. 10.1093/nar/gkr020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Le LTT, Swingler TE, Crowe N, et al. The microRNA‐29 family in cartilage homeostasis and osteoarthritis. J Mol Med (Berl). 2016;94(5):583‐596. 10.1007/s00109-015-1374-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Lian, WS , Ko JY, Chen YS, et al. MicroRNA‐29a represses osteoclast formation and protects against osteoporosis by regulating PCAF‐mediated RANKL and CXCL12. Cell Death Dis. 2019;10(10):705. 10.1038/s41419-019-1942-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Liston A, Papadopoulou AS, Danso‐Abeam D, Dooley J. MicroRNA‐29 in the adaptive immune system: setting the threshold. Cell Mol Life Sci. 2012;69(21):3533‐3541. 10.1007/s00018-012-1124-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Ramdas V, McBride M, Denby L, Baker AH. Canonical transforming growth factor‐beta signaling regulates disintegrin metalloprotease expression in experimental renal fibrosis via miR‐29. Am J Pathol. 2013;183(6):1885‐1896. 10.1016/j.ajpath.2013.08.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Wang, H , Wang B, Zhang A, et al. Exosome‐mediated miR‐29 transfer reduces muscle atrophy and kidney fibrosis in mice. Mol Ther. 2019;27(3):571‐583. 10.1016/j.ymthe.2019.01.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Li G, Yin J, Gao J, et al. Subchondral bone in osteoarthritis: insight into risk factors and microstructural changes. Arthritis Res Ther. 2013;15(6):223. 10.1186/ar4405 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Thysen S, Luyten FP, Lories RJ. Targets, models and challenges in osteoarthritis research. Dis Model Mech. 2015;8(1):17‐30. 10.1242/dmm.016881 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Nazempour A, Van Wie BJ. Chondrocytes, mesenchymal stem cells, and their combination in articular cartilage regenerative medicine. Ann Biomed Eng. 2016;44(5):1325‐1354. 10.1007/s10439-016-1575-9 [DOI] [PubMed] [Google Scholar]
- 40. Kobayashi T, Lu J, Cobb BS, et al. Dicer‐dependent pathways regulate chondrocyte proliferation and differentiation. Proc Natl Acad Sci USA. 2008;105(6):1949‐1954. 10.1073/pnas.0707900105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Harada N, Watanabe Y, Sato K, et al. Bone regeneration in a massive rat femur defect through endochondral ossification achieved with chondrogenically differentiated MSCs in a degradable scaffold. Biomaterials. 2014;35(27):7800‐7810. 10.1016/j.biomaterials.2014.05.052 [DOI] [PubMed] [Google Scholar]
- 42. Mobasheri A, Kalamegam G, Musumeci G, Batt ME. Chondrocyte and mesenchymal stem cell‐based therapies for cartilage repair in osteoarthritis and related orthopaedic conditions. Maturitas. 2014;78(3):188‐198. 10.1016/j.maturitas.2014.04.017 [DOI] [PubMed] [Google Scholar]
- 43. Barter MJ, Tselepi M, Gómez R, et al. Genome‐wide microRNA and gene analysis of mesenchymal stem cell chondrogenesis identifies an essential role and multiple targets for miR‐140‐5p. Stem Cells. 2015;33(11):3266‐3280. 10.1002/stem.2093 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Yan C, Wang Y, Shen XY, et al. MicroRNA regulation associated chondrogenesis of mouse MSCs grown on polyhydroxyalkanoates. Biomaterials. 2011;32(27):6435‐6444. 10.1016/j.biomaterials.2011.05.031 [DOI] [PubMed] [Google Scholar]
- 45. Guérit D, Brondello JM, Chuchana P, et al. FOXO3A regulation by miRNA‐29a controls chondrogenic differentiation of mesenchymal stem cells and cartilage formation. Stem Cells Dev. 2014;23(11):1195‐1205. 10.1089/scd.2013.0463 [DOI] [PubMed] [Google Scholar]
- 46. Wright E, Hargrave MR, Christiansen J, et al. The Sry‐related gene Sox9 is expressed during chondrogenesis in mouse embryos. Nat Genet. 1995;9(1):15‐20. 10.1038/ng0195-15 [DOI] [PubMed] [Google Scholar]
- 47. Cucchiarini M, Thurn T, Weimer A, Kohn D, Terwilliger EF, Madry H. Restoration of the extracellular matrix in human osteoarthritic articular cartilage by overexpression of the transcription factor SOX9. Arthritis Rheum. 2007;56(1):158‐167. 10.1002/art.22299 [DOI] [PubMed] [Google Scholar]
- 48. Akiyama H, Chaboissier MC, Martin JF, Schedl A, de Crombrugghe B. The transcription factor Sox9 has essential roles in successive steps of the chondrocyte differentiation pathway and is required for expression of Sox5 and Sox6. Genes Dev. 2002;16(21):2813‐2828. 10.1101/gad.1017802 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Pittenger MF. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284(5411):143‐147. 10.1126/science.284.5411.143 [DOI] [PubMed] [Google Scholar]
- 50. Furumatsu T, Tsuda M, Taniguchi N, Tajima Y, Asahara H. Smad3 induces chondrogenesis through the activation of SOX9 via CREB‐binding protein/p300 recruitment. J Biol Chem. 2005;280(9):8343‐8350. 10.1074/jbc.M413913200 [DOI] [PubMed] [Google Scholar]
- 51. Massague J, Wotton D. Transcriptional control by the TGF‐beta/Smad signaling system. EMBO J. 2000;19(8):1745‐1754. 10.1093/emboj/19.8.1745 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Miyazawa K, Shinozaki M, Hara T, Furuya T, Miyazono K. Two major Smad pathways in TGF‐beta superfamily signalling. Genes Cells. 2002;7(12):1191‐1204. 10.1046/j.1365-2443.2002.00599.x [DOI] [PubMed] [Google Scholar]
- 53. Ferguson CM, Schwarz EM, Reynolds PR, Puzas JE, Rosier RN, O'Keefe RJ. Smad2 and 3 mediate transforming growth factor‐beta1‐induced inhibition of chondrocyte maturation. Endocrinology. 2000;141(12):4728‐4735. 10.1210/endo.141.12.7848 [DOI] [PubMed] [Google Scholar]
- 54. Ko JY, Lee MS, Lian WS, et al. MicroRNA‐29a counteracts synovitis in knee osteoarthritis pathogenesis by targeting VEGF. Sci Rep. 2017;7(1):3584. 10.1038/s41598-017-03616-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Chen, L , Li Q, Wang J, et al. MiR‐29b‐3p promotes chondrocyte apoptosis and facilitates the occurrence and development of osteoarthritis by targeting PGRN. J Cell Mol Med. 2017;21(12):3347‐3359. 10.1111/jcmm.13237 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Iliopoulos D, Malizos KN, Oikonomou P, Tsezou A. Integrative microRNA and proteomic approaches identify novel osteoarthritis genes and their collaborative metabolic and inflammatory networks. PLoS One. 2008;3(11):e3740. 10.1371/journal.pone.0003740 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Borgonio Cuadra VM, Gonzalez‐Huerta NC, Romero‐Cordoba S, Hidalgo‐Miranda A, Miranda‐Duarte A. Altered expression of circulating microRNA in plasma of patients with primary osteoarthritis and in silico analysis of their pathways. PLoS One. 2014;9(6):e97690. 10.1371/journal.pone.0097690 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Li YH, Tavallaee G, Tokar T, et al. Identification of synovial fluid microRNA signature in knee osteoarthritis: differentiating early‐ and late‐stage knee osteoarthritis. Osteoarthritis Cartilage. 2016;24(9):1577‐1586. 10.1016/j.joca.2016.04.019 [DOI] [PubMed] [Google Scholar]
- 59. Kapoor M, Martel‐Pelletier J, Lajeunesse D, Pelletier JP, Fahmi H. Role of proinflammatory cytokines in the pathophysiology of osteoarthritis. Nat Rev Rheumatol. 2011;7(1):33‐42. 10.1038/nrrheum.2010.196 [DOI] [PubMed] [Google Scholar]
- 60. Tanigawa S, Aida Y, Kawato T, et al. Interleukin‐17F affects cartilage matrix turnover by increasing the expression of collagenases and stromelysin‐1 and by decreasing the expression of their inhibitors and extracellular matrix components in chondrocytes. Cytokine. 2011;56(2):376‐386. 10.1016/j.cyto.2011.08.015 [DOI] [PubMed] [Google Scholar]
- 61. Santini P, Politi L, Vedova PD, Scandurra R, Scotto d'Abusco A. The inflammatory circuitry of miR‐149 as a pathological mechanism in osteoarthritis. Rheumatol Int. 2014;34(5):711‐716. 10.1007/s00296-013-2754-8 [DOI] [PubMed] [Google Scholar]
- 62. Akhtar N, Rasheed Z, Ramamurthy S, Anbazhagan AN, Voss FR, Haqqi TM. MicroRNA‐27b regulates the expression of matrix metalloproteinase 13 in human osteoarthritis chondrocytes. Arthritis Rheum. 2010;62(5):1361‐1371. 10.1002/art.27329 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Daheshia M, Yao JQ. The interleukin 1beta pathway in the pathogenesis of osteoarthritis. J Rheumatol. 2008;35(12):2306‐2312. 10.3899/jrheum.080346 [DOI] [PubMed] [Google Scholar]
- 64. Kobayashi M, Squires GR, Mousa A, et al. Role of interleukin‐1 and tumor necrosis factor alpha in matrix degradation of human osteoarthritic cartilage. Arthritis Rheum. 2005;52(1):128‐135. 10.1002/art.20776 [DOI] [PubMed] [Google Scholar]
- 65. Marcu KB, Otero M, Olivotto E, Borzi RM, Goldring MB. NF‐kappaB signaling: multiple angles to target OA. Curr Drug Targets. 2010;11(5):599‐613. 10.2174/138945010791011938 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Assirelli E, Pulsatelli L, Dolzani P, et al. Human osteoarthritic cartilage shows reduced in vivo expression of IL‐4, a chondroprotective cytokine that differentially modulates IL‐1beta‐stimulated production of chemokines and matrix‐degrading enzymes in vitro. PLoS One. 2014;9(5):e96925. 10.1371/journal.pone.0096925 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Maurer B, Stanczyk J, Jüngel A, et al. MicroRNA‐29, a key regulator of collagen expression in systemic sclerosis. Arthritis Rheum. 2010;62(6):1733‐1743. 10.1002/art.27443 [DOI] [PubMed] [Google Scholar]
- 68. Kawaguchi H. Regulation of osteoarthritis development by Wnt‐beta‐catenin signaling through the endochondral ossification process. J Bone Miner Res. 2009;24(1):8‐11. 10.1359/jbmr.081115 [DOI] [PubMed] [Google Scholar]
- 69. Luyten FP, Tylzanowski P, Lories RJ. Wnt signaling and osteoarthritis. Bone. 2009;44(4):522‐527. 10.1016/j.bone.2008.12.006 [DOI] [PubMed] [Google Scholar]
- 70. Bhanot P, Brink M, Samos CH, et al. A new member of the frizzled family from drosophila functions as a wingless receptor. Nature. 1996;382(6588):225‐230. 10.1038/382225a0 [DOI] [PubMed] [Google Scholar]
- 71. Rattner A, Hsieh JC, Smallwood PM, et al. A family of secreted proteins contains homology to the cysteine‐rich ligand‐binding domain of frizzled receptors. Proc Natl Acad Sci USA. 1997;94(7):2859‐2863. 10.1073/pnas.94.7.2859 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Semenov M, Tamai K, He X. SOST is a ligand for LRP5/LRP6 and a Wnt signaling inhibitor. J Biol Chem. 2005;280(29):26770‐26775. 10.1074/jbc.M504308200 [DOI] [PubMed] [Google Scholar]
- 73. Glinka A, Wu W, Delius H, Monaghan AP, Blumenstock C, Niehrs C. Dickkopf‐1 is a member of a new family of secreted proteins and functions in head induction. Nature. 1998;391(6665):357‐362. 10.1038/34848 [DOI] [PubMed] [Google Scholar]
- 74. Li J, Xue J, Jing Y, et al. SOST deficiency aggravates osteoarthritis in mice by promoting sclerosis of subchondral bone. BioMed Res Int. 2019;2019:7623562‐7623568. 10.1155/2019/7623562 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Oh H, Chun CH, Chun JS. Dkk‐1 expression in chondrocytes inhibits experimental osteoarthritic cartilage destruction in mice. Arthritis Rheum. 2012;64(8):2568‐2578. 10.1002/art.34481 [DOI] [PubMed] [Google Scholar]
- 76. Tarride JE, Burke N, Leslie WD, et al. Loss of health related quality of life following low‐trauma fractures in the elderly. BMC Geriatr. 2016;16:84. 10.1186/s12877-016-0259-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. van den Bergh JP, van Geel TA, Geusens PP. Osteoporosis, frailty and fracture: implications for case finding and therapy. Nat Rev Rheumatol. 2012;8(3):163‐172. 10.1038/nrrheum.2011.217 [DOI] [PubMed] [Google Scholar]
- 78. Center JR, Nguyen TV, Schneider D, Sambrook PN, Eisman JA. Mortality after all major types of osteoporotic fracture in men and women: an observational study. Lancet. 1999;353(9156):878‐882. 10.1016/S0140-6736(98)09075-8 [DOI] [PubMed] [Google Scholar]
- 79. Frost SA, Nguyen ND, Center JR, Eisman JA, Nguyen TV. Excess mortality attributable to hip‐fracture: a relative survival analysis. Bone. 2013;56(1):23‐29. 10.1016/j.bone.2013.05.006 [DOI] [PubMed] [Google Scholar]
- 80. Kapinas K, Delany AM. MicroRNA biogenesis and regulation of bone remodeling. Arthritis Res Ther. 2011;13(3):220. 10.1186/ar3325 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Feng Q, Zheng S, Zheng J. The emerging role of microRNAs in bone remodeling and its therapeutic implications for osteoporosis. Biosci Rep. 2018;38(3), 10.1042/BSR20180453 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Husain A, Jeffries MA. Epigenetics and Bone Remodeling. Curr Osteoporos Rep. 2017;15(5):450‐458. 10.1007/s11914-017-0391-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Karsenty G, Wagner EF. Reaching a genetic and molecular understanding of skeletal development. Dev Cell. 2002;2(4):389‐406. 10.1016/s1534-5807(02)00157-0 [DOI] [PubMed] [Google Scholar]
- 84. Komori T. Regulation of osteoblast differentiation by transcription factors. J Cell Biochem. 2006;99(5):1233‐1239. 10.1002/jcb.20958 [DOI] [PubMed] [Google Scholar]
- 85. Asagiri M, Takayanagi H. The molecular understanding of osteoclast differentiation. Bone. 2007;40(2):251‐264. 10.1016/j.bone.2006.09.023 [DOI] [PubMed] [Google Scholar]
- 86. Ralston SH, de Crombrugghe B. Genetic regulation of bone mass and susceptibility to osteoporosis. Genes Dev. 2006;20(18):2492‐2506. 10.1101/gad.1449506 [DOI] [PubMed] [Google Scholar]
- 87. Khosla S, Oursler MJ, Monroe DG. Estrogen and the skeleton. Trends Endocrinol Metab. 2012;23(11):576‐581. 10.1016/j.tem.2012.03.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Garnero P, Sornay‐Rendu E, Chapuy MC, Delmas PD. Increased bone turnover in late postmenopausal women is a major determinant of osteoporosis. J Bone Miner Res. 1996;11(3):337‐349. 10.1002/jbmr.5650110307 [DOI] [PubMed] [Google Scholar]
- 89. Hu H, He X, Zhang Y, et al. MicroRNA alterations for diagnosis, prognosis, and treatment of osteoporosis: a comprehensive review and computational functional survey. Front Genet. 2020;11:181. 10.3389/fgene.2020.00181 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Compston JE, McClung MR, Leslie WD. Osteoporosis. Lancet. 2019;393(10169):364‐376. 10.1016/S0140-6736(18)32112-3 [DOI] [PubMed] [Google Scholar]
- 91. Saag, KG , Petersen J, Brandi ML, et al. Romosozumab or alendronate for fracture prevention in women with osteoporosis. N Engl J Med. 2017;377(15):1417‐1427. 10.1056/NEJMoa1708322 [DOI] [PubMed] [Google Scholar]
- 92. Yavropoulou MP, Anastasilakis AD, Makras P, Tsalikakis DG, Grammatiki M, Yovos JG. Expression of microRNAs that regulate bone turnover in the serum of postmenopausal women with low bone mass and vertebral fractures. Eur J Endocrinol. 2017;176(2):169‐176. 10.1530/EJE-16-0583 [DOI] [PubMed] [Google Scholar]
- 93. Kocijan R, Muschitz C, Geiger E, et al. Circulating microRNA signatures in patients with idiopathic and postmenopausal osteoporosis and fragility fractures. J Clin Endocrinol Metab. 2016;101(11):4125‐4134. 10.1210/jc.2016-2365 [DOI] [PubMed] [Google Scholar]
- 94. Shetty S, Kapoor N, Bondu JD, Thomas N, Paul TV. Bone turnover markers: emerging tool in the management of osteoporosis. Indian J Endocrinol Metab. 2016;20(6):846‐852. 10.4103/2230-8210.192914 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Feichtinger X, Muschitz C, Heimel P, et al. Bone‐related Circulating MicroRNAs miR‐29b‐3p, miR‐550a‐3p, and miR‐324‐3p and their Association to Bone Microstructure and Histomorphometry. Sci Rep. 2018;8(1):4867. 10.1038/s41598-018-22844-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Bandyopadhyay A, Tsuji K, Cox K, Harfe BD, Rosen V, Tabin CJ. Genetic analysis of the roles of BMP2, BMP4, and BMP7 in limb patterning and skeletogenesis. PLoS Genet. 2006;2(12):e216. 10.1371/journal.pgen.0020216 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Bodine PV, Komm BS. Wnt signaling and osteoblastogenesis. Rev Endocr Metab Disord. 2006;7(1‐2):33‐39. 10.1007/s11154-006-9002-4 [DOI] [PubMed] [Google Scholar]
- 98. Yamashita M, Ying SX, Zhang GM, et al. Ubiquitin ligase Smurf1 controls osteoblast activity and bone homeostasis by targeting MEKK2 for degradation. Cell. 2005;121(1):101‐113. 10.1016/j.cell.2005.01.035 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Gaur, T , Lengner CJ, Hovhannisyan H, et al. Canonical WNT signaling promotes osteogenesis by directly stimulating Runx2 gene expression. J Biol Chem. 2005;280(39):33132‐33140. 10.1074/jbc.M500608200 [DOI] [PubMed] [Google Scholar]
- 100. Ge C, Xiao G, Jiang D, Franceschi RT. Critical role of the extracellular signal‐regulated kinase‐MAPK pathway in osteoblast differentiation and skeletal development. J Cell Biol. 2007;176(5):709‐718. 10.1083/jcb.200610046 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Greenblatt MB, Shim JH, Zou W, et al. The p38 MAPK pathway is essential for skeletogenesis and bone homeostasis in mice. J Clin Invest. 2010;120(7):2457‐2473. 10.1172/JCI42285 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Jeon, EJ , Lee KY, Choi NS, et al. Bone morphogenetic protein‐2 stimulates Runx2 acetylation. J Biol Chem. 2006;281(24):16502‐16511. 10.1074/jbc.M512494200 [DOI] [PubMed] [Google Scholar]
- 103. Maeda S, Hayashi M, Komiya S, Imamura T, Miyazono K. Endogenous TGF‐beta signaling suppresses maturation of osteoblastic mesenchymal cells. EMBO J. 2004;23(3):552‐563. 10.1038/sj.emboj.7600067 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Verdin E, Dequiedt F, Kasler HG. Class II histone deacetylases: versatile regulators. Trends Genet. 2003;19(5):286‐293. 10.1016/S0168-9525(03)00073-8 [DOI] [PubMed] [Google Scholar]
- 105. Zhu J, Shimizu E, Zhang X, Partridge NC, Qin L. EGFR signaling suppresses osteoblast differentiation and inhibits expression of master osteoblastic transcription factors Runx2 and Osterix. J Cell Biochem. 2011;112(7):1749‐1760. 10.1002/jcb.23094 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Kang JS, Alliston T, Delston R, Derynck R. Repression of Runx2 function by TGF‐beta through recruitment of class II histone deacetylases by Smad3. EMBO J. 2005;24(14):2543‐2555. 10.1038/sj.emboj.7600729 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Kapinas K, Kessler C, Ricks T, Gronowicz G, Delany AM. miR‐29 modulates Wnt signaling in human osteoblasts through a positive feedback loop. J Biol Chem. 2010;285(33):25221‐25231. 10.1074/jbc.M110.116137 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Kapinas K, Kessler CB, Delany AM. miR‐29 suppression of osteonectin in osteoblasts: regulation during differentiation and by canonical Wnt signaling. J Cell Biochem. 2009;108(1):216‐224. 10.1002/jcb.22243 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Ko JY, Chuang PC, Chen MW, et al. MicroRNA‐29a ameliorates glucocorticoid‐induced suppression of osteoblast differentiation by regulating beta‐catenin acetylation. Bone. 2013;57(2):468‐475. 10.1016/j.bone.2013.09.019 [DOI] [PubMed] [Google Scholar]
- 110. Ko JY, Chuang PC, Ke HJ, Chen YS, Sun YC, Wang FS. MicroRNA‐29a mitigates glucocorticoid induction of bone loss and fatty marrow by rescuing Runx2 acetylation. Bone. 2015;81:80‐88. 10.1016/j.bone.2015.06.022 [DOI] [PubMed] [Google Scholar]
- 111. Li Z, Hassan MQ, Jafferji M, et al. Biological functions of miR‐29b contribute to positive regulation of osteoblast differentiation. J Biol Chem. 2009;284(23):15676‐15684. 10.1074/jbc.M809787200 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Trompeter HI, Dreesen J, Hermann E, et al. MicroRNAs miR‐26a, miR‐26b, and miR‐29b accelerate osteogenic differentiation of unrestricted somatic stem cells from human cord blood. BMC Genomics. 2013;14:111. 10.1186/1471-2164-14-111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Wang, FS , Chung PC, Lin CL, et al. MicroRNA‐29a protects against glucocorticoid‐induced bone loss and fragility in rats by orchestrating bone acquisition and resorption. Arthritis Rheum. 2013;65(6):1530‐1540. 10.1002/art.37948 [DOI] [PubMed] [Google Scholar]
- 114. Stein GS, Lian JB. Molecular mechanisms mediating proliferation/differentiation interrelationships during progressive development of the osteoblast phenotype. Endocr Rev. 1993;14(4):424‐442. 10.1210/edrv-14-4-424 [DOI] [PubMed] [Google Scholar]
- 115. Kalajzic I, Staal A, Yang WP, et al. Expression profile of osteoblast lineage at defined stages of differentiation. J Biol Chem. 2005;280(26):24618‐24626. 10.1074/jbc.M413834200 [DOI] [PubMed] [Google Scholar]
- 116. Burger C, Zhou H, Wang H, et al. Lateral packing of mineral crystals in bone collagen fibrils. Biophys J. 2008;95(4):1985‐1992. 10.1529/biophysj.107.128355 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Roberto VP, Tiago DM, Silva IA, Cancela ML. MiR‐29a is an enhancer of mineral deposition in bone‐derived systems. Arch Biochem Biophys. 2014;564:173‐183. 10.1016/j.abb.2014.09.006 [DOI] [PubMed] [Google Scholar]
- 118. Tanaka S, Nakamura K, Takahasi N, Suda T. Role of RANKL in physiological and pathological bone resorption and therapeutics targeting the RANKL‐RANK signaling system. Immunol Rev. 2005;208:30‐49. 10.1111/j.0105-2896.2005.00327.x [DOI] [PubMed] [Google Scholar]
- 119. Del Fattore A, Teti A, Rucci N. Osteoclast receptors and signaling. Arch Biochem Biophys. 2008;473(2):147‐160. 10.1016/j.abb.2008.01.011 [DOI] [PubMed] [Google Scholar]
- 120. Takayanagi H, Kim S, Koga T, et al. Induction and activation of the transcription factor NFATc1 (NFAT2) integrate RANKL signaling in terminal differentiation of osteoclasts. Dev Cell. 2002;3(6):889‐901. 10.1016/s1534-5807(02)00369-6 [DOI] [PubMed] [Google Scholar]
- 121. Tanaka S, Nakamura I, Inoue J, Oda H, Nakamura K. Signal transduction pathways regulating osteoclast differentiation and function. J Bone Miner Metab. 2003;21(3):123‐133. 10.1007/s007740300021 [DOI] [PubMed] [Google Scholar]
- 122. Franzoso G, Carlson L, Xing L, et al. Requirement for NF‐kappaB in osteoclast and B‐cell development. Genes Dev. 1997;11(24):3482‐3496. 10.1101/gad.11.24.3482 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Grigoriadis AE, Wang ZQ, Cecchini MG, et al. c‐Fos: a key regulator of osteoclast‐macrophage lineage determination and bone remodeling. Science. 1994;266(5184):443‐448. 10.1126/science.7939685 [DOI] [PubMed] [Google Scholar]
- 124. Iotsova V, Caamano J, Loy J, Yang Y, Lewin A, Bravo R. Osteopetrosis in mice lacking NF‐kappaB1 and NF‐kappaB2. Nat Med. 1997;3(11):1285‐1289. 10.1038/nm1197-1285 [DOI] [PubMed] [Google Scholar]
- 125. Ishida N, Hayashi K, Hoshijima M, et al. Large scale gene expression analysis of osteoclastogenesis in vitro and elucidation of NFAT2 as a key regulator. J Biol Chem. 2002;277(43):41147‐41156. 10.1074/jbc.M205063200 [DOI] [PubMed] [Google Scholar]
- 126. Kong, YY , Yoshida H, Sarosi I, et al. OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph‐node organogenesis. Nature. 1999;397(6717):315‐323. 10.1038/16852 [DOI] [PubMed] [Google Scholar]
- 127. Hofbauer LC, Khosla S, Dunstan CR, Lacey DL, Boyle WJ, Riggs BL. The roles of osteoprotegerin and osteoprotegerin ligand in the paracrine regulation of bone resorption. J Bone Miner Res. 2000;15(1):2‐12. 10.1359/jbmr.2000.15.1.2 [DOI] [PubMed] [Google Scholar]
- 128. Schett G. Effects of inflammatory and anti‐inflammatory cytokines on the bone. Eur J Clin Invest. 2011;41(12):1361‐1366. 10.1111/j.1365-2362.2011.02545.x [DOI] [PubMed] [Google Scholar]
- 129. Sugatani T, Hruska KA. Impaired micro‐RNA pathways diminish osteoclast differentiation and function. J Biol Chem. 2009;284(7):4667‐4678. 10.1074/jbc.M805777200 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Miller CH, Smith SM, Elguindy M, et al. RBP‐J‐regulated miR‐182 promotes TNF‐alpha‐induced osteoclastogenesis. J Immunol. 2016;196(12):4977‐4986. 10.4049/jimmunol.1502044 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Franceschetti T, Kessler CB, Lee SK, Delany AM. miR‐29 promotes murine osteoclastogenesis by regulating osteoclast commitment and migration. J Biol Chem. 2013;288(46):33347‐33360. 10.1074/jbc.M113.484568 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Sul OJ, Rajasekaran M, Park HJ, Suh JH, Choi HS. MicroRNA‐29b enhances osteoclast survival by targeting BCL‐2‐modifying factor after lipopolysaccharide stimulation. Oxid Med Cell Longev. 2019;2019:6018180‐11. 10.1155/2019/6018180 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Rossi M, Pitari MR, Amodio N, et al. miR‐29b negatively regulates human osteoclastic cell differentiation and function: implications for the treatment of multiple myeloma‐related bone disease. J Cell Physiol. 2013;228(7):1506‐1515. 10.1002/jcp.24306 [DOI] [PubMed] [Google Scholar]
- 134. Buckley L, Humphrey MB. Glucocorticoid‐induced osteoporosis. N Engl J Med. 2018;379(26):2547‐2556. 10.1056/NEJMcp1800214 [DOI] [PubMed] [Google Scholar]
- 135. Reynolds RM, Dennison EM, Walker BR, et al. Cortisol secretion and rate of bone loss in a population‐based cohort of elderly men and women. Calcif Tissue Int. 2005;77(3):134‐138. 10.1007/s00223-004-0270-2 [DOI] [PubMed] [Google Scholar]
- 136. Compston J. Glucocorticoid‐induced osteoporosis: an update. Endocrine. 2018;61(1):7‐16. 10.1007/s12020-018-1588-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Wu RW, Lian WS, Chen YS, et al. MicroRNA‐29a counteracts glucocorticoid induction of bone loss through repressing TNFSF13b modulation of osteoclastogenesis. Int J Mol Sci. 2019;20(20):5141. 10.3390/ijms20205141 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Ronco C, Haapio M, House AA, Anavekar N, Bellomo R. Cardiorenal syndrome. J Am Coll Cardiol. 2008;52(19):1527‐1539. 10.1016/j.jacc.2008.07.051 [DOI] [PubMed] [Google Scholar]
- 139. Ronco C, House AA, Haapio M. Cardiorenal and renocardiac syndromes: the need for a comprehensive classification and consensus. Nat Clin Pract Nephrol. 2008;4(6):310‐311. 10.1038/ncpneph0803 [DOI] [PubMed] [Google Scholar]
- 140. Wynn TA, Ramalingam TR. Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nat Med. 2012;18(7):1028‐1040. 10.1038/nm.2807 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Zeisberg M, Kalluri R. Cellular mechanisms of tissue fibrosis. 1. Common and organ‐specific mechanisms associated with tissue fibrosis. Am J Physiol Cell Physiol. 2013;304(3):C216‐C225. 10.1152/ajpcell.00328.2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Zannad F, Rossignol P. Cardiorenal syndrome revisited. Circulation. 2018;138(9):929‐944. 10.1161/CIRCULATIONAHA.117.028814 [DOI] [PubMed] [Google Scholar]
- 143. Ronco C, Di Lullo L. Cardiorenal syndrome in western countries: epidemiology, diagnosis, and management approaches. Kidney Dis (Basel). 2017;2(4):151‐163. 10.1159/000448749 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. van den Borne SW, Diez J, Blankesteijn WM, Verjans J, Hofstra L, Narula J. Myocardial remodeling after infarction: the role of myofibroblasts. Nat Rev Cardiol. 2010;7(1):30‐37. 10.1038/nrcardio.2009.199 [DOI] [PubMed] [Google Scholar]
- 145. Khan R, Sheppard R. Fibrosis in heart disease: understanding the role of transforming growth factor‐beta in cardiomyopathy, valvular disease and arrhythmia. Immunology. 2006;118(1):10‐24. 10.1111/j.1365-2567.2006.02336.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. MacKenna D, Summerour SR, Villarreal FJ. Role of mechanical factors in modulating cardiac fibroblast function and extracellular matrix synthesis. Cardiovasc Res. 2000;46(2):257‐263. 10.1016/s0008-6363(00)00030-4 [DOI] [PubMed] [Google Scholar]
- 147. Camelliti P, Borg TK, Kohl P. Structural and functional characterisation of cardiac fibroblasts. Cardiovasc Res. 2005;65(1):40‐51. 10.1016/j.cardiores.2004.08.020 [DOI] [PubMed] [Google Scholar]
- 148. van Rooij E, Olson EN. Searching for miR‐acles in cardiac fibrosis. Circ Res. 2009;104(2):138‐140. 10.1161/CIRCRESAHA.108.192492 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. da Costa Martins PA, Bourajjaj M, Gladka M, et al. Conditional dicer gene deletion in the postnatal myocardium provokes spontaneous cardiac remodeling. Circulation. 2008;118(15):1567‐1576. 10.1161/CIRCULATIONAHA.108.769984 [DOI] [PubMed] [Google Scholar]
- 150. Harvey SJ, Jarad G, Cunningham J, et al. Podocyte‐specific deletion of dicer alters cytoskeletal dynamics and causes glomerular disease. J Am Soc Nephrol. 2008;19(11):2150‐2158. 10.1681/ASN.2008020233 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Ho J, Ng KH, Rosen S, Dostal A, Gregory RI, Kreidberg JA. Podocyte‐specific loss of functional microRNAs leads to rapid glomerular and tubular injury. J Am Soc Nephrol. 2008;19(11):2069‐2075. 10.1681/ASN.2008020162 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Ho JJ, Marsden PA. Dicer cuts the kidney. J Am Soc Nephrol. 2008;19(11):2043‐2046. 10.1681/ASN.2008090986 [DOI] [PubMed] [Google Scholar]
- 153. Zhao, Y , Ransom JF, Li A, et al. Dysregulation of cardiogenesis, cardiac conduction, and cell cycle in mice lacking miRNA‐1‐2. Cell. 2007;129(2):303‐317. 10.1016/j.cell.2007.03.030 [DOI] [PubMed] [Google Scholar]
- 154. Qin W, Chung ACK, Huang XR, et al. TGF‐beta/Smad3 signaling promotes renal fibrosis by inhibiting miR‐29. J Am Soc Nephrol. 2011;22(8):1462‐1474. 10.1681/ASN.2010121308 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. van Rooij E, Sutherland LB, Liu N, et al. 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. 10.1073/pnas.0608791103 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. van Rooij E, Sutherland LB, Thatcher JE, et al. 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. 10.1073/pnas.0805038105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Cheng Y, Ji R, Yue J, et al. MicroRNAs are aberrantly expressed in hypertrophic heart: do they play a role in cardiac hypertrophy? Am J Pathol. 2007;170(6):1831‐1840. 10.2353/ajpath.2007.061170 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Kanasaki K, Shi S, Kanasaki M, et al. Linagliptin‐mediated DPP‐4 inhibition ameliorates kidney fibrosis in streptozotocin‐induced diabetic mice by inhibiting endothelial‐to‐mesenchymal transition in a therapeutic regimen. Diabetes. 2014;63(6):2120‐2131. 10.2337/db13-1029 [DOI] [PubMed] [Google Scholar]
- 159. Heid, J , Cencioni C, Ripa R, et al. Age‐dependent increase of oxidative stress regulates microRNA‐29 family preserving cardiac health. Sci Rep. 2017;7(1):16839. 10.1038/s41598-017-16829-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160. Abonnenc M, Nabeebaccus AA, Mayr U, et al. Extracellular matrix secretion by cardiac fibroblasts: role of microRNA‐29b and microRNA‐30c. Circ Res. 2013;113(10):1138‐1147. 10.1161/CIRCRESAHA.113.302400 [DOI] [PubMed] [Google Scholar]
- 161. Dawson K, Wakili R, Ördög B, et al. MicroRNA29: a mechanistic contributor and potential biomarker in atrial fibrillation. Circulation. 2013;127(14):1466‐1475.1475e1461‐1428. 10.1161/CIRCULATIONAHA.112.001207 [DOI] [PubMed] [Google Scholar]
- 162. Zhang S, Yin Z, Dai FF, et al. miR‐29a attenuates cardiac hypertrophy through inhibition of PPARdelta expression. J Cell Physiol. 2019;234(8):13252‐13262. 10.1002/jcp.27997 [DOI] [PubMed] [Google Scholar]
- 163. Li M, Wang N, Zhang J, et al. MicroRNA‐29a‐3p attenuates ET‐1‐induced hypertrophic responses in H9c2 cardiomyocytes. Gene. 2016;585(1):44‐50. 10.1016/j.gene.2016.03.015 [DOI] [PubMed] [Google Scholar]
- 164. Han W, Han Y, Liu X, Shang X. Effect of miR‐29a inhibition on ventricular hypertrophy induced by pressure overload. Cell Biochem Biophys. 2015;71(2):821‐826. 10.1007/s12013-014-0269-x [DOI] [PubMed] [Google Scholar]
- 165. Huang Y, Tang S, Huang C, et al. Circulating miRNA29 family expression levels in patients with essential hypertension as potential markers for left ventricular hypertrophy. Clin Exp Hypertens. 2017;39(2):119‐125. 10.1080/10641963.2016.1226889 [DOI] [PubMed] [Google Scholar]
- 166. Roncarati R, Viviani Anselmi C, Losi MA, et al. Circulating miR‐29a, among other up‐regulated microRNAs, is the only biomarker for both hypertrophy and fibrosis in patients with hypertrophic cardiomyopathy. J Am Coll Cardiol. 2014;63(9):920‐927. 10.1016/j.jacc.2013.09.041 [DOI] [PubMed] [Google Scholar]
- 167. Sassi Y, Avramopoulos P, Ramanujam D, et al. Cardiac myocyte miR‐29 promotes pathological remodeling of the heart by activating Wnt signaling. Nat Commun. 2017;8(1):1614. 10.1038/s41467-017-01737-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Shi JY, Chen C, Xu X, Lu Q. miR‐29a promotes pathological cardiac hypertrophy by targeting the PTEN/AKT/mTOR signalling pathway and suppressing autophagy. Acta Physiol. 2019;227(2):e13323. 10.1111/apha.13323 [DOI] [PubMed] [Google Scholar]
- 169. Wang, L , Niu X, Hu J, et al. After myocardial ischemia‐reperfusion, miR‐29a, and Let7 could affect apoptosis through regulating IGF‐1. BioMed Res Int. 2015;2015:245412‐245417. 10.1155/2015/245412 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Ye Y, Hu Z, Lin Y, Zhang C, Perez‐Polo JR. Downregulation of microRNA‐29 by antisense inhibitors and a PPAR‐gamma agonist protects against myocardial ischaemia‐reperfusion injury. Cardiovasc Res. 2010;87(3):535‐544. 10.1093/cvr/cvq053 [DOI] [PubMed] [Google Scholar]
- 171. Srivastava SP, Hedayat AF, Kanasaki K, Goodwin JE. microRNA crosstalk influences epithelial‐to‐mesenchymal, endothelial‐to‐mesenchymal, and macrophage‐to‐mesenchymal transitions in the kidney. Front Pharmacol. 2019;10:904. 10.3389/fphar.2019.00904 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Wynn TA. Cellular and molecular mechanisms of fibrosis. J Pathol. 2008;214(2):199‐210. 10.1002/path.2277 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. Chung AC, Lan HY. MicroRNAs in renal fibrosis. Front Physiol. 2015;6:50. 10.3389/fphys.2015.00050 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Fang, Y , Yu X, Liu Y, et al. miR‐29c is downregulated in renal interstitial fibrosis in humans and rats and restored by HIF‐alpha activation. Am J Physiol Renal Physiol. 2013;304(10):F1274‐F1282. 10.1152/ajprenal.00287.2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Wang, B , Komers R, Carew R, et al. Suppression of microRNA‐29 expression by TGF‐beta1 promotes collagen expression and renal fibrosis. J Am Soc Nephrol. 2012;23(2):252‐265. 10.1681/ASN.2011010055 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176. Wang, B , Wang J, He W, et al. Exogenous miR‐29a attenuates muscle atrophy and kidney fibrosis in unilateral ureteral obstruction mice. Hum Gene Ther. 2020;31(5‐6):367‐375. 10.1089/hum.2019.287 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Sarnak MJ, Levey AS, Schoolwerth AC, et al. Kidney disease as a risk factor for development of cardiovascular disease: a statement from the American Heart Association Councils on kidney in cardiovascular disease, high blood pressure research, clinical cardiology, and epidemiology and prevention. Circulation. 2003;108(17):2154‐2169. 10.1161/01.CIR.0000095676.90936.80 [DOI] [PubMed] [Google Scholar]
- 178. Drummond CA, Hill MC, Shi H, et al. Na/K‐ATPase signaling regulates collagen synthesis through microRNA‐29b‐3p in cardiac fibroblasts. Physiol Genomics. 2016;48(3):220‐229. 10.1152/physiolgenomics.00116.2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Kennedy DJ, Vetteth S, Periyasamy SM, et al. Central role for the cardiotonic steroid marinobufagenin in the pathogenesis of experimental uremic cardiomyopathy. Hypertension. 2006;47(3):488‐495. 10.1161/01.HYP.0000202594.82271.92 [DOI] [PubMed] [Google Scholar]
- 180. Drummond CA, Fan X, Haller ST, Kennedy DJ, Liu J, Tian J. Na/K‐ATPase signaling mediates miR‐29b‐3p regulation and cardiac fibrosis formation in mice with chronic kidney disease. PLoS One. 2018;13(5):e0197688. 10.1371/journal.pone.0197688 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181. Jia G, Hill MA, Sowers JR. Diabetic cardiomyopathy: an update of mechanisms contributing to this clinical entity. Circ Res. 2018;122(4):624‐638. 10.1161/CIRCRESAHA.117.311586 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182. Haugen E, Gan LM, Isic A, Skommevik T, Fu M. Increased interleukin‐6 but not tumour necrosis factor‐alpha predicts mortality in the population of elderly heart failure patients. Exp Clin Cardiol. 2008;13(1):19‐24. [PMC free article] [PubMed] [Google Scholar]
- 183. Liu F, Guo J, Zhang Q, et al. The expression of tristetraprolin and its relationship with urinary proteins in patients with diabetic nephropathy. PLoS One. 2015;10(10):e0141471. 10.1371/journal.pone.0141471 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184. Zhang Y, Wang JH, Zhang YY, et al. Deletion of interleukin‐6 alleviated interstitial fibrosis in streptozotocin‐induced diabetic cardiomyopathy of mice through affecting TGFbeta1 and miR‐29 pathways. Sci Rep. 2016;6:23010. 10.1038/srep23010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185. Arnold N, Koppula PR, Gul R, Luck C, Pulakat L. Regulation of cardiac expression of the diabetic marker microRNA miR‐29. PLoS One. 2014;9(7):e103284. 10.1371/journal.pone.0103284 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Murphy M, Docherty NG, Griffin B, et al. IHG‐1 amplifies TGF‐beta1 signaling and is increased in renal fibrosis. J Am Soc Nephrol. 2008;19(9):1672‐1680. 10.1681/ASN.2007101080 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. Chen HY, Huang XR, Wang W, et al. The protective role of Smad7 in diabetic kidney disease: mechanism and therapeutic potential. Diabetes. 2011;60(2):590‐601. 10.2337/db10-0403 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Fraser D, Brunskill N, Ito T, Phillips A. Long‐term exposure of proximal tubular epithelial cells to glucose induces transforming growth factor‐beta 1 synthesis via an autocrine PDGF loop. Am J Pathol. 2003;163(6):2565‐2574. 10.1016/s0002-9440(10)63611-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Sheetz MJ, King GL. Molecular understanding of hyperglycemia's adverse effects for diabetic complications. JAMA. 2002;288(20):2579‐2588. 10.1001/jama.288.20.2579 [DOI] [PubMed] [Google Scholar]
- 190. Chen HY, Zhong X, Huang XR, et al. MicroRNA‐29b inhibits diabetic nephropathy in db/db mice. Mol Ther. 2014;22(4):842‐853. 10.1038/mt.2013.235 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191. Zampetaki A, Kiechl S, Drozdov I, et al. Plasma microRNA profiling reveals loss of endothelial miR‐126 and other microRNAs in type 2 diabetes. Circ Res. 2010;107(6):810‐817. 10.1161/CIRCRESAHA.110.226357 [DOI] [PubMed] [Google Scholar]
- 192. Hsu YC, Chang PJ, Ho C, et al. Protective effects of miR‐29a on diabetic glomerular dysfunction by modulation of DKK1/Wnt/beta‐catenin signaling. Sci Rep. 2016;6:30575. 10.1038/srep30575 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193. Lin CL, Lee PH, Hsu YC, et al. MicroRNA‐29a promotion of nephrin acetylation ameliorates hyperglycemia‐induced podocyte dysfunction. J Am Soc Nephrol. 2014;25(8):1698‐1709. 10.1681/ASN.2013050527 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194. Long J, Wang Y, Wang W, Chang BH, Danesh FR. MicroRNA‐29c is a signature microRNA under high glucose conditions that targets Sprouty homolog 1, and its in vivo knockdown prevents progression of diabetic nephropathy. J Biol Chem. 2011;286(13):11837‐11848. 10.1074/jbc.M110.194969 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195. Guo J, Li J, Zhao J, et al. MiRNA‐29c regulates the expression of inflammatory cytokines in diabetic nephropathy by targeting tristetraprolin. Sci Rep. 2017;7(1):2314. 10.1038/s41598-017-01027-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196. Carvajal G, Rodríguez‐Vita J, Rodrigues‐Díez R, et al. Angiotensin II activates the Smad pathway during epithelial mesenchymal transdifferentiation. Kidney Int. 2008;74(5):585‐595. 10.1038/ki.2008.213 [DOI] [PubMed] [Google Scholar]
- 197. Huang XR, Chung ACK, Yang F, et al. Smad3 mediates cardiac inflammation and fibrosis in angiotensin II‐induced hypertensive cardiac remodeling. Hypertension. 2010;55(5):1165‐1171. 10.1161/HYPERTENSIONAHA.109.147611 [DOI] [PubMed] [Google Scholar]
- 198. Ma F, Li Y, Jia L, et al. Macrophage‐stimulated cardiac fibroblast production of IL‐6 is essential for TGF beta/Smad activation and cardiac fibrosis induced by angiotensin II. PLoS One. 2012;7(5):e35144. 10.1371/journal.pone.0035144 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. Ruiz‐Ortega M, Ruperez M, Esteban V, et al. Angiotensin II: a key factor in the inflammatory and fibrotic response in kidney diseases. Nephrol Dial Transplant. 2006;21(1):16‐20. 10.1093/ndt/gfi265 [DOI] [PubMed] [Google Scholar]
- 200. Wang W, Koka V, Lan HY. Transforming growth factor‐beta and Smad signalling in kidney diseases. Nephrology (Carlton). 2005b;10(1):48‐56. 10.1111/j.1440-1797.2005.00334.x [DOI] [PubMed] [Google Scholar]
- 201. Wang, W , Huang XR, Li AG, et al. Signaling mechanism of TGF‐beta1 in prevention of renal inflammation: role of Smad7. J Am Soc Nephrol. 2005a;16(5):1371‐1383. 10.1681/ASN.2004121070 [DOI] [PubMed] [Google Scholar]
- 202. Liu GX, Li YQ, Huang XR, et al. Disruption of Smad7 promotes ANG II‐mediated renal inflammation and fibrosis via Sp1‐TGF‐beta/Smad3‐NF.kappaB‐dependent mechanisms in mice. PLoS One. 2013;8(1):e53573. 10.1371/journal.pone.0053573 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203. Wei LH, Huang XR, Zhang Y, et al. Smad7 inhibits angiotensin II‐induced hypertensive cardiac remodelling. Cardiovasc Res. 2013;99(4):665‐673. 10.1093/cvr/cvt151 [DOI] [PubMed] [Google Scholar]
- 204. Palmer SC, Mavridis D, Navarese E, et al. Comparative efficacy and safety of blood pressure‐lowering agents in adults with diabetes and kidney disease: a network meta‐analysis. Lancet. 2015;385(9982):2047‐2056. 10.1016/S0140-6736(14)62459-4 [DOI] [PubMed] [Google Scholar]
- 205. Srivastava SP, Goodwin JE, Kanasaki K, Koya D. Inhibition of angiotensin‐converting enzyme ameliorates renal fibrosis by mitigating DPP‐4 level and restoring antifibrotic microRNAs. Genes (Basel). 2020;11(2). 10.3390/genes11020211 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206. Hu H, Hu S, Xu S, Gao Y, Zeng F, Shui H. miR‐29b regulates Ang II‐induced EMT of rat renal tubular epithelial cells via targeting PI3K/AKT signaling pathway. Int J Mol Med. 2018;42(1):453‐460. 10.3892/ijmm.2018.3579 [DOI] [PubMed] [Google Scholar]
- 207. Zhang Y, Huang XR, Wei LH, Chung AC, Yu CM, Lan HY. miR‐29b as a therapeutic agent for angiotensin II‐induced cardiac fibrosis by targeting TGF‐beta/Smad3 signaling. Mol Ther. 2014;22(5):974‐985. 10.1038/mt.2014.25 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208. Papadopoulou AS, Dooley J, Linterman MA, et al. The thymic epithelial microRNA network elevates the threshold for infection‐associated thymic involution via miR‐29a mediated suppression of the IFN‐alpha receptor. Nat Immunol. 2011;13(2):181‐187. 10.1038/ni.2193 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209. Fang J, Hao Q, Liu L, et al. Epigenetic changes mediated by microRNA miR29 activate cyclooxygenase 2 and lambda‐1 interferon production during viral infection. J Virol. 2012;86(2):1010‐1020. 10.1128/JVI.06169-11 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210. Haralambieva IH, Kennedy RB, Simon WL, et al. Differential miRNA expression in B cells is associated with inter‐individual differences in humoral immune response to measles vaccination. PLoS One. 2018;13(1):e0191812. 10.1371/journal.pone.0191812 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211. Houzet L, Jeang KT. MicroRNAs and human retroviruses. Biochim Biophys Acta. 2011;1809(11‐12):686‐693. 10.1016/j.bbagrm.2011.05.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212. Miller JF. Immunological function of the thymus. Lancet. 1961;2(7205):748‐749. 10.1016/s0140-6736(61)90693-6 [DOI] [PubMed] [Google Scholar]
- 213. Murphy KM, Reiner SL. The lineage decisions of helper T cells. Nat Rev Immunol. 2002;2(12):933‐944. 10.1038/nri954 [DOI] [PubMed] [Google Scholar]
- 214. Chandiran K, Lawlor R, Pannuti A, et al. Notch1 primes CD4 T cells for T helper type I differentiation through its early effects on miR‐29. Mol Immunol. 2018;99:191‐198. 10.1016/j.molimm.2018.05.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215. Sambandam A, Maillard I, Zediak VP, et al. Notch signaling controls the generation and differentiation of early T lineage progenitors. Nat Immunol. 2005;6(7):663‐670. 10.1038/ni1216 [DOI] [PubMed] [Google Scholar]
- 216. Smith KM, Guerau‐de‐Arellano M, Costinean S, et al. miR‐29ab1 deficiency identifies a negative feedback loop controlling Th1 bias that is dysregulated in multiple sclerosis. J Immunol. 2012;189(4):1567‐1576. 10.4049/jimmunol.1103171 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217. Steiner DF, Thomas MF, Hu JK, et al. MicroRNA‐29 regulates T‐box transcription factors and interferon‐gamma production in helper T cells. Immunity. 2011;35(2):169‐181. 10.1016/j.immuni.2011.07.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218. van Nieuwenhuijze A, Dooley J, Humblet‐Baron S, et al. Defective germinal center B‐cell response and reduced arthritic pathology in microRNA‐29a‐deficient mice. Cell Mol Life Sci. 2017;74(11):2095‐2106. 10.1007/s00018-017-2456-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219. Mott JL, Kobayashi S, Bronk SF, Gores GJ. mir‐29 regulates Mcl‐1 protein expression and apoptosis. Oncogene. 2007;26(42):6133‐6140. 10.1038/sj.onc.1210436 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220. Efanov A, Zanesi N, Nazaryan N, et al. CD5+CD23+ leukemic cell populations in TCL1 transgenic mice show significantly increased proliferation and Akt phosphorylation. Leukemia. 2010;24(5):970‐975. 10.1038/leu.2010.46 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221. Filip D, Mraz M. The role of MYC in the transformation and aggressiveness of 'indolent' B‐cell malignancies. Leuk Lymphoma. 2020;61(3):510‐524. 10.1080/10428194.2019.1675877 [DOI] [PubMed] [Google Scholar]
- 222. Zhang X, Zhao X, Fiskus W, et al. Coordinated silencing of MYC‐mediated miR‐29 by HDAC3 and EZH2 as a therapeutic target of histone modification in aggressive B‐Cell lymphomas. Cancer Cell. 2012;22(4):506‐523. 10.1016/j.ccr.2012.09.003 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 223. Luzzi A, Morettini F, Gazaneo S, et al. HIV‐1 Tat induces DNMT over‐expression through microRNA dysregulation in HIV‐related non Hodgkin lymphomas. Infect Agent Cancer. 2014;9:41. 10.1186/1750-9378-9-41 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224. Malpeli G, Barbi S, Tosadori G, et al. MYC‐related microRNAs signatures in non‐Hodgkin B‐cell lymphomas and their relationships with core cellular pathways. Oncotarget. 2018;9(51):29753‐29771. 10.18632/oncotarget.25707 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225. Nijhuis A, Curciarello R, Mehta S, et al. MCL‐1 is modulated in Crohn's disease fibrosis by miR‐29b via IL‐6 and IL‐8. Cell Tissue Res. 2017;368(2):325‐335. 10.1007/s00441-017-2576-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226. Xu L, Xu Y, Jing Z, et al. Altered expression pattern of miR‐29a, miR‐29b and the target genes in myeloid leukemia. Exp Hematol Oncol. 2014;3:17. 10.1186/2162-3619-3-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227. Chitnis T. The role of CD4 T cells in the pathogenesis of multiple sclerosis. Int Rev Neurobiol. 2007;79:43‐72. 10.1016/S0074-7742(07)79003-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228. Ma F, Xu S, Liu X, et al. The microRNA miR‐29 controls innate and adaptive immune responses to intracellular bacterial infection by targeting interferon‐gamma. Nat Immunol. 2011;12(9):861‐869. 10.1038/ni.2073 [DOI] [PubMed] [Google Scholar]
- 229. Hecker M, Thamilarasan M, Koczan D, et al. MicroRNA expression changes during interferon‐beta treatment in the peripheral blood of multiple sclerosis patients. Int J Mol Sci. 2013;14(8):16087‐16110. 10.3390/ijms140816087 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230. Hojati Z, Kay M, Dehghanian F. Chapter 15—mechanism of action of interferon beta in treatment of multiple sclerosis. In: Minagar A, ed. Multiple Sclerosis. Academic Press; 2016:365‐392. [Google Scholar]
- 231. Lu J, Liu J, Li L, Lan Y, Liang Y. Cytokines in type 1 diabetes: mechanisms of action and immunotherapeutic targets. Clin Transl Immunology. 2020;9(3):e1122. 10.1002/cti2.1122 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232. Bravo‐Egana V, Rosero S, Molano RD, et al. Quantitative differential expression analysis reveals miR‐7 as major islet microRNA. Biochem Biophys Res Commun. 2008;366(4):922‐926. 10.1016/j.bbrc.2007.12.052 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233. Roggli E, Gattesco S, Caille D, et al. Changes in microRNA expression contribute to pancreatic beta‐cell dysfunction in prediabetic NOD mice. Diabetes. 2012;61(7):1742‐1751. 10.2337/db11-1086 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234. Beltrami C, Simpson K, Jesky M, et al. Association of elevated urinary miR‐126, miR‐155, and miR‐29b with diabetic kidney disease. Am J Pathol. 2018;188(9):1982‐1992. 10.1016/j.ajpath.2018.06.006 [DOI] [PubMed] [Google Scholar]
- 235. Aghaei Zarch SM, Dehghan Tezerjani M, Talebi M, Vahidi Mehrjardi MY. Molecular biomarkers in diabetes mellitus (DM). Med J Islam Repub Iran. 2020;34:28. 10.34171/mjiri.34.28 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236. Ghosh N, Katare R. Molecular mechanism of diabetic cardiomyopathy and modulation of microRNA function by synthetic oligonucleotides. Cardiovasc Diabetol. 2018;17(1):43. 10.1186/s12933-018-0684-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237. Nathans R, Chu CY, Serquina AK, Lu CC, Cao H, Rana TM. Cellular microRNA and P bodies modulate host‐HIV‐1 interactions. Mol Cell. 2009;34(6):696‐709. 10.1016/j.molcel.2009.06.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238. Triboulet R, Mari B, Lin YL, et al. Suppression of microRNA‐silencing pathway by HIV‐1 during virus replication. Science. 2007;315(5818):1579‐1582. 10.1126/science.1136319 [DOI] [PubMed] [Google Scholar]
- 239. Yeung ML, Bennasser Y, Myers TG, Jiang G, Benkirane M, Jeang KT. Changes in microRNA expression profiles in HIV‐1‐transfected human cells. Retrovirology. 2005;2:81. 10.1186/1742-4690-2-81 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240. Houzet L, Yeung ML, de Lame V, Desai D, Smith SM, Jeang KT. MicroRNA profile changes in human immunodeficiency virus type 1 (HIV‐1) seropositive individuals. Retrovirology. 2008;5:118. 10.1186/1742-4690-5-118 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241. Witwer KW, Watson AK, Blankson JN, Clements JE. Relationships of PBMC microRNA expression, plasma viral load, and CD4+ T‐cell count in HIV‐1‐infected elite suppressors and viremic patients. Retrovirology. 2012;9:5. 10.1186/1742-4690-9-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242. Ortega PAS, Saulle I, Mercurio V, et al. Interleukin 21 (IL‐21)/microRNA‐29 (miR‐29) axis is associated with natural resistance to HIV‐1 infection. AIDS. 2018;32(17):2453‐2461. 10.1097/QAD.0000000000001938 [DOI] [PubMed] [Google Scholar]
- 243. Adoro S, Cubillos‐Ruiz JR, Chen X, et al. IL‐21 induces antiviral microRNA‐29 in CD4 T cells to limit HIV‐1 infection. Nat Commun. 2015;6:7562. 10.1038/ncomms8562 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244. Ahluwalia JK, Khan S, Soni K, et al. Human cellular microRNA hsa‐miR‐29a interferes with viral nef protein expression and HIV‐1 replication. Retrovirology. 2008;5:117. 10.1186/1742-4690-5-117 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245. Matsumoto Y, Itami S, Kuroda M, Yoshizato K, Kawada N, Murakami Y. MiR‐29a assists in preventing the activation of human stellate cells and promotes recovery from liver fibrosis in mice. Mol Ther. 2016;24(10):1848‐1859. 10.1038/mt.2016.127 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246. Millar NL, Gilchrist DS, Akbar M, et al. MicroRNA29a regulates IL‐33‐mediated tissue remodelling in tendon disease. Nat Commun. 2015;6:6774. 10.1038/ncomms7774 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247. Watts AE, Millar NL, Platt J, et al. MicroRNA29a treatment improves early tendon injury. Mol Ther. 2017;25(10):2415‐2426. 10.1016/j.ymthe.2017.07.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
