Review Open Access
Copyright ©The Author(s) 2023. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Stem Cells. May 26, 2023; 15(5): 369-384
Published online May 26, 2023. doi: 10.4252/wjsc.v15.i5.369
Stimulating factors for regulation of osteogenic and chondrogenic differentiation of mesenchymal stem cells
Jia-Qi Zhou, Hao-Yang Wan, Zi-Xuan Wang, Nan Jiang, Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
ORCID number: Nan Jiang (0000-0003-2416-1653).
Author contributions: Zhou JQ and Wan HY contributed equally to this study; Jiang N conceived and designed the study; Zhou JQ searched the literature; Zhou JQ and Wan HY drew the figure; Zhou JQ and Wang ZX drafted the article; Wan HY and Jiang N made critical revisions; all the authors approved the final version of the submitted article.
Conflict-of-interest statement: All authors declare that they have no competing interests.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Nan Jiang, MD, Researcher, Surgeon, Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No.1838 Guangzhou Avenue North, Baiyun District, Guangzhou 510515, Guangdong Province, China. hnxyjn@smu.edu.cn
Received: January 8, 2023
Peer-review started: January 8, 2023
First decision: February 2, 2023
Revised: February 21, 2023
Accepted: March 29, 2023
Article in press: March 29, 2023
Published online: May 26, 2023

Abstract

Mesenchymal stem cells (MSCs), distributed in many tissues in the human body, are multipotent cells capable of differentiating in specific directions. It is usually considered that the differentiation process of MSCs depends on specialized external stimulating factors, including cell signaling pathways, cytokines, and other physical stimuli. Recent findings have revealed other underrated roles in the differentiation process of MSCs, such as material morphology and exosomes. Although relevant achievements have substantially advanced the applicability of MSCs, some of these regulatory mechanisms still need to be better understood. Moreover, limitations such as long-term survival in vivo hinder the clinical application of MSCs therapy. This review article summarizes current knowledge regarding the differentiation patterns of MSCs under specific stimulating factors.

Key Words: Mesenchymal stem cells, Differentiation, Osteogenic, Chondrogenic, Literature review.

Core Tip: Mesenchymal stem cells (MSCs) are multipotent cells capable of differentiating in specific directions. The differentiation process of MSCs depends on diverse specialized external stimulating factors. The results from recent studies have revealed other underrated roles in the differentiation process of MSCs. However, several questions remain to be solved prior to stable and effective clinical treatment. Our review explores the differentiation patterns of MSCs and summarizes the relevant research according to stimulus types. Finally, future prospects are discussed with regard to their clinical applications.



INTRODUCTION

Mesenchymal stem cells (MSCs), which were originally identified in the bone marrow, are adult stem cells with multilineage differentiation potential. Under specific induction conditions, MSCs could differentiate into bone, adipose, muscle, neural, and endothelial tissue cells[1]. With the development of research, MSCs have been obtained from other tissues, including adipose, peripheral blood, umbilical cord blood, and periodontal membrane tissue[2-5]. Due to their multilineage differentiation potential and rich tissue sources, the application of MSCs in research on regenerative medicine is virtually limitless[6]. However, a specific number of MSCs are necessary for tissue regeneration; hence, there is a requirement for MSC amplification before therapy[7]. The question of how the differentiation of MSCs are controlled in vitro and in vivo remains unanswered, which has limited the effectiveness of MSCs in the application of regenerative medicine research. Recently, various external stimulus factors, such as biochemical stimuli, hypoxia, physical stimuli, material properties, and exosomes, have been found to have an impact on the differentiation process of MSCs (Figure 1). The purpose of this review is to discuss a variety of recent findings regarding the important external stimulus factors that influence the self-renewal and osteogenic and chondrogenic differentiation potential of MSCs.

Figure 1
Figure 1 Overview of stimulating factors in differentiation of mesenchymal stem cells. MSC: Mesenchymal stem cell; IL: Interleukin; FGF: Fibroblast growth factor; TGF-β: Transforming growth factor-β.
BIOCHEMICAL STIMULI

Growth factors, cytokines, and miRNAs are examples of biochemical stimuli that have typically been employed to control the destiny of MSCs. Growth factors and cytokines bind to the corresponding receptors and transfer signals, while miRNAs degrade mRNAs or inhibit the translation of mRNAs to regulate gene expression and thus influence the differentiation fate of MSCs. Numerous studies have examined the effects of various growth factors, cytokines, and miRNAs on the proliferation and differentiation of MSCs into other cellular phenotypes (Table 1).

Table 1 Growth factors, cytokines, and their effects on the differentiation of mesenchymal stem cells.
Factors
Amount/types
Concentration
Cell dource
Results
Ref.
FGFFGF-210 ng/mlBM-PACsFGF-2 did not lead to cell differentiation into a chondrogenic lineageEndo et al[9]
bFGF5 ng/mlSMSCsPromoted SMSCs chondrogenic differentiationOkamura et al[10]
bFGF0-40 ng/mlUC-MSCsbFGF did not alter osteogenic nor adipogenic differentiation potentialRamasamy et al[12]
bFGF20 ng/mlBMSCsbFGF pretreatment inhibited osteogenic differentiation of BMSCs at early stage, promoted it in the medium phase, and maintained it in the later stage during osteogenic inductionWang et al[13]
TGF-βTGF-β310 ng/mlSF-MSCsIncreased the expression levels of COL2A1, SOX9, ACAN, COL10A1Jia et al[15]
TGF-β10 ng/mlADSCsPromoted ADSCs chondrogenic differentiation but led to early hypertrophic maturationHesari et al[2]
TGF-β11, 10, 20 or 50 ng/mlBMSCsLow concentration of TGF-β1 (1 ng/ml) promoted osteogenic differentiation of BMSCs while high concentration of TGF-β1 (10 to 50 ng/ml) significantly inhibited osteogenesisXu et al[17]
TGF-β5 ng/mlBMSCsPromoted osteogenic differentiation of BMSCs but suppressed the maturation of ostroblastic MSC differentiation at the last stage of osteogenic processIgarashi et al[18]
TGF-β310 μg/LPDLSCsInduced chondrogenesisChoi et al[5]
ILIL-6100 ng/mLBMSCsPromoted BMSCs osteogenic differentiationXie et al[21]
IL-17A5-40 ng/mlBMSCsPromoted the neuronal-associated gene expression of BMSCsChen et al[24]
IL-1750 ng/mLMouse MSCsEnhanced the osteogenic differentiation of mMSCsLiao et al[22]
IL-6100 ng/mLhMSCsIL-6/soluble IL-6R promoted chondrogenic differentiation of MSCsKondo et al[20]
IL-17A50 ng/mlBMSCsInhibited osteogenic differentiation of BMSCsWang et al[23]
IL-2210 ng/mlMSCsUpregulated osteogenic and adipogenic transcription factorsEl-Zayadi et al[25]
Growth factors

Growth factors, including fibroblast growth factor (FGF), transforming growth factor (TGF), platelet-derived growth factor, hepatocyte growth factor, granulocyte colony-stimulating factor and bone morphogenetic protein (BMP), are a class of peptides that regulate cell growth and other cell functions by binding to specific cell membrane receptors[8].

FGF-2, also known as basic bFGF, has been the subject of the majority of FGF research to date. In a concentration-dependent manner, bFGF might promote the proliferation of MSCs from several tissue sources, including bone marrow peri-adipocyte cells[9], synovial MSCs[10], adipose-derived stem cells (ADSCs)[11], umbilical cord-derived MSCs[12], and bone MSCs (BMSCs)[13,14]. Ramasamy et al[12] reported that cell proliferation increased accordingly with increasing bFGF concentrations in the range of 0-40 ng/mL. However, Ma et al[11] and Wang et al[14] observed that the proliferation efficiency of cells at 5 ng/mL of bFGF was higher than that at 10 ng/mL. As a result, the use of 5 ng/mL of bFGF appeared to be an appropriate choice to promote the proliferation of different MSCs. In addition to enhancing MSC proliferation, bFGF has been shown to maintain stemness, support cartilage differentiation, and influence osteogenic differentiation[9,10,13]. Intriguingly, Wang et al[14] reported that bFGF pretreatment inhibited osteogenic differentiation at the early stage, but promoted it in the medium phase[13]. This finding might indicate that the addition of different growth factors at different phases of osteogenesis induction could successfully promote osteogenic differentiation. Therefore, more studies are needed to clarify the mechanism of action of bFGF at different stages of osteogenic differentiation, as well as to identify the best combination of growth factors to effectively promote the osteogenic differentiation of MSCs.

Previous research has demonstrated the involvement of TGF-β in inducing chondrogenic differentiation[5]. However, while promoting cartilage differentiation, TGF-β also led to early hypertrophic maturation and the eventual formation of nonfunctional fibrocartilage[2,15]. In addition, TGF-β was also found to promote the proliferation of MSCs and their effect on osteogenic differentiation[16,17]. MSC osteogenic differentiation was influenced by TGF-β in a dose-dependent manner. According to research by Xu et al[17], low concentrations of TGF-β (1 ng/mL) promoted the osteogenic development of BMSCs, whereas high concentrations (10–50 ng/mL) of TGF-β inhibited osteogenic differentiation. Igarashi et al[18] showed that 5 ng/mL of TGF-β regulated the phenotypic differentiation of BMSCs toward osteoblasts but seemed to inhibit osteogenic differentiation at the late stage, suggesting that additional cellular signals were necessary for the osteogenic differentiation of some types of MSCs. Therefore, it is crucial to determine how to prevent hypertrophy during TGF-β promoted cartilage differentiation.

Cytokines

The fate of MSCs might be influenced by many cytokines, such as interleukin (IL), tumor necrosis factor (TNF) and interferons (IFN). Studies have previously examined how various cytokines affected osteogenic differentiation. IL-10, IL-11, IL-18, and IFN-γ promoted osteogenesis, while TNF-α, TNF-β, IL-1α, IL-4, IL-7, IL-12, IL-13, IL-23, IFN-α and IFN-β inhibited osteogenesis[19]. In this article, we focus on recently discovered cytokines such as IL-6, IL-17, and IL-22 that have the potential to affect the fate of MSCs.

MSCs both produced IL-6 and reacted to it. Furthermore, the gradual reduction in IL-6 secretion by MSCs during chondrogenic differentiation suggested that IL-6 was one of the distinguishing characteristics of undifferentiated MSCs[20]. Nevertheless, the addition of exogenous IL-6 was found to be effective in promoting the osteogenic differentiation and chondrogenic differentiation of MSCs[20,21]. In contrast to previous studies, Xie et al[21] discovered that IL-6 secretion by BMSCs increased rather than decreased with osteogenic differentiation. The effect of IL-17A on the osteogenic differentiation of MSCs also seemed to be contradictory. Liao et al[22] reported that IL-17A inhibited the osteogenic differentiation of MSCs as well as pre-osteoblast cell lines. However, the study by Wang et al[23] showed the opposite. The appearance of these phenomena might be due to different microenvironments and cellular sources. Additionally, different concentrations of IL-17A have been shown to promote neuronal differentiation, with the best effect at 20 ng/mL[24]. The effect of IL-22 on the proliferation and differentiation of MSCs was first reported by scholars in 2017, which showed that IL-22 alone could upregulate the levels of osteogenic and lipogenic transcription factors but needed to be combined with IFN-γ and TNF to promote the proliferation of MSCs[25].

Cytokines must bind to specific receptors to transmit signals. The amount of the relevant receptor for cytokines appeared to be the rate-limiting element regulating the differentiation of MSCs[20]. Therefore, more studies are required to determine how cytokines affect the growth and differentiation of MSCs. Moreover, a fresh approach will be to look for factors that may raise the number of cytokine receptors on the surfaces of MSCs.

miRNAs

Small non-coding RNAs (approximately 20–25 nucleotides) called miRNAs are a subclass that could bind to complementary target sites in mRNA molecules to inhibit translation or decrease mRNA stability, which controls gene expression[26]. In this case, miRNAs could regulate the expression of key genes during the differentiation of MSCs in specific lineages to influence the direction of differentiation of MSCs (Table 2).

Table 2 Micro RNA and their effects on the differentiation of mesenchymal stem cells.
Amount/types
Expression
Cell source
Results
Ref.
micro-RNA-1286Over expressionhMSCs↓ Osteogenic differentiationZhou et al[27]
micro-RNA-223-3pLow expressionBMSCs↑ Osteogenic differentiationLong et al[28]
micro-RNA-346-5pOver expressionBMSCs↓ Osteogenic differentiationZhang et al[29]
micro-RNA-21Over expressionhucMSCs↑ Osteogenic differentiationMeng et al[4]
micro-RNA-130aOver expressionBMSCs↑ Osteogenic differentiation ↓ adipogenic differentiationLin et al[30]
micro-RNA-130bLow expressionBMSCs↑ Chondrogenic differentiationZhang et al[31]
micro-RNA-218Over expressionSDSCs↑ Chondrogenic differentiation during the eraly stageChen et al[32]
micro-RNA-495Over expressionhMSCs↓ Chondrogenic differentiationLee et al[33]
micro-RNA-30aOver expressionBMSCs↑ Chondrogenic differentiationTian et al[34]
micro-RNA-145Low expressionADSCs↑ Endothelial differentiationArderiu et al[26]
micro-RNA-124Over expressionADSCs↑ Neuronal differentiationMondanizadeh et al[35]
micro-RNA-10-5pLow expressionBMSCs↑ Myocardial differentiationLi et al[36]
micro-RNA-499a-5pOver expressionBMSCs↑ Cardiomyogenic differentiationNeshati et al[37]

The osteogenic differentiation of MSCs was found to be regulated by micro RNA-1286[27], micro RNA-223-3p[28], micro RNA-346-5p[29], micro RNA-21[4] and micro RNA-130a[30], whereas the chondrogenic differentiation of MSCs was found to be regulated by micro RNA-130b[31], micro RNA-218[32], micro RNA-495[33] and micro RNA-30a[34]. In addition to this, some miRNAs also exhibited roles in regulating the adipogenic differentiation[30], endothelial differentiation[26], neuronal differentiation[35], and myocardial differentiation[36,37] of MSCs.

In conclusion, investigating the impact of biochemical stimuli on the growth and differentiation of MSCs has aided our understanding of the patterns of the aberrant differentiation of MSCs in diseased situations and aided in identifying novel therapeutic targets. It appears to be a promising avenue to examine the impact of the combination of diverse biochemical stimuli on the fate of MSCs, since distinct biochemical stimuli in the microenvironment in which MSCs are positioned function in a compound manner. Additionally, since the functions of cytokines and growth factors are dependent on binding to the appropriate receptors and some studies have indicated that receptor expression might be the rate-limiting factor, it would be preferable to determine methods to boost receptor expression as opposed to raising cytokine and growth factor concentrations.

PHYSICAL STIMULI

In addition to the previously mentioned biochemical stimuli, physical stimuli such as electromagnetic fields (EMF), microgravity (MG), fluid shear stress (FSS), and hydrostatic pressure (HP) could also have an impact on the proliferation and differentiation of MSCs (Table 3). EMF, a non-invasive biophysical therapy, is a combination of electric and magnetic fields and has been widely used in the treatment of bone diseases[38,39]. Exposure to sinusoidal EMF (1mT,15Hz,4h/d) promoted the proliferation and osteogenic and chondrogenic differentiation of BMSCs[40]. In contrast, Wang et al[41] found that EMF also promoted the osteogenic differentiation of MSCs but did not inhibit their proliferation under the same parameters. With the exception of 75 Hz square EMF, Asadian et al[42] discovered that EMFs of various frequencies and waveforms (25, 50 Hz square, and sinusoidal waveform EMFs) enabled the suppression of BMSC growth. This might imply that MSCs from different sources had different sensitivities to EMFs. Distinct EMFs had different responses to MSCs. It is crucial to investigate the most appropriate EMF parameters for the proliferation or directed differentiation of MSCs from various sources. For instance, MSCs exposed for a brief period of time to low-amplitude and low-frequency pulsed EMF could be encouraged to differentiate into chondrogenic cells[43], while sinusoidal EMF at 1 mT, 15 Hz, 4 h/d was favorable for MSCs to differentiate into osteogenic cells[40,41], and higher-frequency EMF could also encourage MSCs to differentiate into neuronal cells[42].

Table 3 Physical stimuli and their effects on the differentiation of mesenchymal stem cells.
Physical stimuli
Parameters
Cell source
Results
Ref.
EMF1 mT, 15 Hz, 4 h/dayBMSCsBMSCs pretreated with EMF exhibited stronger osteogenic and chondrogenic differentiation potential and weaker adipogenesis capacityTu et al[40]
25, 50, 75Hz square and sinusoidal waveform EMFBMSCsEMF induced BMSCs differentiation to neuron cells in all treatment groupsAsadian et al[42]
1 mT, 15 Hz, 4 h/dayRabbit MSCsEMF enhanced the osteogenic potential of MSCsWang et al[41]
PEMFMSCsBrief exposure to low amplitude PEMFs enhanced the ability of MSCs to produce and secrete paracrine factors capable of promoting cartilage regenerationParate et al[43]
SMG30 g for 72 h or 10 daysAdult rat MSCsA shorter period of SMG promoted MSCs to differentiate into endothelial, neuronal and adipogenic cells. In comparison, a longer period of SMG promoted MSCs to differentiate into osteoblastsXue et al[48]
10 rpm, 72 h, 0.001 GBMSCsInhibited osteogenic differentiation of MSCsLiu et al[47]
30 rpm clinorotation, 3 dAdult rat MSCsPromoted the neuronal differentiation of rat MSCsChen et al[44]
7 rpm, 21 dhMSCsLowered the chondrogenic potential of hMSCsMayer-Wagner et al[50]
Microgravity0.001 GhMSCsmicrogravity-cultured hMSCs showed a better ability to differentiate into osteoblasts and adipocytes compared to cells cultured under natural gravity conditionsNakaji-Hirabayashi et al[46]]
Spare microgravityhMSCsSpare microgravity reduced the osteogenic differentiation of hMSCs and shifted the osteogenesis of hMSCs into adipogenesis, even during ostergenic inductionZhang et al[49]
FSS0.375 dyn/cm2, 2 h/dBMSCsPromoted osteogenesis-related genes and proteins in BMSCsJiang et al[54]
0.06 dyn/cm2, 6 h/dBMSCsProper FSS stimulation obviously enhanced BMSCs osteogenesis, while the expressions of osteogenic genes decreased with higher intensity of FSSJing et al[51]
0.5, 0.8 Pa, 3 h/dMSCsPromoted MSCs ostegenesisJiao et al[55]
3-7 dynes/cm2hMSCsEnhanced osteogenic differentiation Zhao et al[52]
4.2 dynes/cm2hMSCsFSS could lead to the osteogenic differentiation of hMSCsLiu et al[53]
ΔSS from 0 dyn/cm2 to 10 dyn/cm2MSCsFast ΔSS (0–0′) profits the chondrogenic differentiation, while Slow ΔSS (0–2′) advances osteogenic differentiationYue et al[57]
ΔSS from 0 dyn/cm2 to 10 dyn/cm2MSCsFast ΔSS (0–0′) profits the chondrogenic differentiation, while Slow ΔSS (0–2′) advances osteogenic differentiationLu et al[56]
HP10 MPa, 1 Hz, 4 h/d, 5 d/w, 3 wBMSCsHP promoted BMSCs chondrogenic differentiationSteward et al[60]
0-0.5 MPa, 0.5 HzhMSCsHP promoted the differentiation of the hMSCs toward osteogenesisHuang et al[59]
270 kPa, 1 Hz, 1 h/d, 5 d/w, 3 wBMSCsHP promoted chondrogenic differentiation of BMSCsLuo et al[64]
100 psiADSCsHP significantly increased osteogenic differentiation of AMSCsRu et al[65]
90 kPa, 1 hBMSCsHP promoted chondrogenic differentiation of BMSCsZhao et al[61]
90 kPa, 1 hBMSCsHP promoted the expression of marker genes for early osteogenic differentiation and chondrogenic differentiation of the BMSCsZhao et al[62]

Another independent factor influencing the destiny of MSCs has been identified as MG. Most of the research was thus for only conducted in a simulated MG (SMG) environment produced by a clinostat or rotating vessel, since examining the proliferation and differentiation patterns of MSCs in an actual MG environment led to some technical and budgetary challenges[44]. Quynh et al[45] found that SMG inhibited the proliferation of human umbilical cord MSCs by blocking the cell cycle; in contrast, a study by Nakaji-Hirabayashi et al[46] revealed a proliferative effect. The various SMG action times could be responsible for this circumstance. Shorter SMG treatments appeared to inhibit osteogenesis[47-49] and promote endothelial cell differentiation[48], neuronal differentiation[44,48], and adipogenic differentiation[48,49]. However, extended SMG decreased the potential for chondrogenic differentiation in MSCs[50] and encouraged their differentiation toward osteogenesis[46,48]. Different SMG action times had different effects on the cytoskeleton and could even lead to the aforementioned changes through different signal transduction pathways. In this regard, further studies are needed to determine the appropriate SMG treatment time in regulating the specific lineage differentiation of MSCs.

FSS refers to the mechanical force caused by the friction of fluid flow on the apical cell membrane. It has been demonstrated that the proliferation and differentiation of MSCs are significantly influenced by the strength, timing, and rate of FSS. Jing et al[51] discovered that the proliferation of BMSCs could be effectively induced by 0.06 dyn/cm2 of FSS stimulation, but as the intensity of the FSS increased, cell proliferation gradually decreased or was even inhibited. Meanwhile, Zhao et al[52] revealed that FSS regulated cell proliferation in a rate- and time-dependent manner, with high FSS (9–20 dyn/cm2) and the continuous effect of low FSS both inhibiting MSC proliferation, but the intermittent effect of low FSS (1–9 dyn/cm2) appeared to have little or no effect. Liu et al[53] shown that FSS (4.2 dyn/cm2) could promote the proliferation of MSCs implanted on 3D poly(lactic-co-glycolic acid) scaffolds. Although the effects of FSS on the proliferation of MSCs were differently stated, its promotion of osteogenic differentiation[52-55] seemed to be consistent. Regarding how the rate of FSS (ΔSS) affects MSCs, it was observed that quick ΔSS (From 0 dyn/cm2 in 0 min) was more beneficial for MSCs' chondrogenic development, whereas slow ΔSS (From 0 dyn/cm2 in 2 mins) encouraged their osteogenic differentiation[56,57]. Clearly, more research is required to confirm the impact of FSS on MSC proliferation, as well as the appropriate stimulus parameters for osteogenic differentiation and MSC proliferation.

HP, unlike other physical stimuli, applies homogeneous tension without causing cellular deformation[58]. Physiological load (0.1-10 mPa) did not affect the proliferation of MSCs[59,60], whereas a load of 90 kPa effectively promoted the proliferation of MSCs[61,62], a possibility that resulted from the initiation of the cell cycle by HP[62]. Studies conducted in the past have indicated that HP at low loads (1–50 kPa) has an osteogenic impact on MSCs, whereas HP at physiological loads efficiently promoted chondrogenic differentiation[63]. This concept was also supported by several recent research works[60,64]. Some investigations, however, discovered a facilitative effect of physiological loading on MSCs' osteogenic differentiation[59,65], and a chondrogenic effect of low loading on MSCs[61,62]. Additionally, the study by Zhao et al[62] discovered that HP (70 kPa) could not only stimulate RhoA activation, which in turn promoted the expression of early osteogenic differentiation genes in BMSCs, but could also upregulate Rac1 and downregulate RhoA, which further promoted cartilage development in BMSCs. These findings suggested that further studies are needed to determine the effects of different loads of HP on the spectral differentiation of MSCs and their complex mechanisms.

Overall, physical stimuli did influence MSCs’ proliferation and differentiation to varying degrees, but there is still no consensus on the parameters that are most conducive to the proliferation and specific lineages’ differentiation of MSCs. Cell heterogeneity, various stemness potentials, culture conditions, and techniques that simulated physical stimulation might all be contributing factors in this issue. Therefore, more studies are needed to determine the appropriate parameters of physical stimuli that promote the differentiation of MSCs. In fact, the actual microenvironment in which cells were exposed was multifactorial. Therefore, some studies are now starting to consider the effect of compound factors[50,55,61,66] on the behavior of MSCs. Compound factors could have synergistic effects that increase the benefits for MSCs or counteract the drawbacks of a single factor. This might emerge as a new trend.

HYPOXIA

In most studies, MSCs were cultured under atmospheric oxygen tension (20%-21% O2)[67]. However, MSCs in different ecological niches encounter oxygen concentrations that are significantly lower than 20% (Table 4). For instance, the O2 concentration that MSCs experienced varied from 1% to 5%[68] in adipose tissue and from 1% to 7%[69] in bone marrow. As a result, MSCs from different tissue sources were in a hypoxia microenvironment in vivo. Hypoxia activated various signaling pathways within a cell, which could lead to either cell death or cell adaptation[70]. Theoretically, culturing MSCs at physiological oxygen concentrations facilitated their proliferation, differentiation, and the secretion of cytokines and growth factors. Ciapetti et al[71] discovered that hypoxic circumstances greatly boosted BMSCs’ proliferation and colony-forming capacity, as well as the expression of genes relevant to bone, such as alkaline phosphatase and osteocalcin, supporting the above idea. In contrast, in a study by Xu et al[72], hypoxia inhibited the osteogenic differentiation of BMSCs by activating the Notch pathway. Therefore, we focus on the effect of hypoxia on the behavior of MSCs and try to explain the contradictory findings in different studies.

Table 4 Hypoxia and their effects on the differentiation of mesenchymal stem cells.
Conditions
Cell source
Results
Ref.
Hypoxic culture (5%O2)BMSCs↑ Chondrogenic differentiation; ↑ adipogenic differentiationElabd et al[75]
Hypoxic culture (5.5%-6.5%O2)Balb/c mouse clonal MSCs↑ Osteogenic differentiationKim et al[74]
Hypoxic culture (50 μM CoCl2 simulation)Mice MSCs↑ Osteogenic differentiationYu et al[77]
Hypoxic culture (5%O2)ADSCs↑ Chondrogenic differentiationLee et al[76]
Hypoxic culture (1%O2)PBMSCs↑ Osteogenic differentiationYang et al[3]
Hypoxic culture (1%O2)BMSCs↓ Osteogenic differentiation; ↓ adipogenic differentiation; ↓ chondrogenic differentiationCicione et al[78]
Hypoxic culture (1%O2)BMSCs↑ Neuronal differentiationWang et al[80]
Hypoxic culture (1%O2)BMSCs↓ Osteogenic differentiationXu et al[72]
Hypoxic culture (2%O2)ADSCs↑ Tenocyte differentiationYu et al[73]
Hypoxic culture (2%O2)ADSCs↓ Osteogenic differentiationKim et al[79]
Hypoxic culture (2%O2)BMSCs↑ Osteogenic differentiationCiapetti et al[71]

The two primary techniques used nowadays to create in vitro hypoxic settings are anaerobic chambers[73] and simulation utilizing different chemicals[74]. In a study by Elabd et al[75], moderate hypoxia (5% O2) circumstances promoted the chondrogenic and adipogenic differentiation of BMSCs but had no effect on proliferation or osteogenic differentiation. At the same oxygen concentration, Lee et al[76] showed that hypoxia promoted MSC proliferation and increased the chondrogenic differentiation potential. The proliferation of MSCs was also effectively promoted at a 5.5%-6.5% O2 concentration simulated by 10 μM CoCl2 and 4.0 mmol/L Na2SO3[74]. In contrast, Yu et al[77] demonstrated that a 50 M CoCl2-simulated hypoxia environment appeared to prevent the growth of MSCs. Consistently, the osteogenic differentiation of MSCs was promoted in hypoxia environments simulated using different concentrations of CoCl2[74,77]. Cicione et al[78] investigated the changes in the trilineage differentiation potential of BMSCs under severe hypoxia (1% O2) and showed that the trilineage differentiation potential of BMSCs was inhibited to different degrees. Additional research demonstrated that the activation of the Notch pathway may be responsible for the suppression of the osteogenic differentiation of MSCs by severe hypoxia (1% O2)[3,72]. In addition, Kim et al[79] found that hypoxia could inhibit the osteogenic differentiation of ADSCs by upregulating insulin-like growth factor binding-protein-3. Hypoxia has also been shown to encourage the tendon[73] and neural[80] differentiation of MSCs.

Compared to the laboratory culture environment (20%-21% O2), hypoxia is more representative of the oxygen concentration in the ecological niche of MSCs. Varied oxygen concentrations had extremely different impacts on MSCs. Moderate hypoxia environment enhanced MSCs’ proliferation, osteogenic differentiation, and chondrogenic differentiation. The differentiation capability of all three lineages of MSCs was, however, somewhat hindered under severe hypoxia. The contradictory behavior in the aforementioned research might potentially be connected to the cell source of MSCs and whether they were differentiated under hypoxia conditions. In view of current studies generally focusing on hypoxia exposure either in the phase of expansion or differentiation, which have not been fully grasped, further research is necessary to comprehend the effects on MSCs specifically in these two culture forms.

MATRIX STIFFNESS AND SURFACE TOPOGRAPHY

Two crucial material physical characteristics that have a significant impact on MSC behavior are matrix stiffness and surface topography. Matrix stiffness is a passive mechanical parameter that the cell can not directly sense. By exerting traction pressures on the cytoskeleton through focal adhesion, cells might deform the extracellular matrix (ECM), reflecting matrix stiffness[81]. Materials with ECM properties are currently being designed to simulate the actual microenvironment of cells. The ECMs of different native tissues, such as bone, cartilage, nerves, or blood vessels, are composed of micro- and nanoscale topographic patterns[82]. As a result, an increasing number of researchers have begun to look into how the substrate surface topography affects MSC behavior. Size and surface roughness are the two most fundamental parameters of surface topography[83], and the effects of these two factors, as well as substrate stiffness, on the proliferation and differentiation of MSCs are also mainly explored here.

Matrix stiffness

Stiffness is one of the most common metrics in assessing a material's mechanical properties[81], and it is typically expressed in terms of Young's modulus. Matrix stiffness has been shown in many studies to affect the proliferation and differentiation of MSCs. MSCs exhibited higher proliferative behavior under a higher substrate stiffness, and Winer et al[84] found that MSCs inoculated in 250-Pa polyacrylamide gels that mimicked the elasticity of bone marrow and adipose tissue exhibited cell cycle arrest, but these arrested cells re-entered the cell cycle when a stiff substrate was present[84]. In comparison to lower-stiffness gels, higher-stiffness matrices could increase the number of cells by a factor of 10[85]. With fibronectin-coated polyacrylamide hydrogels, Sun et al[86] controlled the mechanical environment of BMSCs and discovered that BMSCs’ proliferation increased with increasing stiffness. However, as opposed to firmer substrates, Lin et al[87] discovered that MSCs cultivated on softer substrates had greater cell proliferation rates. Gelma hydrogels with different concentrations not only had different hardness, but also showed different porosity as well. Moreover, the pore size also seemed to be one of the influencing factors for the proliferation and differentiation of MSCs. Indeed, many studies have focused on the effect of matrix stiffness on the direction of differentiation of MSCs. MSCs exhibited the upregulation of biomarkers matching tissue stiffness on polyacrylamide gels of different stiffness, such as neurogenic (0.1-1 kPa, brain), myogenic (8-17 kPa, muscle), and osteogenic (25-40 kPa, bone) markers[88]. BMSCs could be driven to develop into an osteogenic phenotype and expressed increased quantities of bone-derived biomarkers including Runt-related transcription factor 2 (RUNX2), alkaline phosphatase (ALP), and bone-bridging proteins when grown on polyacrylamide hydrogels (62-68 kPa)[86]. Rowlands et al[85] found that the osteogenic differentiation of MSCs occurred mainly on polyacrylamide gels of 80 kPa stiffness and that RUNX2 was also expressed at high levels. This might be due to the fact that the 80 kPa collagen I coating could well simulate the microenvironment of the bone tissue. Without an induction medium, the stiffness of the hydrogel itself had a substantial impact in controlling MSC differentiation early on, with softer substrates encouraging the adipogenic differentiation of MSCs, while harder substrates encouraged the osteogenic differentiation of MSCs[89]. However, this effect seemed to be gradually attenuated by biochemical effects in the culture medium, implying that the effects of different factors on the differentiation behavior of MSCs might occur at different stages of differentiation. On 22 kPa gels, as opposed to softer matrices, MSCs produced larger quantities of ALP, which was consistent with the effect of matrix stiffness on osteogenic fractionation shown in the previous work[90]. Although more disagreement has emerged regarding the effect of softer matrices on the differentiation fate of MSCs, such as adipogenic differentiation[84,90-92], myogenic differentiation[85,88], neurogenic differentiation[88], and endothelial differentiation[87], there seems to be a consensus on the osteogenic role of harder matrices for MSCs. The Stiffer matrix enabled cells to produce more cytoskeletal tension and sent differentiation signals via transmembrane proteins such as integrins[81,85], which promoted osteogenic differentiation. Furthermore, the nuclear localization of Yes-associated protein (a key mediator of mechano-transduction) and RUNX2 could be impacted by the substrate stiffness[89,90].

Surface topography

Zhao et al[93] produced nanotubes of various sizes and micro- and nano-hybrid topographies with ECM-like micro/nanostructures and examined their effects on the proliferation and osteogenic differentiation of MSCs. They discovered that larger-sized nanotubes hindered the early proliferation of MSCs, but the micro- and nano-morphology group had a greater cell number. Additionally, they discovered that MSC osteogenic differentiation might be induced by micro/nanotopographies, even in the absence of osteogenic inducers[93]. Similar results were obtained by Chen et al[94], who discovered that the micron/submicron hybrid topography of Ti surfaces promoted osteogenic and chondrogenic differentiation in the early stages of induced differentiation. By introducing nanoengineered topographic glass substrates with different surface roughness, Qian et al[95] investigated the impact of surface morphology on the osteogenic differentiation of MSCs. They found that surface roughness could replace the osteogenic inducer dexamethasone and worked in concert with ascorbic acid and β-glycerophosphate to jointly promote the osteogenic differentiation of MSCs[95]. In the past, it was generally agreed that surface roughness seemed to have a positive effect on osteogenic differentiation[95-97]. The osteogenic differentiation of MSCs, however, was more strongly influenced by the nanopore size than by the surface roughness, according to several recent studies[83,98]. The frequent coupling of size and surface roughness in many studies makes it difficult to state the degree of influence of each factor on the behavior of MSCs[83]. Moreover, the methods used to prepare rough surfaces in these studies differ, such as randomly rough surfaces produced by treatments such as mechanical polishing, acid etching, etc., where cells form focal attachments that differ from those seen on surfaces of the same roughness[98]. Therefore, more research is required to demonstrate how size and surface roughness affect MSC proliferation and differentiation, respectively. Through a variety of pathways, including the control of adhesion, cytoskeletal tension, and nuclear localization of transcription factors[95], MSCs appeared to be able to detect and respond to the surface topography, which in turn influenced their behavior such as proliferation and differentiation. At this stage, it has been reported that micro- and nano-surface topographies inhibit the proliferation of MSCs and promote osteogenic differentiation to some extent. However, there is no detailed elaboration on their respective effects on MSCs in terms of size and surface roughness.

EXOSOMES

Various cells jointly create the microenvironment by secreting functional molecules, which leads to the sharing of stimuli between multiple cell lineages[99]. In addition to the ECM and growth factors, exosomes were considered to be an important component of the microenvironment[100]. Exosomes are small vesicles with a diameter of 30-150 nm that are released by cells through cytosolic action. The released exosomes could interact with target cells and translocated proteins, lipids, mRNAs and miRNAs to the cytoplasm of target cells[101]. Crosstalk existed between MSCs-osteoblasts and monocytes-macrophages and researchers used this to regulate bone homeostasis[99]. In vitro, BMSCs’ behaviors were influenced variably by cell-conditioned media produced by variously polarized macrophages[102]. Previous studies had suggested that cytokines were the main contributors to the function exercised by macrophages. However, Song et al[103] found that lipopolysaccharide (LPS)-activated macrophage-derived exosomes inhibited the osteogenic differentiation of BMSCs by mediating inflammatory stimuli. Therefore, the effect of exosomes secreted by monocytes-macrophages on the differentiation of MSCs should be considered (Table 5).

Table 5 Exosomes of different cell sources and their effects on the differentiation of mesenchymal stem cells.
Source and kind
Specific cargo
Target
Results
Ref.
M1 macrophages-EVsmiRNA-21a-5pBMSCs↑ Osteogenic differentiationLiu et al[104]
M0 macrophages-EVsBMSCs↓ Chondrogenic differentiationXia et al[105]
M1 macrophages-EVsBMSCs↑ Osteogenic differentiation; ↑ adipogenic differentiation; ↓ chondrogenic differentiation
M2 macrophages-EVsBMSCs↓ Chondrogenic differentiation
M2 macrophages-EVsmiRNA-5106BMSCs;SIK2 and SIK3↑ Osteogenic differentiationXiong et al[106]
M2 macrophages-EVsmiRNA-690BMSCs↑ Osteogenic differentiation; ↓ adipogenic differentiationLi et al[107]
M0 macrophages-EVsMSCs↑ Osteogenic differentiationKang et al[108]
M1 macrophages-EVsmiRNA-155MSCs↓ Osteogenic differentiation
M2 macrophages-EVsmiRNA-378aMSCs↑ Osteogenic differentiation
M2 macrophages-EVsmiRNA-26a-5pBMSCs↑ Osteogenic differentiation; ↓ adipogenic differentiationBin-bin et al[109]
Macrophages-EVsBMSCs↓ Osteogenic differentiationSong et al[103]
Monocytes-EVsMSCs↑ Osteogenic differentiationEkström et al[110]
Osteoclasts-EVsmiRNA-324BMSCs↑ Osteogenic differentiationLiang et al[111]

According to Liu et al[104], miR-21a-5p found in M1 macrophage-derived exosomes directed BMSCs toward an osteoblastic fate during the early stages of osteogenesis[104]. In their investigation of the effects of MO, M1, and M2 macrophage-derived exosomes on BMSCs, Xia et al[105] discovered that M1 macrophage-derived exosomes efficiently enhanced the proliferation, osteogenic differentiation, and adipogenic differentiation of BMSCs, but M2 macrophage-derived exosomes were harmful to the proliferation of BMSCs and, curiously, all three hindered the chondrogenic differentiation of BMSCs. Xiong et al[106] noticed that miRNA-5106, enriched in M2 macrophage-derived exosomes, promoted the osteogenic differentiation of BMSCs by suppressing the expression of salt-inducible kinase 2 (SIK2) and SIK3, which was consistent with the role of M2 macrophage-derived exosomes in promoting osteogenesis in a study by Li et al[107]. Kang et al[108] demonstrated that M0 and M2 macrophage-derived exosomes were positive for BMSC osteogenesis while M1 macrophage-derived exosomes lowered BMP expression and inhibited the osteogenic differentiation of BMSCs[108]. Despite being enriched in distinct miRNAs, primary extraction M2 macrophages[109] and RAW264.7 mouse monocyte-macrophage leukemia cell[107] derived exosomes both showed osteogenesis-promoting and lipogenic differentiation-inhibiting effects. Current research has indicated the impact of exosomes produced from monocytes[110], osteoclasts[111], and osteoblasts[112] on BMSCs, in addition to exosomes released by macrophages. Ekström et al[110] found that exosomes released from LPS-stimulated monocytes could be ingested by MSCs and encouraged the osteogenic differentiation of MSCs. Liang et al[111] showed that osteoclast-released exosomes promoted osteogenic differentiation and facilitated osteogenic mineralization by inhibiting Rho GTPase activating protein 1. This might imply that active osteoclasts release large amounts of extracellular vesicles during the resorption phase, promoting the osteogenesis of MSCs for better stabilization and bridging the transition between bone resorption and formation. The addition of osteoblast exosomes could further enhance the expression of RUNX2 and osterix, thereby promoting osteogenic differentiation, and, in addition, osteoblast exosomes could even alter adipocyte ECM-mediated lineage differentiation[112].

Exosomes, one of the recently identified microenvironment components, have unique benefits, such as a nano size, non-toxicity, low immunogenicity, biocompatibility, and versatility of use, drawing widespread attention[113]. The current work appeared to demonstrate the beneficial influence of M2 macrophage-derived exosomes on the osteogenic differentiation of MSCs. As for MO and M1 macrophage-derived exosomes, further research is required to understand their impacts on MSC differentiation and the processes at play. At the same time, research has been conducted progressively on the influence of exosomes released by cells in the same microenvironment as BMSCs on the differentiation of BMSCs, which might represent a new avenue.

CONCLUSION

MSCs play important roles in pathological and physiological processes because of their self-renewal, migration, and pluripotency. Especially due to their multi-differentiation potential, MSCs have been considered as a new therapeutic agent in regenerative medicine. Since the detailed mechanisms involved in these regulation processes has not been fully revealed, research on intrinsic and extrinsic factors regulating MSCs’ differentiation may provide new methods in manipulating the cell fate of MSCs. Here, we discussed multiple chemical and mechanical factors affecting the osteogenic and chondrogenic differentiation of MSCs, including typical differentiation promoting patterns, cell environmental factors, and other interesting research areas, such as material morphology and exosomes. After sensing these differentiation-stimulating factors, MSCs from various sources are able to differentiate into specific cell lineages. With the rising demand for MSCs in clinical treatment, noble strategies have been developed that aim at inducing the stable and directional differentiation of stem cells, and further providing efficient methods of MSC regulation in basic research and clinical application.

Meanwhile, there is much more to discover in stem cell research. Due to some limitations of MSCs, such as homing efficiency and long-term survival in vivo, most of the research has achieved its results at the cellular level in vitro. Moreover, discrepancies remain between single-factor experiments and synergistic effects by multiple factors. At present, extensive research on factors stimulating MSCs’ differentiation has promoted our understanding of cell functional alterations. However, mechanisms involved in manipulating MSCs’ cell fate have so far been incomplete. With the deepening of stem cell research alongside technology improvements, the synergistic effect of multiple factors inducing MSC differentiation is increasingly likely to be clarified, as well as providing new patterns in clinical stem cell therapy.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Cell and tissue engineering

Country/Territory of origin: China

Peer-review report’s scientific quality classification

Grade A (Excellent): A, A

Grade B (Very good): 0

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Shengwen Calvin Li, United States; Cheraqpour K, Iran; Exbrayat JM, France S-Editor: Liu GL L-Editor: A P-Editor: Liu GL

References
1.  Caplan AI. Mesenchymal stem cells. J Orthop Res. 1991;9:641-650.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3539]  [Cited by in F6Publishing: 3155]  [Article Influence: 95.6]  [Reference Citation Analysis (0)]
2.  Hesari R, Keshvarinia M, Kabiri M, Rad I, Parivar K, Hoseinpoor H, Tavakoli R, Soleimani M, Kouhkan F, Zamanluee S, Hanaee-Ahvaz H. Comparative impact of platelet rich plasma and transforming growth factor-β on chondrogenic differentiation of human adipose derived stem cells. Bioimpacts. 2020;10:37-43.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 9]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
3.  Yang M, Liu H, Wang Y, Wu G, Qiu S, Liu C, Tan Z, Guo J, Zhu L. Hypoxia reduces the osteogenic differentiation of peripheral blood mesenchymal stem cells by upregulating Notch-1 expression. Connect Tissue Res. 2019;60:583-596.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
4.  Meng YB, Li X, Li ZY, Zhao J, Yuan XB, Ren Y, Cui ZD, Liu YD, Yang XJ. microRNA-21 promotes osteogenic differentiation of mesenchymal stem cells by the PI3K/β-catenin pathway. J Orthop Res. 2015;33:957-964.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 87]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
5.  Choi S, Cho TJ, Kwon SK, Lee G, Cho J. Chondrogenesis of periodontal ligament stem cells by transforming growth factor-β3 and bone morphogenetic protein-6 in a normal healthy impacted third molar. Int J Oral Sci. 2013;5:7-13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 29]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
6.  Zhong Y, Li X, Wang F, Wang S, Wang X, Tian X, Bai S, Miao D, Fan J. Emerging Potential of Exosomes on Adipogenic Differentiation of Mesenchymal Stem Cells. Front Cell Dev Biol. 2021;9:649552.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 4]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
7.  Tsutsumi S, Shimazu A, Miyazaki K, Pan H, Koike C, Yoshida E, Takagishi K, Kato Y. Retention of multilineage differentiation potential of mesenchymal cells during proliferation in response to FGF. Biochem Biophys Res Commun. 2001;288:413-419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 456]  [Cited by in F6Publishing: 466]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
8.  Lu H, Wu PF, Ma DL, Zhang W, Sun M. Growth Factors and Their Roles in Multiple Sclerosis Risk. Front Immunol. 2021;12:768682.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
9.  Endo K, Fujita N, Nakagawa T, Nishimura R. Effect of Fibroblast Growth Factor-2 and Serum on Canine Mesenchymal Stem Cell Chondrogenesis. Tissue Eng Part A. 2019;25:901-910.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 12]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
10.  Okamura G, Ebina K, Hirao M, Chijimatsu R, Yonetani Y, Etani Y, Miyama A, Takami K, Goshima A, Yoshikawa H, Ishimoto T, Nakano T, Hamada M, Kanamoto T, Nakata K. Promoting Effect of Basic Fibroblast Growth Factor in Synovial Mesenchymal Stem Cell-Based Cartilage Regeneration. Int J Mol Sci. 2020;22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 8]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
11.  Ma Y, Kakudo N, Morimoto N, Lai F, Taketani S, Kusumoto K. Fibroblast growth factor-2 stimulates proliferation of human adipose-derived stem cells via Src activation. Stem Cell Res Ther. 2019;10:350.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 23]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
12.  Ramasamy R, Tong CK, Yip WK, Vellasamy S, Tan BC, Seow HF. Basic fibroblast growth factor modulates cell cycle of human umbilical cord-derived mesenchymal stem cells. Cell Prolif. 2012;45:132-139.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 38]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
13.  Wang R, Liu W, Du M, Yang C, Li X, Yang P. The differential effect of basic fibroblast growth factor and stromal cellderived factor1 pretreatment on bone morrow mesenchymal stem cells osteogenic differentiation potency. Mol Med Rep. 2018;17:3715-3721.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 9]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
14.  Wang JJ, Liu YL, Sun YC, Ge W, Wang YY, Dyce PW, Hou R, Shen W. Basic Fibroblast Growth Factor Stimulates the Proliferation of Bone Marrow Mesenchymal Stem Cells in Giant Panda (Ailuropoda melanoleuca). PLoS One. 2015;10:e0137712.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 13]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
15.  Jia Z, Wang S, Liang Y, Liu Q. Combination of kartogenin and transforming growth factor-β3 supports synovial fluid-derived mesenchymal stem cell-based cartilage regeneration. Am J Transl Res. 2019;11:2056-2069.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Sun J, Zhou Y, Ye Z, Tan WS. Transforming growth factor-β1 stimulates mesenchymal stem cell proliferation by altering cell cycle through FAK-Akt-mTOR pathway. Connect Tissue Res. 2019;60:406-417.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
17.  Xu J, Liu J, Gan Y, Dai K, Zhao J, Huang M, Huang Y, Zhuang Y, Zhang X. High-Dose TGF-β1 Impairs Mesenchymal Stem Cell-Mediated Bone Regeneration via Bmp2 Inhibition. J Bone Miner Res. 2020;35:167-180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 32]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
18.  Igarashi Y, Chosa N, Sawada S, Kondo H, Yaegashi T, Ishisaki A. VEGF-C and TGF-β reciprocally regulate mesenchymal stem cell commitment to differentiation into lymphatic endothelial or osteoblastic phenotypes. Int J Mol Med. 2016;37:1005-1013.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 35]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
19.  Amarasekara DS, Kim S, Rho J. Regulation of Osteoblast Differentiation by Cytokine Networks. Int J Mol Sci. 2021;22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 108]  [Article Influence: 36.0]  [Reference Citation Analysis (0)]
20.  Kondo M, Yamaoka K, Sakata K, Sonomoto K, Lin L, Nakano K, Tanaka Y. Contribution of the Interleukin-6/STAT-3 Signaling Pathway to Chondrogenic Differentiation of Human Mesenchymal Stem Cells. Arthritis Rheumatol. 2015;67:1250-1260.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 72]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
21.  Xie Z, Tang S, Ye G, Wang P, Li J, Liu W, Li M, Wang S, Wu X, Cen S, Zheng G, Ma M, Wu Y, Shen H. Interleukin-6/interleukin-6 receptor complex promotes osteogenic differentiation of bone marrow-derived mesenchymal stem cells. Stem Cell Res Ther. 2018;9:13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 67]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
22.  Liao C, Zhang C, Jin L, Yang Y. IL-17 alters the mesenchymal stem cell niche towards osteogenesis in cooperation with osteocytes. J Cell Physiol. 2020;235:4466-4480.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
23.  Wang Z, Jia Y, Du F, Chen M, Dong X, Chen Y, Huang W. IL-17A Inhibits Osteogenic Differentiation of Bone Mesenchymal Stem Cells via Wnt Signaling Pathway. Med Sci Monit. 2017;23:4095-4101.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 24]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
24.  Chen H, Li S, Xu W, Hong Y, Dou R, Shen H, Liu X, Wu T, He JC. Interleukin-17A promotes the differentiation of bone marrow mesenchymal stem cells into neuronal cells. Tissue Cell. 2021;69:101482.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
25.  El-Zayadi AA, Jones EA, Churchman SM, Baboolal TG, Cuthbert RJ, El-Jawhari JJ, Badawy AM, Alase AA, El-Sherbiny YM, McGonagle D. Interleukin-22 drives the proliferation, migration and osteogenic differentiation of mesenchymal stem cells: a novel cytokine that could contribute to new bone formation in spondyloarthropathies. Rheumatology (Oxford). 2017;56:488-493.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 59]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
26.  Arderiu G, Peña E, Aledo R, Juan-Babot O, Crespo J, Vilahur G, Oñate B, Moscatiello F, Badimon L. MicroRNA-145 Regulates the Differentiation of Adipose Stem Cells Toward Microvascular Endothelial Cells and Promotes Angiogenesis. Circ Res. 2019;125:74-89.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 46]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
27.  Zhou JG, Hua Y, Liu SW, Hu WQ, Qian R, Xiong L. MicroRNA-1286 inhibits osteogenic differentiation of mesenchymal stem cells to promote the progression of osteoporosis via regulating FZD4 expression. Eur Rev Med Pharmacol Sci. 2020;24:1-10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 6]  [Reference Citation Analysis (0)]
28.  Long C, Cen S, Zhong Z, Zhou C, Zhong G. FOXO3 is targeted by miR-223-3p and promotes osteogenic differentiation of bone marrow mesenchymal stem cells by enhancing autophagy. Hum Cell. 2021;34:14-27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 18]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
29.  Zhang Y, Sun Y, Liu J, Han Y, Yan J. MicroRNA-346-5p Regulates Differentiation of Bone Marrow-Derived Mesenchymal Stem Cells by Inhibiting Transmembrane Protein 9. Biomed Res Int. 2020;2020:8822232.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 4]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
30.  Lin Z, He H, Wang M, Liang J. MicroRNA-130a controls bone marrow mesenchymal stem cell differentiation towards the osteoblastic and adipogenic fate. Cell Prolif. 2019;52:e12688.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 94]  [Article Influence: 18.8]  [Reference Citation Analysis (0)]
31.  Zhang P, Gao G, Zhou Z, He X. microRNA-130b downregulation potentiates chondrogenic differentiation of bone marrow mesenchymal stem cells by targeting SOX9. Braz J Med Biol Res. 2021;54:e10345.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
32.  Chen S, Xu Z, Shao J, Fu P, Wu H. MicroRNA-218 promotes early chondrogenesis of mesenchymal stem cells and inhibits later chondrocyte maturation. BMC Biotechnol. 2019;19:6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
33.  Lee S, Yoon DS, Paik S, Lee KM, Jang Y, Lee JW. microRNA-495 inhibits chondrogenic differentiation in human mesenchymal stem cells by targeting Sox9. Stem Cells Dev. 2014;23:1798-1808.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 62]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
34.  Tian Y, Guo R, Shi B, Chen L, Yang L, Fu Q. MicroRNA-30a promotes chondrogenic differentiation of mesenchymal stem cells through inhibiting Delta-like 4 expression. Life Sci. 2016;148:220-228.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 36]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
35.  Mondanizadeh M, Arefian E, Mosayebi G, Saidijam M, Khansarinejad B, Hashemi SM. MicroRNA-124 regulates neuronal differentiation of mesenchymal stem cells by targeting Sp1 mRNA. J Cell Biochem. 2015;116:943-953.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 45]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
36.  Li M, Zhang YL, Huang H, Xiong Y. MicroRNA-10-5p regulates differentiation of bone marrow mesenchymal stem cells into cardiomyocytes by targeting TBX5. Eur Rev Med Pharmacol Sci. 2019;23:479-485.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
37.  Neshati V, Mollazadeh S, Fazly Bazzaz BS, de Vries AAF, Mojarrad M, Naderi-Meshkin H, Neshati Z, Mirahmadi M, Kerachian MA. MicroRNA-499a-5p Promotes Differentiation of Human Bone Marrow-Derived Mesenchymal Stem Cells to Cardiomyocytes. Appl Biochem Biotechnol. 2018;186:245-255.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 14]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
38.  Hamid HA, Sarmadi VH, Prasad V, Ramasamy R, Miskon A. Electromagnetic field exposure as a plausible approach to enhance the proliferation and differentiation of mesenchymal stem cells in clinically relevant scenarios. J Zhejiang Univ Sci B. 2022;23:42-57.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 12]  [Reference Citation Analysis (0)]
39.  Lin HY, Lu KH. Repairing large bone fractures with low frequency electromagnetic fields. J Orthop Res. 2010;28:265-270.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 18]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
40.  Tu C, Xiao Y, Ma Y, Wu H, Song M. The legacy effects of electromagnetic fields on bone marrow mesenchymal stem cell self-renewal and multiple differentiation potential. Stem Cell Res Ther. 2018;9:215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
41.  Wang H, Tang X, Li W, Chen J, Li H, Yan J, Yuan X, Wu H, Liu C. Enhanced osteogenesis of bone marrow stem cells cultured on hydroxyapatite/collagen I scaffold in the presence of low-frequency magnetic field. J Mater Sci Mater Med. 2019;30:89.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 10]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
42.  Asadian N, Jadidi M, Safari M, Jadidi T, Gholami M. EMF frequency dependent differentiation of rat bone marrow mesenchymal stem cells to astrocyte cells. Neurosci Lett. 2021;744:135587.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 2]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
43.  Parate D, Kadir ND, Celik C, Lee EH, Hui JHP, Franco-Obregón A, Yang Z. Pulsed electromagnetic fields potentiate the paracrine function of mesenchymal stem cells for cartilage regeneration. Stem Cell Res Ther. 2020;11:46.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 46]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
44.  Chen J, Liu R, Yang Y, Li J, Zhang X, Wang Z, Ma J. The simulated microgravity enhances the differentiation of mesenchymal stem cells into neurons. Neurosci Lett. 2011;505:171-175.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 42]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
45.  Quynh Chi HN, Nghia Son H, Chinh Chung D, Huan LD, Hong Diem T, Long LT. Simulated microgravity reduces proliferation and reorganizes the cytoskeleton of human umbilical cord mesenchymal stem cells. Physiol Res. 2020;69:897-906.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
46.  Nakaji-Hirabayashi T, Matsumura K, Ishihara R, Ishiguro T, Nasu H, Kanno M, Ichida S, Hatashima T. Enhanced proliferation and differentiation of human mesenchymal stem cells in the gravity-controlled environment. Artif Organs. 2022;46:1760-1770.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
47.  Liu L, Cheng Y, Wang J, Ding Z, Halim A, Luo Q, Song G. Simulated Microgravity Suppresses Osteogenic Differentiation of Mesenchymal Stem Cells by Inhibiting Oxidative Phosphorylation. Int J Mol Sci. 2020;21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
48.  Xue L, Li Y, Chen J. Duration of simulated microgravity affects the differentiation of mesenchymal stem cells. Mol Med Rep. 2017;15:3011-3018.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 33]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
49.  Zhang C, Li L, Jiang Y, Wang C, Geng B, Wang Y, Chen J, Liu F, Qiu P, Zhai G, Chen P, Quan R, Wang J. Space microgravity drives transdifferentiation of human bone marrow-derived mesenchymal stem cells from osteogenesis to adipogenesis. FASEB J. 2018;32:4444-4458.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 39]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
50.  Mayer-Wagner S, Hammerschmid F, Blum H, Krebs S, Redeker JI, Holzapfel BM, Jansson V, Müller PE. Effects of single and combined low frequency electromagnetic fields and simulated microgravity on gene expression of human mesenchymal stem cells during chondrogenesis. Arch Med Sci. 2018;14:608-616.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
51.  Jing L, Fan S, Yao X, Zhang Y. Effects of compound stimulation of fluid shear stress plus ultrasound on stem cell proliferation and osteogenesis. Regen Biomater. 2021;8:rbab066.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
52.  Zhao Y, Richardson K, Yang R, Bousraou Z, Lee YK, Fasciano S, Wang S. Notch signaling and fluid shear stress in regulating osteogenic differentiation. Front Bioeng Biotechnol. 2022;10:1007430.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 9]  [Reference Citation Analysis (0)]
53.  Liu L, Zong C, Li B, Shen D, Tang Z, Chen J, Zheng Q, Tong X, Gao C, Wang J. The interaction between β1 integrins and ERK1/2 in osteogenic differentiation of human mesenchymal stem cells under fluid shear stress modelled by a perfusion system. J Tissue Eng Regen Med. 2014;8:85-96.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 36]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
54.  Jiang M, Shen Q, Zhou Y, Ren W, Chai M, Tan WS. Fluid shear stress and endothelial cells synergistically promote osteogenesis of mesenchymal stem cells via integrin β1-FAK-ERK1/2 pathway. Turk J Biol. 2021;45:683-694.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
55.  Jiao F, Xu J, Zhao Y, Ye C, Sun Q, Liu C, Huo B. Synergistic effects of fluid shear stress and adhesion morphology on the apoptosis and osteogenesis of mesenchymal stem cells. J Biomed Mater Res A. 2022;110:1636-1644.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
56.  Lu J, Fan Y, Gong X, Zhou X, Yi C, Zhang Y, Pan J. The Lineage Specification of Mesenchymal Stem Cells Is Directed by the Rate of Fluid Shear Stress. J Cell Physiol. 2016;231:1752-1760.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
57.  Yue D, Zhang M, Lu J, Zhou J, Bai Y, Pan J. The rate of fluid shear stress is a potent regulator for the differentiation of mesenchymal stem cells. J Cell Physiol. 2019;234:16312-16319.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
58.  Pattappa G, Zellner J, Johnstone B, Docheva D, Angele P. Cells under pressure - the relationship between hydrostatic pressure and mesenchymal stem cell chondrogenesis. Eur Cell Mater. 2019;37:360-381.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 33]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
59.  Huang C, Ogawa R. Effect of hydrostatic pressure on bone regeneration using human mesenchymal stem cells. Tissue Eng Part A. 2012;18:2106-2113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 32]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
60.  Steward AJ, Thorpe SD, Vinardell T, Buckley CT, Wagner DR, Kelly DJ. Cell-matrix interactions regulate mesenchymal stem cell response to hydrostatic pressure. Acta Biomater. 2012;8:2153-2159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 62]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
61.  Zhao Y, Yi FZ, Zhao YH, Chen YJ, Ma H, Zhang M. The Distinct Effects of Estrogen and Hydrostatic Pressure on Mesenchymal Stem Cells Differentiation: Involvement of Estrogen Receptor Signaling. Ann Biomed Eng. 2016;44:2971-2983.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
62.  Zhao YH, Lv X, Liu YL, Zhao Y, Li Q, Chen YJ, Zhang M. Hydrostatic pressure promotes the proliferation and osteogenic/chondrogenic differentiation of mesenchymal stem cells: The roles of RhoA and Rac1. Stem Cell Res. 2015;14:283-296.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 55]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
63.  Higuera GA, van Boxtel A, van Blitterswijk CA, Moroni L. The physics of tissue formation with mesenchymal stem cells. Trends Biotechnol. 2012;30:583-590.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
64.  Luo L, Foster NC, Man KL, Brunet M, Hoey DA, Cox SC, Kimber SJ, El Haj AJ. Hydrostatic pressure promotes chondrogenic differentiation and microvesicle release from human embryonic and bone marrow stem cells. Biotechnol J. 2022;17:e2100401.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 10]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
65.  Ru J, Guo L, Ji Y, Niu Y. Hydrostatic pressure induces osteogenic differentiation of adipose-derived mesenchymal stem cells through increasing lncRNA-PAGBC. Aging (Albany NY). 2020;12:13477-13487.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
66.  Elashry MI, Baulig N, Wagner AS, Klymiuk MC, Kruppke B, Hanke T, Wenisch S, Arnhold S. Combined macromolecule biomaterials together with fluid shear stress promote the osteogenic differentiation capacity of equine adipose-derived mesenchymal stem cells. Stem Cell Res Ther. 2021;12:116.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
67.  Choi JR, Pingguan-Murphy B, Wan Abas WA, Yong KW, Poon CT, Noor Azmi MA, Omar SZ, Chua KH, Xu F, Wan Safwani WK. In situ normoxia enhances survival and proliferation rate of human adipose tissue-derived stromal cells without increasing the risk of tumourigenesis. PLoS One. 2015;10:e0115034.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 48]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
68.  Choi JR, Yong KW, Wan Safwani WKZ. Effect of hypoxia on human adipose-derived mesenchymal stem cells and its potential clinical applications. Cell Mol Life Sci. 2017;74:2587-2600.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 54]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
69.  Fehrer C, Brunauer R, Laschober G, Unterluggauer H, Reitinger S, Kloss F, Gülly C, Gassner R, Lepperdinger G. Reduced oxygen tension attenuates differentiation capacity of human mesenchymal stem cells and prolongs their lifespan. Aging Cell. 2007;6:745-757.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 362]  [Cited by in F6Publishing: 362]  [Article Influence: 21.3]  [Reference Citation Analysis (0)]
70.  Buravkova LB, Andreeva ER, Gogvadze V, Zhivotovsky B. Mesenchymal stem cells and hypoxia: where are we? Mitochondrion. 2014;19 Pt A:105-112.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 91]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
71.  Ciapetti G, Granchi D, Fotia C, Savarino L, Dallari D, Del Piccolo N, Donati DM, Baldini N. Effects of hypoxia on osteogenic differentiation of mesenchymal stromal cells used as a cell therapy for avascular necrosis of the femoral head. Cytotherapy. 2016;18:1087-1099.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 34]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
72.  Xu N, Liu H, Qu F, Fan J, Mao K, Yin Y, Liu J, Geng Z, Wang Y. Hypoxia inhibits the differentiation of mesenchymal stem cells into osteoblasts by activation of Notch signaling. Exp Mol Pathol. 2013;94:33-39.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 67]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
73.  Yu Y, Zhou Y, Cheng T, Lu X, Yu K, Hong J, Chen Y. Hypoxia enhances tenocyte differentiation of adipose-derived mesenchymal stem cells by inducing hypoxia-inducible factor-1α in a co-culture system. Cell Prolif. 2016;49:173-184.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 29]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
74.  Kim H, Kwon S. Dual effects of hypoxia on proliferation and osteogenic differentiation of mouse clonal mesenchymal stem cells. Bioprocess Biosyst Eng. 2021;44:1831-1839.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
75.  Elabd C, Ichim TE, Miller K, Anneling A, Grinstein V, Vargas V, Silva FJ. Comparing atmospheric and hypoxic cultured mesenchymal stem cell transcriptome: implication for stem cell therapies targeting intervertebral discs. J Transl Med. 2018;16:222.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 40]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
76.  Lee J, Byeon JS, Lee KS, Gu NY, Lee GB, Kim HR, Cho IS, Cha SH. Chondrogenic potential and anti-senescence effect of hypoxia on canine adipose mesenchymal stem cells. Vet Res Commun. 2016;40:1-10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 9]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
77.  Yu X, Wan Q, Ye X, Cheng Y, Pathak JL, Li Z. Cellular hypoxia promotes osteogenic differentiation of mesenchymal stem cells and bone defect healing via STAT3 signaling. Cell Mol Biol Lett. 2019;24:64.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 45]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
78.  Cicione C, Muiños-López E, Hermida-Gómez T, Fuentes-Boquete I, Díaz-Prado S, Blanco FJ. Effects of severe hypoxia on bone marrow mesenchymal stem cells differentiation potential. Stem Cells Int. 2013;2013:232896.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 60]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
79.  Kim JH, Yoon SM, Song SU, Park SG, Kim WS, Park IG, Lee J, Sung JH. Hypoxia Suppresses Spontaneous Mineralization and Osteogenic Differentiation of Mesenchymal Stem Cells via IGFBP3 Up-Regulation. Int J Mol Sci. 2016;17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 24]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
80.  Wang JP, Liao YT, Wu SH, Chiang ER, Hsu SH, Tseng TC, Hung SC. Mesenchymal stem cells from a hypoxic culture improve nerve regeneration. J Tissue Eng Regen Med. 2020;14:1804-1814.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
81.  Wang L, Zheng F, Song R, Zhuang L, Yang M, Suo J, Li L. Integrins in the Regulation of Mesenchymal Stem Cell Differentiation by Mechanical Signals. Stem Cell Rev Rep. 2022;18:126-141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 16]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
82.  Nguyen AT, Sathe SR, Yim EK. From nano to micro: topographical scale and its impact on cell adhesion, morphology and contact guidance. J Phys Condens Matter. 2016;28:183001.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 173]  [Cited by in F6Publishing: 159]  [Article Influence: 19.9]  [Reference Citation Analysis (0)]
83.  Xia J, Yuan Y, Wu H, Huang Y, Weitz DA. Decoupling the effects of nanopore size and surface roughness on the attachment, spreading and differentiation of bone marrow-derived stem cells. Biomaterials. 2020;248:120014.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 41]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
84.  Winer JP, Janmey PA, McCormick ME, Funaki M. Bone marrow-derived human mesenchymal stem cells become quiescent on soft substrates but remain responsive to chemical or mechanical stimuli. Tissue Eng Part A. 2009;15:147-154.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 279]  [Cited by in F6Publishing: 259]  [Article Influence: 17.3]  [Reference Citation Analysis (0)]
85.  Rowlands AS, George PA, Cooper-White JJ. Directing osteogenic and myogenic differentiation of MSCs: interplay of stiffness and adhesive ligand presentation. Am J Physiol Cell Physiol. 2008;295:C1037-C1044.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 408]  [Cited by in F6Publishing: 412]  [Article Influence: 25.8]  [Reference Citation Analysis (0)]
86.  Sun M, Chi G, Li P, Lv S, Xu J, Xu Z, Xia Y, Tan Y, Li L, Li Y. Effects of Matrix Stiffness on the Morphology, Adhesion, Proliferation and Osteogenic Differentiation of Mesenchymal Stem Cells. Int J Med Sci. 2018;15:257-268.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 142]  [Cited by in F6Publishing: 141]  [Article Influence: 23.5]  [Reference Citation Analysis (0)]
87.  Lin CH, Su JJ, Lee SY, Lin YM. Stiffness modification of photopolymerizable gelatin-methacrylate hydrogels influences endothelial differentiation of human mesenchymal stem cells. J Tissue Eng Regen Med. 2018;12:2099-2111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 27]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
88.  Engler AJ, Sen S, Sweeney HL, Discher DE. Matrix elasticity directs stem cell lineage specification. Cell. 2006;126:677-689.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9969]  [Cited by in F6Publishing: 9125]  [Article Influence: 506.9]  [Reference Citation Analysis (0)]
89.  Liu Y, Li Z, Li J, Yang S, Zhang Y, Yao B, Song W, Fu X, Huang S. Stiffness-mediated mesenchymal stem cell fate decision in 3D-bioprinted hydrogels. Burns Trauma. 2020;8:tkaa029.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 26]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
90.  Mao AS, Shin JW, Mooney DJ. Effects of substrate stiffness and cell-cell contact on mesenchymal stem cell differentiation. Biomaterials. 2016;98:184-191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 168]  [Cited by in F6Publishing: 174]  [Article Influence: 21.8]  [Reference Citation Analysis (0)]
91.  Gungordu HI, Bao M, van Helvert S, Jansen JA, Leeuwenburgh SCG, Walboomers XF. Effect of mechanical loading and substrate elasticity on the osteogenic and adipogenic differentiation of mesenchymal stem cells. J Tissue Eng Regen Med. 2019;13:2279-2290.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
92.  Wu L, Magaz A, Darbyshire A, Howkins A, Reynolds A, Boyd IW, Song H, Song JH, Loizidou M, Emberton M, Birchall M, Song W. Thermoresponsive Stiffness Softening of Hierarchically Porous Nanohybrid Membranes Promotes Niches for Mesenchymal Stem Cell Differentiation. Adv Healthc Mater. 2019;8:e1801556.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
93.  Zhao L, Liu L, Wu Z, Zhang Y, Chu PK. Effects of micropitted/nanotubular titania topographies on bone mesenchymal stem cell osteogenic differentiation. Biomaterials. 2012;33:2629-2641.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 249]  [Cited by in F6Publishing: 202]  [Article Influence: 16.8]  [Reference Citation Analysis (0)]
94.  Chen P, Aso T, Sasaki R, Tsutsumi Y, Ashida M, Doi H, Hanawa T. Micron/Submicron Hybrid Topography of Titanium Surfaces Influences Adhesion and Differentiation Behaviors of the Mesenchymal Stem Cells. J Biomed Nanotechnol. 2017;13:324-336.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 14]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
95.  Qian W, Gong L, Cui X, Zhang Z, Bajpai A, Liu C, Castillo AB, Teo JCM, Chen W. Nanotopographic Regulation of Human Mesenchymal Stem Cell Osteogenesis. ACS Appl Mater Interfaces. 2017;9:41794-41806.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 42]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
96.  Gittens RA, Olivares-Navarrete R, McLachlan T, Cai Y, Hyzy SL, Schneider JM, Schwartz Z, Sandhage KH, Boyan BD. Differential responses of osteoblast lineage cells to nanotopographically-modified, microroughened titanium-aluminum-vanadium alloy surfaces. Biomaterials. 2012;33:8986-8994.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 119]  [Cited by in F6Publishing: 127]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
97.  Olivares-Navarrete R, Hyzy SL, Gittens RA 1st, Schneider JM, Haithcock DA, Ullrich PF, Slosar PJ, Schwartz Z, Boyan BD. Rough titanium alloys regulate osteoblast production of angiogenic factors. Spine J. 2013;13:1563-1570.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 89]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
98.  Deligianni DD, Katsala N, Ladas S, Sotiropoulou D, Amedee J, Missirlis YF. Effect of surface roughness of the titanium alloy Ti-6Al-4V on human bone marrow cell response and on protein adsorption. Biomaterials. 2001;22:1241-1251.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 622]  [Cited by in F6Publishing: 436]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
99.  Huang X, Lan Y, Shen J, Chen Z, Xie Z. Extracellular Vesicles in Bone Homeostasis: Emerging Mediators of Osteoimmune Interactions and Promising Therapeutic Targets. Int J Biol Sci. 2022;18:4088-4100.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 8]  [Reference Citation Analysis (0)]
100.  Bjørge IM, Kim SY, Mano JF, Kalionis B, Chrzanowski W. Extracellular vesicles, exosomes and shedding vesicles in regenerative medicine - a new paradigm for tissue repair. Biomater Sci. 2017;6:60-78.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 165]  [Article Influence: 27.5]  [Reference Citation Analysis (0)]
101.  Pitt JM, Kroemer G, Zitvogel L. Extracellular vesicles: masters of intercellular communication and potential clinical interventions. J Clin Invest. 2016;126:1139-1143.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 271]  [Cited by in F6Publishing: 325]  [Article Influence: 40.6]  [Reference Citation Analysis (0)]
102.  He XT, Li X, Yin Y, Wu RX, Xu XY, Chen FM. The effects of conditioned media generated by polarized macrophages on the cellular behaviours of bone marrow mesenchymal stem cells. J Cell Mol Med. 2018;22:1302-1315.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 31]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
103.  Song X, Xue Y, Fan S, Hao J, Deng R. Lipopolysaccharide-activated macrophages regulate the osteogenic differentiation of bone marrow mesenchymal stem cells through exosomes. PeerJ. 2022;10:e13442.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 13]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
104.  Liu K, Luo X, Lv ZY, Zhang YJ, Meng Z, Li J, Meng CX, Qiang HF, Hou CY, Hou L, Liu FZ, Zhang B. Macrophage-Derived Exosomes Promote Bone Mesenchymal Stem Cells Towards Osteoblastic Fate Through microRNA-21a-5p. Front Bioeng Biotechnol. 2021;9:801432.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 23]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
105.  Xia Y, He XT, Xu XY, Tian BM, An Y, Chen FM. Exosomes derived from M0, M1 and M2 macrophages exert distinct influences on the proliferation and differentiation of mesenchymal stem cells. PeerJ. 2020;8:e8970.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 24]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
106.  Xiong Y, Chen L, Yan C, Zhou W, Yu T, Sun Y, Cao F, Xue H, Hu Y, Chen D, Mi B, Liu G. M2 Macrophagy-derived exosomal miRNA-5106 induces bone mesenchymal stem cells towards osteoblastic fate by targeting salt-inducible kinase 2 and 3. J Nanobiotechnology. 2020;18:66.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 98]  [Article Influence: 24.5]  [Reference Citation Analysis (0)]
107.  Li Z, Wang Y, Li S, Li Y. Exosomes Derived From M2 Macrophages Facilitate Osteogenesis and Reduce Adipogenesis of BMSCs. Front Endocrinol (Lausanne). 2021;12:680328.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 32]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
108.  Kang M, Huang CC, Lu Y, Shirazi S, Gajendrareddy P, Ravindran S, Cooper LF. Bone regeneration is mediated by macrophage extracellular vesicles. Bone. 2020;141:115627.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 91]  [Article Influence: 22.8]  [Reference Citation Analysis (0)]
109.  Bin-Bin Z, Da-Wa ZX, Chao L, Lan-Tao Z, Tao W, Chuan L, Chao-Zheng L, De-Chun L, Chang F, Shu-Qing W, Zu-Nan D, Xian-Wei P, Zhang ZX, Ke-Wen L. M2 macrophagy-derived exosomal miRNA-26a-5p induces osteogenic differentiation of bone mesenchymal stem cells. J Orthop Surg Res. 2022;17:137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
110.  Ekström K, Omar O, Granéli C, Wang X, Vazirisani F, Thomsen P. Monocyte exosomes stimulate the osteogenic gene expression of mesenchymal stem cells. PLoS One. 2013;8:e75227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 158]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
111.  Liang M, Yin X, Zhang S, Ai H, Luo F, Xu J, Dou C, Dong S, Ma Q. Osteoclast-derived small extracellular vesicles induce osteogenic differentiation via inhibiting ARHGAP1. Mol Ther Nucleic Acids. 2021;23:1191-1203.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 23]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
112.  Narayanan K, Kumar S, Padmanabhan P, Gulyas B, Wan ACA, Rajendran VM. Lineage-specific exosomes could override extracellular matrix mediated human mesenchymal stem cell differentiation. Biomaterials. 2018;182:312-322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 56]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
113.  Ailuno G, Baldassari S, Lai F, Florio T, Caviglioli G. Exosomes and Extracellular Vesicles as Emerging Theranostic Platforms in Cancer Research. Cells. 2020;9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 38]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]