Open access peer-reviewed chapter

Mesenchymal Stem/Stromal Cells and Hydrogel Scaffolds for Tissue Engineering

Written By

Leisheng Zhang and Zhihai Han

Submitted: 14 November 2021 Reviewed: 27 November 2021 Published: 09 January 2022

DOI: 10.5772/intechopen.101793

From the Edited Volume

Hydrogels - From Tradition to Innovative Platforms with Multiple Applications

Edited by Lăcrămioara Popa, Mihaela Violeta Ghica and Cristina-Elena Dinu-Pîrvu

Chapter metrics overview

343 Chapter Downloads

View Full Metrics

Abstract

Hydrogels are splendid biomaterials and play a critical role in multiple applications for disease management via offering a microenvironment for drug metabolism and exerting the bonding effect attribute to the preferable physical and chemical properties. State-of-the-art renewal has indicated the combination of hydrogels with mesenchymal stem/stromal cells (MSCs), which are heterogeneous populations with unique hematopoietic-supporting and immunoregulatory properties. For decades, we and other investigators have demonstrated the promising prospects of MSCs in regenerative medicine, and in particular, for the administration of recurrent and refractory disease. Very recently, we took advantage of the hydrogel/MSC composite for the applications in osteoarthritis, burn wounds, and refractory wounds associated with diabetic foot as well. Strikingly, the composite showed superiority in continuous improvement of the biological functions of the injured areas over hydrogels or MSCs, respectively. Collectively, hydrogel-based biomaterials are of importance for disease treatment and the accompanied regenerative medicine. Therefore, in this chapter, we will summarize the latest updates of hydrogel/MSCs composite in tissue engineering and put forward the direction of hotspot issues in the future including hydrogel/MSC and hydrogel/MSC-exosome in preclinical and clinical studies.

Keywords

  • mesenchymal stem/stromal cells
  • exosomes
  • hydrogel scaffolds
  • tissue engineering
  • regenerative medicine

1. Introduction

Mesenchymal stem/stromal cells (MSCs) have been acknowledged as medicinal signaling cells as well as the most important niche cells in the microenvironment, and possess advantaged properties such as immunomodulatory capacity, hematopoietic-supporting effect and multi-lineage differentiation potential towards adipocytes, osteoblasts and chondrocytes, which thus hold promising prospects for tissue engineering and regenerative medicine [1, 2, 3]. MSCs were first isolated from bone marrow in the 1960s, and followed by various stromal fractions of adult tissues [4, 5], perinatal tissues [6, 7, 8], and even derived from stem cells [9, 10, 11]. For decades, due to the limitation of unique biomarkers and the wide range of cell sources, MSCs are recognized as heterogeneous populations with great heterogeneity in cellular phenotypes and transcriptome characteristics [12, 13, 14]. Generally, MSCs with diverse origins mainly function via direct- or trans-differentiation, paracrine or autocrine, homing, dual immunomodulation, neovascularization, and constitutive microenvironment [4, 15, 16]. To date, more than 1340 MSC-based clinical trials have been registered for various disease treatment according to the ClinicalTrials.gov website. For instance, we and other investigators have indicated the therapeutic effects of MSCs upon multiple refractory and recurrent disorders including acute graft-versus-host diseases (aGVHD) [17], aplastic anemia [18, 19], osteoarthritis [11, 20], critical limb ischemia (CLI) [9], acute-on-chronic liver failure (ACLF) [21], Parkinson’s syndrome [22], acute myocardial infarction (AMI) [23], rheumatoid arthritis (RA) [24], and coronavirus disease 2019 (COVID-19) [15, 25]. It’s noteworthy that the variation of the therapeutic efficacy of MSCs upon acute liver failure and aGVHD has also been respectively observed by Zhang et al. and us, which further indicated the necessity and urgency of developing tissue engineering including biomaterials, three-dimensional (3D) printing, and MSC-based gene therapy [26, 27].

Simultaneously, state-of-the-art updates have further suggested the preferable application of biomaterial/MSC composite as well [11, 28, 29]. Of the current natural extracellular matrices (ECMs), hydrogels have been regarded as the most promising alternative biomaterials attribute to their excellent swelling property and the resemblance to soft tissues [11, 30]. In particular, synthetic biomimetic hydrogels with appropriate mechanical behavior and predictable biodegradation property can be easily synthesized and modulated for facilitating the biological phenotypes and bioapplications of the encapsulated MSCs such as adhesion, migration, differentiation, proliferation, and apoptosis [30]. For instance, Gwon et al. and Huang et al. reported the influence of heparin-hyaluronic acid (HA) hydrogel upon cellular activity and hydrogel scaffolds for the differentiation of adipose-derived stem cells, respectively [30, 31]. Very recently, we took advantage of the HA hydrogel/MSC composite and demonstrated the reinforced cell vitality of human pluripotent stem cell-derived MSCs (hPSC-MSCs) over monolayer-cultured MSCs for chondrogenesis and the management of osteoarthritis rabbits [11].

Herein, we summarize the current progress in MSCs or MSC-derived exosomes and hydrogel scaffold for tissue engineering, and in particular, the potentially reinforcing or attenuating effects of hydrogel scaffold with unique biochemical and biophysical properties upon the MSC-based cytotherapy for regenerative medicine.

Advertisement

2. The cell sources of MSCs for tissue engineering

Not until 2006, the International Society for Cellular Therapy (ISCT) defined the preliminary criteria of defining multipotent MSCs including adherent property, multi-lineage differentiation capacities in vitro towards adipocytes, osteoblasts, and chondrocytes, together with high-levels of mesenchymal biomarker expression (CD73, CD90, and CD105) whereas minimal expression of hematopoietic or endothelial markers (CD31, CD34, and CD45) [32]. After that, numerous studies aiming at dissecting the similarities and differences in biological phenotypes and biofunctions as well as transcriptome characteristics of MSCs derived from adult tissue, perinatal tissue, and PSCs have been extensively conducted (Table 1).

ClassificationType of scaffoldDiseaseApplicationsReference
Adult tissue-derived MSCsPF-127 hydrogel/AD-MSCDiabetic wound healingPreclinical studyKaisang et al. [33]
AdhHG hydrogel/G-MSCsCraniofacial bone tissue regenerationPreclinical studyHasani-Sadrabadi et al. [34]
HPCH-PCL-nHA hydrogel/BM-MSCMassive bone defectsPreclinical studyJi et al. [35]
Perinatal tissue-derived MSCsPF-127 hydrogel/UC-MSC-exoChronic diabetic wound healingPreclinical studyYang et al. [36]
Chitosan hydrogel/P-MSC-exoHindlimb ischemiaPreclinical studyZhang et al. [37]
Collagen/UC-MSCsPOFClinical studyDing et al. [38]
Pluripotent stem cell-derived MSCsHA hydrogel/PSC-MSCsOsteoarthritisPreclinical studyZhang et al. [11]
Hydrogel/iPSC-MSCsEndometrial injuryPreclinical studyJi et al. [39]

Table 1.

Representative applications of HA/MSC-based scaffold in tissue engineering.

2.1 Adult tissue-derived MSCs

As mentioned above, adult tissue-derived MSCs hold vast prospect in tissue repairing and organ reconstruction [3]. To date, massive literatures have reported the isolation and identification of MSCs from various adult tissues such as adipose tissue, bone marrow, synovium, dental pulp, peripheral blood, muscle tendon, and menstrual blood [40, 41]. According to the ClinicalTrials.gov website, a total of 1096 trials have been registered worldwide against disorders such as acute respiratory distress syndrome (ARDS), CLI, AMI, anoxic or hypoxic brain injury, moderate-to-severe Crohn’s disease, idiopathic pulmonary fibrosis (IPF), and COVID-19. For example, a phase I interventional trial was led by Dr. Jesus JV Vaquero Crespo in Puerta de Hierro University Hospital was aiming to evaluate the security of local administration of autologous bone marrow-derived MSCs (BM-MSCs) in traumatic injuries of the spinal cord (NCT01909154), which was consistent with another study by Geffner and their colleagues [42]. In details, 12 participants received 1 × 108 BM-MSCs by intrathecal injection (subarachnoid and intramedullary), and another 3 × 107 BM-MSCs by subarachnoid administration after 3 months depending on centromedullary post-traumatic injury. The safety outcomes of the patients were evaluated according to vital signs (ECG, blood pressure, and heart rate) and possibility of adverse reaction (headache, meningeal irritation, and infectious complications). The secondary outcomes were quantized from the view of sensitivity recovery (e.g., surface sensitivity and pain sensitivity), level of chronic pain, neurophysiological parameters, maximum cystometric capacity, and the decrease in volume and hyperintensity of intramedullary lesions. Very recently, Oraee-Yazdani et al. further verified that BM-MSCs in combination with autologous Schwann cell co-transplantation was safe and effective for treating 11 patients of spinal cord injury (SCI), and in particular for spinal cord regeneration during subacute period [43].

Notably, cutting-edge advances have also put forward the variations and limitations of adult tissue-derived MSCs in both preclinical and clinical studies [1, 14, 44]. For example, among the indicated adult tissue-derived MSCs, BM-MSCs are recognized as the widest application in clinical practices whereas with inherent disadvantages such as ethical risk, pathogenic risk, invasive pain, replicative senescence and individual diversity for cell source, and in particular, the limitation in healthy donors and declined long-term ex vivo amplification further restrict the large-scale application in future [11, 44]. Interestingly, despite the variations in signatures and functions, we recently verified the potential conservative properties in adipose tissue-derived stem cells (AD-MSCs) from type 2 diabetics and healthy donors [4]. However, multifaceted diversity among BM-MSCs, AD-MSCs, dental pulp stem cells (DPSCs), and supernumerary teeth-derived apical papillary stem cells (SCAP-Ss) were observed by investigators in the field [16, 45, 46, 47].

2.2 Perinatal tissue-derived MSCs

Perinatal tissues are abundant sources of MSCs and extracellular matrix with a wide range of therapeutic purposes in tissue engineering, which thus act as particularly interesting candidates for regenerative medicine [48, 49]. To date, a variety of perinatal tissue-derived MSCs have been identified such as placental-derived MSCs (P-MSCs), umbilical cord-derived MSCs (UC-MSCs), cord blood-derived MSCs (CB-MSCs) [49, 50], amniotic-derived MSCs (A-MSCs) [51], amniotic fluid-derived MSCs (AF-MSCs) [52], decidua-derived MSCs (D-MSCs) [53], and chorionic villi-derive MSCs (CV-MSCs) [54]. Of the aforementioned perinatal tissue-derived MSCs, UC-MSCs are promising sources with preferable properties in long-term proliferation in vitro and immunoregulation, and most of all, without ethical risks and limitation in supply, and thus hold great prospect for large-scale clinical investigation and investigational new drug (IND) purposes [18, 44]. Up to November 11th of 2021, a total of 317 interventional clinical trials have been registered for the administration of numerous refractory diseases by UC-MSC infusion such as diabetic nephropathy, heart failure, perianal fistulas with Crohn’s disease, lumbar discogenic pain, chronic obstructive pulmonary disease (COPD), Duchenne muscular dystrophy (DMD), and cerebral hemorrhage sequela (CHS) according to ClinicalTrials.gov website.

Similarly, other types of MSC sources are of equal importance in offering “seeds” for tissue engineering and regenerative medicine (e.g., umbilical cord, placenta, amniotic membrane, and amniotic fluid). For instance, Liu and the colleagues took advantage of A-MSCs and conducted intragastric administration and intraperitoneal injection for the management of hydrogen peroxide-induced premature ovarian failure (POF) model. As expected, POF mice with A-MSC transfusion in bilateral ovaries revealed increased estrogen levels, decreased follicle-stimulating hormone level, and evaluated ovarian index and fertility rate, which collectively suggested the ameliorative effects of MSCs in improving the follicular microenvironment and recovering ovarian function in POF [55].

2.3 Pluripotent stem cell-derived MSCs

Pluripotent stem cells (PSCs), including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), possess self-renewal and multi-lineage differentiation potential, which thus provide advantaged “seeds” for disease modeling and drug validation as well as unprecedented opportunities for cytotherapy against intractable diseases [56, 57, 58]. Since the year of 2005, a number of literatures have reported the generation of MSCs from ESCs and iPSCs [59, 60]. Strikingly, the PSC-derived MSCs (PSC-MSCs) revealed multifaceted superiority over those derived from adult tissues such as unlimited source, homogeneity, large-scale generation without pathogenic or ethical risks, and in particular, PSC-MSCs could be used for exploring the early development and molecular mechanism of MSCs [10, 53, 61, 62]. Notably, current studies have suggested the considerable efficacy of MSCs or MSC-derived exosomes in preclinical application including experimental inflammatory bowel disease (IBD) [63, 64], allergic tracheal inflammation (e.g., asthma and anaphylactic rhinitis) [65], experimental autoimmune encephalitis (EAE) of multiple sclerosis [66], lupus nephritis [67], acute colitis [68], kidney fibrosis [69], and hematopoietic reconstitution [70].

Generally, there are three strategies for PSC-MSC generation including monolayer induction, PSCs and stromal cell coculture and the embryoid body (EB) models. However, most of the existing strategies with drawbacks such as laborious manipulations (e.g., handpicking and scraping), time-consuming (3–8 weeks), low efficacy (approximately 5–20%), cell sorting (e.g., CD73+ and CD105+), and serial passages [10, 71, 72]. For instance, Wei et al., Deng et al., Vainieri et al., Wang et al., and Tran et al. reported the elevated generation of PSC-MSCs by modulating intracellular JAK-STAT [9], IKK/NF-κB [73], PDGF-BB [74], bone morphogenetic protein 4 (BMP) [68], and ABB (activin A, 6-bromoindirubin-3′-oxime, and BMP4) [75] signaling pathways in feeder or serum-free model, respectively. Notably, we recently took advantage of the Msh homeobox 2 (MSX2) and small molecule library-based cell programming strategies for high-efficient induction of PSC-MSCs within 2 weeks, respectively [9, 10, 11]. Even though the convenience in practice as well as the promising prospects in tissue engineering and regenerative medicine [76, 77], the potential risks of PSCs attribute to genome editing and their inherent characteristics such as tumorigenicity, heterogeneity, and immunogenicity should cause enough attention [56, 78, 79].

Advertisement

3. Hydrogels/MSCs scaffolds for tissue engineering

The therapeutic effects of MSC transplantation largely attributed to the paracrine including soluble factors and extracellular vehicles (e.g., exosome and secretome), which could be rapidly sequestered and cleared [80]. Hydrogels are ideally suited for MSCs cultivation and are adequate to offer splendid delivery platforms, enhance vehicles retention rates and thus enhance immunomodulation and tissue regeneration after in vivo transplantation (Figure 1, Table 1) [80, 81, 82].

Figure 1.

Illustration of hydrogel/MSC-based cytotherapy for tissue engineering.

3.1 Hydrogels/MSCs scaffolds for wound healing

Wound healings are regulated by series of events with overlapping phases, which represent an intractable issue in clinical practice [83, 84]. State-of-the-art renewal has indicated the prospective applications of hydrogel in combination with MSCs or MSC-derived exosomes in skin wound healing, and in particular, the recurrent and refractory cutaneous types (Table 1) [83, 85]. Of them, chronic refractory wounds are disorders attribute to multifactorial comorbidity with characteristics of inflammation and impaired vascular networks, which eventually result in unfavorable prognosis due to the lack of effective treatments [36, 86]. Recently, Yang and the colleagues topically applied UC-MSC-derived exosomes encapsulated into the thermosensitive PF-127 hydrogel (hydrogel/MSC-exo) and demonstrated that hydrogel/MSC-exo scaffold significantly upregulated expression of multiple cytokines (e.g., VEGF and TGFβ-1), enhanced regeneration of granulation tissue and accelerated wound closure rate in a streptozotocin-induced diabetic rat model, which was further verified by another study in a streptozotocin-induced diabetic model with hydrogel/AD-MSC composite [33, 36]. Marusina et al. and Xin et al. reported the influence of tunable bio-inert poly (ethylene glycol)-based hydrogels and microporous annealed particle hydrogels on MSCs and the optimization of cell-degradable hydrogels/MSCs delivery for wound re-epithelialization [81, 87]. Zhang et al. took advantage of the bioluminescence imaging (BLI) technology and further demonstrated the therapeutic effects of prostaglandin E2 (PGE2) and chitosan (CS) hydrogel (PGE2 + CS hydrogel) in a murine wound healing model via modulating the M1 and M2 paradigms of macrophage activation [88]. Meanwhile, a full sheet consisting of A-MSCs on thermoresponsive polymers have been considered as advantaged skin substitute for the management of burn wounds [89]. Collectively, these studies suggested that hydrogel-based MSC/MSC-exo therapy represent a novel therapeutic approach for refractory cutaneous regeneration of chronic wounds.

3.2 Hydrogels/MSCs scaffolds for osteoarticular diseases

Despite the dramatic progress in bone reconstruction, the osteoarticular diseases and bone regeneration in clinical practices are still challenging [82]. Hydrogels have been extensively investigated in numerous osteoarticular diseases (e.g., osteoarthritis) and bone regeneration (e.g., craniofacial bone tissue) largely attribute to the high cell compatibility [34]. For example, Ji et al. recently combined MSCs with a newly synthesized hybrid scaffold consisting of thermosensitive hydroxypropyl chitin hydrogel (HPCH) and 3D-printed nano-hydroxyapatite (nHA)/poly (ε-caprolactone) (PCL) for bone regeneration. Strikingly, they found the vascularization and osteogenesis and immunomodulation of encapsulated MSCs as well as cytokine secretion of macrophages were collectively orchestrated in bone defect mice model [35].

Osteoarthritis (OA) is recognized as the most prevalent chronic joint disease, which increases in prevalence with age and resultant in functional loss or decline in quality of life, and in particular, act as a major socioeconomic cost worldwide and a leading musculoskeletal cause of impaired mobility in individuals over 65-year-old [90, 91]. Despite joint replacement is an effective strategy for symptomatic end-stage disease, yet most of the functional outcomes are poor and the lifespans of prostheses are largely limited [91, 92]. Current studies have shown that MSC-based cytotherapy are promising for osteoarticular disease administration. For example, Portron et al. and Merceron et al. found that the in vivo chondrogenic potential of AD-MSCs encapsulated in a cellulose-based self-setting hydrogel (Si-HPMC) preconditioned by hypoxia (5% oxygen) was significantly enhanced compare to that in the control (20% oxygen) group, which was confirmed by subcutaneous transplantation of AD-MSCs with an injectable hydrogel in rabbits [93, 94]. Very recently, our group also found that the application of hyaluronic acid (HA) hydrogel/PSC-MSCs and HA hydrogel/hydroxyapatite/UC-MSC (HA/HAP/UC-MSC) composite with reinforced efficacy upon OA rabbits and mice, respectively (Table 1) [11]. It’s noteworthy that Chung and their colleagues have systematically explored and detailed dissected the efficacy of articular cartilage repair in vivo by combining UC-MSCs with various hydrogels such as alginate, pluronic, HA, and chitosan. They finally concluded that HA hydrogel/UC-MSC composites resulted in preferable cartilage repair and collagen organization pattern, which were similar to adjacent uninjured articular cartilage [95]. Additionally, the gingival MSC-laden photocrosslinkable hydrogels were also confirmed with preferable biocompatibility, biodegradability, and osteoconductivity for craniofacial bone tissue engineering in rat peri-implantitis model as well [34]. Taken together, the biodegradable and biocompatible hydrogels can serve as advantaged scaffolds and supply structural integrity for cellular organization and morphogenic guidance of hydrogel scaffold-laden MSCs [88].

3.3 Hydrogels/MSCs scaffolds for reproductive diseases

Premature ovarian failure (POF) is a refractory disorder with declined fertility in females [96, 97]. In 2019, Yang and their colleagues took advantage of collagen scaffold loaded with UC-MSCs (collagen/UC-MSCs) and verified the efficacy in POF mice via increasing estrogen (E2) and ovarian volume, and promoting granulosa cell proliferation and ovarian angiogenesis [96]. Similarly, Ding et al. reported the rescue of E2 concentrations and activation of follicles in the dormant ovaries of premature ovarian failure (POF) patients with long history of infertility after transplantation of collagen/UC-MSC scaffold (Table 1) [38].

As to premature ovarian insufficiency (POI), an intractable endocrine disease that severely restricts the reproductive and physiological function of females and resultant in menopausal symptoms, a series of literatures have suggested the ameliorative effect of hydrogel/MSC composite or hydrogel/MSC-derived microvesicles/secretomes via facilitating angiogenesis, enhancing granulosa cell generation and steroidogenesis, and accelerating follicular regeneration [98, 99, 100, 101]. Notably, Li et al. have summarized the current renewal of the therapeutic effects and molecular mechanisms of MSC-based cytotherapy in both preclinical research and clinical trials [102].

3.4 Hydrogels/MSCs scaffolds for vascular diseases

Peripheral arterial diseases (PAD) are severe medical conditions, which are characterized by blood vascular blockage and low limb Doppler signals and commonly associated with hind-limb ischemia or critical limb ischemia (CLI) [103]. For decades, we and other investigators have primarily suggested the therapeutic of MSCs or MSC-derived exosomes in hind limb ischemia models by alleviating the severity, promoting angiogenesis, and enhancing immunomodulation [9, 104, 105]. In recently years, a certain number of outstanding researchers turned to injectable hydrogels such as self-assembled Nap-GFFYK-Thiol hydrogel, nitric oxide-releasing hydrogels, and the novel hydrogel composed of pooled platelet lysate (PL) to enhance the efficacy of MSCs or derivations upon peripheral artery diseases (PADs) [37, 106, 107, 108]. For example, Lee et al. found that fucoidan was adequate to improve the bioactivity and vasculogenic potential of MSCs in hind limb ischemia murine with chronic kidney disease (CKD) whereas Nammian et al. further compared the variations of efficacy between BM-MSCs and AD-MSCs for CLI [109, 110]. Notably, Ding and their colleagues systematically dissected an injectable nanocomposite hydrogel consisting of chitosan, gelatin, β-glycerophosphate and Arg-Gly-Asp (RGD) peptide for potential applications of facilitating vascularization and tissue engineering [111]. Collectively, the aforementioned studies suggest that hydrogel/MSC-based composites occupy a greater angiogenic potential over single hydrogel- or MSC-based treatment for PADs.

3.5 Hydrogels/MSCs scaffolds for digestive diseases

Gastroparesis is characterized by pyloric dysfunction, vomiting, severe nausea, delayed gastric emptying and impaired fundamental structures, which is related with consume of enteric neurons and interstitial cells of Cajal [112]. Meanwhile, stem cell therapy has also been extensively explored in inflammatory bowel diseases (IBDs) including ulcerative colitis (UC) and Crohn’s disease (CD) in both preclinical studies and clinical trials [113, 114, 115]. State-of-the-art updates have indicated the mitigatory effects of MSCs in gastrointestinal diseases such as acute ulcerative colitis and perianal CD [8, 10, 68, 116]. For example, we recently reported the spatio-temporal metabolokinetics and efficacy of placenta-derived MSCs (P-MSCs) on intractable CD with enterocutaneous fistula in mice via simultaneously accelerating neovascularization and downregulating reactive oxygen species (ROS) [8]. Interestingly, Joddar et al. conducted delivery of the MSC-alginate/gelatin/poly-l-lysine hydrogel atop stomach grafts facing the luminal side, and confirmed the significant advance towards the entire tissue-engineered “microgels” or “gastric patch” [112]. Of note, the therapeutic effects of MSCs via systemic administration are still contradictory largely due to the localization in the lungs, which is confirmed by the outcomes of two clinical trials with BM-MSC transplantation [117, 118]. Therefore, the local administration of hydrogel/MSC or hydrogel/MSC-exosomes are promising alternatives for resolving refractory digestive diseases.

Advertisement

4. Conclusions

Tissue engineering is an inveterate and promising area in the field of regenerative medicine, which also has long-lasting limitations in engineering and regenerating tissues. MSCs of different origins are splendid “seeds” for the efficient administration of various refractory and recurrent diseases. As mentioned above, MSCs as well as the released extracellular vesicles (EVs) reveal substantial therapeutic effect in numerous pathophysiological conditions and potentially reconstructing an extensive range of diseased or damaged tissues and organs in tissue regeneration engineering, which have been predominantly demonstrated from pre-clinical or clinical in vitro and in vivo studies. MSC-encapsulated hydrogel scaffolds demonstrate enhanced cell vitality and committed differentiation, prolonged fundamental and operational consistency, which thus hold promising prospects for tissue engineering and the resultant regenerative medicine. Meanwhile, the exosomes and other nano-scale secretions released from the multivesicular MSCs encapsulated into appropriative hydrogel formulations (e.g., HA, nHAP, PLGA, pDA, and FHE) have manifested higher therapeutic potential in both fundamental research and clinical application. Overall, the current progress of regenerative medicine will extensively benefit from the “advantaged” artificial hydrogel/MSC-based cytotherapy in the near future.

Advertisement

Acknowledgments

The authors would like to thank the members in National Postdoctoral Research Station of Gansu Provincial Hospital, Institute of Biology & Hefei Institutes of Physical Science, Chinese Academy of Sciences, and Institute of Health-Biotech, Health-Biotech (Tianjin) Stem Cell Research Institute Co., Ltd. for their technical support. We also thank the staff in Beijing Yunwei Biotechnology Development Co., Ltd. for their language editing service. This study was supported by grants from China Postdoctoral Science Foundation (2019 M661033), Science and Technology projects of Guizhou Province (QKH-J-ZK[2021]-107), the National Science and Technology Major Projects of China for “Major New Drugs Innovation and Development” (2014ZX09508002-003), Major Program of the National Natural Science Foundation of China (81330015), the project Youth Fund funded by Shandong Provincial Natural Science Foundation (ZR2020QC097), Natural Science Foundation of Tianjin (19JCQNJC12500), Jiangxi Provincial Novel Research & Development Institutions of Shangrao City (2020AB002), the project Youth Fund funded by Jiangxi Provincial Natural Science Foundation (20212BAB216073), Jiangxi Provincial Key New Product Incubation Program from Technical Innovation Guidance Program of Shangrao city (2020G002).

Advertisement

Conflict of interest

The authors declare no conflict of interest.

Advertisement

Notes/thanks/other declarations

Not applicable.

Advertisement

Appendices and nomenclature

MSCs

mesenchymal stem/stromal cells

aGVHD

acute graft-versus-host diseases

CLI

critical limb ischemia

ACLF

acute-on-chronic liver failure

AMI

acute myocardial infarction

RA

rheumatoid arthritis

COVID-19

coronavirus disease 2019

ECMs

extracellular matrices

HA

hyaluronic acid

PSC-MSCs

pluripotent stem cell-derived MSCs

BM-MSCs

bone marrow-derived MSCs

ARDS

acute respiratory distress syndrome

IPF

idiopathic pulmonary fibrosis

DPSCs

dental pulp stem cells

UC-MSCs

umbilical cord-derived MSCs

P-MSCs

placental-derived MSCs

AF-MSCs

amniotic fluid-derived MSCs

A-MSCs

amniotic-derived MSCs

D-MSCs

decidua-derived MSCs

CV-MSCs

chorionic villi-derive MSCs

COPD

chronic obstructive pulmonary disease

CHS

cerebral hemorrhage sequela

DMD

Duchenne muscular dystrophy

POF

premature ovarian failure

EAE

experimental autoimmune encephalitis

IBD

inflammatory bowel disease

ESCs

embryonic stem cells

iPSCs

induced pluripotent stem cells

nHA

nano-hydroxyapatite

CKD

chronic kidney disease

PADs

peripheral artery diseases

CD

Crohn’s disease

UC

ulcerative colitis

IUA

intrauterine adhesions

PGE2

prostaglandin E2

ROS

reactive oxygen species

References

  1. 1. Gao F, Chiu SM, Motan DA, Zhang Z, Chen L, Ji HL, et al. Mesenchymal stem cells and immunomodulation: Current status and future prospects. Cell Death & Disease. 2016;7:e2062. DOI: 10.1038/cddis.2015.327
  2. 2. Peltzer J, Aletti M, Frescaline N, Busson E, Lataillade JJ, Martinaud C. Mesenchymal stromal cells based therapy in systemic sclerosis: Rational and challenges. Frontiers in Immunology. 2018;9:2013. DOI: 10.3389/fimmu.2018.02013
  3. 3. Kfoury Y, Scadden DT. Mesenchymal cell contributions to the stem cell niche. Cell Stem Cell. 2015;16(3):239-253. DOI: 10.1016/j.stem.2015.02.019
  4. 4. Wang L, Zhang L, Liang X, Zou J, Liu N, Liu T, et al. Adipose tissue-derived stem cells from type 2 diabetics reveal conservative alterations in multidimensional characteristics. International Journal of Stem Cells. 2020;13(2):268-278. DOI: 10.15283/ijsc20028
  5. 5. Xu L, Liu Y, Sun Y, Wang B, Xiong Y, Lin W, et al. Tissue source determines the differentiation potentials of mesenchymal stem cells: A comparative study of human mesenchymal stem cells from bone marrow and adipose tissue. Stem Cell Research & Therapy. 2017;8(1):275. DOI: 10.1186/s13287-017-0716-x
  6. 6. Shi L, Huang H, Lu X, Yan X, Jiang X, Xu R, et al. Effect of human umbilical cord-derived mesenchymal stem cells on lung damage in severe COVID-19 patients: A randomized, double-blind, placebo-controlled phase 2 trial. Signal Transduction and Targeted Therapy. 2021;6(1):58. DOI: 10.1038/s41392-021-00488-5
  7. 7. Kim KH, Kim EY, Kim GJ, Ko JJ, Cha KY, Koong MK, et al. Human placenta-derived mesenchymal stem cells stimulate ovarian function via miR-145 and bone morphogenetic protein signaling in aged rats. Stem Cell Research & Therapy. 2020;11(1):472. DOI: 10.1186/s13287-020-01988-x
  8. 8. Hou H, Zhang L, Duan L, Liu Y, Han Z, Li Z, et al. Spatio-temporal metabolokinetics and efficacy of human placenta-derived mesenchymal stem/stromal cells on mice with refractory Crohn’s-like enterocutaneous fistula. Stem Cell Reviews and Reports. 2020;16(6):1292-1304. DOI: 10.1007/s12015-020-10053-2
  9. 9. Wei Y, Hou H, Zhang L, Zhao N, Li C, Huo J, et al. JNKi- and DAC-programmed mesenchymal stem/stromal cells from hESCs facilitate hematopoiesis and alleviate hind limb ischemia. Stem Cell Research & Therapy. 2019;10(1):186. DOI: 10.1186/s13287-019-1302-1
  10. 10. Zhang L, Wang H, Liu C, Wu Q, Su P, Wu D, et al. MSX2 initiates and accelerates mesenchymal stem/stromal cell specification of hPSCs by regulating TWIST1 and PRAME. Stem Cell Reports. 2018;11(2):497-513. DOI: 10.1016/j.stemcr.2018.06.019
  11. 11. Zhang L, Wei Y, Chi Y, Liu D, Yang S, Han Z, et al. Two-step generation of mesenchymal stem/stromal cells from human pluripotent stem cells with reinforced efficacy upon osteoarthritis rabbits by HA hydrogel. Cell & Bioscience. 2021;11(1):6. DOI: 10.1186/s13578-020-00516-x
  12. 12. Aydin S, Sagrac D, Sahin F. Differentiation potential of mesenchymal stem cells into pancreatic beta-cells. Advances in Experimental Medicine and Biology. 2020;1247:135-156. DOI: 10.1007/5584_2019_476
  13. 13. Fan W, Li J, Wang Y, Pan J, Li S, Zhu L, et al. CD105 promotes chondrogenesis of synovium-derived mesenchymal stem cells through Smad2 signaling. Biochemical and Biophysical Research Communications. 2016;474(2):338-344. DOI: 10.1016/j.bbrc.2016.04.101
  14. 14. Nombela-Arrieta C, Ritz J, Silberstein LE. The elusive nature and function of mesenchymal stem cells. Nature Reviews. Molecular Cell Biology. 2011;12(2):126-131. DOI: 10.1038/nrm3049
  15. 15. Zhang LS, Yu Y, Yu H, Han ZC. Therapeutic prospects of mesenchymal stem/stromal cells in COVID-19 associated pulmonary diseases: From bench to bedside. World Journal of Stem Cells. 2021;13(8):1058-1071. DOI: 10.4252/wjsc.v13.i8.1058
  16. 16. Yao J, Chen N, Wang X, Zhang L, Huo J, Chi Y, et al. Human supernumerary teeth-derived apical papillary stem cells possess preferable characteristics and efficacy on hepatic fibrosis in mice. Stem Cells International. 2020;2020:6489396. DOI: 10.1155/2020/6489396
  17. 17. Zhao C, Zhang L, Kong W, Liang J, Xu X, Wu H, et al. Umbilical cord-derived mesenchymal stem cells inhibit cadherin-11 expression by fibroblast-like synoviocytes in rheumatoid arthritis. Journal of Immunology Research. 2015;2015:137695. DOI: 10.1155/2015/137695
  18. 18. Huo J, Zhang L, Ren X, Li C, Li X, Dong P, et al. Multifaceted characterization of the signatures and efficacy of mesenchymal stem/stromal cells in acquired aplastic anemia. Stem Cell Research & Therapy. 2020;11(1):59. DOI: 10.1186/s13287-020-1577-2
  19. 19. Wei Y, Zhang L, Chi Y, Ren X, Gao Y, Song B, et al. High-efficient generation of VCAM-1(+) mesenchymal stem cells with multidimensional superiorities in signatures and efficacy on aplastic anaemia mice. Cell Proliferation. 2020;53(8):e12862. DOI: 10.1111/cpr.12862
  20. 20. Jo CH, Lee YG, Shin WH, Kim H, Chai JW, Jeong EC, et al. Intra-articular injection of mesenchymal stem cells for the treatment of osteoarthritis of the knee: A proof-of-concept clinical trial. Stem Cells. 2014;32(5):1254-1266. DOI: 10.1002/stem.1634
  21. 21. Saleh M, Taher M, Sohrabpour AA, Vaezi AA, Nasiri Toosi M, Kavianpour M, et al. Perspective of placenta derived mesenchymal stem cells in acute liver failure. Cell & Bioscience. 2020;10:71. DOI: 10.1186/s13578-020-00433-z
  22. 22. Staff NP, Jones DT, Singer W. Mesenchymal stromal cell therapies for neurodegenerative diseases. Mayo Clinic Proceedings. 2019;94(5):892-905. DOI: 10.1016/j.mayocp.2019.01.001
  23. 23. Lim M, Wang W, Liang L, Han ZB, Li Z, Geng J, et al. Intravenous injection of allogeneic umbilical cord-derived multipotent mesenchymal stromal cells reduces the infarct area and ameliorates cardiac function in a porcine model of acute myocardial infarction. Stem Cell Research & Therapy. 2018;9(1):129. DOI: 10.1186/s13287-018-0888-z
  24. 24. Wang L, Huang S, Li S, Li M, Shi J, Bai W, et al. Efficacy and safety of umbilical cord mesenchymal stem cell therapy for rheumatoid arthritis patients: A prospective phase I/II study. Drug Design, Development and Therapy. 2019;13:4331-4340. DOI: 10.2147/DDDT.S225613
  25. 25. Aitong W, Leisheng Z, Hao Y. Visualized analyses of investigations upon mesenchymal stem/stromal cell-based cytotherapy and underlying mechanisms for COVID-19 associated ARDS. Current Stem Cell Research & Therapy. 2021. DOI: 10.2174/1574888X16666210712212421 [Online ahead of print]
  26. 26. Mesure B, Menu P, Venkatesan JK, Cucchiarini M, Velot E. Biomaterials and gene therapy: A smart combination for MSC musculoskeletal engineering. Current Stem Cell Research & Therapy. 2019;14(4):337-343. DOI: 10.2174/1574888X14666181205121658
  27. 27. Chen P, Zheng L, Wang Y, Tao M, Xie Z, Xia C, et al. Desktop-stereolithography 3D printing of a radially oriented extracellular matrix/mesenchymal stem cell exosome bioink for osteochondral defect regeneration. Theranostics. 2019;9(9):2439-2459. DOI: 10.7150/thno.31017
  28. 28. Yu H, Cauchois G, Louvet N, Chen Y, Rahouadj R, Huselstein C. Comparison of MSC properties in two different hydrogels. Impact of mechanical properties. Bio-medical Materials and Engineering. 2017;28(s1):S193-S200. DOI: 10.3233/BME-171641
  29. 29. Lv B, Zhang X, Yuan J, Chen Y, Ding H, Cao X, et al. Biomaterial-supported MSC transplantation enhances cell-cell communication for spinal cord injury. Stem Cell Research & Therapy. 2021;12(1):36. DOI: 10.1186/s13287-020-02090-y
  30. 30. Huang Q, Zou Y, Arno MC, Chen S, Wang T, Gao J, et al. Hydrogel scaffolds for differentiation of adipose-derived stem cells. Chemical Society Reviews. 2017;46(20):6255-6275. DOI: 10.1039/c6cs00052e
  31. 31. Gwon K, Kim E, Tae G. Heparin-hyaluronic acid hydrogel in support of cellular activities of 3D encapsulated adipose derived stem cells. Acta Biomaterialia. 2017;49:284-295. DOI: 10.1016/j.actbio.2016.12.001
  32. 32. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8(4):315-317. DOI: 10.1080/14653240600855905
  33. 33. Kaisang L, Siyu W, Lijun F, Daoyan P, Xian CJ, Jie S. Adipose-derived stem cells seeded in Pluronic F-127 hydrogel promotes diabetic wound healing. The Journal of Surgical Research. 2017;217:63-74. DOI: 10.1016/j.jss.2017.04.032
  34. 34. Hasani-Sadrabadi MM, Sarrion P, Pouraghaei S, Chau Y, Ansari S, Li S, et al. An engineered cell-laden adhesive hydrogel promotes craniofacial bone tissue regeneration in rats. Science Translational Medicine. 2020;12(534):eaay6853. DOI: 10.1126/scitranslmed.aay6853
  35. 35. Ji X, Yuan X, Ma L, Bi B, Zhu H, Lei Z, et al. Mesenchymal stem cell-loaded thermosensitive hydroxypropyl chitin hydrogel combined with a three-dimensional-printed poly(epsilon-caprolactone)/nano-hydroxyapatite scaffold to repair bone defects via osteogenesis, angiogenesis and immunomodulation. Theranostics. 2020;10(2):725-740. DOI: 10.7150/thno.39167
  36. 36. Yang J, Chen Z, Pan D, Li H, Shen J. Umbilical cord-derived mesenchymal stem cell-derived exosomes combined pluronic F127 hydrogel promote chronic diabetic wound healing and complete skin regeneration. International Journal of Nanomedicine. 2020;15:5911-5926. DOI: 10.2147/IJN.S249129
  37. 37. Zhang K, Zhao X, Chen X, Wei Y, Du W, Wang Y, et al. Enhanced therapeutic effects of mesenchymal stem cell-derived exosomes with an injectable hydrogel for hindlimb ischemia treatment. ACS Applied Materials & Interfaces. 2018;10(36):30081-30091. DOI: 10.1021/acsami.8b08449
  38. 38. Ding L, Yan G, Wang B, Xu L, Gu Y, Ru T, et al. Transplantation of UC-MSCs on collagen scaffold activates follicles in dormant ovaries of POF patients with long history of infertility. Science China. Life Sciences. 2018;61(12):1554-1565. DOI: 10.1007/s11427-017-9272-2
  39. 39. Ji W, Hou B, Lin W, Wang L, Zheng W, Li W, et al. 3D bioprinting a human iPSC-derived MSC-loaded scaffold for repair of the uterine endometrium. Acta Biomaterialia. 2020;116:268-284. DOI: 10.1136/gut.2010.215152
  40. 40. Samsonraj RM, Raghunath M, Nurcombe V, Hui JH, van Wijnen AJ, Cool SM. Concise review: Multifaceted characterization of human mesenchymal stem cells for use in regenerative medicine. Stem Cells Translational Medicine. 2017;6(12):2173-2185. DOI: 10.1002/sctm.17-0129
  41. 41. Shi Y, Wang Y, Li Q, Liu K, Hou J, Shao C, et al. Immunoregulatory mechanisms of mesenchymal stem and stromal cells in inflammatory diseases. Nature Reviews. Nephrology. 2018;14(8):493-507. DOI: 10.1038/s41581-018-0023-5
  42. 42. Geffner LF, Santacruz P, Izurieta M, Flor L, Maldonado B, Auad AH, et al. Administration of autologous bone marrow stem cells into spinal cord injury patients via multiple routes is safe and improves their quality of life: Comprehensive case studies. Cell Transplantation. 2008;17(12):1277-1293. DOI: 10.3727/096368908787648074
  43. 43. Oraee-Yazdani S, Akhlaghpasand M, Golmohammadi M, Hafizi M, Zomorrod MS, Kabir NM, et al. Combining cell therapy with human autologous Schwann cell and bone marrow-derived mesenchymal stem cell in patients with subacute complete spinal cord injury: Safety considerations and possible outcomes. Stem Cell Research & Therapy. 2021;12(1):445. DOI: 10.1186/s13287-021-02515-2
  44. 44. Zhao Q, Zhang L, Wei Y, Yu H, Zou L, Huo J, et al. Systematic comparison of hUC-MSCs at various passages reveals the variations of signatures and therapeutic effect on acute graft-versus-host disease. Stem Cell Research & Therapy. 2019;10(1):354. DOI: 10.1186/s13287-019-1478-4
  45. 45. Jeyaraman M, Muthu S, Ganie PA. Does the source of mesenchymal stem cell have an effect in the management of osteoarthritis of the knee? Meta-analysis of randomized controlled trials. Cartilage. 2020;13(1_suppl):1532S-1547S. DOI: 10.1177/1947603520951623
  46. 46. El-Sayed M, El-Feky MA, El-Amir MI, Hasan AS, Tag-Adeen M, Urata Y, et al. Immunomodulatory effect of mesenchymal stem cells: Cell origin and cell quality variations. Molecular Biology Reports. 2019;46(1):1157-1165. DOI: 10.1007/s11033-018-04582-w
  47. 47. Sugiyama Y, Sato Y, Kitase Y, Suzuki T, Kondo T, Mikrogeorgiou A, et al. Intravenous administration of bone marrow-derived mesenchymal stem cell, but not adipose tissue-derived stem cell, ameliorated the neonatal hypoxic-ischemic brain injury by changing cerebral inflammatory state in rat. Frontiers in Neurology. 2018;9:757. DOI: 10.3389/fneur.2018.00757
  48. 48. Deus IA, Mano JF, Custodio CA. Perinatal tissues and cells in tissue engineering and regenerative medicine. Acta Biomaterialia. 2020;110:1-14. DOI: 10.1016/j.actbio.2020.04.035
  49. 49. Torre P, Flores AI. Current status and future prospects of perinatal stem cells. Genes (Basel). 2020;12(1):6. DOI: 10.3390/genes12010006
  50. 50. Xiang E, Han B, Zhang Q, Rao W, Wang Z, Chang C, et al. Human umbilical cord-derived mesenchymal stem cells prevent the progression of early diabetic nephropathy through inhibiting inflammation and fibrosis. Stem Cell Research & Therapy. 2020;11(1):336. DOI: 10.1186/s13287-020-01852-y
  51. 51. Lange-Consiglio A, Gusmara C, Manfredi E, Idda A, Soggiu A, Greco V, et al. Antimicrobial effects of conditioned medium from amniotic progenitor cells in vitro and in vivo: Toward tissue regenerative therapies for bovine mastitis. Frontiers in Veterinary Science. 2019;6:443. DOI: 10.3389/fvets.2019.00443
  52. 52. Subhan BS, Kwong J, Kuhn JF, Monas A, Sharma S, Rabbani PS. Amniotic fluid-derived multipotent stromal cells drive diabetic wound healing through modulation of macrophages. Journal of Translational Medicine. 2021;19(1):16. DOI: 10.1186/s12967-020-02674-5
  53. 53. Shofuda T, Kanematsu D, Fukusumi H, Yamamoto A, Bamba Y, Yoshitatsu S, et al. Human decidua-derived mesenchymal cells are a promising source for the generation and cell banking of human induced pluripotent stem cells. CELLMED. 2013;4(3):125-147. DOI: 10.3727/215517912X658918
  54. 54. Kusuma GD, Menicanin D, Gronthos S, Manuelpillai U, Abumaree MH, Pertile MD, et al. Ectopic bone formation by mesenchymal stem cells derived from human term placenta and the decidua. PLoS One. 2015;10(10):e0141246. DOI: 10.1371/journal.pone.0141246
  55. 55. Liu R, Zhang X, Fan Z, Wang Y, Yao G, Wan X, et al. Human amniotic mesenchymal stem cells improve the follicular microenvironment to recover ovarian function in premature ovarian failure mice. Stem Cell Research & Therapy. 2019;10(1):299. DOI: 10.1186/s13287-019-1315-9
  56. 56. Yamanaka S. Pluripotent stem cell-based cell therapy-promise and challenges. Cell Stem Cell. 2020;27(4):523-531. DOI: 10.1016/j.stem.2020.09.014
  57. 57. Lu SJ, Feng Q. CAR-NK cells from engineered pluripotent stem cells: Off-the-shelf therapeutics for all patients. Stem Cells Translational Medicine. 2021;10(Suppl 2):S10-S17. DOI: 10.1002/sctm.21-0135
  58. 58. Sampaziotis F, de Brito MC, Madrigal P, Bertero A, Saeb-Parsy K, Soares FAC, et al. Cholangiocytes derived from human induced pluripotent stem cells for disease modeling and drug validation. Nature Biotechnology. 2015;33(8):845-852. DOI: 10.1038/nbt.3275
  59. 59. Barberi T, Willis LM, Socci ND, Studer L. Derivation of multipotent mesenchymal precursors from human embryonic stem cells. PLoS Medicine. 2005;2(6):e161. DOI: 10.1371/journal.pmed.0020161
  60. 60. Choo A, Lim SK. Derivation of mesenchymal stem cells from human embryonic stem cells. Methods in Molecular Biology. 2011;690:175-182. DOI: 10.1007/978-1-60761-962-8_12
  61. 61. Sfougataki I, Varela I, Stefanaki K, Karagiannidou A, Roubelakis MG, Kalodimou V, et al. Proliferative and chondrogenic potential of mesenchymal stromal cells from pluripotent and bone marrow cells. Histology and Histopathology. 2020;35(12):1415-1426. DOI: 10.14670/HH-18-259
  62. 62. Luzzani CD, Miriuka SG. Pluripotent stem cells as a robust source of mesenchymal stem cells. Stem Cell Reviews and Reports. 2017;13(1):68-78. DOI: 10.1007/s12015-016-9695-z
  63. 63. Kagia A, Tzetis M, Kanavakis E, Perrea D, Sfougataki I, Mertzanian A, et al. Therapeutic effects of mesenchymal stem cells derived from bone marrow, umbilical cord blood, and pluripotent stem cells in a mouse model of chemically induced inflammatory bowel disease. Inflammation. 2019;42(5):1730-1740. DOI: 10.1007/s10753-019-01033-x
  64. 64. da Costa Goncalves F, Paz AH. Cell membrane and bioactive factors derived from mesenchymal stromal cells: Cell-free based therapy for inflammatory bowel diseases. World Journal of Stem Cells. 2019;11(9):618-633. DOI: 10.4252/wjsc.v11.i9.618
  65. 65. Sanchez L, Gutierrez-Aranda I, Ligero G, Rubio R, Munoz-Lopez M, Garcia-Perez JL, et al. Enrichment of human ESC-derived multipotent mesenchymal stem cells with immunosuppressive and anti-inflammatory properties capable to protect against experimental inflammatory bowel disease. Stem Cells. 2011;29(2):251-262. DOI: 10.1002/stem.569
  66. 66. Wang X, Kimbrel EA, Ijichi K, Paul D, Lazorchak AS, Chu J, et al. Human ESC-derived MSCs outperform bone marrow MSCs in the treatment of an EAE model of multiple sclerosis. Stem Cell Reports. 2014;3(1):115-130. DOI: 10.1016/j.stemcr.2014.04.020
  67. 67. Thiel A, Yavanian G, Nastke MD, Morales P, Kouris NA, Kimbrel EA, et al. Human embryonic stem cell-derived mesenchymal cells preserve kidney function and extend lifespan in NZB/W F1 mouse model of lupus nephritis. Scientific Reports. 2015;5:17685. DOI: 10.1038/srep17685
  68. 68. Wang X, Lazorchak AS, Song L, Li E, Zhang Z, Jiang B, et al. Immune modulatory mesenchymal stem cells derived from human embryonic stem cells through a trophoblast-like stage. Stem Cells. 2016;34(2):380-391. DOI: 10.1002/stem.2242
  69. 69. Liu L, Wu Y, Wang P, Shi M, Wang J, Ma H, et al. PSC-MSC-derived exosomes protect against kidney fibrosis in vivo and in vitro through the SIRT6/beta-catenin signaling pathway. International Journal of Stem Cells. 2021;14(3):310-319. DOI: 10.15283/ijsc20184
  70. 70. Li O, Tormin A, Sundberg B, Hyllner J, Le Blanc K, Scheding S. Human embryonic stem cell-derived mesenchymal stroma cells (hES-MSCs) engraft in vivo and support hematopoiesis without suppressing immune function: Implications for off-the shelf ES-MSC therapies. PLoS One. 2013;8(1):e55319. DOI: 10.1371/journal.pone.0055319
  71. 71. Boyd NL, Robbins KR, Dhara SK, West FD, Stice SL. Human embryonic stem cell-derived mesoderm-like epithelium transitions to mesenchymal progenitor cells. Tissue Engineering. Part A. 2009;15(8):1897-1907. DOI: 10.1089/ten.tea.2008.0351
  72. 72. Fukuta M, Nakai Y, Kirino K, Nakagawa M, Sekiguchi K, Nagata S, et al. Derivation of mesenchymal stromal cells from pluripotent stem cells through a neural crest lineage using small molecule compounds with defined media. PLoS One. 2014;9(12):e112291. DOI: 10.1371/journal.pone.0112291
  73. 73. Deng P, Zhou C, Alvarez R, Hong C, Wang CY. Inhibition of IKK/NF-kappaB signaling enhances differentiation of mesenchymal stromal cells from human embryonic stem cells. Stem Cell Reports. 2016;6(4):456-465. DOI: 10.1016/j.stemcr.2016.02.006
  74. 74. Vainieri ML, Lolli A, Kops N, D’Atri D, Eglin D, Yayon A, et al. Evaluation of biomimetic hyaluronic-based hydrogels with enhanced endogenous cell recruitment and cartilage matrix formation. Acta Biomaterialia. 2020;101:293-303. DOI: 10.1016/j.actbio.2019.11.015
  75. 75. Tran NT, Trinh QM, Lee GM, Han YM. Efficient differentiation of human pluripotent stem cells into mesenchymal stem cells by modulating intracellular signaling pathways in a feeder/serum-free system. Stem Cells and Development. 2012;21(7):1165-1175. DOI: 10.1089/scd.2011.0346
  76. 76. Li E, Zhang Z, Jiang B, Yan L, Park JW, Xu RH. Generation of mesenchymal stem cells from human embryonic stem cells in a complete serum-free condition. International Journal of Biological Sciences. 2018;14(13):1901-1909. DOI: 10.7150/ijbs.25306
  77. 77. Jiang B, Yan L, Wang X, Li E, Murphy K, Vaccaro K, et al. Concise review: Mesenchymal stem cells derived from human pluripotent cells, an unlimited and quality-controllable source for therapeutic applications. Stem Cells. 2019;37(5):572-581. DOI: 10.1002/stem.2964
  78. 78. Hockemeyer D, Jaenisch R. Induced pluripotent stem cells meet genome editing. Cell Stem Cell. 2016;18(5):573-586. DOI: 10.1016/j.stem.2016.04.013
  79. 79. Yamanaka S. Induced pluripotent stem cells: Past, present, and future. Cell Stem Cell. 2012;10(6):678-684. DOI: 10.1016/j.stem.2012.05.005
  80. 80. Brennan MA, Layrolle P, Mooney DJ. Biomaterials functionalized with MSC secreted extracellular vesicles and soluble factors for tissue regeneration. Advanced Functional Materials. 2020;30(37):1909125. DOI: 10.1002/adfm.201909125
  81. 81. Marusina AI, Merleev AA, Luna JI, Olney L, Haigh NE, Yoon D, et al. Tunable hydrogels for mesenchymal stem cell delivery: Integrin-induced transcriptome alterations and hydrogel optimization for human wound healing. Stem Cells. 2020;38(2):231-245. DOI: 10.1002/stem.3105
  82. 82. Khayambashi P, Iyer J, Pillai S, Upadhyay A, Zhang Y, Tran SD. Hydrogel encapsulation of mesenchymal stem cells and their derived exosomes for tissue engineering. International Journal of Molecular Sciences. 2021;22(2):684. DOI: 10.3390/ijms22020684
  83. 83. An Y, Lin S, Tan X, Zhu S, Nie F, Zhen Y, et al. Exosomes from adipose-derived stem cells and application to skin wound healing. Cell Proliferation. 2021;54(3):e12993. DOI: 10.1111/cpr.12993
  84. 84. Weiliang Z, Lili G. Research advances in the application of adipose-derived stem cells derived exosomes in cutaneous wound healing. Annals of Dermatology. 2021;33(4):309-317. DOI: 10.5021/ad.2021.33.4.309
  85. 85. Qiu H, Liu S, Wu K, Zhao R, Cao L, Wang H. Prospective application of exosomes derived from adipose-derived stem cells in skin wound healing: A review. Journal of Cosmetic Dermatology. 2020;19(3):574-581. DOI: 10.1111/jocd.13215
  86. 86. Bray ER, Oropallo AR, Grande DA, Kirsner RS, Badiavas EV. Extracellular vesicles as therapeutic tools for the treatment of chronic wounds. Pharmaceutics. 2021;13(10):1543. DOI: 10.3390/pharmaceutics13101543
  87. 87. Xin S, Gregory CA, Alge DL. Interplay between degradability and integrin signaling on mesenchymal stem cell function within poly(ethylene glycol) based microporous annealed particle hydrogels. Acta Biomaterialia. 2020;101:227-236. DOI: 10.1016/j.actbio.2019.11.009
  88. 88. Zhang S, Liu Y, Zhang X, Zhu D, Qi X, Cao X, et al. Prostaglandin E2 hydrogel improves cutaneous wound healing via M2 macrophages polarization. Theranostics. 2018;8(19):5348-5361. DOI: 10.7150/thno.27385
  89. 89. Kitala D, Klama-Baryla A, Kraut M, Labus W, Glik J, Kawecki M, et al. Amniotic stem cells cultured on thermoresponsive polymers allow obtaining a full cell sheet. Transplantation Proceedings. 2020;52(7):2198-2203. DOI: 10.1016/j.transproceed.2020.02.118
  90. 90. Xia B, Di C, Zhang J, Hu S, Jin H, Tong P. Osteoarthritis pathogenesis: A review of molecular mechanisms. Calcified Tissue International. 2014;95(6):495-505. DOI: 10.1007/s00223-014-9917-9
  91. 91. Glyn-Jones S, Palmer AJ, Agricola R, Price AJ, Vincent TL, Weinans H, et al. Osteoarthritis. Lancet. 2015;386(9991):376-387. DOI: 10.1016/S0140-6736(14)60802-3
  92. 92. Bijlsma JW, Berenbaum F, Lafeber FP. Osteoarthritis: An update with relevance for clinical practice. Lancet. 2011;377(9783):2115-2126. DOI: 10.1016/S0140-6736(11)60243-2
  93. 93. Portron S, Merceron C, Gauthier O, Lesoeur J, Sourice S, Masson M, et al. Effects of in vitro low oxygen tension preconditioning of adipose stromal cells on their in vivo chondrogenic potential: Application in cartilage tissue repair. PLoS One. 2013;8(4):e62368. DOI: 10.1371/journal.pone.0062368
  94. 94. Merceron C, Portron S, Masson M, Lesoeur J, Fellah BH, Gauthier O, et al. The effect of two- and three-dimensional cell culture on the chondrogenic potential of human adipose-derived mesenchymal stem cells after subcutaneous transplantation with an injectable hydrogel. Cell Transplantation. 2011;20(10):1575-1588. DOI: 10.3727/096368910X557191
  95. 95. Chung JY, Song M, Ha CW, Kim JA, Lee CH, Park YB. Comparison of articular cartilage repair with different hydrogel-human umbilical cord blood-derived mesenchymal stem cell composites in a rat model. Stem Cell Research & Therapy. 2014;5(2):39. DOI: 10.1186/scrt427
  96. 96. Yang Y, Lei L, Wang S, Sheng X, Yan G, Xu L, et al. Transplantation of umbilical cord-derived mesenchymal stem cells on a collagen scaffold improves ovarian function in a premature ovarian failure model of mice. In Vitro Cellular & Developmental Biology. Animal. 2019;55(4):302-311. DOI: 10.1007/s11626-019-00337-4
  97. 97. Zhang H, Luo Q, Lu X, Yin N, Zhou D, Zhang L, et al. Effects of hPMSCs on granulosa cell apoptosis and AMH expression and their role in the restoration of ovary function in premature ovarian failure mice. Stem Cell Research & Therapy. 2018;9(1):20. DOI: 10.1186/s13287-017-0745-5
  98. 98. Vural B, Duruksu G, Vural F, Gorguc M, Karaoz E. Effects of VEGF (+) mesenchymal stem cells and platelet-rich plasma on inbred rat ovarian functions in cyclophosphamide-induced premature ovarian insufficiency model. Stem Cell Reviews and Reports. 2019;15(4):558-573. DOI: 10.1007/s12015-019-09892-5
  99. 99. Park HS, Ashour D, Elsharoud A, Chugh RM, Ismail N, El Andaloussi A, et al. Towards cell free therapy of premature ovarian insufficiency: Human bone marrow mesenchymal stem cells secretome enhances angiogenesis in human ovarian microvascular endothelial cells. HSOA Journal of Stem Cells Research, Development & Therapy. 2019;5(2):019. DOI: 10.24966/srdt-2060/100019
  100. 100. Zhao Y, Ma J, Yi P, Wu J, Zhao F, Tu W, et al. Human umbilical cord mesenchymal stem cells restore the ovarian metabolome and rescue premature ovarian insufficiency in mice. Stem Cell Research & Therapy. 2020;11(1):466. DOI: 10.1186/s13287-020-01972-5
  101. 101. Park HS, Chugh RM, El Andaloussi A, Hobeika E, Esfandyari S, Elsharoud A, et al. Human BM-MSC secretome enhances human granulosa cell proliferation and steroidogenesis and restores ovarian function in primary ovarian insufficiency mouse model. Scientific Reports. 2021;11(1):4525. DOI: 10.1038/s41598-021-84216-7
  102. 102. Li Z, Zhang M, Tian Y, Li Q, Huang X. Mesenchymal stem cells in premature ovarian insufficiency: Mechanisms and prospects. Frontiers in Cell and Development Biology. 2021;9:718192. DOI: 10.3389/fcell.2021.718192
  103. 103. Kim HJ, Yang H, Jung DH, Hwang JT, Ko BS. Ameliorating effects of Cuscuta chinensis Lamak extract on hindlimb ischemia, and angiogenic or inflammatory associated factors in ovariectomized mice. Molecular Medicine Reports. 2019;19(4):3321-3329. DOI: 10.3892/mmr.2019.9977
  104. 104. Hu GW, Li Q, Niu X, Hu B, Liu J, Zhou SM, et al. Exosomes secreted by human-induced pluripotent stem cell-derived mesenchymal stem cells attenuate limb ischemia by promoting angiogenesis in mice. Stem Cell Research & Therapy. 2015;6:10. DOI: 10.1186/scrt546
  105. 105. Lian Q, Zhang Y, Zhang J, Zhang HK, Wu X, Zhang Y, et al. Functional mesenchymal stem cells derived from human induced pluripotent stem cells attenuate limb ischemia in mice. Circulation. 2010;121(9):1113-1123. DOI: 10.1161/CIRCULATIONAHA.109.898312
  106. 106. Huang A, Liu D, Qi X, Yue Z, Cao H, Zhang K, et al. Self-assembled GFFYK peptide hydrogel enhances the therapeutic efficacy of mesenchymal stem cells in a mouse hindlimb ischemia model. Acta Biomaterialia. 2019;85:94-105. DOI: 10.1016/j.actbio.2018.12.015
  107. 107. Robinson ST, Douglas AM, Chadid T, Kuo K, Rajabalan A, Li H, et al. A novel platelet lysate hydrogel for endothelial cell and mesenchymal stem cell-directed neovascularization. Acta Biomaterialia. 2016;36:86-98. DOI: 10.1016/j.actbio.2016.03.002
  108. 108. Zhang K, Chen X, Li H, Feng G, Nie Y, Wei Y, et al. A nitric oxide-releasing hydrogel for enhancing the therapeutic effects of mesenchymal stem cell therapy for hindlimb ischemia. Acta Biomaterialia. 2020;113:289-304. DOI: 10.1016/j.actbio.2020.07.011
  109. 109. Lee JH, Ryu JM, Han YS, Zia MF, Kwon HY, Noh H, et al. Fucoidan improves bioactivity and vasculogenic potential of mesenchymal stem cells in murine hind limb ischemia associated with chronic kidney disease. Journal of Molecular and Cellular Cardiology. 2016;97:169-179. DOI: 10.1016/j.yjmcc.2016.05.011
  110. 110. Nammian P, Asadi-Yousefabad SL, Daneshi S, Sheikhha MH, Tabei SMB, Razban V. Comparative analysis of mouse bone marrow and adipose tissue mesenchymal stem cells for critical limb ischemia cell therapy. Stem Cell Research & Therapy. 2021;12(1):58. DOI: 10.1186/s13287-020-02110-x
  111. 111. Ding Y, Zhao AS, Liu T, Wang YN, Gao Y, Li JA, et al. An injectable nanocomposite hydrogel for potential application of vascularization and tissue repair. Annals of Biomedical Engineering. 2020;48(5):1511-1523. DOI: 10.1007/s10439-020-02471-7
  112. 112. Joddar B, Tasnim N, Thakur V, Kumar A, McCallum RW, Chattopadhyay M. Delivery of mesenchymal stem cells from gelatin-alginate hydrogels to stomach lumen for treatment of gastroparesis. Bioengineering. 2018;5(1):12. DOI: 10.3390/bioengineering5010012
  113. 113. Ocansey DKW, Qiu W, Wang J, Yan Y, Qian H, Zhang X, et al. The achievements and challenges of mesenchymal stem cell-based therapy in inflammatory bowel disease and its associated colorectal cancer. Stem Cells International. 2020;2020:7819824. DOI: 10.1155/2020/7819824
  114. 114. Irhimeh MR, Cooney J. Management of inflammatory bowel disease using stem cell therapy. Current Stem Cell Research & Therapy. 2016;11(1):72-77. DOI: 10.2174/1574888x10666150728121738
  115. 115. Hoffman AM, Dow SW. Concise review: Stem cell trials using companion animal disease models. Stem Cells. 2016;34(7):1709-1729. DOI: 10.1002/stem.2377
  116. 116. Carvello M, Lightner A, Yamamoto T, Kotze PG, Spinelli A. Mesenchymal stem cells for perianal Crohn’s disease. Cell. 2019;8(7):764. DOI: 10.3390/cells8070764
  117. 117. Dhere T, Copland I, Garcia M, Chiang KY, Chinnadurai R, Prasad M, et al. The safety of autologous and metabolically fit bone marrow mesenchymal stromal cells in medically refractory Crohn’s disease—A phase 1 trial with three doses. Alimentary Pharmacology & Therapeutics. 2016;44(5):471-481. DOI: 10.1111/apt.13717
  118. 118. Duijvestein M, Vos AC, Roelofs H, Wildenberg ME, Wendrich BB, Verspaget HW, et al. Autologous bone marrow-derived mesenchymal stromal cell treatment for refractory luminal Crohn’s disease: Results of a phase I study. Gut. 2010;59(12):1662-1669. DOI: 10.1136/gut.2010.215152

Written By

Leisheng Zhang and Zhihai Han

Submitted: 14 November 2021 Reviewed: 27 November 2021 Published: 09 January 2022