Skip to main content

Table 2 Mechanisms of non-coding RNA in mesenchymal stromal cell-based therapy for knee osteoarthritis

From: Mesenchymal stromal cell-based therapy for cartilage regeneration in knee osteoarthritis

Source

Target

Amount

Axis/signaling pathway

Function

References

Human AT-MSCs

In vitro

400 µg/mL

miR-145 and miR-221

Downregulated the expression of pro-inflammatory markers IL-6, NF-κB, and TNF-α, while upregulated the expression of the anti‐inflammatory cytokine IL‐10

[109]

Human OA cartilage-derived MSCs and BM-MSCs

Mice

NR

miR-365

Activation of aggrecan and collagen type 2a1 gene expression. MiR-365 expression was activated by chondrogenic induction in both OA-MSCs and BM-MSCs

[139]

Human BM-MSCs-Exos

In vitro

NR

miR-520d-5p/HDAC1

MiR-520d-5p promoted MSCs chondrogenesis and regulates chondrocyte metabolism through targeting HDAC1

[140]

Human BM-MSCs

In vitro

NR

miR-410/Wnt3a

MiR-410 was elevated during TGF-β3-induced chondrogenic differentiation of MSCs, and regulated the Wnt signaling pathway

[135]

Rat BM-MSCs-Exos

Rat

NR

miR-9-5p/syndecan 1

Anti-inflammatory and chondroprotective effects of BM-MSC-derived exosomal miR-9-5p on KOA via regulation of syndecan 1

[110]

Human BM-MSCs-Exos

Rat

250 ng/5 µL

miR-26a-5p/Cox2

Human BM-MSC-Exos overexpressing miR-26a-5p serve as a repressor for damage of synovial fibroblasts via Cox2 in KOA

[112]

Human SMSCs-Exos

Rat

30 µL, 1011 particles/mL

miR-26a-5p/PTEN/IL-1β

SMSC-exos enhanced IL-1β-induced cell proliferation, whereas inhibited apoptosis and inflammation. MiR-26a-5p targeted PTEN, for which overexpression spoiled the protection of exosomes against IL-1β-induced cell damage

[111]

SMSCs-Exos

Mice

5 µL

miR-31/KDM2A/E2F1/PTTG1

SMSC-Exos and Exos from miR-31-overexpressed SMSCs alleviated cartilage damage and inflammation in KOA in vivo

[91]

Human AT-MSCs-Exos

Mice

10 µL, 1010 particles/mL

miR-100-5p/mTOR

The level of miR-100-5p decreased the luciferase activity of mTOR 3′UTR, while inhibition of miR-100-5p could reverse the MSC-Exos-decreased mTOR signaling pathway

[97]

Rat BM-MSCs-Exos

Nude mice

20 µg

miR-127-3p/ CDH11/Wnt/β-catenin

MiR-127-3p targeted CDH11 and over-expressed CDH11 in chondrocytes weakened the therapeutic effect of exosomes. IL-1β treatment resulted in the activation of the Wnt/β-catenin pathway in chondrocytes

[96]

Rat BM-MSCs-Exos

Rat

100 µL, 1011 particles/mL

miR-135b/MAPK6

MiR-135b promoted M2 polarization of synovial macrophages through targeting MAPK6

[117]

Rat MSCs-Exos

Rat

100 µL, 1011 particles/mL

miR-135b/Sp1/TGF-β1

TGF-β1 stimulation enhanced miR-135b expression in MSC-exosomes, and MSC-exosomes-derived miR-135b increased the cell viability of C5.18 cells via downregulated Sp1 expression

[92]

Human BM-MSCs-Exos

Mice

100 µL, 1011 particles/mL

miRNA-136-5p/ELF3

An increased ELF3 expression and reduced miR-136-5p expression were detected in the clinical samples of traumatic OA cartilage tissues. BM-MSC-derived exosomal miR-136-5p could promote chondrocyte migration in vitro and inhibit cartilage degeneration in vivo

[101]

Human AT-MSCs-Exos

Mice

NR

miR-124/NF-κB and miR-143/ ROCK1/TLR9

MiR-143 and miR-124 inhibited the expression of NF-κB and ROCK1 in OA cells. In addition, the 3’ UTRs of NF-κB and ROCK1 were proven to contain the binding sites for miR-143 and miR-124, respectively

[138]

Rat BM-MSCs-Exos

Rat

200 µg

miR-216a-5p/JAK2/STAT3

Hypoxic-Exos promoted the proliferation and migration of chondrocytes and inhibited their apoptosis by targeting functional miR-216a-5p to chondrocytes and then downregulating JAK2. In addition, HIF-1α induces hypoxic BM-MSCs to release Exos

[93]

Human BM-MSCs-Exos

Rat

2 µg

miR-361-5p/DDX20/NF-κB

MiR-361-5p was verified to directly target DDX20. Additionally, human BM-MSC-Exos-transferred miR-361-5p alleviates chondrocyte damage and inhibits the NF-κB signaling pathway

[137]

Human SMSCs-Exos

BALB/C mouse

30 µL, 1011 particles/mL

miR-155-5p/Runx2

The SMSC-155-5p-Exos prevented KOA. Overexpression of Runx2 partially reversed the effect of the SMSC-155-5p-Exos

[136]

MSCs-Exos

In vitro, co-culture with mouse chondrocytes

NR

circRNA_HIPK3/miR-124-3p/MYH9

MSCs-Exos overexpressing circHIPK3 improved IL-1β-induced chondrocyte injury. Mechanistically, circHIPK3 could directly bind to miR-124-3p and subsequently elevate the expression of the target gene MYH9

[143]

Human MSCs

In vitro

NR

lncRNA HOTAIRM1-1/miR-125b/ BMPR2; JNK/MAPK/ERK pathway

HOTAIRM1-1 was downregulated in KOA cartilages and may inhibit MSCs viability, induce apoptosis, and suppress differentiation via regulating miR-125b/BMPR2 axis JNK/MAPK/ERK pathway may be a possible downstream mechanism to mediate the role of HOTAIRM1-1 in OA development

[146]

Human BM-MSCs-Exos

In vitro

NR

lncRNA HOTTIP/miR-455-3p/CCL3

HOTTIP negatively regulated miR-455-3p and increased CCL3 levels in human chondrocytes

[147]

Human AT-MSCs

In vitro

NR

circRNA_ATRNL1/miR‐145‐5p/SOX9

Circ_ATRNL1 regulated the promotion of SOX9 expression to promote chondrogenic differentiation of human AT-MSCs mediated by miR‐145‐5p

[141]

Human BM-MSCs-Exos

Mice

10 µL, 500 µg/mL

circRNA_0001236/miR-3677-3p/Sox9

Exosomal circRNA_0001236 enhanced the expression of Col2α1 and SOX9, but inhibited MMP13 in chondrogenesis via targeting miR-3677-3p and Sox9

[142]

Human BM-MSCs

In vitro

NR

lncRNA GRASLND

Silencing of lncRNA GRASLND resulted in lower accumulation of cartilage-like extracellular matrix, while GRASLND overexpression significantly enhanced cartilage matrix production

[144]

Human SMSCs

In vitro

NR

lncRNA MEG3/EZH2-mediated H3K27me3/TRIB2

LncRNA MEG3 regulated chondrogenic differentiation by inhibiting TRIB2 expression through EZH2-mediated H3K27me3

[145]

Human and mouse MSCs

In vitro

NR

lncRNA EPB41L4A‐AS1 and lncRNA SNHG7/miR‐146a

MiR‐146a significantly inhibited BM-MSCs proliferation partly interacting with lncRNA EPB41L4A‐AS1 and lncRNA SNHG7

[148]

Human BM-MSCs-Exos

In vitro

NR

lncRNA LYRM4-AS1/GRPR/miR-6515-5p

IL-1β significantly decreased cell viability, promoted apoptosis, and upregulated the expression of MMP3, AKT, and GRPR, while Exos reversed the changes

[149]

  1. OA Osteoarthritis, KOA knee osteoarthritis, MSCs mesenchymal stromal cells, BM bone marrow, AT adipose tissue, Exos exosomes, NR not reported, IL interleukin, NF-κB nuclear factor-kappaB, TNF-α tumor necrosis factor-α, TGF-β transforming growth factor-β, UTR untranslated regions of mRNA, CDH11 cadherin-11, SMSCs synovial-derived mesenchymal stromal cells, Runx2 Runt-related transcription factor 2, MAPK mitogen-activated protein kinases, DDX20 Asp-Glu-Ala-Asp (DEAD)-box polypeptide 20, ROCK1 Rho-associated kinase 1, TLR9 Toll-like receptor 9, mTOR mechanistic target of rapamycin, JAK2 Janus kinase 2, STAT3 signal transducer and activator of transcription 3, PTEN phosphatase and tensin homolog, HDAC1 histone deacetylase 1, Cox2 cyclooxygenase-2, ELF3 E74-like factor 3, SOX9 sex-determining region of Y chromosome-box transcription factor 9, Col2α1 α-1 chain of procollagen type 2, MMP matrix metalloproteinase, MYH9 myosin heavy chain 9, TRIB2 tribbles homolog 2, BMPR2 bone morphogenetic protein receptor 2, JNK p38 and c-jun N-terminal kinase, ERK extracellular signal-regulated kinase, CCL3 macrophage inflammatory protein 1-α, AKT protein kinase B, GRPR gastrin-releasing peptide receptor