Skip to main content
. 2024 Mar 2;36:126–156. doi: 10.1016/j.bioactmat.2024.02.021

Table 5.

Recent advances in the applications of exosomes in regenerative medicine.

Disorder Exosome source Aim of using exosomes Regenerative medicine methodology In vivo/In vitro Ref.
Heart Cardiosphere-derived cells Improving cardiac functions after myocardial hypertrophy treatment Enhancing accumulation of exosomes by expressing heart homing peptide, miRNA-148a delivery, and inhibition of β-MHC, BNP, GP130, p-STAT3, p-ERK1/2, and p-AKT In vitro, in vivo [313]
Enhancing endocytosis of exosomes by binding cardiomyocyte-specific peptide Ligation of modified exosomes to cardiomyocyte-specific peptide In vitro, in vivo [314]
Modifying injured skeletal and cardiac muscle function Transcriptome profile reversion and increasing cardio myogenesis In vivo [315]
Improving the cardiac functions in DMD patients Reduction in collagen I and III levels, increase in cardiomyocyte proliferation and MYOD levels, restoration of dystrophin levels In vivo [316]
Cardiac regeneration Derived exosomes enriched in miR-146a, enhancing cell survival and angiogenesis In vitro, in vivo [317]
Hypoxia-pretreated Cardiosphere-derived cells Cardio-protection Upregulation miR-210, miR-130a, and miR-126 and angiogenesis In vitro [318]
MSCs Cardio-protection after ischemic injury HSF1 overexpressing MSCs and isolating miRNAs' enriched exosomes In vivo [319]
Reduction of infarct size Increasing ATP and NADH levels and phosphorylated-Akt and phosphorylated-GSK-3β, and decreasing oxidative stress and phosphorylated-c-JNK In vivo [320]
Human UCMSCs Myocardial protection by preventing apoptosis of myocardial cells Increasing in Bcl-2 expression In vitro, in vivo [321]
Cardiac regeneration after acute myocardial infarction Exosomal TGF-β3 could expand angiogenesis, diminish myocardial fibrosis, and preserve the heart function In vitro, in vivo [322]
Cardiac progenitor cells Apoptosis inhibitor Enriching in miRNAs that inhibit apoptosis or help the formation of the endothelial tube such as miR-210, miR-132, miR-146a-3p, and miR-181 In vitro, in vivo [323]
Cardiomyocytes Angiogenesis HSP20 association with Akt and ERK signaling pathways and VEGFR2 activation In vitro, in vivo [324]
HT1080 and cardiosphere-derived cells Targeting exosomes by cardiac homing peptide Target delivery of infracted heart, improve survival of neonatal rat cardiomyocytes, and vascularization In vitro, in vivo [325]
Atorvastatin-pretreated MSCs Cardio-protection IL-6 and TNF-α inhibition, regulation of miR-675 expression, activation of vascular endothelial growth factor, improve lncRNA H19 expression In vitro, in vivo [326]
Transduced MSCs with GATA-4 The effects of GATA-4 transduction on levels of miRs Increasing in miR-19a expression, decreasing in PTEN levels, activation of Akt and ERK signaling In vitro, in vivo [327]
Blood Cardio-protection HSP70 and toll-like receptor 4 communication and HSP27 activation Ex vivo [328]
Central nervous system Rat multipotent MSCs Improve hippocampal neurogenesis in rats of TBI Exosomes carrying miRNA-124 are correlated with M2 polarization of microglia via the TLR4 pathway In vivo [329]
Stimulate neurite outgrowth after stroke Exosomal transfer of miRNA-133b to neural cells In vitro [330]
Promote endogenous angiogenesis and neurogenesis and reduce neuroinflammation Correlated with suppression of activated microglia and macrophages by exosomes In vivo [331]
Human BMSCs Promote endogenous angiogenesis and neurogenesis and reduce neuroinflammation Correlated with suppression of activated microglia and macrophages by exosomes In vivo [332]
Promote retinal ganglion cells' survival and regeneration of their axons Knockout of Argonaute-2, a key miRNA effector molecule In vitro, in vivo [333]
Human UCMSCs Inhibition neural apoptosis, reduced inflammation and promoted neurological regeneration in rats after TBI. Suppression of NF-kB signaling pathway In vivo [334]
Peripheral nervous system Rat ASCs PNS regeneration, by reducing apoptosis Upregulation the anti‐apoptotic Bcl‐2 mRNA expression and downregulating the pro‐apoptotic Bax mRNA expression In vitro [335]
Promote regeneration of the myelin sheath Kpna2 downregulation via miR-25b In vitro, in vivo [336]
Murine ASCs Enhancing nerve regeneration after nerve crush injury Might be correlated with HDAC, APP and ITGB1, candidates involved in exosomes-mediated nerve regeneration In vivo [337]
Modulate the microenvironment in neuro-inflammatory and neurodegenerative disorders. Associated with inhibition of apoptotic cascade In vitro [338]
Human ASCs Promote neural survival and proliferation MALAT1 protein mediates the splicing of pkcδII, an anti-apoptotic protein In vitro [339]
Rat BMSCs Stimulate peripheral nerves' regeneration Closely related to expression of VEGFA and S100b genes via a miRNA-mediated mechanism In vitro, in vivo [340]
Dedifferentiated Schwann cells Increased axonal regeneration in vitro and enhanced regeneration after sciatic nerve injury in rat Inhibition of GTPase Rhoa activity, thereby inhibiting axonal elongation and promoting growth cone collapse after activation In vitro, in vivo [341]
Endothelial cells Boosting and maintaining the repair phenotypes of Schwann cells Stimulation of PI3K/AKT/PTEN signaling pathway In vitro, in vivo [342]
Gingiva-derived MSCs Peripheral nerve regeneration Closely related to activation of c-JUN activity, and upregulation of Notch1, GFAP and SOX2 In vivo [343]
Pericytes Promote angiogenesis and cavernous nerve regeneration under diabetic conditions Might be correlated with Lcn2 which acts activating MAP kinase and PI3K/Akt and suppressing P53 signaling In vivo [344]
Skin ASCs Molecular mechanism in skin wound healing Long noncoding RNA H19 targets miR-15 b y and this, in turn, targets SOX9, which activates the Wnt/β-catenin pathway In vitro, in vivo [345]
Skin wound healing miR-19b exosome regular the TFG-β pathway by CCL1 In vitro, in vivo [346]
Photoaging by UVB irradiation Upregulate the expression of type I collagen mRNA and downregulate the expression of type III collagen, MMP-1, and MMP-3 mRNA In vitro, in vivo [347]
Inflammatory response and skin wound healing Reduces the lipopolysaccharide-induced inflammatory mRNA and M1-type macrophage-specific marker expression and increases cytokines IL-10, VEGF, and TGF-β and M2-type macrophage marker Arg1 expression In vitro, in vivo [348]
Human platelet lysate Skin aging Reduces MMP-1 levels and, consequently, increases collagen levels In vivo [349]
Bovine milk Aging and skin hydration Hydrating effect on keratinocytes through the increase of filaggrin and CD44 receptor.
Hydrating effect on fibroblasts through the increase of HAS2. Prevents the decrease of type II and III collagen after exposure to UVB rays.
In vitro, in vivo [350]
Bovine colostrum Improves on aging and UV-induced damage to various skin cells Antioxidant effect on keratinocytes by reducing intracellular ROS through the glutathione oxidation pathway. Effect on elasticity by decreasing MMP2 expression. In vitro [351]
Solanum tuberosum Photoaging by UVB irradiation Inhibition action on MMP1, 2, and 9, as well as on the cytokines IL6 and TNF-α in keratinocytes. Antioxidant effect through the expression of glutathione S-transferase α 4 In vitro [352]
Lactobacillus plantarum Skin aging Decrease MMP-1 mRNA expression and elastase activity and increase filaggrin mRNA expression and HAS2 protein expression. In vitro, in vivo [353]
Apple Skin aging and reparation Negative effect on the activity of Toll-like Receptor 4 and NF-κB pro-inflammatory pathway In vitro [354]
Human amniotic fluid-derived stem cells Skin wound healing Decreased secretion of inflammation-associated cytokines through CXCR4 In vivo [355]
Human UCMSCs Molecular mechanism in skin wound healing PI3K/AKT pathway is regulated by phosphatase and tensin homolog In vitro [356]
BMSCs Photoaging by UVB irradiation Suppressed the mRNA of MMP-2 In vitro [357]
Hypoxia-pretreated ASCs Regenerative effects in UVB-induced skin injury circ-Ash 1l targets miR-700-5p and GPX4, and miR-700-5p target GPX4. In vitro, in vivo [358]
Epidermal stem cells Skin wound healing May inhibit the differentiation of fibroblasts to myofibroblasts via suppressing TGF-β1 expression via miR-425-5p and miR-142-3p In vivo [359]
Human dermal fibroblast Photoaging by UVB irradiation Increased procollagen type I expression and a decrease in MMP-1 expression, mainly through the downregulation of TNF-α and the upregulation of TGF-β In vitro, in vivo [360]
Acellular gelatinous Wharton's jelly of the human umbilical cord Mechanism of action in skin wound healing. Could be related to the paracrine effects of alpha-2-macroglobulin In vitro, in vivo [361]
Human pluripotent stem cells Photoaging by UVB irradiation and natural senescence Decreases mRNA expression of MMP-1 and MMP-3
Increases the expression of type I collagen mRNA.
In vitro [362]
Muscle ASCs Regeneration of skeletal muscle defect Enhancing myocyte proliferation as well as MYOG and MYOD genes In vitro, in vivo [363]
Muscle regeneration Increasing the number of centrally located nuclei In vivo [364]
C2C12 myoblasts Promoting the musculoskeletal repair and regeneration Exosomes derived from mechanically strained C2C12 cells improved cell proliferation and differentiation In vitro [365]
Reveal the mechanism of interactions between exosomes and muscle regeneration Increasing the levels of Pax7; an increase on day 3 and decrease on day 5 of peroxisome proliferator-activated receptor γ (PPARγ) levels, α-SMA, Collagen-1 In vivo [366]
Detecting endocrine signal role of exosomes Regulating miRNAs, which are important for cell differentiation and growth control as well as myogenesis In vitro [367]
M2 macrophages The role of exosomes derived from M2 macrophage on muscle regeneration Exosomes enriched in miR-501, targeting Yin Yang 1, and increasing the levels of MyHC and MyoG In vitro, in vivo [368]
PRPs and MSCs Functional recovery of injured muscle Increase in the expression of MYOG in PRP-exosome group; reduction of TGF-β in MSC-exosomes group; no effects on MYOD and IL-1β levels In vivo [369]
MSCs Muscle regeneration Exosomes enriched in miR-21, miR-1, miR-133, miR-206, and miR-494 enhance tube formation and improve vascularization; increase myofibers diameter and centrally located nuclei; decrease fibrotic area; increase MYOD and MYOG expressions In vitro, in vivo [370]
Human skeletal myoblasts, differentiating to myotube Muscle regeneration Enrich in growth factors such as IGFs, VEGF, HGF, NT-3, FGF2, and PDGF-AA; upregulation of FGF2, TNF, MYOD1, DAG1, DES, MYH1/2, and TNNT1 In vitro and in vivo [371]
Liver
Hepatocyte-derived cells Promoting liver regeneration after acute liver failure The miR-183-5p uptake leads to the activation of FoxO1/Akt/GSK3β/β-catenin signaling In vitro, in vivo [372]
Hepatocytes Hepatocyte proliferation and regeneration after acute hepatic injury Exosomal transfer of SK2 to target hepatocytes In vitro, in vivo [373]
Human UCMSCs Antioxidant and antiapoptotic effects, rescuing liver from failure Might be correlated with GPX1 In vivo [374]
Human BMSCs Promote anti-fibrosis by stimulating hepatocyte regeneration Suppression of Wnt/β-catenin signaling components (PPARγ, Wnt3a, Wnt10b, β-catenin, WISP1, Cyclin D1) In vivo [375]
ASCs Liver regeneration after hepatic ischemia-reperfusion in rats Activation of Wnt/β-catenin signaling In vivo [376]
Human Placenta-derived MSCs
Upregulate angiogenesis and liver regeneration Might be related with Wnt/β-catenin signaling triggered by C-reactive protein In vitro, in vivo [377]
Promote cell proliferation and liver regeneration after hepatectomy
Related with the exosomal circ-RBM23 mopping miR-139-5p, activating eIF4G expression and AKT/mTOR pathway
In vitro, in vivo
[378]
Vessel
Endothelial progenitor cells Reendothelialization elevating angiogenesis genes levels such as HIF-1a, VEGF family, eNOS, E-selectin, IL8, ANG1, CXCL family; down-regulation of MMP-9 and PDFGB In vitro, in vivo [379]
Investigating the mechanism of vascularization by derived exosomes Delivering miR-21-5p and targeting angiogenesis inhibitor Thrombospondin-1 In vitro, in vivo [380]
Induced vascular progenitor cells angiogenesis Enhance endothelial tube formation, cell migration, and proliferation; enrich in miR-143-3p, miR-291b, miR-20b-5p, and IGF-binding protein In vitro, ex vivo, and in vivo [381]
MSCs cultured on titanium Vascular regeneration Improve VEGFR2 level and cell migration; downregulation of miR-15b-5p, miR-16-5p, miR-155-5p, miR-24-3p, miR-32-5p, mir-125b-5p, miR-146a-5p, and miR-320a In vitro [382]
MSCs Identifying the angiogenesis factors of exosomes Induce proangiogenic via PDGF, EGF, FGF, and NFkB pathways In vitro [383]
ASCs Improving angiogenesis properties of EVs by PDGF Enrich in proangiogenic factors such as MMP, c-kit, and SCF; enhance endothelial tube formation In vitro, in vivo [384]
Annulus fibrous cells Investigation of vascularization mechanism Enhance endothelial cells migration and increase IL-6, TNF-α, MMP-3, MMP-13 and VEGF expressions In vitro [385]
hypoxia-resistant multiple myeloma cell line Improving angiogenesis Upregulation of miR-210 and miR-135b, which miR-135b targets inhibiting hypoxia-inducible factor-1 In vitro, in vivo [386]
Leukemia cells in hypoxia Endothelial tube formation Elevation of several miRNAs such as miR-18b and miR-210 In vitro [387]
Lung cancer cells in hypoxia Reveal the exosomal communication of cancer cells and endothelial cells Elevation of miR-23a, suppression of t prolyl hydroxylase 1 and 2, increase in hypoxia-inducible factor-1α (HIF-1α) level, and downregulation of tight junction protein ZO-1 In vitro, in vivo [388]
Overexpressed miR-21 ASCs
The role of miR-21 on vascularization
Increasing in HIF-1α, VEGF, SDF-1, p-Akt, p-ERK1/2 and decreasing in PTEN levels
In vitro
[389]
Cartilage MSCs Enhance proliferation, attenuate apoptosis, and modulate immune reactivity Activation of AKT and ERK signaling, and higher infiltration of CD163+ M2 macrophages In vitro, in vivo [390]
Synovial MSCs Enhance proliferation and migration in vitro and prevent osteoarthritis Exosomal transfer of miRNA-140-5p to target cells. Might also be related with Wnt signaling In vitro, in vivo [391]
Human BMSCs Inhibit inflammatory mediators, promoting cartilage regeneration in vitro Abolishment of TNF-alpha-mediated upregulation of COX2 In vitro [392]
Subcutaneous MSCs Ameliorate the pathological severity degree of cartilage Delivery of miR-199a-3p-mediates mTOR-autophagy pathway In vitro, in vivo [393]
KGN-pre-treated BMSCs Chondral matrix formation and cartilage repair Could be related to targeting C-myc and further regulating the MAPK signaling pathway In vitro, in vivo [394]
Ovary Amniotic fluid stem cells POI miR-369-3p inhibition of expression YAF2 and PDCD5/p53 in OGCs In vitro, in vivo [395]
POI Anti-apoptosis and proliferative effect in OGCs by PI3K/AKT/mTOR pathway In vitro, in vivo [396]
POI Anti-apoptosis on OGCs through regulation of miR-146a and miR-10a pathway In vitro, in vivo [397]
BMSCs POI Anti-apoptosis effect on OGCs through miR-144-5p suppressing PTE gene expression and this in turn increases PI3K/AKT pathway In vitro, in vivo [398]
POI Anti-apoptosis effect on OGCs through miR-664-5p inhibiting p53 luciferase activity In vitro, in vivo [399]
Amniotic fluid POI Antifibrotic effect by increasing SMAD6, which in turn inhibits the TGF-β signaling pathway In vivo [400]
Ovarian tissue POI Anti-apoptosis effect in OGC by regulating BCL9 expression by miR-122-5p inhibitor In vitro, in vivo [401]
Menstrual blood-derived stromal cells POI and mechanism ovulation Follicular development through increased A-azoospermia Like, proliferation of OGCs through increased forkhead box L2 In vitro, in vivo [402]
Human UCMSCs Inhibited apoptosis, increased OR, and recoopered the function of POI Anti-apoptosis and proliferation effect in OGC through miR-17-5p inhibiting SIRT7 and target gene (γH2AX, PARP1 and XRCC6) In vitro, in vivo [403]
Human ASCs POI Ovarian function is regulated through the SMAD pathway, which in turn inhibits the expression of apoptosis genes (Fas, FasL, caspase-3, and caspase-8) In vitro, in vivo [404]
Skeleton MSCs osteogenesis Improve the bone formation and expressions of angiogenesis genes such as VEGF, ANG 1, ANG2, COL1, and ALP In vitro, in vivo [405]
Fabricating cell-free scaffold for bone regeneration Increasing the levels of osteogenic factors such as osteopontin, ALP, Hsa-miR-146a-5p, HsamiR-503-5p, Hsa-miR-483-3p, Hsa-miR-129-5p; decreasing the levels of anti-osteogenic miRNAs such as Hsa-miR-32-5p, Hsa-miR-133a-3p, and Hsa-miR-204-5p; activation of PI3K/Akt and MAPK signaling pathways In vitro [406]
Improving bone healing Enrich in miR-4532, -125b-5p, −4516, -338-3p, and −548aa In vitro, in vivo [407]
BMSCs Osteogenesis and bone tissue targeting Enhancing the osteogenic activity of BMSCs due to elevation of miR-26a, −29a, −218, −34a, and −3960; enhancing bone localization of exosomes due to conjugation with BMSC-specific aptamer In vitro, in vivo [408]
Promoting angiogenesis and regulating of osteoclast-related activities during bone remodeling process miR-26a can influence bone formation through the Tob gene, which acts as a negative regulator of the BMP/SMAD pathway In vitro, in vivo [409]
human UCMSCs Reduction of cell apoptosis Elevation of miR-186, miR-1304, miR144, miR-1263, and miR-302b levels; regulation of apoptosis by inhibition Hippo signaling pathway In vitro, in vivo [410]
Fracture healing Elevation of β-catenin, Wnt3a, Col1, OPN, and RUNX2 In vivo [411]
ASCs Osteogenesis effects of cell-free scaffold incorporated with exosomes Improve cell migration, RUNX2, ALP, COL1A1 expressions of MSCs cultured in the osteogenic medium; enhance new bone formation In vitro, in vivo [412]
Noggin-suppressed MSCs Increasing bone healing efficacy Downregulation of noggin; upregulation of ALP, RUNX2, Osterix, and OCN; inhibition of miR-26 In vitro, in vivo [413]
miR‐375‐overexpressing ASCs Improving bone regeneration with miR-375 Exosomes were enriched in miR-375; improved osteogenesis of BMSCs, increase ALP, COL1A1, and RUNX2 levels as well as AZR quantification In vitro, in vivo [414]
miR-21 transfected Human Wharton's jelly of UCMSCs Decreasing osteocytes apoptosis Elevation of miR-21-PTEN-AKT signaling pathway In vitro, in vivo [415]
HIF-α overexpressing BMSCs Improving bone healing with HIF-α Increasing the levels of HIF-α, ALP, and OCN In vitro, in vivo [416]
MSCs derived from human induced pluripotent stem cells Improving osteogenesis and angiogenesis Improving osteogenic differentiation and vascularization; Increase in ALP, RUNX-2, and COL1A1 levels In vitro, in vivo [417]
MSCs derived from human induced pluripotent stem cells Angiogenesis Increasing microvessels and cell necrosis inhibition; upregulation of PI3K/Akt signaling pathway In vitro, in vivo [418]
TNF-α preconditioned ASCs Improving bone regeneration efficacy of exosomes Upregulation of Wnt3a, RUNX2, osteopontin, and bone sialoprotein In vitro [419]
Endothelial progenitor cells Bone healing via angiogenesis Enrich in miR-126; downregulation of SPRED1; regulation of Raf/ERK signaling pathway In vitro, in vivo [420]

Abbreviations: Adipose-derived stem cells, ASCs; Bone marrow mesenchymal stem cells, BMSCs; Mesenchymal stem cells, MSCs; Umbilical cord mesenchymal stem cells, UCMSCs; Traumatic brain injury, TBI; Chemokine CC motif ligand 1, CCL1; Duchenne muscular dystrophy, DMD; Hyaluronidase 2, HAS2; Interleukin 10, IL-10; Heat shock proteins, HSP; heat shock factor 1, HSF1; Matrix metalloproteinase-1, MMP1; Matrix metalloproteinase-2, MMP2; Matrix metalloproteinase-3, MMP3; insulin-like growth factors, IGFs; vascular endothelial growth factor, VEGF; hepatocyte growth factor, HGF; neurotrophin-3, NT-3; fibroblast growth factor-2, FGF2; Platelet-derived growth factor-AA, PDGF-AA; Reactive oxygen species, ROS; Transforming growth factor beta, TGF-β; Tumor necrosis factor-alpha, TNF-α; Interleukin-6, IL-6; Bone morphogenetic proteins, BMP; Ultraviolet radiation, UV; Ultraviolet B, UVB; vascular endothelial growth factor, VEGF; microARN, miARN; ovarian granulosa cells, OGCs; Premature ovarian insufficiency, POI; Platelet-Rich Plasma, PRP; mother against decapentaplegic-related proteins, SMADs; SMAD family member 6, SMAD6.