Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 2998 word(s) 2998 2022-03-06 09:16:05 |
2 FORMAT CHANGE Meta information modification 2998 2022-03-07 10:05:33 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Ding, J. Mesenchymal Stem Cell-Based Regenerative Medicine. Encyclopedia. Available online: https://encyclopedia.pub/entry/20264 (accessed on 11 September 2024).
Ding J. Mesenchymal Stem Cell-Based Regenerative Medicine. Encyclopedia. Available at: https://encyclopedia.pub/entry/20264. Accessed September 11, 2024.
Ding, Jianxun. "Mesenchymal Stem Cell-Based Regenerative Medicine" Encyclopedia, https://encyclopedia.pub/entry/20264 (accessed September 11, 2024).
Ding, J. (2022, March 07). Mesenchymal Stem Cell-Based Regenerative Medicine. In Encyclopedia. https://encyclopedia.pub/entry/20264
Ding, Jianxun. "Mesenchymal Stem Cell-Based Regenerative Medicine." Encyclopedia. Web. 07 March, 2022.
Mesenchymal Stem Cell-Based Regenerative Medicine
Edit

In recent decades, the biomedical applications of mesenchymal stem cells (MSCs) have attracted increasing attention. MSCs are easily extracted from the bone marrow, fat, and synovium, and differentiate into various cell lineages according to the requirements of specific biomedical applications. As MSCs do not express significant histocompatibility complexes and immune stimulating molecules, they are not detected by immune surveillance and do not lead to graft rejection after transplantation. These properties make them competent biomedical candidates, especially in tissue engineering.

mesenchymal stem cell extraction cell differentiation growth factor scaffold biomaterial tissue engineering regenerative medicine biomedical application

1. Introduction

Since the discovery of spindle-shaped, bone marrow-derived plastic-adherent cells in the mid-1970s [1], science has come a long way, and studies have found that these cells could differentiate into osteoblasts and chondrocytes [2][3]. Techniques for extraction, culture, and induction of mesenchymal stem cells (MSCs) have improved, with almost all MSC types derived from various tissues now capable of differentiation into osteocytes and end-stage lineages [4]. The rapid development of molecular biology and transplantation techniques has benefitted MSC applications in regenerative medicine.
MSCs are an ideal cell source for tissue regeneration, owing to the excellent properties as follows. MSCs exist in almost all tissues, including bone marrow, adipose, and synovium [5], and are easily extracted. MSCs can differentiate into almost any end-stage lineage cells to enable their seeding in specific scaffolds (Figure 1) [6]. Their immunological properties, including anti-inflammatory, immunoregulatory, and immunosuppressive capacities, contribute to their potential role as immune tolerant agents [7][8].
Figure 1. Schematic diagram of regenerative medicine based on mesenchymal stem cells (MSCs). The MSCs can be easily extracted from varies tissues, and the multilineage differentiation and immunoregulatory properties of MSCs make them an ideal cell therapeutic candidate.
Numerous studies have explored MSCs for tissue regeneration in several animal models in vitro; trials have not been limited to preclinical validation. Several clinical reports verify the potential efficacy of MSC-based cell therapy; although its effectiveness remains limited, the outcomes are inspiring.

2. Bone Regeneration

Bone defects frequently accompany recovery from trauma, revision arthroplasty, or tumor resection surgeries. Autologous bone grafting represents the gold standard therapeutic strategy, despite its many drawbacks, including (1) the limited supply of autologous bone, (2) increased operation time and blood loss, (3) temporary disruption of bone structure in the donor site, and (4) donor site morbidity [9]. Allografting carries a risk of disease and/or infection [10]. Therefore, MSC-based bone regeneration is considered an optimal approach [11].
The MSC osteoblast-differentiation capacity has been identified [2][3], with BMSCs representing the most frequently applied cells for osteoblast differentiation [2]. Comparative studies evaluating the osteogenic ability of other MSC types yielded no definitive conclusions. By contrast, UCB-MSCs show better angiogenic capacity, supporting more abundant blood supply during bone regeneration [12], which promotes rapid tissue reconstruction. In addition to BMSCs, human dental pulp stem cells (hDPSCs) show excellent vascular differentiation potential while differentiating into osteoblasts, which subsequently support bone regeneration [13]. However, these hDPSCs are screened from a stromal vascular dental pulp fraction; therefore, this represents a limited source for further research and application. Since ADSCs can be routinely isolated from lipoaspirate with a high degree of purity with minimal donor site morbidity or patient discomfort, ADSCs are considered to have the most significant potential as a primary source for clinical bone tissue engineering [14][15][16]. Additional comparative and screening studies are necessary to identify other cell sources with applications in bone reconstruction.
Stimulating factors play an important role in directing MSC differentiation into target cells in vitro. The most commonly used inducing factor for osteogenesis is the bone morphogenetic protein-2 (BMP-2), which is usually immobilized on scaffolds to promote osteoblast differentiation. BMP-2 exhibits a strong osteogenic ability, which can be tested by the osteoblast activity and/or expression of bone markers, such as alkaline phosphatase (ALP), osteopontin (OPN), and osteocalcin (OCN) [17][18][19]. BMP-7 activates the transforming growth factor-β (TGF-β) /SMAD signaling in CD105+ MSCs to enhance the expression of osteogenesis-related genes [20]; Wnt11 enhances the osteogenic potential of BMP-9 [21]. Nano-hydroxyapatite [17] and strontium [22] are used as osteogenic regulators in tissue engineering to promote osteogenic differentiation of MSCs while changing the physical properties of the scaffolds.
Studies of MSC-based cell therapy for bone defects and the use of novel scaffolds describe inspiring advances in vitro and in vivo [23][24]. Clinical applications of MSCs in bone reconstruction have been described, including those involving implantation of scaffolds seeded with MSCs into bone defect sites. Specifically, dentists have used this technique to address alveolar cleft defects, jaw defect reconstruction, and maxillary sinus augmentation, with excellent outcomes [25][26][27]. Defects in or non-union of human tubular bone have been tentatively treated via local implantation of MSCs with or without scaffolds [28][29].

3. Cartilage Repair

Cartilage defect repair is one of the significant challenges faced by orthopedic surgeons. Due to the inherent avascular nature of cartilage and the proliferation of mature chondrocytes, cartilage is greatly limited in its ability to repair itself. Currently, the clinically applied cartilage repair techniques, such as bone marrow stimulation and osteochondral transplantation, have their limitations. Fibrocartilage produced by bone marrow stimulation is not strong enough, and grafts for osteochondral transplantation are challenging to integrate.
MSCs offer a new strategy for the repair of damaged cartilage, as they can differentiate into chondrocytes [2][3]. An integrated cartilage reconstruction unit comprises cells, scaffolds, and stimulatory factors, with BMSCs [30], ADSCs [15], and SMSCs [31], used as the primary cell sources. Among these, BMSCs displayed better chondrogenic capacity in vitro and in vivo [32][33][30], although SMSCs show better proliferation and differentiation potential and less hypertrophy than BMSCs and ADSCs [31]. Cartilage reconstruction requires a combination of multiple stimulating factors; co-culture of chondrocytes and MSCs would achieve better results than the application of MSCs alone [34].
Novel bioactive three-dimensional (3D) scaffolds, such as hydrogels [13] and electrospun scaffolds [35], have undergone constant improvement. These scaffolds provide an optimal 3D microenvironment for cartilage regeneration. Moreover, hydrogels can regulate MSC proliferation and differentiation due to their high water content and biocompatibility and similar properties to the extracellular matrix (ECM) [13]. Injectable hydrogels enable minimally invasive treatment of large areas of cartilage defects [36]; thus, hydrogels loaded with MSCs and stimulating factors are highly efficacious for the repair of cartilage damage. The development of electrospun scaffolds suggests that the arrangement of nanofibers also affects cell differentiation and provides a different approach to cartilage repair [35]. Stimulating factors are necessary for cartilage engineering and responsible for inducing, accelerating, and/or enhancing cartilage formation. Common stimulating factors include BMP-2/-4, insulin-like growth factor (IGF)-1, and TGF-β1/-β3 [37][38][39]. Moreover, physical stimuli, such as hydrostatic pressure and dynamic compression, have been explored to induce MSC-mediated cartilage formation [40].
The first preclinical trial of MSC application for cartilage repair occurred in 1994 [41]. MSCs were seeded into a collagen (Col) gel to treat a full-thickness defect in rabbit femoral cartilage, resulting in better outcomes than those observed in a control group. This defect model was subsequently used as a classical cartilage defect model for cartilage regeneration; subsequently, numerous trials have been conducted in both animal models and humans to evaluate MSC-based therapy for cartilage damage [42].
Despite clinical trials being conducted, there are no commercially available products for MSC-based cartilage reconstruction [43][44]. Several studies investigated the effects of expanded MSCs in vivo on damage to human articular cartilage. Transplantation of expanded autologous BMSCs improved cartilage quality in patients with chronic knee osteoarthritis [44], although the clinical improvement was not significant [43]. Other studies reported the injection of allogeneic MSCs into joints in the presence or absence of pre-mixing with autologous chondrocytes [42]. All the clinical outcomes indicated the safety of these therapeutic approaches, and their ability to relieve some symptoms, although their ability to repair the effects of cartilage damage was not always apparent. MSC transplantation showed better results for early lesions [42].
There remain many challenges for MSC-based cartilage regeneration, including the identification of optimal cell sources. Additional studies are needed to enable the use of MSC-based materials as commercial products for implantation to promote cartilage regeneration.

4. Regeneration of Other Musculoskeletal Tissues

Recent studies investigated the MSC-mediated regeneration of musculoskeletal tissues outside the bone and cartilage, including the meniscus, tendons and ligaments, and intervertebral discs (IVD).
Meniscus regeneration has received increasing attention. Intra-articular administration of MSCs to promote meniscal regeneration was first performed with favorable outcomes [45]. Similar to its use for cartilage regeneration, hydrogels [46] and electrospun scaffolds [47] loaded with MSCs were used to reconstruct the meniscus. Moreover, the meniscus-derived decellularized matrix shows better histocompatibility and is more capable of inducing MSC differentiation as compared with natural or synthetic polymer materials [48]. Scaffold-free tissue-engineered constructs show promise as an MSC-based implantation technique to repair meniscal lesions [49]. Tarafder et al. [50] proposed the recruitment of synovial MSCs through connective tissue TGF and TGF-β3 to repair meniscus injury, thereby avoiding the disadvantages of cell-based techniques. Mechanical stimulation is crucial for meniscus growth and maintenance, with mechanical stimuli, such as dynamic compression and tensile loads applied for meniscus repair [51]. Although satisfactory results were obtained in animal models, there remains a lack of evidence in humans regarding the capability of MSCs for forming durable tissues similar to the meniscus [52].
Tendon injury is a common problem associated with sports [53]. BMP-14 induces myogenic differentiation of BMSCs via the sirtuin-1−Janus N-terminal kinase (JNK)/SMAD1-peroxisome proliferator-activated receptor-γ signaling pathway [54]. Studies describing tendogenic differentiation of MSCs were not limited to stimulating factors [55] and scaffolds [56] but also referred to mechanical stimuli that play essential roles in MSC differentiation into tendon lineages. Uniaxial cyclic stretching promoted tendogenic differentiation of MSCs in vitro and in vivo [57]; however, MSCs did not repair tendon injury but only delayed lesion progression [58].
With the increasing age of populations, IVD degeneration has become prevalent. MSCs represent promising candidates for disc regeneration; scaffolds made of Col provide readily available support for chondrogenic differentiation of MSCs in vitro, although the phenotype of the differentiated MSCs is not yet equivalent to that of nucleus pulposus (NP) cells [59]. The acellular matrices derived from NP cells stimulated by TGF-β3 also enhance MSC differentiation [60]. Transfection of adenoviral expression of SOX-9 and BMP-2 in BMSCs increased Col II and aggrecan expression, and promoted IVD repair [61]. Varma et al. [62] used a hydrogel loaded with two different concentrations of MSCs to repair NP, and showed that MSC inoculation at a lower density resulted in a better NP-specific matrix phenotype. A systematic review of MSC-based cell therapy for IVD indicated the safety and effectiveness of short-term MSC transplantation, as well as the necessity for human-based clinical trials [63]. In 2011, expanded autologous BMSCs injected into patients with lumbar disc degeneration revealed several advantages and better prognosis relative to the current gold standard treatments [64]. Clinical percutaneous injection of autologous bone marrow concentrate cells into a patient with degenerative IVD resulted in decreased lumbar discogenic pain within 13 years [65]. Clinical studies [66] indicated that MSC transplantation represents a safe treatment option for degenerative IVD; the specific effects need verification by additional clinical trials.

5. Central Nervous System Rebuilding

The adult central nervous system (CNS) lacks the ability to repair damaged neurons, so the damage of CNS is irreversible, and there is currently no effective repair method for CNS injury in clinical practice repair. In the area of CNS regeneration, MSC-based therapy mainly focuses on two areas: Damage or injury of the CNS caused by severe trauma and continuous ischemia and CNS dysfunction caused by neurologic diseases. To date, BMSCs and ADSCs are the most extensively studied cell sources for CNS repair, with each showing similar neuronal differentiation potential [67][68]. BMSCs can reduce scar formation around spinal cord injury (SCI) lesions and promoted axonal regeneration [69]; however, ADSCs might represent a more suitable cell source owing to their easy extraction and abundant sources. ADSCs inhibit inflammation of the nervous system and improve the recovery of function from traumatic brain injury via neural stem cells [70]. UCB-MSCs can be induced to differentiate into neuron-like cells in vitro [71]; DPSCs can differentiate into neurons and express multiple factors that promote neuronal and axonal regeneration [72].
MSC expression of neuronal or astrocytic markers has been observed in vitro [73] and in vivo [74]. To promote MSC-based CNS restoration, gene-modified MSCs, such as neurotrophin-3-transferred BMSCs, showed improved neuronal differentiation in vivo [75]. Persistent release of specific cytokines and growth factors, which can facilitate neurogenesis, angiogenesis, and synaptogenesis, creates a favorable microenvironment for angiogenesis or remyelination during reconstruction [76]. IGF-1-transfected spinal cord-derived neural stem cells displayed higher viability and the ability to differentiate into oligodendrocytes [77]. Moreover, MSCs can induce T cell tolerance and release of paracrine anti-inflammatory factors, such as TGF-β, that promote neuroprotective effects [78].
Animal models of traumatic and ischemic brain injury or SCI have been used to evaluate MSC-based therapy [79][80]. A meta-analysis of 1568 rats with traumatic SCI showed that MSC therapy provided a substantial beneficial effect on locomotor recovery [80]. Clinical studies indicated MSC-based therapy as a safe and feasible technique for patients with SCI and/or traumatic brain injury [81][82]. Migration of MSC pretreated under hypoxic conditions to the peri-cerebral injury area of cerebral hemorrhagic stroke victimized rats resulted in the release of various growth factors to promote neurogenesis and neurological recovery [83]. For neurological diseases, non-expanded or expanded MSCs have been widely used for the treatment of multiple sclerosis [84], amyotrophic lateral sclerosis [85], ischemic stroke [86], and Parkinson’s disease [87]. Most of the beneficial effects of MSCs on neurological diseases are associated with their immunomodulatory and neuroprotective properties exerted following local injection of non-expanded or expanded autologous MSCs, with clinical trials assessing their ability to achieve promising outcomes and different degrees of remission.

6. Peripheral Nervous System Rebuilding

Peripheral nervous system (PNS) injury is mainly caused by severe trauma, usually accompanied by bone fracture and vascular damage. Autologous nerve grafting (autologous nerve bridging) is the gold standard for peripheral nerve repair; however, limited donor nerve resources and other issues preclude the search for new therapeutic strategies. Schwann cells, neurotrophic factors, and anti-inflammatory cells work together to promote peripheral nerve regeneration, with this process involving axonal sprouting and fiber myelination.
There are few comparative studies concerning the effects of different MSC types on animal models of peripheral nerve injury. ADSCs are more suitable cell sources for neural regeneration in vitro [67], and a nerve growth factor transcript has been identified in ADSC-secreted nanovesicles that promotes synaptic growth in vitro and repair of sciatic nerve damage in vivo [88]. Sun et al. [89] proposed a new protocol called intermittent induction that alternates complete and incomplete induction media to induce ADSC differentiation into SLCs. Compared with traditional protocols, SLCs obtained by intermittent induction secrete neurotrophic factors and promote axonal growth in vitro and more effectively repair rat sciatic nerve injury in vivo. Notably, SLCs seeded in acellular nerve grafts show better functional recovery as compared with MSCs [90]. BMSCs [75], ADSCs [91], and UCB-MSCs [92] have also been seeded onto a variety of biodegradable scaffolds, with almost all resulting in better recovery relative to controls. MSCs form a neuroblast-like sheath following transplantation at the site of nerve injury and secrete neurotrophic factors that provide physical and chemical barriers for the inner nerve fibers [74]. Compared with polymers, acellular neural matrix hydrogels show better biocompatibility and tissue specificity and support Schwann cell proliferation in vitro and repair rat sciatic nerve defects in vivo [93]. Furthermore, 3D-bioprinting technology has enabled the development of 3D scaffolds with complex structures to address the challenges of nerve tissue regeneration [94].
For nerve regeneration studies, sciatic nerve crush and nerve gap animal models were established [95][96], and the effects of local implantation [97] or intravenous administration [98][99] of neural stem cells or MSCs in peripheral nerve injury models have been investigated, resulting in excellent outcomes relative to controls. ADSCs displayed relevant therapeutic potential not only via their direct release of growth factors but also through the indirect modulation of neurocyte behavior in an animal model of acute axonal injury [98]. Moreover, intravenously infused MSCs ameliorated function recovery post-acute peripheral nerve injury in a sciatic nerve crush model [99].
Although preclinical studies show the feasibility of MSC-based therapy in animal models of peripheral nerve injury, there are few reports of its clinical application [90].

7. Myocardium Restoration

Cardiac disease is characterized by substantial morbidity and mortality, and serious adverse consequences. In addition to congenital heart disease, almost all cardiac diseases involve insufficient blood supply to critical regions, resulting in myocardial damage and necrosis. Although myocardium has limited regenerative capacity, restoration of severe damage to cardiomyocytes due to catastrophic myocardial infarction or other myocardial diseases is inadequate.
A role for MSC in attenuating myocardium damage was first reported in 2002 [100]; purified BMSCs engrafted in the murine myocardium appeared to differentiate into cardiomyocytes. Several subsequent studies evaluated the potential of different MSC sources to differentiate into cardiomyocytes [101][102][103], finding ADSCs as the most suitable. Spraying was found to be a more cost-effective and less invasive method for transferring ADSCs into a pig heart infarction model to promote cardiac function recovery [104]. MSC-specific mechanisms associated with the repair of damaged cardiomyocytes involve three factors: (1) myogenic and angiogenic capacities; (2) the ability to supply massive amounts of angiogenic, anti-apoptotic, and mitogenic factors; and (3) the inhibition of myocardial fibrosis (Figure 2) [105][106]. Butler et al. [107] demonstrated the safety of MSC therapy, and that it improved the left ventricular ejection fraction in patients with non-ischemic cardiomyopathy via its immunomodulatory effects. Co-culture of MSCs with cardiomyocytes promotes resistance to high oxidative stress in heart tissue after myocardial infarction [108]. 5-Azacytidine is an effective factor for inducing MSC differentiation into cardiomyocytes [109]. IGF-1-transfected MSCs protected the myocardium from fibrosis and cardiomyocyte apoptosis and reduced infarct size after myocardial infarction in rats [110]. Interleukin (IL)-7 enhances the fusion of MSCs with cardiomyocytes to improve cardiac function, with this attributed to the ability of IL-7 to promote cell proliferation and support damaged myocardial regeneration [111]. In addition to their ability to differentiate into cardiomyocytes, MSCs promoted angiogenesis by secreting vascular endothelial growth factor (VEGF) in a critical limb ischemia model [112], resulting in cardiac reconstruction.
Figure 2. Schematic mechanisms of mesenchymal stem cells (MSCs) for cardiac regeneration. Angiogenesis, vasculogenesis, and cardiomyocytes differentiation capacities of MSCs make them possible for cardiac repair. Moreover, the paracrine effects of MSCs provide different kinds of growth and anti-inflammatory factors for the immunoregulation after ischemia of heart [106].
MSC-based therapy is a feasible strategy to improve cardiac function, according to preclinical and clinical findings [113]. Furthermore, MSC therapy has been intensively investigated as a treatment for myocardial infarction [114], peripheral ischemic vascular diseases [115], dilated cardiomyopathy [105], and pulmonary hypertension [116].

References

  1. Friedenstein, A.J.; Gorskaja, U.F.; Kulagina, N.N. Fibroblast precursors in normal and irradiated mouse hematopoietic organs. Exp. Hematol. 1976, 4, 267–274.
  2. Ashton, B.A.; Allen, T.D.; Howlett, C.; Eaglesom, C.; Hattori, A.; Owen, M. Formation of bone and cartilage by marrow stromal cells in diffusion chambers in vivo. Clin. Orthop. Relat. Res. 1980, 151, 294–307.
  3. Owen, M.; Friedenstein, A. Stromal stem cells: Marrow-derived osteogenic precursors. Ciba. Found Symp. 1988, 136, 42–60.
  4. Salgado, A.J.; Oliveira, J.M.; Martins, A.; Teixeira, F.G.; Silva, N.A.; Neves, N.M.; Sousa, N.; Reis, R.L. Tissue engineering and regenerative medicine: Past, present, and future. Int. Rev. Neurobiol. 2013, 108, 1–33.
  5. Campagnoli, C.; Roberts, I.A.G.; Kumar, S.; Bennett, P.R.; Bellantuono, I.; Fisk, N.M. Identification of mesenchymal stem/progenitor cells in human first-trimester fetal blood, liver, and bone marrow. Blood 2001, 98, 2396–2402.
  6. Chen, Y.; Shao, J.Z.; Xiang, L.X.; Dong, X.J.; Zhang, G.R. Mesenchymal stem cells: A promising candidate in regenerative medicine. Int. J. Biochem. Cell Biol. 2008, 40, 815–820.
  7. Gao, F.; Chiu, S.M.; Motan, D.A.L.; Zhang, Z.; Chen, L.; Ji, H.L.; Tse, H.F.; Fu, Q.L.; Lian, Q. Mesenchymal stem cells and immunomodulation: Current status and future prospects. Cell Death Dis. 2016, 7, e2062.
  8. Aggarwal, S.; Pittenger, M.F. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood 2005, 105, 1815–1822.
  9. Younger, E.M.; Chapman, M.W. Morbidity at bone graft donor sites. J. Orthop. Trauma. 1989, 3, 192–195.
  10. Lozano-Calderon, S.A.; Swaim, S.O.; Federico, A.; Anderson, M.E.; Gebhardt, M.C. Predictors of soft-tissue complications and deep infection in allograft reconstruction of the proximal tibia. J. Surg. Oncol. 2016, 113, 811–817.
  11. Jiang, Y.H.; Jahagirdar, B.N.; Reinhardt, R.L.; Schwartz, R.E.; Keene, C.D.; Ortiz-Gonzalez, X.R.; Reyes, M.; Lenvik, T.; Lund, T.; Blackstad, M.; et al. Pluripotency of mesenchymal stem cells derived from adult marrow. Nature 2002, 418, 41–49.
  12. Kargozar, S.; Mozafari, M.; Hashemian, S.J.; Milan, P.B.; Hamzehlou, S.; Soleimani, M.; Joghataei, M.T.; Gholipourmalekabadi, M.; Korourian, A.; Mousavizadeh, K.; et al. Osteogenic potential of stem cells-seeded bioactive nanocomposite scaffolds: A comparative study between human mesenchymal stem cells derived from bone, umbilical cord Wharton’s jelly, and adipose tissue. J. Biomed. Mater. Res. B 2018, 106, 61–72.
  13. Yang, J.Z.; Zhang, Y.S.; Yue, K.; Khademhosseini, A. Cell-laden hydrogels for osteochondral and cartilage tissue engineering. Acta Biomater. 2017, 57, 1–25.
  14. Lin, H.; Sohn, J.; Shen, H.; Langhans, M.T.; Tuan, R.S. Bone marrow mesenchymal stem cells: Aging and tissue engineering applications to enhance bone healing. Biomaterials 2018, 203, 96–110.
  15. Bougioukli, S.; Sugiyama, O.; Pannell, W.; Ortega, B.; Tan, M.H.; Tang, A.H.; Yoho, R.; Oakes, D.A.; Lieberman, J.R. Genetherapy for bone repair using human cells: Superior osteogenic potential of bone morphogenetic protein 2-transduced mesenchymal stem cells derived from adipose tissue compared to bone marrow. Hum. Gene Ther. 2018, 29, 507–519.
  16. Burrow, K.L.; Hoyland, J.A.; Richardson, S.M. Human adipose-derived stem cells exhibit enhanced proliferative capacity and retain multipotency longer than donor-matched bone marrow mesenchymal stem cells during expansion in vitro. Stem Cells Int. 2017, 15, 2541275.
  17. Ye, K.Q.; Liu, D.H.; Kuang, H.Z.; Cai, J.Y.; Chen, W.M.; Sun, B.B.; Xia, L.G.; Fang, B.; Morsi, Y.; Mo, X.M. Three-dimensional electrospun nanofibrous scaffolds displaying bone morphogenetic protein-2-derived peptides for the promotion of osteogenic differentiation of stem cells and bone regeneration. J. Colloid Interface Sci. 2019, 534, 625–636.
  18. Aragon, J.; Salerno, S.; De Bartolo, L.; Irusta, S.; Mendoza, G. Polymeric electrospun scaffolds for bone morphogenetic protein 2 delivery in bone tissue engineering. J. Colloid Interface Sci. 2018, 531, 126–137.
  19. Decambron, A.; Devriendt, N.; Larochette, N.; Manassero, M.; Bourguignon, M.; El-Hafci, H.; Petite, H.; Viateau, V.; Logeart-avramoglou, D. Effect of the bone morphogenetic protein-2 doses on the osteogenic potential of human multipotent stromal cells- containing tissue engineered constructs. Tissue Eng. Part A 2019, 25, 642–651.
  20. Chen, F.; Bi, D.; Cheng, C.; Ma, S.; Liu, Y.; Cheng, K. Bone morphogenetic protein 7 enhances the osteogenic differentiation of human dermal-derived CD105+ fibroblast cells through the Smad and MAPK pathways. Int. J. Mol. Med. 2019, 43, 37–46.
  21. Zhu, J.H.; Liao, Y.P.; Li, F.S.; Hu, Y.; Li, Q.; Ma, Y.; Wang, H.; Zhou, Y.; He, B.C.; Su, Y.X. Wnt11 promotes BMP9-induced osteogenic differentiation through BMPs/Smads and p38 MAPK in mesenchymal stem cells. J. Cell Biochem. 2018, 119, 9462–9473.
  22. Bizelli-Silveira, C.; Pullisaar, H.; Abildtrup, L.A.; Andersen, O.Z.; Spin-Neto, R.; Foss, M.; Kraft, D.C.E. Strontium enhances proliferation and osteogenic behavior of periodontal ligament cells in vitro. J. Periodont. Res. 2018, 53, 1020–1028.
  23. Tang, D.; Tare, R.S.; Yang, L.Y.; Williams, D.F.; Ou, K.L.; Oreffo, R.O.C. Biofabrication of bone tissue: Approaches, challenges and translation for bone regeneration. Biomaterials 2016, 83, 363–382.
  24. Westhauser, F.; Senger, A.S.; Reible, B.; Moghaddam, A. In vivo models for the evaluation of the osteogenic potency of bone substitutes seeded with mesenchymal stem cells of human origin: A concise review. Tissue Eng. Part C 2017, 23, 881–888.
  25. Khojasteh, A.; Fahimipour, F.; Jafarian, M.; Sharifi, D.; Jahangir, S.; Khayyatan, F.; Eslaminejad, M.B. Bone engineering in dog mandible: Coculturing mesenchymal stem cells with endothelial progenitor cells in a composite scaffold containing vascular endothelial growth factor. J. Biomed. Mater. Res. B 2017, 105, 1767–1777.
  26. Katagiri, W.; Watanabe, J.; Toyama, N.; Osugi, M.; Sakaguchi, K.; Hibi, H. Clinical study of bone regeneration by conditioned medium from mesenchymal stem cells after maxillary sinus floor elevation. Implant. Dent. 2017, 26, 607–612.
  27. Gjerde, C.; Mustafa, K.; Hellem, S.; Rojewski, M.; Gjengedal, H.; Yassin, M.A.; Feng, X.; Skaale, S.; Berge, T.; Rosen, A.; et al. Cell therapy induced regeneration of severely atrophied mandibular bone in a clinical trial. Stem Cell Res. Ther. 2018, 9, 213.
  28. Liebergall, M.; Schroeder, J.; Mosheiff, R.; Gazit, Z.; Yoram, Z.; Rasooly, L.; Daskal, A.; Khoury, A.; Weil, Y.; Beyth, S. Stem cell-based therapy for prevention of delayed fracture union: A randomized and prospective preliminary study. Mol. Ther. 2013, 21, 1631–1638.
  29. Giannotti, S.; Trombi, L.; Bottai, V.; Ghilardi, M.; D’Alessandro, D.; Danti, S.; Dell’Osso, G.; Guido, G.; Petrini, M. Use of autologous human mesenchymal stromal cell/fibrin clot constructs in upper limb non-unions: Long-term assessment. PloS ONE 2013, 8, e73893.
  30. Honarpardaz, A.; Irani, S.; Pezeshki-Modaress, M.; Zandi, M.; Sadeghi, A. Enhanced chondrogenic differentiation of bone marrow mesenchymal stem cells on gelatin/glycosaminoglycan electrospun nanofibers with different amount of glycosaminoglycan. J. Biomed. Mater. Res. Part A 2019, 107, 38–48.
  31. Fernandes, T.L.; Kimura, H.A.; Pinheiro, C.C.G.; Shimomura, K.; Nakamura, N.; Ferreira, J.R.M.; Gomoll, A.H.; Hernandez, A.J.; Bueno, D.F. Human synovial mesenchymal stem cells good manufacturing practices for articular cartilage regeneration. Tissue Eng. Part C 2018, 24, 709–716.
  32. Park, J.S.; Shim, M.S.; Shim, S.H.; Yang, H.N.; Jeon, S.Y.; Woo, D.G.; Lee, D.R.; Yoon, T.K.; Park, K.H. Chondrogenic potential of stem cells derived from amniotic fluid, adipose tissue, or bone marrow encapsulated in fibrin gels containing TGF-β3. Biomaterials 2011, 32, 8139–8149.
  33. Pievani, A.; Scagliotti, V.; Russo, F.M.; Azario, I.; Rambaldi, B.; Sacchetti, B.; Marzorati, S.; Erba, E.; Giudici, G.; Riminucci, M.; et al. Comparative analysis of multilineage properties of mesenchymal stromal cells derived from fetal sources shows an advantage of mesenchymal stromal cells isolated from cord blood in chondrogenic differentiation potential. Cytotherapy 2014, 16, 893–905.
  34. Zou, J.Y.; Bai, B.; Yao, Y.C. Progress of co-culture systems in cartilage regeneration. Expert. Opin. Biol. Ther. 2018, 18, 1151–1158.
  35. Girao, A.F.; Semitela, A.; Ramalho, G.; Completo, A.; Marques, P. Mimicking nature: Fabrication of 3D anisotropic electrospun polycaprolactone scaffolds for cartilage tissue engineering applications. Composites, Part B 2018, 154, 99–107.
  36. Liu, M.; Zeng, X.; Ma, C.; Yi, H.; Ali, Z.; Mou, X.B.; Li, S.; Deng, Y.; He, N.Y. Injectable hydrogels for cartilage and bone tissue engineering. Bone Res. 2017, 5, 20.
  37. Legendre, F.; Ollitrault, D.; Gomez-Leduc, T.; Bouyoucef, M.; Hervieu, M.; Gruchy, N.; Mallein-Gerin, F.; Leclercq, S.; Demoor, M.; Galera, P. Enhanced chondrogenesis of bone marrow-derived stem cells by using a combinatory cell therapy strategy with BMP-2/TGF-β1, hypoxia, and COL1A1/HtrA1 siRNAs. Sci. Rep. 2017, 7, 3406.
  38. Crecente-Campo, J.; Borrajo, E.; Vidal, A.; Garcia-Fuentes, M. New scaffolds encapsulating TGF-β3/BMP-7 combinations driving strong chondrogenic differentiation. Eur. J. Pharm. Biopharm. 2017, 114, 69–78.
  39. Gugjoo, M.B.; Amarpal; Abdelbaset-Ismail, A.; Aithal, H.P.; Kinjavdekar, P.; Pawde, A.M.; Kumar, G.S.; Sharma, G.T. Mesenchymal stem cells with IGF-1 and TGF-β1 in laminin gel for osteochondral defects in rabbits. Biomed. Pharmacother. 2017, 93, 1165–1174.
  40. Fahy, N.; Alini, M.; Stoddart, M.J. Mechanical stimulation of mesenchymal stem cells: Implications for cartilage tissue engineering. J. Orthop. Res. 2018, 36, 52–63.
  41. Wakitani, S.; Goto, T.; Pineda, S.J.; Young, R.G.; Mansour, J.M.; Caplan, A.I.; Goldberg, V.M. Mesenchymal cell-based repair of large, full-thickness defects of articular cartilage. J. Bone Joint Surg. Am. 1994, 76, 579–592.
  42. de Windt, T.S.; Vonk, L.A.; Slaper-Cortenbach, I.C.; van den Broek, M.P.; Nizak, R.; van Rijen, M.H.; de Weger, R.A.; Dhert, W.J.; Saris, D.B. Allogeneic mesenchymal stem cells stimulate cartilage regeneration and are safe for single-stage cartilage repair in humans upon mixture with recycled autologous chondrons. Stem Cells 2017, 35, 256–264.
  43. Wakitani, S.; Imoto, K.; Yamamoto, T.; Saito, M.; Murata, N.; Yoneda, M. Human autologous culture expanded bone marrow mesenchymal cell transplantation for repair of cartilage defects in osteoarthritic knees. Osteoarthr. Cartilage 2002, 10, 199–206.
  44. Orozco, L.; Munar, A.; Soler, R.; Alberca, M.; Soler, F.; Huguet, M.; Sentis, J.; Sanchez, A.; Garcia-Sancho, J. Treatment of knee osteoarthritis with autologous mesenchymal stem cells: A pilot study. Transplantation 2013, 95, 1535–1541.
  45. Shapiro, S.A.; Kazmerchak, S.E.; Heckman, M.G.; Zubair, A.C.; O’Connor, M.I. A prospective, single-blind, placebo-controlled trial of bone marrow aspirate concentrate for knee osteoarthritis. Am. J. Sports Med. 2017, 45, 82–90.
  46. Sasaki, H.; Rothrauff, B.B.; Alexander, P.G.; Lin, H.; Gottardi, R.; Fu, F.H.; Tuan, R.S. In vitro repair of meniscal radial tear with hydrogels seeded with adipose stem cells and TGF-β3. Am. J. Sports Med. 2018, 46, 2402–2413.
  47. Shimomura, K.; Rothrauff, B.B.; Hart, D.A.; Hamamoto, S.; Kobayashi, M.; Yoshikawa, H.; Tuan, R.S.; Nakamura, N. Enhanced repair of meniscal hoop structure injuries using an aligned electrospun nanofibrous scaffold combined with a mesenchymal stem cell-derived tissue engineered construct. Biomaterials 2018, 192, 346–354.
  48. Liang, Y.; Idrees, E.; Szojka, A.R.A.; Andrews, S.H.J.; Kunze, M.; Mulet-Sierra, A.; Jomha, N.M.; Adesida, A.B. Chondrogenic differentiation of synovial fluid mesenchymal stem cells on human meniscus-derived decellularized matrix requires exogenous growth factors. Acta Biomater. 2018, 80, 131–143.
  49. Toratani, T.; Nakase, J.; Numata, H.; Oshima, T.; Takata, Y.; Nakayama, K.; Tsuchiya, H. Scaffold-free tissue-engineered allogenic adipose-derived stem cells promote meniscus healing. Arthroscopy 2017, 33, 346–354.
  50. Tarafder, S.; Gulko, J.; Sim, K.H.; Yang, J.; Cook, J.L.; Lee, C.H. Engineered healing of avascular meniscus tears by stem cell recruitment. Sci. Rep. 2018, 8, 8150.
  51. Chen, M.X.; Guo, W.M.; Gao, S.; Hao, C.X.; Shen, S.; Zhang, Z.Z.; Wang, Z.H.; Li, X.; Jing, X.G.; Zhang, X.L.; et al. Biomechanical stimulus based strategies for meniscus tissue engineering and regeneration. Tissue Eng. Part B 2018, 24, 392–402.
  52. Chew, E.; Prakash, R.; Khan, W. Mesenchymal stem cells in human meniscal regeneration: A systematic review. Ann. Med. Surg. 2017, 24, 3–7.
  53. Clanton, T.O.; Coupe, K.J. Hamstring strains in athletes: Diagnosis and treatment. J. Am. Acad. Orthop. Surg. 1998, 6, 237–248.
  54. Wang, D.; Jiang, X.H.; Lu, A.Q.; Tu, M.; Huang, W.; Huang, P. BMP14 induces tenogenic differentiation of bone marrow mesenchymal stem cells in vitro. Exp. Ther. Med. 2018, 16, 1165–1174.
  55. Aktas, E.; Chamberlain, C.S.; Saether, E.E.; Duenwald-Kuehl, S.E.; Kondratko-Mittnacht, J.; Stitgen, M.; Lee, J.S.; Clements, A.E.; Murphy, W.L.; Vanderby, R. Immune modulation with primed mesenchymal stem cells delivered via biodegradable scaffold to repair an Achilles tendon segmental defect. J. Orthop. Res. 2017, 35, 269–280.
  56. Park, S.H.; Choi, Y.J.; Moon, S.W.; Lee, B.H.; Shim, J.H.; Cho, D.W.; Wang, J.H. Three-dimensional bio-printed scaffold sleeves with mesenchymal stem cells for enhancement of tendon-to-bone healing in anterior cruciate ligament reconstruction using soft-tissue tendon graft. Arthroscopy 2018, 34, 166–179.
  57. Wang, T.; Thien, C.; Wang, C.; Ni, M.; Gao, J.J.; Wang, A.; Jiang, Q.; Tuan, R.S.; Zheng, Q.J.; Zheng, M.H. 3D uniaxial mechanical stimulation induces tenogenic differentiation of tendon-derived stem cells through a PI3K/AKT signaling pathway. FASEB J. 2018, 32, 4804–4814.
  58. Gulecyuz, M.F.; Macha, K.; Pietschmann, M.F.; Ficklscherer, A.; Sievers, B.; Rossbach, B.P.; Jansson, V.; Muller, P.E. Allogenic myocytes and mesenchymal stem cells partially improve fatty rotator cuff degeneration in a rat model. Stem Cell Rev. Rep. 2018, 14, 847–859.
  59. Bertolo, A.; Mehr, M.; Aebli, N.; Baur, M.; Ferguson, S.J.; Stoyanov, J.V. Influence of different commercial scaffolds on the in vitro differentiation of human mesenchymal stem cells to nucleus pulposus-like cells. Eur. Spine J. 2012, 21 Suppl 6, S826–838.
  60. Zhou, X.; Wang, J.; Huang, X.; Fang, W.; Tao, Y.; Zhao, T.; Liang, C.; Hua, J.; Chen, Q.; Li, F. Injectable decellularized nucleus pulposus-based cell delivery system for differentiation of adipose-derived stem cells and nucleus pulposus regeneration. Acta Biomater. 2018, 81, 115–128.
  61. Hou, Y.; Liang, L.; Shi, G.D.; Shi, J.G.; Sun, J.C.; Xu, G.H.; Guo, Y.F.; Yuan, W. In vivo evaluation of the adenovirus-mediated Sox9 and BMP2 double gene transduction on treatment of intervertebral disc degeneration in rabbit annular puncture model. J. Biomater. Tissue Eng. 2018, 8, 1091–1099.
  62. Varma, D.M.; DiNicolas, M.S.; Nicoll, S.B. Injectable, redox-polymerized carboxymethylcellulose hydrogels promote nucleus pulposus-like extracellular matrix elaboration by human MSCs in a cell density-dependent manner. J. Biomater. Appl. 2018, 33, 576–589.
  63. Kraus, P.; Lufkin, T. Implications for a stem cell regenerative medicine based approach to human intervertebral disk degeneration. Front. Cell Dev. Biol. 2017, 5, 17.
  64. Orozco, L.; Soler, R.; Morera, C.; Alberca, M.; Sanchez, A.; Garcia-Sancho, J. Intervertebral disc repair by autologous mesenchymal bone marrow cells: A pilot study. Transplantation 2011, 92, 822–828.
  65. Pettine, K.A.; Suzuki, R.K.; Sand, T.T.; Murphy, M.B. Autologous bone marrow concentrate intradiscal injection for the treatment of degenerative disc disease with three-year follow-up. Int. Orthop. 2017, 41, 2097–2103.
  66. Kumar, H.; Ha, D.H.; Lee, E.J.; Park, J.H.; Shim, J.H.; Ahn, T.K.; Kim, K.T.; Ropper, A.E.; Sohn, S.; Kim, C.H.; et al. Safety and tolerability of intradiscal implantation of combined autologous adipose-derived mesenchymal stem cells and hyaluronic acid in patients with chronic discogenic low back pain: 1-year follow-up of a phase I study. Stem Cell Res. Ther. 2017, 8, 262.
  67. Chung, C.S.; Fujita, N.; Kawahara, N.; Yui, S.; Nam, E.; Nishimura, R. A comparison of neurosphere differentiation potential of canine bone marrow-derived mesenchymal stem cells and adipose-derived mesenchymal stem cells. J. Vet. Med. Sci. 2013, 75, 879–886.
  68. Bae, K.S.; Park, J.B.; Kim, H.S.; Kim, D.S.; Park, D.J.; Kang, S.J. Neuron-like differentiation of bone marrow-derived mesenchymal stem cells. Yonsei. Med. J. 2011, 52, 401–412.
  69. Kim, M.; Kim, K.H.; Song, S.U.; Yi, T.G.; Yoon, S.H.; Park, S.R.; Choi, B.H. Transplantation of human bone marrow-derived clonal mesenchymal stem cells reduces fibrotic scar formation in a rat spinal cord injury model. J. Tissue Eng. Regen. Med. 2018, 12, E1034–E1045.
  70. Jahan-Abad, A.J.; Negah, S.S.; Ravandi, H.H.; Ghasemi, S.; Borhani-Haghighi, M.; Stummer, W.; Gorji, A.; Ghadiri, M.K. Human neural stem/progenitor cells derived from epileptic human brain in a self-assembling peptide nanoscaffold improve traumatic brain injury in rats. Mol. Neurobiol. 2018, 55, 9122–9138.
  71. Hou, L.L.; Cao, H.; Wang, D.M.; Wei, G.R.; Bai, C.X.; Zhang, Y.; Pei, X.T. Induction of umbilical cord blood mesenchymal stem cells into neuron-like cells in vitro. Int. J. Hematol. 2003, 78, 256–261.
  72. Luo, L.H.; He, Y.; Wang, X.Y.; Key, B.; Lee, B.H.; Li, H.Q.; Ye, Q.S. Potential roles of dental pulp stem cells in neural regeneration and repair. Stem Cells Int. 2018, 2018, 1731289.
  73. Fesharaki, M.; Razavi, S.; Ghasemi-Mobarakeh, L.; Behjati, M.; Yarahmadian, R.; Kazemi, M.; Hejazi, H. Differentiation of human scalp adipose-derived mesenchymal stem cells into mature neural cells on electrospun nanofibrous scaffolds for nerve tissue engineering applications. Cell J. 2018, 20, 168–176.
  74. Ma, Y.H.; Zeng, X.; Qiu, X.C.; Wei, Q.S.; Che, M.T.; Ding, Y.; Liu, Z.; Wu, G.H.; Sun, J.H.; Pang, M.; et al. Perineurium-like sheath derived from long-term surviving mesenchymal stem cells confers nerve protection to the injured spinal cord. Biomaterials 2018, 160, 37–55.
  75. Comar, M.; Delbue, S.; Zanotta, N.; Valencic, E.; Piscianz, E.; Del Savio, R.; Tesser, A.; Tommasini, A.; Ferrante, P. In vivo detection of polyomaviruses JCV and SV40 in mesenchymal stem cells from human umbilical cords. Pediatr. Blood Cancer 2014, 61, 1347–1349.
  76. van Velthoven, C.T.J.; Kavelaars, A.; Heijnen, C.J. Mesenchymal stem cells as a treatment for neonatal ischemic brain damage. Pediatr. Res. 2012, 71, 474–481.
  77. Shi, B.; Ding, J.X.; Liu, Y.; Zhuang, X.M.; Zhuang, X.L.; Chen, X.S.; Fu, C.F. ERK1/2 pathway-mediated differentiation of IGF-1-transfected spinal cord-derived neural stem cells into oligodendrocytes. PloS ONE 2014, 9, e106038.
  78. de Araujo Farias, V.; Carrillo-Galvez, A.B.; Martin, F.; Anderson, P. TGF-β and mesenchymal stromal cells in regenerative medicine, autoimmunity and cancer. Cytokine Growth Factor Rev. 2018, 43, 25–37.
  79. Zhang, Y.L.; Chopp, M.; Zhang, Z.G.; Katakowski, M.; Xin, H.Q.; Qu, C.S.; Ali, M.; Mahmood, A.; Xiong, Y. Systemic administration of cell-free exosomes generated by human bone marrow derived mesenchymal stem cells cultured under 2D and 3D conditions improves functional recovery in rats after traumatic brain injury. Neurochem. Int. 2017, 111, 69–81.
  80. Oliveri, R.S.; Bello, S.; Biering-Sorensen, F. Mesenchymal stem cells improve locomotor recovery in traumatic spinal cord injury: Systematic review with meta-analyses of rat models. Neurobiol. Dis. 2014, 62, 338–353.
  81. Oh, S.K.; Choi, K.H.; Yoo, J.Y.; Kim, D.Y.; Kim, S.J.; Jeon, S.R. A phase III clinical trial showing limited efficacy of autologous mesenchymal stem cell therapy for spinal cord injury. Neurosurgery 2016, 78, 436–447.
  82. Wang, S.; Cheng, H.B.; Dai, G.H.; Wang, X.D.; Hua, R.R.; Liu, X.B.; Wang, P.S.; Chen, G.M.; Yue, W.; An, Y.H. Umbilical cord mesenchymal stem cell transplantation significantly improves neurological function in patients with sequelae of traumatic brain injury. Brain Res. 2013, 1532, 76–84.
  83. Hu, C.X.; Li, L.J. Preconditioning influences mesenchymal stem cell properties in vitro and in vivo. J. Cell Mol. Med. 2018, 22, 1428–1442.
  84. Harris, V.K.; Stark, J.; Vyshkina, T.; Blackshear, L.; Joo, G.; Stefanova, V.; Sara, G.; Sadiq, S.A. Phase I trial of intrathecal mesenchymal stem cell-derived neural progenitors in progressive multiple sclerosis. EBioMedicine 2018, 29, 23–30.
  85. Sykova, E.; Rychmach, P.; Drahoradova, I.; Konradova, S.; Ruzickova, K.; Vorisek, I.; Forostyak, S.; Homola, A.; Bojar, M. Transplantation of mesenchymal stromal cells in patients with amyotrophic lateral sclerosis: Results of phase I/IIa clinical trial. Cell Transplant. 2017, 26, 647–658.
  86. Shichinohe, H.; Kawabori, M.; Iijima, H.; Teramoto, T.; Abumiya, T.; Nakayama, N.; Kazumata, K.; Terasaka, S.; Arato, T.; Houkin, K. Research on advanced intervention using novel bone marrOW stem cell (RAINBOW): A study protocol for a phase I, open-label, uncontrolled, dose-response trial of autologous bone marrow stromal cell transplantation in patients with acute ischemic stroke. BMC Neurol. 2017, 17, 179.
  87. Venkatesh, K.; Sen, D. Mesenchymal stem cells as a source of dopaminergic neurons: A potential cell based therapy for Parkinson’s disease. Curr. Stem Cell Res. T. 2017, 12, 326–347.
  88. Bucan, V.; Vaslaitis, D.; Peck, C.T.; StrauSs, S.; Vogt, P.M.; Radtke, C. Effect of exosomes from rat adipose-derived mesenchymal stem cells on neurite outgrowth and sciatic nerve regeneration after crush injury. Mol. Neurobiol. 2019, 56, 1812–1824.
  89. Sun, X.; Zhu, Y.; Yin, H.Y.; Guo, Z.Y.; Xu, F.; Xiao, B.; Jiang, W.L.; Guo, W.M.; Meng, H.Y.; Lu, S.B.; et al. Differentiation of adipose-derived stem cells into Schwann cell-like cells through intermittent induction: Potential advantage of cellular transient memory function. Stem Cell Res. Ther. 2018, 9, 133.
  90. Fan, L.H.; Yu, Z.F.; Li, J.; Dang, X.Q.; Wang, K.Z. Schwann-like cells seeded in acellular nerve grafts improve nerve regeneration. BMC Musculoskel. Dis. 2014, 15, 165.
  91. Hu, F.H.; Zhang, X.F.; Liu, H.X.; Xu, P.; Doulathunnisa; Teng, G.J.; Xiao, Z.D. Neuronally differentiated adipose-derived stem cells and aligned PHBV nanofiber nerve scaffolds promote sciatic nerve regeneration. Biochem. Biophys. Res. Commun. 2017, 489, 171–178.
  92. Papa, S.; Vismara, I.; Mariani, A.; Barilani, M.; Rimondo, S.; De Paola, M.; Panini, N.; Erba, E.; Mauri, E.; Rossi, F.; et al. Mesenchymal stem cells encapsulated into biomimetic hydrogel scaffold gradually release CCL2 chemokine in situ preserving cytoarchitecture and promoting functional recovery in spinal cord injury. J. Controlled Release 2018, 278, 49–56.
  93. Lin, T.; Liu, S.; Chen, S.H.; Qiu, S.; Rao, Z.L.; Liu, J.H.; Zhu, S.; Yan, L.W.; Mao, H.Q.; Zhu, Q.T.; et al. Hydrogel derived from porcine decellularized nerve tissue as a promising biomaterial for repairing peripheral nerve defects. Acta Biomater. 2018, 73, 326–338.
  94. Lee, S.J.; Esworthy, T.; Stake, S.; Miao, S.; Zuo, Y.Y.; Harris, B.T.; Zhang, L.G. Advances in 3D bioprinting for neural tissue engineering. Adv. Biosyst. 2018, 2, 18.
  95. Decosterd, I.; Woolf, C.J. Spared nerve injury: An animal model of persistent peripheral neuropathic pain. Pain 2000, 87, 149–158.
  96. Angius, D.; Wang, H.; Spinner, R.J.; Gutierrez-Cotto, Y.; Yaszemski, M.J.; Windebank, A.J. A systematic review of animal models used to study nerve regeneration in tissue-engineered scaffolds. Biomaterials 2012, 33, 8034–8039.
  97. O’Rourke, C.; Day, A.G.E.; Murray-Dunning, C.; Thanabalasundaram, L.; Cowan, J.; Stevanato, L.; Grace, N.; Cameron, G.; Drake, R.A.L.; Sinden, J.; et al. An allogeneic ‘off the shelf’ therapeutic strategy for peripheral nerve tissue engineering using clinical grade human neural stem cells. Sci. Rep. 2018, 8, 11.
  98. Thomson, A.A.; Cunha, G.R. Prostatic growth and development are regulated by FGF10. Development 1999, 126, 3693–3701.
  99. Matthes, S.M.; Reimers, K.; Janssen, I.; Liebsch, C.; Kocsis, J.D.; Vogt, P.M.; Radtke, C. Intravenous transplantation of mesenchymal stromal cells to enhance peripheral nerve regeneration. Biomed. Res. Int. 2013, 2013, 573169.
  100. Toma, C.; Pittenger, M.F.; Cahill, K.S.; Byrne, B.J.; Kessler, P.D. Human mesenchymal stem cells differentiate to a cardiomyocyte phenotype in the adult murine heart. Circulation 2002, 105, 93–98.
  101. Lei, H.; Yu, B.; Huang, Z.; Yang, X.; Liu, Z.; Mao, X.; Tian, G.; He, J.; Han, G.; Chen, H.; et al. Comparative analysis of mesenchymal stem cells from adult mouse adipose, muscle, and fetal muscle. Mol. Biol. Rep. 2013, 40, 885–892.
  102. Xu, W.; Zhang, X.; Qian, H.; Zhu, W.; Sun, X.; Hu, J.; Zhou, H.; Chen, Y. Mesenchymal stem cells from adult human bone marrow differentiate into a cardiomyocyte phenotype in vitro. Exp. Biol. Med. 2004, 229, 623–631.
  103. Yang, J.; Song, T.; Wu, P.; Chen, Y.; Fan, X.; Chen, H.; Zhang, J.; Huang, C. Differentiation potential of human mesenchymal stem cells derived from adipose tissue and bone marrow to sinus node-like cells. Mol. Med. Rep. 2012, 5, 108–113.
  104. Mori, D.; Miyagawa, S.; Yajima, S.; Saito, S.; Fukushima, S.; Ueno, T.; Toda, K.; Kawai, K.; Kurata, H.; Nishida, H.; et al. Cell spray transplantation of adipose-derived mesenchymal stem cell recovers ischemic cardiomyopathy in a porcine model. Transplantation 2018, 102, 2012–2024.
  105. Nagaya, N.; Kangawa, K.; Itoh, T.; Iwase, T.; Murakami, S.; Miyahara, Y.; Fujii, T.; Uematsu, M.; Ohgushi, H.; Yamagishi, M.; et al. Transplantation of mesenchymal stem cells improves cardiac function in a rat model of dilated cardiomyopathy. Circulation 2005, 112, 1128–1135.
  106. Gnecchi, M.; Danieli, P.; Cervio, E. Mesenchymal stem cell therapy for heart disease. Vascul. Pharmacol. 2012, 57, 48–55.
  107. Butler, J.; Epstein, S.E.; Greene, S.J.; Quyyumi, A.A.; Sikora, S.; Kim, R.J.; Anderson, A.S.; Wilcox, J.E.; Tankovich, N.I.; Lipinski, M.J.; et al. Intravenous allogeneic mesenchymal stem cells for nonischemic cardiomyopathy safety and efficacy results of a phase II-a randomized trial. Circ. Res. 2017, 120, 332–340.
  108. Choe, G.; Park, J.; Jo, H.; Kim, Y.S.; Ahn, Y.; Lee, J.Y. Studies on the effects of microencapsulated human mesenchymal stem cells in RGD-modified alginate on cardiomyocytes under oxidative stress conditions using in vitro biomimetic co-culture system. Int. J. Biol. Macromol. 2018, 123, 512–520.
  109. Jiang, S.; Zhang, S. Differentiation of cardiomyocytes from amniotic fluid-derived mesenchymal stem cells by combined induction with transforming growth factor β1 and 5-azacytidine. Mol. Med. Rep. 2017, 16, 5887–5893.
  110. Jung, S.; Kim, J.H.; Yim, C.; Lee, M.; Kang, H.J.; Choi, D. Therapeutic effects of a mesenchymal stem cell-based insulin-like growth factor-1/enhanced green fluorescent protein dual gene sorting system in a myocardial infarction rat model. Mol. Med. Rep. 2018, 18, 5563–5571.
  111. Haneef, K.; Ali, A.; Khan, I.; Naeem, N.; Jamall, S.; Salim, A. Role of IL-7 in fusion of rat bone marrow mesenchymal stem cells with cardiomyocytes in vitro and improvement of cardiac function in vivo. Cardiovasc. Ther. 2018, 36, e12479.
  112. Fujita, Y.; Kawamoto, A. Stem cell-based peripheral vascular regeneration. Adv. Drug Deliv. Rev. 2017, 120, 25–40.
  113. Shafei, A.E.; Ali, M.A.; Ghanem, H.G.; Shehata, A.I.; Abdelgawad, A.A.; Handal, H.R.; Talaat, K.A.; Ashaal, A.E.; El-Shal, A.S. Mesenchymal stem cell therapy: A promising cell-based therapy for treatment of myocardial infarction. J. Gene. Med. 2017, 19, e2995.
  114. Tse, H.F.; Kwong, Y.L.; Chan, J.K.; Lo, G.; Ho, C.L.; Lau, C.P. Angiogenesis in ischaemic myocardium by intramyocardial autologous bone marrow mononuclear cell implantation. Lancet 2003, 361, 47–49.
  115. Eng, G.; Lee, B.W.; Parsa, H.; Chin, C.D.; Schneider, J.; Linkov, G.; Sia, S.K.; Vunjak-Novakovic, G. Assembly of complex cell microenvironments using geometrically docked hydrogel shapes. Proc. Natl. Acad. Sci. USA 2013, 110, 4551–4556.
  116. Cheng, G.S.; Wang, X.Y.; Li, Y.X.; He, L. Let-7a-transfected mesenchymal stem cells ameliorate monocrotaline-induced pulmonary hypertension by suppressing pulmonary artery smooth muscle cell growth through STAT3-BMPR2 signaling. Stem Cell Res. Ther. 2017, 8, 11.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 601
Revisions: 2 times (View History)
Update Date: 29 Mar 2022
1000/1000
ScholarVision Creations