Editorial
Copyright ©The Author(s) 2024.
World J Stem Cells. Jun 26, 2024; 16(6): 623-640
Published online Jun 26, 2024. doi: 10.4252/wjsc.v16.i6.623
Table 1 Typical pre-clinical and clinical studies involving mesenchymal stem cells primed using different strategies
Ref.
Model type
Cell source and pre-treatment
Main findings
Pre-treatment of MSCs by physical manipulation
Kim et al[17], 2009In vitro studiesBM-MSCs pre-treated with IP (with two cycles of 30 min anoxia/reoxygenation)IP significantly reduced apoptosis in MSCs through activation of Akt [Ser(473)] and ERK1/2 [Thr(202)/Tyr(204)] and nuclear translocation of HIF-α. There was simultaneous induction of miR-210 in the IP MSCs. RT-PCR array for rat apoptotic genes, computational target gene analyses, and luciferase reporter assay identified FLICE-associated huge protein/caspase-8-associated protein-2 in (PC)MSCs as the target gene of miR-210, responsible for improved cell survival
Fang et al[24], 2019In vitro studyStretch pre-treatment of AD-MSCsHuman ADSCs were subjected to cyclic stretch stimulation, significantly promoting their proliferation, adhesion, and migration. It also reduced cellular apoptosis but inhibited adipogenesis and increased osteogenesis. Long-term stretch promotes cell aging but without any size or morphological changes. Stretching also caused the induction of PI3K/AKT and MAPK pathways
Bianconi et al[28], 2023A femur defect rat modelElecS in 2D and 3D culturesA femur defect rat model was used to assess the healing effects of MSCs pre-treated by ElecS in 2D and 3D cultures for one hour/day for seven days. The bone healing effect was evaluated at one, four-, and eight weeks post-surgery. In all groups, the percentage of new bone increased, while fibrous tissue and CD68+ cell count were reduced. However, these and other healing features, like mineral density, bending stiffness, the amount of new bone and cartilage, and the gene expression of osteogenic markers, did not significantly differ between groups
Li et al[29], 2023In vitro studyUC-MSCs pre-exposed to (2% O2) hypoxia + treatment with IL-1β, TNF-α, INF-γ for 24 hCombined pretreatment with hypoxia and inflammatory factors changed UC-MSC morphology without changing viability, proliferation, or size. Also, pretreatment did not alter surface marker expression or mitochondrial function and integrity. However, pretreatment promoted UC-MSC apoptosis and senescence. Interestingly, immune regulation-related genes and protein expression significantly increased. These molecular changes increased peripheral blood mononuclear cell and NK cell proliferation and reduced NK cell-induced toxicity
Liu et al[30], 2021Mice model of ICH-induced brain injury using collagenase IVHypoxic OM-MSCs treated with hypoxia (3% O2) for 48 hThis study investigated the neuroprotective effects of hypoxia-preconditioned OM-MSCs in treating ICH in mice. Hypoxia-pretreated OM-MSCs reduced microglial activation and IL-1β and TNF-α. Also, there was a significant reduction in pyroptosis and pyroptosis-associated proteins in peri-hematoma brain tissues. Molecular studies showed reduced microglial NLRP3 expression, caspase-1, and reduced membrane pores on microglia after ICH
Dong et al[31], 2021In vitro studyMacrophages co-cultured with BM-derived MSCs with mechanical stretchMacrophages co-cultured with MSCs with mechanical stretch efficiently induced osteogenic differentiation of MSCs. Cyclical stretch caused macrophages to be polarized to the anti-inflammatory M2 phenotype, inducing IL-10 and TGF-β expression. YAP activation and nuclear translocation also caused BMP2 expression to facilitate MSC osteogenesis
Romanek et al[32], 2018In vitro studyPorcine MSCs under HHP of 20, 30, 40, 50, or 60 MPa (1 h at 24 °C)Porcine BM-MSCs were cultured in vitro and, before cryopreservation, subjected to HHP, i.e., 20, 30, 40, 50, or 60 MPa for 1 h at 24 °C. Immediately after thawing and on day 8, the cells were assessed for survival and proliferation. MSCs subjected to 40, 50, and 60 MPa showed improved survival vs the control, while cells exposed to 40 MPa HHP had higher proliferation than the control group after eight days of culture
Hu et al[33], 2016In vitro and in vivo studyCynomolgus monkey MSCs were exposed to 0.5% oxygen for 24 hMSCs were exposed to 0.5% oxygen (HP-MSCs) for 24 h and later used to treat MI in cynomolgus monkeys. Hypoxia pretreatment increased the expression of pro-survival/pro-angiogenic factors in vitro. Post-transplantation, they reduced the infarct size and LVEF on day 90 after treatment compared to the control group. This was also accompanied by higher cardiomyocyte proliferation, blood vessel density, myocardial glucose uptake, and engraftment of the transplanted cells. There were no arrhythmogenic changes
Sun et al[34], 2016In vitro studyBM-derived MSCs and ligament tissue-derived fibroblast under the uniflex/bioflex culture systemThe cells were uniaxially or radially stretched under 5%, 10%, and 15% strains at 0.1, 0.5, and 1.0 Hz. Exposure to uniaxial stretch (15% at 0.5 Hz; 10% at 1.0 Hz) increased proliferation and collagen production in fibroblast. On the other hand, the uniaxial strains (5%, 10%, and 15%) at 0.5 Hz and 10% strain at 1.0 Hz were favorable for MSCs. Radial strain did not affect fibroblast, but radial strains 5%, 10%, and 15% at 0.1 Hz increase MSC proliferation. The data supported the differential response of the cells depending on cell type under different types of mechanical stress
Wang et al[35], 2013In vitro studyCyclical compressive stress on rat BM-derived MSCsDynamic cyclical compressive stress remarkably increased MSCs quantity and viability during early chondrogenesis, increasing cyclin D1, CDK4, and Col2α1. MEK/ERK and p38 MAPK were not activated, but BMP signaling was activated in mechanotransduction for chondrogenic proliferation
Pre-treatment with chemicals and pharmacological agents
Qazi et al[36], 2022Rat model of MI3D-cultured rat BM-MSCs and treated with zebularineMSCs were cultured on a collagen scaffold and, after treatment with zebularine for cardiac differentiation, were used to treat the rat MI model. Compared to the MI control, there was a significant improvement in cardiac function in the zebularine-treated scaffold-cultured group. A significant reduction in a fibrotic scar and an improvement in LV wall thickness preserved LV remodeling. Blood vessel density in and around the infarct area was also improved
Aslam et al[37], 2020In vitro studiesUC-MSCs pre-treated with IHThe scratch assay showed a decreased scratch area in the case of IH-treated MSCs at 24 h, extending to complete closure of the scratch area at 48 h. Histological analysis showed reduced inflammation and completely remodeled epidermis and dermis without scar formation. There was a time-dependent reduction in IL-1β and IL-6. There was a simultaneous increase in Bcl-2 and TGF-β, VEGF, Bcl-2, and MMP-9, with increased angiogenesis and reduced inflammation and apoptosis
Li et al[38], 2015Rat model of AMIAtorvastatin-treated rat BM-MSCsAtorvastatin pretreatment induced CXCR4 expression in MSCs and supported their emigrational potential. When delivered intravenously in a rat model of AMI, the cells homed into the infarcted myocardium, participated in myocardial repair, and preserved global cardiac function
Liu et al[39], 2014Rat model of AMIHIF-α prolyl hydroxylase inhibitor DMOGDMOG pre-treatment of BM-MSCs significantly enhanced the expression of pro-survival and pro-angiogenic factors, including HIF-1α, VEGF, glucose transporter 1, and phospho-Akt. DMOG-treated MSCs also survived better than naïve MSCs post-engraftment in the rat model of AMI, in addition to increasing blood vessel density in and around the infarcted myocardium
Shinmura et al[40], 2011Nude rat model of AMIPioglitazone pre-treated human MSCsMSCs pretreated with pioglitazone were injected two weeks after MI. Pretreatment with pioglitazone significantly improved change in LVFS. Immunohistochemistry showed increased cardiomyogenic transdifferentiation of the transplanted cells and improved global cardiac function
Suzuki et al[41], 2010In vitro studiesDiazoxide pre-treated BM-MSCsTreatment of MSCs with DZ (200 μM) induced NF-κB-dependent miR-146a expression to support cell survival. Abrogation of miR-146a expression using an antisense miR-146a inhibitor abolished DZ-induced cytoprotective effects. The computational analysis demonstrated a consensus putative target site of miR-146a in the 3’ untranslated region of Fas mRNA regulating cell apoptosis. A Luciferase reporter assay revealed forced expression of miR-146a downregulated Fas expression
Pre-treatment of MSCs with cytokines and growth factors
Chen et al[42], 2023In vitro studieshUC-MSCs pre-treated with IFN-γ & TNF-α alone or combined in a colitis mice modelTreatment with IFN-γ alone increased PD-L1 in hUC-MSCs, while TNF-α alone did not. Co-treatment with IFN-γ and TNF-α increased PD-L1 expression. IFN-γ also activated the JAK/STAT1 signaling pathway, increased the IRF1 transcription factor, promoted the binding of IRF1 and the PD-L1 gene promoter, and finally increased PD-L1 mRNA. TNF-α significantly enhanced IFN-γ-induced JAK/STAT1/IRF1 activation. TNF-α increased IFN-γ receptors via the NF-κB signaling pathway, significantly enhancing IFN-γ signaling. Finally, co-treatment inhibited lymphocyte proliferation, reduced mucosal damage, inflammatory cell infiltration, and up-regulation of inflammatory factors in colitis mice
Chen et al[43], 2023In vitro studiesRat BM-MSCs treated with SDF-1αCXCR4 expression was observed on BM-MSCs by immunofluorescence staining. Treatment with SDF-1α increased collagen X and MMP13 expression during cartilage differentiation but with no change in collagen II or aggrecan. SDF-1α treated MSCs were validated in primary chondrocytes. SDF-1α increased p-GSK3β and β-catenin in MSCs. Abrogation of this pathway using ICG-001 (5 μmol/L) abrogated the SDF-1α-mediated up-regulation of collagen X and MMP13 expression in MSCs
Esmaeili et al[44], 2021Rat model of AMIRat BM-MSCs overexpressing VEGF and pre-treated with SDF-1αSDF-1α pre-treatment significantly reduced LDH release in MSCs, significantly thus increasing their survival more than the naïve control MSCs. The LVEF was improved considerably with a concomitant reduction in the infarct size in the animals in SDF-1α pretreated cells compared to the other treatment groups of animals
Lu et al[45], 2009In vitro studies and Rat model of AMIBM-derived Sca-1+ cells exposed to OGD and pre-treatment with IGF-1Exposure to OGD for up to 12 h activated Erk1/2 in Sca-1(+) cells. Moreover, higher intracellular calcium with simultaneous PKC activation was also observed. Pretreatment with nifedipine or dantrolene reduced cellular calcium, abrogated PKC, and Erk1/2 activation. Pretreatment with 100 nM IGF-1 increased cell resistance to ischemia via Erk1/2 activation to improve their survival under OGD and post-engraftment of the infarcted heart
Hahn et al[46], 2008Rat model of AMIRat BM-MSCs pre-treated with FGF-2 and IGF-1Pre-treatment of MSCs with bFGF + IGF1 increased the expression of cardiac transcription factors and survival. Transplantation of the pre-treated cells in a rat myocardial infarction model reduced infarct size and improved global cardiac function compared to untreated MSCs. Pre-treatment with growth factors enhanced gap junction formation in the transplanted MSCs without any arrhythmias
Pre-treatment of MSCs by genetic manipulation
Li et al[47], 2018Isoproterenol-induced heart failure model in ratsMSCs overexpressing ADMTransplantation of ADM-MSCs significantly improved heart function and reduced the size of the fibrotic area. Fluorescence microscopy revealed that ADM-MSCs survived considerably better in the heart. ADM-MSC treatment also improved heart function through enhanced antifibrotic activity
Gómez-Mauricio et al[48], 2016Porcine heart model of MIPorcine adipose tissue-derived MSCs genetically modified for HGF-1 and IGF-1I/M delivery of MSCs with IGF-1 and HGF-1 was safe. Inflammation was significantly reduced in some myocardial sections analyzed. There was a significant increase in blood vessel density in ischemic tissue. Although cardiac function parameters were not significantly improved, cell retention and IGF-1 overexpression were confirmed within the myocardium. Concomitant IGF-1- and HGF overexpression promoted a synergistic effect
Gnecchi et al[49], 2009Rat model of acute MIMSCs overexpressing Akt1Akt-MSCs, or PBS, were used to treat rats with experimental MI. High energy metabolism and basal 2-DG uptake were evaluated on isolated hearts using phosphorus-31 NMR 72 h and two weeks after MI. Treatment with Akt-MSCs increased 2-DG uptake in the residual intact myocardium vs PBS or the naïve MSC treatment. Also, Akt-MSC-treated hearts had normal pH and functional recovery after MI, thus showing that Akt-MSCs preserved normal metabolism and pH in the surviving myocardium
Haider et al[50], 2008Rat model of acute MIRat BM-MSCs overexpressing IGF-1Overexpression of IGF-1 led to enhanced phosphoinositide 3-kinase, Akt, and Bcl-xL and inhibition of glycogen synthase kinase 3beta besides the release of SDF-1α in BM-MSCs. Intramyocardially transplantation of IGF-expressing MSCs of the transplanted MSCs with massive mobilization and homing of ckit(+), MDR1(+), CD31(+), and CD34(+) cells into the infarcted heart. Infarction size was significantly reduced vs control. There was extensive angiomyogenesis in the infarcted heart and improved LVEF
Pre-treatment of stem cells in clinical trials
Bartunek et al[51], 2013C-CURE Clinicaltrial.gov ID: NCT00810238hBM-MSCs pre-treated with a cocktail of bioactive moleculesIn 100% of cases, treatment using MSCs pre-treated with a cardiopoietic cocktail was without complications. Cardiopoietic cell therapy did not induce systemic toxicity. LVEF was significantly improved compared to the standard therapy without cell treatment. Cell therapy also increased the 6-min walk distance, improving the New York Heart Association functional class, quality of life, and physical performance
Bartunek et al[52], 2016CHART-1 Clinicaltrial.gov ID: NCT01768702Cardiopoietic MSCs C3BS-CQR-1Patients (n = 351) with symptomatic advanced HF with reduced LVEF (< 35%) were randomized to receive C3BS-CQR-1 or a sham procedure. Treatment with C3BS-CQR-1 resulted in a significant progressive reduction in LVEDV and LVESV during a 52-wk follow-up. Interestingly, the most considerable reverse remodeling was observed in the patients receiving moderate injections (< 20)
Qayyum et al[53], 2017MyStromalCell trial. ClinicalTrials.gov ID: NCT01449032VEGF-A165-stimulated adipose-derived stromal cells ASCsThe MyStromalCell trial is a randomized, double-blind, placebo-controlled study in sixty patients with refractory angina, CCS/NYHA class II-III, LVEF > 40%, and at least one significant coronary artery stenosis. ASCs were culture-expanded and primed with VEGF-A165. Six months of follow-up showed that the treatment was safe and improved exercise tolerance compared to placebo
Table 2 Typical examples of growth factors, bioactive molecules, chemicals, and pharmacological agents used to pre-treat mesenchymal stem cells
GF, cytokines, & other bioactive molecules
Transgenes for genetic modulation of MSCs
Chemicals and pharmacological agents
AdrenomedullinAdrenomedullinCobalt chloride
Angiopoietin-1Akt2,4-dinitrophenol
Angiotensin-IIAkt + angiopoietinAliskiren
Basic fibroblast GFBcl2Atorvastatin
Epidermal GFCXCR4, CXCR7Diazoxide
GCSFGATA4Deferoxamine
Hepatocyte GF-1Focal adhesion kinaseDimethyloxalylglycine
Insulin-like GF-1Hepatocyte GF-1Glucagon-like protein-1
Interferon-γHypoxia-inducible factorHydrogen peroxide
Interleukin1-βInsulin-like GF-1Lipopolysaccharide
Interleukin 6Integrin subunit-alpha4Nicorandil
MelatoninKallikerin-1Pioglitazone
OxytocincMycSalvianolic acid
Platelet-derived GFOct4Simvastatin
Stromal cell-derived factor 1αProtein kinase CSevoflurane
Thymosin β4Sox2Sodium butyrate
Transforming GF-βStromal cell-derived factor 1αTadalafil
Tumor necrosis factor-αTumor necrosis factor-α receptorTrimetazidine
Vascular endothelial GFVascular endothelial GFTrace elements like Zn
Vitamin EVarious microRNAsValproic acid
Table 3 Studies for further reading on physical manipulation and genetic priming strategies
Ref.
Model type
Cell source and pre-treatment
Main findings
Pre-treatment of MSCs by physical manipulation
Izadpanah et al[103], 2022In vitro5-Aza treatment + static and microfluidic cell culture systems5-Aza induced cardiac-specific markers in MSCs, but this induction was significantly increased after exposure to both 5-Aza and shear stress, showing their synergistic effects vs 5-Aza or in shear stress-only groups. These results demonstrated that MSCs’ exposure to 5-Aza and shear stress is required for high-level cardiac gene expression
Manjua et al[104], 2021In vitro/in vivo models for angiogenesisMSCs exposed to magnetic pre-treatmentMSCs cultured on polyvinylalcohol and gelatin-based scaffolds containing iron oxide nanoparticles were exposed to a magnetic field. The cells showed significantly increased VEGF-A production and altered their morphology and alignment. MSCs’ angiogenic potential was observed by the increase in angiogenic response using conditioned media in vitro and in vivo
Helms et al[105], 2020In vitroAD-SCs pre-treated with TSB or mechanical stimulation or their combined actionThe study was intended to show if mechanical stimulation can support or replace TSB-induced differentiation of Ad-SCs. ASC or pre-differentiated SMC exposed to pulsatile perfusion for ten days with or without TSB resulted in collagen-I expression and circumferential orientation of the cells around the lumen. Molecular studies showed upregulation of αSMA and calponin expression. On the other hand, contractility and smoothelin expression required both mechanical and TSB stimulation
Vaez et al[106], 2018BM-MSCs in static 2D and microfluidic cell culture systemsThere was a clear but insignificant difference between the beating rate of APCs and CNCs in both 2D and the microfluidic system during 30 d. Data from RT-PCR showed GATA4, Nkx2.5, CX43, cTnI, cTnT, and β-MHC induction during four weeks more in microfluidic chips than those co-cultured in 24-well plates. Combined shear stress and co-culture with cardiomyocytes significantly enhanced the differentiation rate vs co-culture alone
Popa et al[107], 2016In vitrohAD-SCs pre-treated by MNPs integrated in κC hydrogelsThe MNP concentration in the κC hydrogels significantly influenced the cell viability, cell content, and metabolic activity. The optimal MNP concentration was 5% in κC. Exposure to magnetic actuation further altered their gene expression profile, favoring chondrogenic phenotype induction
Shi et al[108], 2011In vitroMSCs’ exposure to CCMTRhoA activity after CCMT stimulation was reduced. Pre-treatment of CCMT-stimulated MSCs with LPA, a RhoA activator, recovered ALP activity and Runx2 expression. In contrast, pre-treatment with C3 toxin, a RhoA inhibitor, reduced ALP activity with a concomitant reduction in Runx2. These results showed inhibition of Runx2 expression after the RhoA-ERK1/2 pathway mediates CCMT stimulation
Liu et al[109], 2011hMSCs under perfusion culture system to produce FSShMSCs subjected to a perfusion culture system to produce FSS, which activated ERK1/2. The pre-treatment enhanced the pro-osteogenic gene expression profile in the cells via activating NF-κB and BMP. FSS inducing the osteogenic differentiation of hMSCs will provide new targets for osteoporosis and related bone-wasting diseases
Kasten et al[110], 2010 In vitroBM-MSCs subjected integrin integrin-induced and inhomogeneous magnetic force exposureExposure to inhomogeneous magnetic forces increased Sox 9 (a marker of chondrogenesis) and decreased ALP expression. Molecular studies showed that VEGF induction induced by physical forces involved Akt activation. The results showed that the biological functions of MSCs can be stimulated by pretreatment with integrin-mediated mechanical forces and inhomogeneous magnetic field exposure
Pre-treatment of MSCs by genetic manipulation
Li et al[111], 2023In vitro and mice model of PDhMSCs overexpressing VEGF189hMSC overexpressing VEGF189-GFP significantly increased VEGF expression and slightly increased viability of the cells vs naïve cells. Transplantation of VEGF expressing MSCs significantly improved mechanical allodynia and inhibited the site’s TRPV1 expression. TRPV1 agonists could partially block such pain relief effects. There was no tumorigenicity or neuron degeneration in hMSCs expressing VEGF189-GFP
Yu et al[112], 2023In vitro and in vivo mice model of alkali-burned corneaAD-MSCs overexpressing IGF-1Treatment with MSCs overexpressing IGF-1 significantly recovered corneal morphology and function vs control and IGF-1 protein eyedrops. The healing of corneal epithelium and limbus, the inhibition of corneal stromal fibrosis, angiogenesis, and lymphangiogenesis, and the repair of corneal nerves were observed. In vitro experiments showed that MSCs with IGF-1 promoted trigeminal ganglion cell activity and maintained limbal stem cells’ stemness
Singh et al[113], 2018 In vitroPharmacological and genetic manipulation of MSCs to enhance survivinInduction of survivin is essential for MSC survival, expansion, lineage commitment, and migrational potential. On the other hand, pharmacological or genetic blockade of survivin expression in mouse and human BM-MSC increased caspase 3 and 7 expression and reduced proliferation, resulting in fewer MSC and clonogenic colony-forming unit-fibroblasts, growth factor (i.e., b-FGF or PDGF)-mediated survivin modulation represents a novel therapeutic strategy
Konoplyannikov et al[114], 2013In vitro and in vivo in rat model of MISimultaneous overexpression of IGF-1, VEGF, sSDF-1a, HGF-1 in SKMOverexpression of four growth factors led to the induction of multiple angiogenic and pro-survival factors, including secreted frizzled-related protein-1,2,4,5, matrix metalloproteinases-3 and 9, connexin-43, netrin-1, Nos-2, Wnt-3, Akt, MAPK42/44, Stat3, NFκB, HIF-1α, and protein kinase C. Transplantation of the genetically modified cells causes extensive neomyogenesis and angiogenesis in the infarcted heart, attenuating infarct size and improving global heart function at eight weeks vs control animals. There was also massive mobilization and homing of stem/progenitor cells from the peripheral circulation, the bone marrow, and the heart for participation in infarcted myocardium repair
Jiang et al[115], 2006In vitro and in vivo study in rat model of MIRat BM-MSCs are co-overexpressing Ang-1 and AktMSCs co-overexpressing Ang-1 and Akt survived better under anoxia vs naïve MSCs. At two weeks after cell transplantation, MAAs survived significantly more than the naïve MSCs in the infarcted heart. The heart function indices were significantly improved LVEF and fractional shortening vs control
Ye et al[116], 2005In vitro and in vivo using a rat model of acute MISKMs genetically modulated to overexpress VEGFThe genetically modified cells expressed copious amounts of VEGF. Transplantation of the cells into the infarcted heart significantly increased blood vessel density compared to control animals. LVEF and fractional shortening were improved considerably compared to control-treated animals, and regional flow improved