Skip to main content

Table 2 Therapeutic effects of mesenchymal stem cell-derived exosomes in different diseases in preclinical experimental models

From: Mesenchymal stem cell derived-exosomes: a modern approach in translational medicine

Condition/disease

Exosome source

Experimental model

Target mechanism(s)

Therapeutic effect(s)

Ref

Cardiovascular diseases

hBM-MSC

HUVEC cell

Improved proliferation, migration, and tube formation of endothelial cells in vitro

Promoted neoangiogenesis in vitro and in vivo

[50]

Rat MI

Improved cardiac indices, i.e. cardiac systolic/diastolic performance and blood flow

Reduced infarct size in vivo

mBM-MSC

Mouse HPH

Inactivated STAT3 pathway

Reduced vascular remodeling and HPH

[51]

Decreased the levels of miR-17 superfamily

Increased miR-204 levels in lung cells

Repressed the hypoxic pulmonary influx of macrophages and the induction of MCP1 and HIMF

rBM-MSC overexpressing CXCR4

Neonatal CM

Upregulated IGF1α and pAkt levels, inhibited caspase 3, and promote VEGF expression and tubulogenesis in vitro

Increased angiogenesis

[52]

Rat MI

Reduced infarct size

Improved cardiac remodeling

rBM-MSC

HUVEC cell

Enhanced tube formation by HUVEC cells

Decreased infarct size; preserved cardiac systolic and diastolic performance; enhanced the density of new functional capillary and blood flow recovery in vivo

[53]

Rat MI

Compromised T cell function by impeding cell proliferation in vitro

mBM-MSC

Mouse MI

miR-22-enriched exosomes were secreted after MI which reduced cardiomyocyte apoptosis by direct targeting of Mecp2

Reduced infarct size and cardiac fibrosis in vivo

[55]

rBM-MSC overexpressing GATA-4

Neonatal rat CM

miR-221-enriched exosomes reduced the expression of p53 while upregulating PUMA

 

[56]

Expression of PUMA was greatly declined in CM cocultured with MSC

rBM-MSC overexpressing GATA-4

Neonatal rat CM

Increased CM survival, reduced CM apoptosis, and preserved mitochondrial membrane potential in vitro

Exosomal miR-19a could restore cardiac contractile function and decreased infarct size in vivo

[57]

Rat MI

Exosomal miR-19a downregulated PTEN and triggered the Akt and ERK signaling pathways

mBM-MSC

HUVEC cell

Enhanced the proliferation, migration and tube formation in vitro

Promoted angiogenesis and cardiac function in vivo

[58]

Mouse MI

The pro-angiogenic effect of exosomes is probably associated with a miR-210-Efna3 dependent mechanism

hEn-MSC

Neonatal CM

Overexpression and shuttling of exosomal miR-21 was attributed to suppression of PTEN, stimulation of Akt, along with Bcl-2 and VEGF upregulation

Restored cardiac function and reduced infarct size

[59]

HUVEC cell

Rat MI

rBM-MSC

Cardiac stem cell

Triggered proliferation, migration, and angiotube formation in vitro probably mediated by a set of microRNAs

Reduced cardiac fibrosis in vivo

[60]

Rat MI

Enhanced capillary density

Restored long‐term cardiac function

Kidney diseases

hBM-MSC

mTEC

Exosomal mRNAs encoding CDC6, CDK8 and CCNB1 influenced cell cycle entry

Improved renal function and morphology

[61]

Mouse AKI

Exosomal miRNAs regulated proliferative/anti-apoptotic pathways and growth factors (HGF and IGF1) that led to renal tubular cell proliferation

hAD-MSC overexpressing GDNF

HUVEC cell

Triggered migration and angiogenesis in vitro

Reduced peritubular capillary rarefaction and renal fibrosis scores in vivo

[62]

Mouse ureteral obstruction

Conferred apoptosis resistance

Enhanced Sirtuin 1 signaling and p-eNOS levels

hBM-MSC

Rat IRI

Enhanced TEC proliferation and survival possibly via exosomal miRNA and mRNA molecules regulating renoprotective signaling routes

 

[63]

Gl-MSC

Mouse IRI

Activated TEC proliferation

Ameliorated kidney function

[64]

Reduced the ischemic damage post IRI

mK-MSC

HUVEC cell

Promoted cell proliferation in vitro and in vivo

Selective engraftment in ischemic tissues and significant improvement of renal function

[65]

Mouse IRI

Improved endothelial tube formation on growth factor reduced Matrigel

Expressed pro-angiogenic mRNA molecules encoding bFGF, IGF1 and VEGF

rAD-MSC

Rat IRI

Decreased expression of TNFα, NF-κB, IL1β, MIF, PAI1, Cox2 pro-inflammatory molecules

Reduced creatinine and BUN level, and improved renal function

[66]

Reduced the levels of NOX1, NOX2, and oxidized protein

Downregulated Smad3 and TGFβ fibrotic proteins

Enhanced Smad1/5 and BMP2 anti-apoptotic proteins

Upregulated CD31, vWF, and angiopoietin angiogenic biomarkers

Enhanced mito-Cyt C levels

rBM-MSC

Rat AKI

Enhanced IL10 levels

Decreased creatinine, urea, FENa, necrosis, apoptosis

[67]

Downregulated TNFα and IL6 expression

Increased cell proliferation

hWJ-MSC

HUVEC cell

Repressed NOX2 and ROS

Reduced fibrosis

[68, 69]

NRK-52E cell

Decreased apoptosis and sNGAL levels

Improved renal function

Rat IRI

Enhanced cell proliferation. Upregulated Nrf2/antioxidant response element and HO1 in vitro and in vivo

hWJ-MSC

NRK-52E cell

Upregulated autophagy-related genes such as ATG5, -7, and LC3B in vitro and in vivo

Improved renal function in vivo

[70]

Rat AKI

Induced mitochondrial apoptosis

Inhibited secretion of TNFα, IL1β, and IL6 pro-inflammatory cytokines in vitro

hWJ-MSC

NRK-52E cell

Reduced apoptosis and necrosis of proximal kidney tubules

Decreased BUN and creatinine levels

[71]

Rat AKI

Decreased production of tubular protein casts through anti-oxidation and anti-apoptosis pathways in vitro and in vivo

Promoted cell proliferation by activating the ERK1/2 pathway

hBM-MSC

PTEC cell

Promoted cell proliferation by carrying IGF1 receptor mRNA, but not IGF1 mRNA

 

[72]

Liver diseases

hWJ-MSC

HL7702 cell

Suppressing epithelial-to-mesenchymal transition in vitro and in vivo

Reduced LF

[73]

Mouse LF

Inactivated the TGFβ1/SMAD2 pathway

Alleviated hepatic inflammation and collagen deposition

Recovered serum AST function

Reduced collagen type I and III

hESC-MSC

TAMH, THLE-2, and HuH-7 cells

Upregulated PCNA and Cyclin D1 cell cycle proteins and anti-apoptotic Bcl-xL gene

Recovered ALI

[74]

Mouse ALI

hCP-MSC

Rat LF

Exosomal miR-125b blocked Smo production and inactivated Hedgehog signaling mode

Reduced expansion of progenitors and regressed LF

[75]

MiR‐122‐modified-hAD-MSC

Mouse LF

Exosomal miR-122 regulated the expression of IGF1R, Cyclin G1 (CCNG1) and P4HA1, which suppress HSC activation and collagen maturation

Suppressed LF development

[76]

Reduced the serum levels of HA, P‐III‐P, ALT, AST and liver hydroxyproline content

mBM-MSC

Mouse ALI

Reduced pro-inflammatory cytokines and apoptosis

Decreased the serum levels of ALT and liver necrotic areas

[77]

Upregulated anti-inflammatory cytokines

Triggered the number of Tregs

hWJ-MSC

Mouse ALI

Exosomal GPX1 cleared H2O2 and reduced apoptosis

Treated liver failure

[78]

h/mBM-MSC

Mouse LI

Exosomal Y-RNA-1 modulated cytokine expression and reduced peripheral inflammatory responses and apoptosis

Reduced hepatic injury and increased survival

[79]

Neurological diseases

hBM-MSC

Mouse stroke

Enhanced angioneurogenesis

Recovered postischemic neurological injury

[80]

Attenuated postischemic immunosuppression (i.e., B cell, NK cell and T cell lymphopenia) in the peripheral blood

Presented long term neuroprotection. Reduced motor coordination impairment

rBM-MSC

Rat stroke

Increased synaptophysin-positive regions in the ischemic boundary zone

Promoted neurovascular remodeling, axonal density and functional recovery

[81]

Enhanced the number of newly formed doublecortin and vW

rBM-MSC

Rat stroke

Exosomal miR-133b decreased the expression of connective tissue growth factor and ras homolog gene family member A

Resulted in neurite remodeling and stroke recovery

[82]

rBM-MSC overexpressing miR-17–92 cluster

Rat stroke

Inhibited PTEN and activated the downstream proteins, protein kinase B and glycogen synthase kinase 3β

Improved neurogenesis, neurite remodeling/neuronal dendrite plasticity and oligodendrogenesis

[83]

hBM-MSC

Rat BI

Attenuated inflammation-induced neuronal cellular degeneration

Improved long-lasting cognitive functions

[84]

Decreased microgliosis and prevented reactive astrogliosis

Restored short term myelination deficits and long term microstructural abnormalities of the white matter

hBM-MSC

Ewe BI

Reduced the neurological sequelae

Promoted brain function via decreasing the total number and duration of seizures

[85]

Did not affect cerebral inflammation

Preserved baroreceptor reflex sensitivity

rBM-MSC

Rat TBI

Enhanced angiogenesis, the number of newborn immature and mature neurons, and decreased neuroinflammation

Improvement of spatial learning

[86]

Recovered sensorimotor function

rB-MSCs

Mouse TBI

Suppressed the expression of pro-apoptotic Bcl-2-associated X protein, TNFα and IL1β

Reduced the lesion size and recovering neurobehavioral performance

[87]

Upregulated anti-apoptotic protein B-cell lymphoma 2

Modulated microglia/macrophage polarization

rBM-MSC

Rat SCI

Regulated macrophage function by targeting M2-type macrophages in the injured sites

 

[88]

rBM-MSC

Rat SCI

Reduced the proportion of A1 astrocytes via blocking the nuclear translocation of the NF-κB p65

Reduced lesion area

[89]

Reduced the percentage of p65 positive nuclei in astrocytes and TUNEL-positive cells in the ventral horn

Downregulated IL1α, IL1β and TNFα

Increased the expression of myelin basic protein, synaptophysin and neuronal nuclei

hBM-MSC

Rat SCI

Showed anti-inflammatory responses in the damaged tissue and disorganization of astrocytes and microglia

Improved locomotor activity

[90]

mBM-MSC

Mouse AD

Normoxic MSC exosomes: Decreased plaque deposition and Aβ levels

Normoxic MSC exosomes: Recovered cognition and memory impairment

[91]

Reduced the activation of astrocytes and microglia

Preconditioned MSC exosomes: Improved learning and memory capabilities

Downregulated TNFα and IL1β and upregulated IL4 and IL10

Deactivated STAT3 and NF-κB

Preconditioned MSC exosomes: Reduced plaque deposition and Aβ levels

Upregulated growth-associated protein 43, synapsin 1, and IL10

Decreased the levels of glial fibrillary acidic protein, ionized calcium-binding adaptor molecule 1, TNFα, IL1β

Deactivated STAT3 and NF-κB

Enhanced miR-21 levels

hAD-MSC

Mouse N2a cell

Exosomes carried enzymatically active neprilysin and decreased both secreted and intracellular Aβ levels

 

[92]

hDP-MSC

ReNcell VM human neural stem cell

Rescued dopaminergic neurons from apoptosis via inducing 6-hydroxy-dopamine

 

[93]

Wound healing

hWJ-MSC

EA.hy926 and HFL1 cells

Triggered propagation, migration, and tube formation in vitro

Improved wound healing in vivo

[94]

Rat skin burn

Stimulated β-catenin nuclear translocation

Upregulated proliferating cell nuclear antigen, cyclin D3, N-cadherin, and β-catenin

Downregulated E-cadherin

hWJ-MSC

Dermal fibroblast and HEK293T cell

Exosomal miR-21, ‐23a, ‐125b, and ‐145 inhibited scar formation and myofibroblast accumulation through TGFβ2/SMAD2 pathway blockade and reduction of collagen deposition in vitro and in vivo

 

[95]

Mouse skin-defect

hiPSC-MSC

HUVEC cell

Upregulated angiogenesis-related biomolecules

Increased microvessel density and blood perfusion

[96]

Mouse femoral artery excision

hWJ-MSC

Rat skin burn

Upregulated collagen I, PCNA and CK19

Resulted in rapid in vivo re-epithelialization

[97]

Exosomal Wnt4 contributed to β-catenin nuclear translocation and promotion of skin cell propagation and migration

Activated AKT pathway which reduced heat stress-induced apoptosis in vivo

hWJ-MSC

Rat skin burn

Decreased TNFα and IL1β levels and increased IL10 levels

 

[98]

Exosomal miR-181c decreased inflammation via suppressing the TLR4 signaling route

hAD-MSC

HUVEC cell

Promoted angiogenesis in vitro and in vivo

 

[99]

Immunodeficient mouse

Exosomal miR-125a acted as a pro-angiogenic factor by downregulating DLL4 and regulating the generation of endothelial tip cells

hiPSC-MSC

HUVEC cell and dermal fibroblast

Promoted collagen maturity and neoangiogenesis

Enhanced re-epithelialization

[100]

Rat skin wound

Triggered cell proliferation and migration in vitro

Decreased scar size

Increased type I, III collagen and elastin mRNA expression and secretion and tube formation in vitro

hBM-MSC

Diabetic wound and normal fibroblasts

Promoted fibroblast propagation and migration

 

[101]

Enhanced tube formation

Triggered Akt, ERK, and STAT3 signaling pathways

Upregulated HGF, IGF1, NGF and SDF1

Other diseases

hWJ-MSC and hBM-MSC

Mouse BPD

Triggered pleiotropic effects on gene expression related with hyperoxia -induced inflammation

Relieving BPD, hyperoxia-associated inflammation, fibrosis, pulmonary hypertension and pulmonary vascular remodeling in the lung tissue

[102]

Modulated the macrophage phenotype fulcrum, repressing the M1 state and promoting a M2-like state

hAD-MSC

Mouse atopic dermatitis

Decreased the levels of eosinophils, IgE, CD86+ and CD206+ cells, and infiltrated mast cells

Ameliorated atopic dermatitis in vivo

[103]

hBM-MSC

C2C12 and HUVEC cells

Exosomal miR-494 improved angiogenesis and myogenesis in vitro and in vivo

Resulted in muscle regeneration

[104]

Mouse muscle injury

hBM-MSC and hWJ-MSC

hPBMC

Enhanced the number of Tregs in vitro

Decreased educed the mean clinical score of EAE mice

[105]

Mouse EAE

Decreased PBMC proliferation and levels of pro-inflammatory Th1 and Th17 cytokines inclusive of IL6, IL12p70, IL17AF, and IL22

Decreased demyelination and neuroinflammation

Enhanced levels of indoleamine 2,3-dioxygenase

  1. amyloid β peptide, AD Alzheimer’s disease, AKI acute kidney injury, ALI acute liver injury, ALT alanine aminotransferase, AST aspartate aminotransferase, bFGF basic fibroblast growth factor, BPD: bronchopulmonary dysplasia, BMP2 bone morphogenetic protein 2, BUN blood urea nitrogen, CM cardiomyocyte, Cox-2 cyclooxygenase 2, DLL4 angiogenic inhibitor delta-like 4, DP-MSC dental pulp-derived MSC, EAE experimental autoimmune encephalomyelitis, ERK extracellular-signal-regulated kinase, FENa fractional excretion of sodium, GDNF glial cell line-derived neurotrophic factor, Gl-MSC glomeroli MSC, GPX1 glutathione peroxidase 1, HA hyaluronic acid, hCP-MSC human chorionic plate-derived MSC, hEn-MSC human endometrium-derived MSC, hESC-MSC human emberyonic stem cell-derived MSC, HGF hepatocyte growth factor, HIMF hypoxia-inducible mitogenic factor, hiPSC-MSC human induced pluripotent stem cell-derived MSC, HO1 heme oxygenase 1, hPBMC human peripheral blood mononuclear cell, HPH hypoxic pulmonary hypertension, HSC hepatic stellate cell, HUVEC human umbilical vein endothelial cell, IGF1α insulin-like growth factor 1α, IL1β interleukin 1β, IRI ischemia reperfusion injury, LF liver fibrosis, MCP1 monocyte chemoattractant protein 1, Mecp2 methyl CpG binding protein 2, MI myocardial infarction, MIF macrophage migration inhibitor factor, mito-Cyt C mitochondrial cytochrome C, mK-MSC mouse kidney-derived MSC, mTEC murine tubular epithelial cells, NF-κB nuclear factor κB protein, NGF nerve growth factor, NOX NADPH oxidase, P4HA1 prolyl-4-hydroxylase α1, PAI-1 protein expression of plasminogen activator inhibitor 1, PCNA proliferating cell nuclear antigen, p-eNOS phosphorylated endothelial nitric oxide synthase, P‐III‐P procollagen III‐N‐peptide, PTEC proximal tubular epithelial cell, PTEN phosphatase and tensin homolog, PUMA p53 upregulated modulator of apoptosis, rAD-MSC rat adipocyte-derived MSC, rB-MSC rat bone-derived MSCs, ROS reactive oxygen specie, SCI spinal cord injury, SDF1 stromal cell-derived factor 1, sNGAL serum neutrophil gelatinase-associated lipocalin, STAT3 signal transducer and activator of transcription 3, TBI traumatic brain injury, TGFβ transforming growth factor β, TNFα tumor necrosis factor α, Treg regulatory T cell, VEGF vascular endothelial growth factor, vWF von Willebrand factor