Skip to main content
MedComm logoLink to MedComm
. 2023 Jun 17;4(4):e291. doi: 10.1002/mco2.291

Application of stem cells in regeneration medicine

Ye Jin 1,#, Shuangyang Li 1,#, Qixuan Yu 1, Tianli Chen 1,, Da Liu 1,
PMCID: PMC10276889  PMID: 37337579

Abstract

Regeneration is a complex process affected by many elements independent or combined, including inflammation, proliferation, and tissue remodeling. Stem cells is a class of primitive cells with the potentiality of differentiation, regenerate with self‐replication, multidirectional differentiation, and immunomodulatory functions. Stem cells and their cytokines not only inextricably linked to the regeneration of ectodermal and skin tissues, but also can be used for the treatment of a variety of chronic wounds. Stem cells can produce exosomes in a paracrine manner. Stem cell exosomes play an important role in tissue regeneration, repair, and accelerated wound healing, the biological properties of which are similar with stem cells, while stem cell exosomes are safer and more effective. Skin and bone tissues are critical organs in the body, which are essential for sustaining life activities. The weak repairing ability leads a pronounced impact on the quality of life of patients, which could be alleviated by stem cell exosomes treatment. However, there are obstacles that stem cells and stem cells exosomes trough skin for improved bioavailability. This paper summarizes the applications and mechanisms of stem cells and stem cells exosomes for skin and bone healing. We also propose new ways of utilizing stem cells and their exosomes through different nanoformulations, liposomes and nanoliposomes, polymer micelles, microspheres, hydrogels, and scaffold microneedles, to improve their use in tissue healing and regeneration.

Keywords: bone regeneration, exosomes, nanoformulations, skin regeneration, stem cells


Regeneration is a complex process affected by many elements independent or combined, including inflammation, proliferation, and tissue remodeling. Stem cells is a class of primitive cells with the potentiality of differentiation, regenerate with self‐replication, multidirectional differentiation, and immunomodulatory functions. Stem cells and their cytokines not only inextricably linked to the regeneration of ectodermal and skin tissues, but also can be used for the treatment of a variety of chronic wounds. Stem cells can produce exosomes in a paracrine manner. Stem cell exosomes play an important role in tissue regeneration, repair, and accelerated wound healing, the biological properties of which are similar with stem cells, while stem cell exosomes are safer and more effective. Skin and bone tissues are critical organs in the body, which are essential for sustaining life activities. The weak repairing ability leads a pronounced impact on the quality of life of patients, which could be alleviated by stem cell exosomes treatment. However, there are obstacles that stem cells and stem cells exosomes trough skin for improved bioavailability. This paper summarizes the applications and mechanisms of stem cells and stem cells exosomes for skin and bone healing. We also propose new ways of utilizing stem cells and their exosomes through different nanoformulations, liposomes and nanoliposomes, polymer micelles, microspheres, hydrogels, and scaffold microneedles, to improve their use in tissue healing and regeneration.

graphic file with name MCO2-4-e291-g004.jpg

1. INTRODUCTION

Regenerative medicine is an interdisciplinary field that activates endogenous stem cells in the body or implants exogenous stem cells, stem cell‐derived cells, or functional tissues and organs. 1 It repairs, replaces, and enhances damaged, diseased, or defective skin, bone, and organs in the human body to achieve disease treatment. 2 Stem cells are primitive, undifferentiated cells with the capacity of self‐replication, multidirectional differentiation, and homing potential and also retain the properties of their parents. 3 According to the physiological requirements of the body, stem cells can differentiate into various cell types, such as nerve cells, cardiomyocytes, and liver cells, which are necessary for tissue regeneration. 4 , 5 , 6 Stem cells are subdivided into totipotent, multipotent, and unipotent stem cells according to their differentiation potential 7 ; among them, pluripotent stem cells exhibit significant tissue repair capacity. 8 , 9 Stem cells have anti‐inflammatory properties, promote epithelial cell proliferation, and inhibit wound scarring. 10 , 11 In addition, stem cells and their exosomes have significant effects on bone tissue and nervous system regeneration. 12 , 13 Therefore, researchers have envisioned the application of stem cells in regenerative medicine to promote skin and bone tissue regeneration. However, due to the difficulties of stem cell transportation and preservation, as well as their tumorigenicity and the difficulty of exosome extraction, nanoformulations of stem cells have been studied and have gradually become a hot research topic for elucidating new ways to repair damaged skin and bone.

As the largest organ in the body, the skin plays a protective role against external damage. 14 Owing to its external exposure and weak self‐healing ability, skin regeneration faces great challenges. 15 , 16 Skin regeneration is a complex and dynamic process 17 , 18 and generally consists of three phases: inflammatory, proliferative, and remodeling. 19 , 20 , 21 The whole process is relatively long and can have a negative psychological impact on the patient. 22 Large, chronic, and hard‐to‐heal skin wounds caused by severe trauma or radiation burns 23 cause physiological dysfunction of the body that is prone to further infection, 24 seriously affecting the quality of life of patients. 25 , 26 Moreover, the healing of bone injury is a major concern in current medical research. In the United States alone, 100,000 fractures result in nonhealing bone injuries annually. 27 The bone plays an important role in supporting movement and protecting organs and is one of the essential organs for maintaining life activities. 28 , 29 , 30 , 31 Although the bone is a highly vascularized organ with a certain ability of regeneration, the external damage beyond its self‐healing range results in nonhealing and scarring of the bone tissue. 32 In such cases, recovery is difficult through autologous bone tissue regeneration alone, and the healing time is increased; therefore, patients experience pain. 33 Therefore, a new treatment method is needed to promote skin and bone tissue regeneration. 34

The first controlled‐release formulation was introduced in 1950. Significant advances in nanodrug delivery technology 35 and the continuous development of nanotechnology 36 have led to its application in the field of drug delivery systems. 37 Nanoformulations can selectively target drug transport to fingertip sites, thus reversing the traditional drug delivery process and enabling higher biocompatibility, 38 safety, eco‐friendliness, specificity, and reduced toxicity while maintaining therapeutic efficacy. 39 In addition, nanoformulations can cross the cellular barrier and activate the transport mechanism. 40 Therefore, drug activity can be maintained to the maximum extent, 41 ensuring fast and stable drug action. When used in skin regeneration, combination with biologically active molecules prevents the drug from being degraded by proteases in the wound, thus improving drug stability. 42 The direct use of easily degradable and metabolically fast stem cells and their exosomes and the use of nanoformulations in skin and bone tissue healing is of great importance for improving the use of stem cells and their exosomes in regenerative medicine. Several studies have shown that nanoformulations such as hydrogels and liposomes can significantly improve the speed and time of bone healing with the use of drugs. 43 , 44

In this paper, we attempt to contribute to the field of regenerative medicine by reviewing the main mechanisms of stem cells and their exosomes for tissue regeneration and the progress of novel nanoformulations loaded with stem cells and exosomes in the field of tissue regeneration. 4 We also describe the modification effect of genetic engineering in stem cells and their exosomes to in the regeneration of skin and bone tissue.

2. STEM CELLS IN REGENERATIVE MEDICINE

In continuous research, the position of stem cells in the field of regenerative medicine have gradually become a hot research topic. 45 Stem cells are widely used in regenerative medicine research because of their strong self‐renewal ability and ease of extraction. 46 Some stem cell treatments have already been in clinical trials, especially for skin and bone regeneration. 47 , 48 , 49

2.1. Introduction to stem cells

Since the 20th century, stem cell and regenerative medicine technology have been one of the hot frontiers in the international biomedical field, which plays an irreplaceable role in safeguarding human life and health in improving the quality of human survival and extending human life expectancy. 50 Stem cells are undifferentiated cells characterized by a high capacity for proliferation and self‐renewal as well as clonality usually derived from a single cell and differentiated into different types of cells and tissues, these properties may vary between stem cells. 51 For example, embryonic stem cells (ESCs) obtained from blastocysts and adult humans have different properties, with ESCs from blastocysts having higher tissue specificity. According to the stage of stem cell development, ESCs and adult stem cells (ASCs) are classified. 52 ESCs can self‐regenerate and differentiate into all tissues in vivo. Numerous animal studies have shown that ESCs promote neural regeneration in neuron‐deficient and chronically denervated rat models. 53 However, the application of ESCs is currently limited due to ethical issues. In addition to this, adult stem cells include cells present in tissues or organs that have the potential for further differentiation, the ability to self‐renew, and the ability to differentiate into the major types of specialized cells of the tissue from which they originate. ASCs mainly include mesenchymal stem cells (MSCs), myogenic stem cells (MDSCs), neural stem cells (NSCs), and urogenic stem cells (USCs). 54

MSCs are one kind of adult stem cells that originate from the early mesoderm and self‐renewing mesodermal cells with multidirectional differentiation potential, which can be isolated from adult bone marrow, dental pulp, adipose tissue, umbilical cord, and other tissues. 55 Among the stem cell family, MSCs are the most widely studied and used stem cells in the field of regenerative medicine. According to different sources, they can be divided into: adipose‐derived MSCs, umbilical cord MSCs, bone marrow MSCs, and so on. 56 There are relevant research tables showing that it has good application prospects in the field of regenerative medicine, and some MSCs have already entered clinical application. 57 MSCs have many advantages such as easy isolation, high in vitro expansion, low immunogenicity, and targeted differentiation into neural tissue cells. 57 MDSCs have long‐lasting self‐renewal and multigerm differentiation ability and can differentiate not only into mesodermal myoblasts, osteoblasts, chondrocytes, adipocytes, endothelial cells, hematopoietic cells, but also into ectodermal neuronal cells. 58 With immunity, donor cells can still be detected weeks after the acute inflammatory response and the damaged nerve can still be functionally regenerated after transplantation of human MDSCs. 59 NSCs transplantation provides a new therapeutic idea for neurological injury diseases and promotes the development of neural regeneration. NSCs not only have the function of cell replacement, but also play the function of immunomodulation, and regulate the activation of complement. 60 Dental stem cells (DSCs) are a population of embryonic neural crest‐derived adult stem cells that are homologous to neural tissue. 61 DSC has stronger proliferation and multidirectional differentiation ability than MSC from other tissues and is one of the ideal stem cell sources for tissue regeneration because it is easy to obtain with features of less invasive and less immune rejection. 62 Urine‐derived stem cells are new stem cells with good expansion capacity, multilineage differentiation potential, and paracrine function. 63 Increasing interests in this aspect shows that it can be isolated from urine samples with the advantages of easy collection, noninvasive sampling, and few ethical issues. 64

Stem cell therapy represents an important trend in the development of medicine for regeneration. The role of stem cell therapy in repairing, restoring or reconstructing human tissue and organ damages is becoming more and more prominent. The application of stem cells and their derivatives has solved many medical problems in recent years. Skin and bone injuries often lead to severe disability, and exploration of promising therapeutic strategies is of great importance. As the largest organ in the body, the skin is vulnerable to injury and weak in self‐repair. Furthermore, the bones are vulnerable to injury causing greater damage, relying solely on self‐repair cycles that leading to more pain on patients. These reasons making skin and bone a current research hotspot in regenerative medicine. 47 , 48 , 49

2.2. Stem cells promote skin tissue regeneration

MSCs were first observed in the bone marrow in 1867 by Kornheim, who found that these cells may be the source of fibroblasts involved in wound repair. 65 Studies have confirmed that stem cells play an important role in the treatment of various skin injuries 66 and numerous scholars have studied their regenerative repair mechanism. 67 , 68 Additionally, stem cells can effectively suppress the inflammatory response and assist in wound repair for skin regeneration through their immunomodulatory effects (Table 1). In this article, we discuss the use of stem cells in several stages of wound healing.

TABLE 1.

Stem cells applied to skin and bone regeneration and their mechanism advantages.

Type Mechanism Dominance Model References
Adipose‐derived stem cells (ADSCs)

Participation in immune regulation

Through paracrine effect

Inhibit scar formation

Rich source

Small damage

Large quantity

Strong proliferative capacity

Low immunogenicity

Cellular models

Rheumatoid arthritis

Mouse model

72 , 88
Umbilical cord‐matrix stem cells (UCMCs)

Control inflammatory response

Promoting granulation angiogenesis and cell proliferation

Inhibit scar formation

Donor risk‐free

Rapid and simple

Painless

Noninvasive sampling

Low potential culture cost

Diabetic mice 75
Bone marrow mesenchymal stem cells (BM‐MSCs)

homing features

Multidirectional differentiation

Immune regulation and immunosuppression

Paracrine effect

Increased anti‐inflammatory expression

Easy to obtain

simple culture technology

Low immunogenicity

Critical limb ischemia rats model

Deep partial‐thickness burns rats model

Collagen‐induced arthritis murine model

diabetic mice

Cranial defect rat model

Bone damage in rats

75 , 76 , 85 , 86 , 89 , 90

The inflammatory phase plays a crucial role in the wound healing process by inducing the recruitment of immune cells to prevent continued pathogenic damage to the organism. 69 , 70 During the hemostatic phase, stem cells migrate to the local wound site and induce vasoconstriction and platelet agglutination to promote blood clotting. 71 To produce hemostatic and anti‐inflammatory effects, a closed wound is temporarily formed on the wound surface. 72 A few hours after wound formation, the inflammatory phase begins, which is characterized by local congestion and plasma exudation. Some studies have used flow cytometry to examine cell proliferation and demonstrated that MSCs have potent immunomodulatory effects and possess antimicrobial properties by regulating the functional properties of T cells, B cells, and dendritic cells. 73 In the early stages of inflammation, the level of wound inflammatory factors increases, including the anti‐inflammatory factors interleukin (IL)‐4, IL‐10, IL‐13, and others. In an experiment on rats, MSCs increased the level of anti‐inflammatory factors, whereas that of proinflammatory factors and tumor necrosis factor (TNF)‐α, gamma‐interferon, IL‐1, IL‐6, IL‐8, intercellular adhesion molecule‐1, and others was decreased. 74

Macrophages play a major immunomodulatory role after 3 days of wound formation and are the key regulators of the local inflammatory microenvironment. After injecting the wound surface of diabetic mice with MSCs, it was found that macrophages were recruited to the wound in large numbers, which in turn induced macrophages to acquire the M2 anti‐inflammatory phenotype. 75 This caused the wound to produce an immune response that inhibited bacterial and other undesirable proliferation and promoted tissue regeneration. Another reported further studies that by experimenting with mouse bone marrow MSCs. Assessment of the immunosuppressive effects of different immune cells. MCSs promote macrophage polarization while inhibiting B‐cell differentiation by regulating IL‐1 receptor antagonists. 76 The anti‐inflammatory effects of MSCs have been well illustrated. Furthermore, MSCs induce the production of IL‐10 by macrophages at inflammatory sites. IL‐10 is a cytokine with anti‐inflammatory properties and induces regeneration of traumatized tissue, which greatly contributes to wound healing and lays the foundation for wound repair.

During the entire repair process, recruited stem cells produce paracrine factors. These paracrine factors exhibit antiapoptotic, proangiogenic, and accelerated cell proliferation effects, which are necessary to promote tissue regeneration. 77 , 78 Exogenous MSCs implanted in the injured areas can produce cytokines, such as fibroblast growth factor (FGF)‐2, hepatocyte growth factor (HGF), vascular endothelial growth factor (VEGF), and transforming growth factor (TGF)‐β through paracrine effects. 79 These cytokines promote angiogenesis as well as fibroblast migration and proliferation. 80 Moreover, they accelerate the deposition of collagen, 81 regulate the inflammatory response in damaged skin tissues, and promote skin tissue regeneration. 82

Reportedly, treatment using paracrine factors (TGF‐β, FGF‐2, angiopoietin‐2, and VEGF‐1) secreted by transplanted bone marrow MSCs after myocardial infarction modeling in rats manipulated the microenvironment and reduced inflammation. 83 From this experiment, it can be concluded that the cytokines produced by stem cells at the infarct site can effectively promote angiogenesis and accelerate cell migration. In addition, a study conducted on mouse spinal cord MSCs by coculturing dermal fibroblasts with insert‐grown bone marrow MSCs and cell scratching experiments showed that the paracrine effect of bone marrow‐derived MSCs could accelerate the proliferation and migration of dermal fibroblasts, apart from the chemotaxis assay. 84 The migration of endogenous MSCs and endothelial progenitor cells has also been considered by researchers as an important mechanism for repairing damaged skin. Moreover, a study reported that MSCs of bone marrow and adipose origin could be isolated from a rat model of severe limb ischemia. 85 In vitro and in vivo experiments revealed that bone marrow‐derived MSCs can promote endothelial cell migration, muscle reorganization, limb function improvement, and neovascularization. In addition, a previous study on rats has reported treatment of deep second‐degree burns with bone marrow MSCs through paracrine effects via the stromal cell‐derived factor (SDF)‐1 alpha/CXCR4 pathway. 86 This approach induces the accumulation of endogenous MSCs and endothelial progenitor cells at the wound site, which can accelerate skin healing. Many of the therapeutic capabilities of stem cells rely on their paracrine actions. Various growth factors regulate the differentiation of fibroblasts and endothelial progenitor cells and promote their transfer to the wound. This mechanism promotes the formation of new blood vessels and granulation tissue, laying the foundation for skin regeneration. 36 Stem cell paracrine factors acting on damaged skin tissue are more effective than direct stem cell differentiation 87 and the healing is faster. 87 The potential clinical applications of cytokines derived from stem cells and the experimental models mentioned in the literature are listed in Table 2.

TABLE 2.

Potential clinical application fields of stem cell active factors.

Stem cell active factor Mechanism Potential clinical application Target organ Model References
PDGF (platelet‐derived growth factor) Anti‐inflammatory immune regulation and angiogenesis Burns skin repair Vascular regeneration Burn‐rats model 36

VEGF (vascular endothelial growth factor)

SDF‐1

(stromal cell‐derived factor‐1)

Promote Mature ECs Migration

Promoting angiogenesis and artery formation

Treatment of acute myocardial infarction, chronic ischemic heart disease, and congestive heart failure

Myocardial regeneration

Vascular regeneration

Deep partial‐thickness burns rats model 80
VEGF‐1 (vascular endothelial growth factor‐1)

Inducing angiogenesis in ischemic tissue

Angiogenesis induced by proliferation of endothelial cells and vascular smooth muscle cells

Stimulate endothelial cells to sprout from existing blood vessels

Treatment of myocardial infarction

Improve myocardial function

Increase angiogenesis in infarcted area

Tumor microenvironment

Embolization site

Animal and preliminary human 83
Ang (angiogenin)

Improving capillary density of ischemic tissue

Promote the formation of new blood vessels

Ischemic disease of hind limbs Ischemic tissue Critical limb ischemia rats model 85
SDF1‐α (stromal cell‐derived factor 1 alpha)

Enhance angiogenesis

Reduce apoptosis

Burns

Skin repair

Vascular regeneration Deep partial‐thickness burns rats model 86

2.3. Stem cells promote bone tissue regeneration

Bone is a mineralized tissue that provides structural support to the vertebrate body. 91 It protects internal organs, attaches to muscles to allow the body to move, and plays an important role in regulating serum calcium and phosphorus levels. 92 The response of bone tissue to trauma is a continuous and complex process that includes an inflammatory response, activation of repair mechanisms, and remodeling of various tissues. 93 If a bone defect is followed by delayed healing or nonhealing that develops into a larger defect, traditional restorative means are no longer sufficient to restore such defects. 94 , 95 Regenerating damaged bone tissue to its predisease state has been a major challenge for clinicians and researchers worldwide. 96 Researchers aim to address this problem with new treatments. 97 Recently, stem cells have been found to be a good medium and option for achieving bone tissue regeneration. 98 The use of bone marrow MSCs in bone tissue engineering has entered the preclinical phase, and the U.S. Food and Drug Administration has approved an in vitro cell manufacturing procedure for obtaining biologically active bone marrow MSCs from high‐quality human bone marrow. 92 Stem cells can migrate to the site of injury and have the ability to differentiate into local components of the injury site and help regenerate the tissue. 99 , 100 , 101 , 102 For example, Kaku et al. 89 isolated MSCs from the bone marrow of rat femurs and transplanted them into a rat model with cranial defects. These results suggest that MSC transplantation may be a new option for cranioplasty. MSCs can not only repair bone tissue but also form suture‐like gaps and promote cranial bone growth.

A direct reflection of the initial stage of bone injury is the formation of hematoma and the onset of an inflammatory response; hematoma can effectively prevent excess bleeding. 103 The fracture hematoma is initially infiltrated by immune cells, mainly neutrophils and macrophages. 104 When the bone tissue is damaged, blood vessels rupture, platelets are activated, and multiple coagulation factors, cytokines, and chemokines are released, triggering a hemostatic effect. Multiple cell types are recruited to migrate to the site of injury, contributing to the initial stage of bone tissue repair. 105 Inflammation is a protective response of tissues against harmful stimuli and leads to the initiation of the healing process. 106 , 107 Transplantation of stem cells into bone injury sites revealed the systemic anti‐inflammatory effects of the cytokines they release. 108 The acute inflammatory phase peaks at 24−48 h after injury and subsides after one week. On the first day of a bone injury, neutrophils arrive at the injury site and release signaling molecules that recruit macrophages to the injury site. 109 Granero‐Molto et al. 108 reported the presence of bone marrow MSCs in the region of new bone formation in a mouse fracture model. This indicated the promoting effect of stem cells on bone tissue regeneration. Macrophages not only engulf necrotic cells and tissue debris at the fracture site, but also initiate the recruitment of MSCs and vascular progenitor cells. 110 , 111 During acute inflammation, macrophages amplify the inflammatory response by phagocytosing invading microbes and recruiting additional immune cells to restore tissue homeostasis. 112 A study demonstrated that MSCs can mediate elevated M1 macrophage marker expression and promote M2 macrophage polarization, tissue vascularization, and bone volume increase in a rat femoral defect model. 113 This study illustrated the anti‐inflammatory and osteogenic effects of stem cells. Furthermore, it was found that BMSCs reversed the pure Lap‐induced polarization of mouse‐derived macrophages RAW 264.7 cells from M1 to M2 and promoted osteogenesis. 114 These results suggested that BMSCs improved Lap‐induced inflammation and enhanced bone formation. BMSCs were isolated from rats and cocultured with macrophages using hydrolyzed fish collagen, which inhibited the proliferation of PBMCs and significantly increased the expression levels of anti‐inflammatory mediators, including IL‐6, TGF‐β1, and PGE2. 115 BMSCs exhibit immunosuppressive capacity through crosstalk with immune cells, significantly limiting the inflammatory response after bone injury. 116 Inflammatory cytokines play important role in regulating bone tissue destruction and promoting bone tissue regeneration. 90 Bone healing after an injury is a complex biological and biomechanical process, 117 and final healing is highly dependent on the initial inflammatory phase, which is influenced by local and systemic responses to the injury stimulus. In addition, immune cells and mesenchymal stromal cells are involved in critical intercellular communication and crosstalk to regulate bone healing. Therefore, understanding the mechanisms of stem cells in bone healing is important for practical application.

It has been found that during the repair process, recruited stem cells secrete various chemokines, cytokines, and growth factors, which are collectively known as paracrine factors and are necessary to promote tissue repair and regeneration or tissue differentiation. 65 , 77 , 87 Cytokines produced by stem cells through their paracrine action, including IL‐1, IL‐6, platelet‐derived growth factor (PDGF), VEGF, and bone morphogenetic protein (BMP), play an essential role in bone tissue regeneration. 118 These cytokines initiate the process of bone tissue formation and generate new blood vessels.

A study conducted by transplanting stem cells to an injured site revealed that the stem cells significantly reduced IL‐6 levels on days 1 and 3 after the fracture, and reduced TNF‐α and IL‐1β levels on day 3. 119 This process inhibits tissue damage and prevents the development of fibrosis, thereby promoting rapid bone tissue regeneration. In our previous study, we reported that the cytokines secreted by BMSCs treated with sinusoidal electromagnetic fields transplanted into a rat cranial defect model could better promote neoangiogenesis and bone immunomodulation, which play important roles in bone regeneration. 120 The osteogenic potential of BMSCs and the facilitative role of the paracrine function of BMSCs in promoting bone regeneration were illustrated. Pearson et al. 121 implanted BMP‐2 in a rat femoral defect model, which significantly and strongly induced neovascularization, and found BMP‐2‐induced angiogenesis through paracrine signals secreted by bone progenitor cells. The presence of cytokines such as insulin‐like growth factor‐1 (IGF‐1), VEGF, TGF‐β1, and HGF in MSC‐CM significantly increased the migration and expression of genes associated with osteogenesis, and bone regeneration was observed in the rat cranial bone. 122 In addition to traditional methods, some emerging cytokine techniques are gradually being developed. For example, Yu et al. 123 evaluated the bone regeneration effect of collagen membranes chemically coupled to SDF‐1α in a rat model and found the chemical coupling to significantly promote the formation of new bone and microvasculature compared with the physical adsorption of SDF‐1α. The study also provided a cell‐free method to shorten bone healing time and improve the success rate of guided bone regeneration.

2.4. Other applications of stem cells in regenerative medicine

Apart from thoughts mentioned above, stem cells can promote regeneration of nerve and hair follicle tissues in addition to skin and bone tissues. Central nervous system diseases, such as stroke, traumatic brain injury, Alzheimer's disease (AD), and Parkinson's disease, are usually accompanied by neuroinflammation and nerve regeneration. 124 Nerve injury is one of the most common types of traumatic injury, commonly occurring in accidents and medically induced injuries. 125 However, traditional treatments are limited, which helps to find an alternative therapy is especially critical, and stem cell therapy is an excellent medium. Studies have shown that transplantation of stem cells are highly effective, both in terms of the number of axons and the restoration of function, showing the safety of this method. 126 In a short period of injury, the myelin sheath of the distal nerve dissociates, macrophages infiltrate and phagocytic debris produce an inflammatory response. Yang et al. 127 found that transplantation of undifferentiated tooth‐derived stem cells (DSCs) could reduce the inflammatory response and promote neuronal regeneration by inhibiting the expression of IL‐1 and member A of the Ras homologue gene family. Moreover, it has been found that transplanted NSCs reduce M1‐type proinflammatory macrophage infiltration, promote M1‐type to M2‐type polarization, increase the release of anti‐inflammatory factors (e.g., IL‐10), and exert neuroprotective effects. 128 After stem cell transplantation, the stem cells are still able to produce and secrete various immune factors and neurotrophic factors to improve neurological function, even if they do not differentiate directly into nerve cells or mature nerve cells differentiated after limited transplantation. 129 Nerve regeneration requires the growth of nerve axons and secreted expression of neurotrophic factors, such as FGF‐2, GDNF, and BDNF. A significant increase in the number of synapses was found after transplantation of stem cells to the injury site. 130 Studies have shown that stem cells are effective in treating neurological disorders, significantly promoting changes in neurological function. 131 Vojnits et al. 132 suggests transplantation of injury‐induced MDSCs or their cell extracts into Duchenne muscular dystrophy mice (i.e., mdx mice) significantly promotes morphological and functional recovery of already injured neuromuscular junctions. In addition, umbilical cord MSCs (UC‐MSCs) have multidirectional differentiation possibilities and improve cognitive dysfunction caused by AD after transplantation. 133 Stem cells for neural regeneration have a wide range of applications, but there are still many challenges for their clinical application, such as the lack of gold standard for cell isolation and culture, and preservation methods, genetic material variation and ethical issues. However, it is believed that these problems will be solved as the research progresses.

Stem cells are also a research hotspot in the field of hair follicle regeneration. Hair follicle is an organ with self‐renewal ability and under normal conditions. The hair growth cycle goes through three phases—anagen phase, apoptosis phase, and resting phase to maintain the cycle for life. 134 However, due to the weak physical resistance of hair follicles and their susceptibility to genetic or environmental factors, leading to irreversible damage to hair follicles. Therefore, hair follicle regeneration has become an extremely important research area in the field of regenerative medicine. The role of stem cells in promoting hair follicle regeneration has become a hot issue. 135 Under normal physiological conditions, stem cells in each region maintain their own tissue renewal. When trauma occurs, stem cells located in the augmentation site can rapidly divide and proliferate to participate in the repair of the hair follicle. 136 ASCs are isolated from adipose tissue and induced to differentiate in dermal papilla formation medium for 30 days by visual and histological observation. The results of growth factor secretion were evaluated by immunohistochemistry and immunoblotting, showing that ASCs dermal papilla‐like tissues (DPLTs) have characteristics of dermal papillae and have a positive effect on hair regeneration by secreting growth factors. 137 Stem cells can produce bioactive factors such as VEGF, IGF, and PDGF through paracrine action. When stem cells are applied to damaged hair follicles, these substances can regulate the hair growth cycle and promote hair growth. 138 Bu et al. 139 conducted a study in which cord blood MSCs were isolated and cultured in vitro and induced to differentiate into CK15 hair follicle cells. High speed induction of high purity differentiation into hair follicle cells was achieved by adding hair follicle extracellular matrix (ECM) to the culture medium. This study provides a new idea to promote the regeneration of hair follicle tissue and has a wide range of application prospects. And Fukuoka et al. 140 reported that ADSCs were isolated from the adipose tissue of healthy women, and the supernatant of ADSCs was collected and made into a lyophilized powder to act on the damaged hair follicles of the patients, and after 1−3 months of treatment, the number of hair follicles in the patients increased significantly. A previous study reported that mouse‐derived pluripotent stem cells were effective in promoting hair follicle development. 141 Stem cells have a broad application prospect in hair follicle regeneration since the development of regenerative medicine.

2.5. Clinical applications of stem cells

Regenerative medicine refers to the use of biological and engineering methods to promote repair and regeneration of damaged tissues, which restores the normal tissue characteristics and functions of an organism. The application of stem cells in regenerative medicine has gained widespread attention over the last 50 years. Because of their strong self‐renewal and dividing abilities, stem cells are the most widely used cells in the field of tissue regeneration therapy and play an important role in tissue healing and regenerative medicine. 142 According to our research, bone marrow MSCs, umbilical cord MSCs, and adipose MSCs have shown good ability to promote skin tissue regeneration. 88 Hassan, a 7‐year‐old Syrian “butterfly” boy, underwent treatment using his own skin cells to replace almost all his skin 143 and has since gained new life (Table 3). Stem cells have low immunogenicity and can be allografted or xenografted for clinical treatment. Exogenous stem cells are implanted to promote wound repair for skin regeneration.

TABLE 3.

Clinical applications of stem cells in tissue regeneration.

Type/intervention/treatment Diseases/conditions Phase Trial number References
Bone marrow stem cells (BMC) Diabetic foot Phase 2 NCT01065337 47
Adipose‐derived stem cells (ADSCs)

Diabetic foot

Venous ulcer

Pressure ulcer

Diabetic foot ulcer

Phase 2

NCT02092870

NCT03754465

48 , 49
Autologous bone marrow stem cells (BMC)

Type IV pressure Ulcers

Chronic wounds

Spinal cord injury

Phase 2 NCT01572376 146
Mesenchymal stem cells (MSCs)

Diabetic foot

Lower limb ischemia

Phase 1 NCT02304588 147
Umbilical cord mesenchymal stem cells (UC‐MSCs) Difficult to healing of skin ulcers Phase 1 NCT02685722 58
Stem cell

Chronic wounds

Pressure sores

Hematopoietic stem cells

Wound healing

Phase 1 NCT00535548 148
Injection the mesenchymal stem cell in nonunion site Nonunion fracture Phase 2 NCT01788059 149
Biological: Application of autologous mesenchymal stem cells Mandibular fractures Phase 3 NCT02755922 150

Biological: Mesenchymal stem cells

Other: culture medium without MSC

Nonunion fracture Phase 2 NCT01429012 151
Biological: MSC

Nonunion fracture

Metaphyseal fibrous defect

Early Phase 1 NCT02307435 152

Bone is a highly vascularized organ, and the skeletal system responds to external stimuli in numerous ways. 144 Repairing bone defects has always been an important clinical challenge, and innovations and improvements in bone repair are changing daily. Compared with traditional means of tissue repair, regenerative cellular and molecular techniques are improving, and the repair effect of stem cells on tissues is demonstrated by the ability to induce and stimulate differentiation into parenchymal cells in vitro to replace damaged areas. 4 , 145 There have been several clinical trials on stem cells for bone regeneration. Thus, the promotion of stem cell paracrine action on bone tissue regeneration is illustrated, and it is noteworthy that the paracrine function of stem cells has recently received more attention than the direct differentiation of stem cells.

3. STEM CELL EXOSOMES IN REGENERATIVE MEDICINE

In 1983, Pan et al. 153 studied the maturation process of transferrin receptors in sheep reticulocytes in vitro and for the first time discovered that cells could release tiny extracellular vesicles (EVs), which were named exosomes in 1987. 154 Exosomes are a class of membranous vesicles that contain biologically active molecules such as lipids, nucleic acids, and proteins. They are secreted extracellularly through a specialized mechanism known as endo‐and exocytotic vesicles, with a diameter of approximately 30−100 nm. 155 They are the smallest known EVs and are secreted by almost all living cells. 156 , 157 Exosomes can be separated from various body fluids, such as urine and blood. if this communicates your intended meaning. 97 , 158 Exosomes are present in the culture medium of most cells. 159

Stem cells produce exosomes in a paracrine manner. Stem cell exosomes are safer and more effective than stem cells with similar biological properties. Additionally, cell transplantation is also not required in this method. Stem cell exosomes represent a new way to achieve tissue regeneration, repair, and accelerate wound healing. Studies have shown that MSC‐derived exosomes (MSC‐Exos) can function well in different environments. MSC‐Exos induced repair in mouse models of wound healing and myocardial infarction. 160 , 161 , 162 EVs of MSCs are rich in MSC‐derived bioactive molecules and regulate the phenotype, function, and homing of immune cells. Application of MSC‐EVs has a significant inhibitory effect on the inflammatory response of an organism, accelerating the survival and regeneration of damaged parenchymal cells. 163 Human umbilical cord MSCs‐derived exosomes are effective in preventing cardiac dysfunction caused by aging. UMSC‐derived exosomes inhibit the NF‐κB/TNF‐α signaling pathway by releasing novel lncRNAs, 164 thus preventing aging‐induced cardiac dysfunction. MicroRNA‐342‐3p has a regulatory role in MSC‐Exos against breast cancer, confirming that microRNA‐342‐3p inhibits metastasis and chemoresistance of cancer cells by targeting ID. 165 Exosomes of stem cells can transfer proteins and DNA molecules between cells via paracrine or endocrine signals; therefore, the whole biological process is changed. 166 MSC exocytosis delivers healing proteins and RNA to recipient cells, 167 thus adding to the vitality of the therapy. The delivery of functional substances from exosomes to receptor cells helps to heal injured or diseased tissues and organs. MSC‐Exos can be manipulated to establish a new cell‐free therapeutic approach and can be used to heal skin wounds. 168

3.1. Stem cell exosomes promote skin regeneration

Similar to stem cells, stem cell exosomes play different roles at different stages of wound healing. Exosomes can promote cell proliferation and neovascularization by regulating the inflammatory response 169 (Table 4). In addition, they can inhibit scarring and promote other mechanisms of effective skin tissue regeneration. 170 , 171 , 172

TABLE 4.

Mesenchymal stem cell exosome‐acting signaling pathways promote skin tissue regeneration.

Type Signaling pathways Dominance References
Bone marrow mesenchymal stem cells exosomes (BMSC‐Exos) Activation of the PTEN/AKT signaling pathway

Inhibits proinflammatory phenotype M1‐type polarization

Promote macrophage anti‐inflammatory phenotype M2‐type polarization

173
Molecular mechanism of human umbilical cord MSC‐derived exosomes (hUCMSC‐Exos) Inhibition of TLR4 signaling pathway Increase the expression level of anti‐inflammatory factors 174
Adipose‐derived stem cells exosomes (ADSC‐Exos)

Activation of PI3K/AKT signaling pathway

Activation of Wnt/β‐catenin pathway

Affects MMP‐2 and TIMP‐1 protein expression

Mediates h2o2‐induced wound healing

175 , 176

The inflammatory response is the initial stage of skin tissue regeneration can produce an immune response that includes proliferation of immune cells together with secretion of inflammatory and chemotactic factors. 177 A previous study reported that diabetic mice wounded by molding were treated by implantation of exogenous MSC exosomes. Exosomes were found to activate the PTEN/AKT signaling pathway to promote macrophage anti‐inflammatory phenotype M2‐type polarization and inhibited proinflammatory phenotype M1‐type polarization. Furthermore, they inhibited the expression of proinflammatory factors IL‐1β and TNF‐α, promoting the expression of the anti‐inflammatory factor IL‐10, which accelerated the healing rate of skin injury in mice. This effectively demonstrated their ability to promote skin tissue regeneration. 173 Several investigators have reported that human exosomes derived from umbilical cord MSCs can mediate miR‐181c expression and were found to effectively inhibit the TLR4 signaling pathway. 174 Exosomes can increase the expression of anti‐inflammatory factors, which, in turn, reduce the inflammatory response to burn‐induced skin damage in rats. Several researchers have conducted studies in which MSC‐Exos were found to act as recruiters and trainers of immune cells, synergistically promoting the levels of beneficial macrophages and regulating T cells in the skin wounds of mice. They not only inhibited the proliferation of T lymphocytes but also promoted the conversion of activated T lymphocytes to regulatory T cells. 178 This inhibits the inflammatory reaction and promotes skin tissue regeneration. A moderate inflammatory response plays a role in fighting infection and removing damaged cells as well as broken cell debris; however, an excessive inflammatory response can prolong the healing cycle of skin wounds, with serious consequences. 179 Therefore, reducing the inflammatory response during wound healing is an important aspect of skin wound repair. 180

As the inflammatory response proceeds, there is a gradual transition from the inflammatory phase to the proliferative phase, in which leukocytes release cytokines, approximately 3−10 days after injury. This results in the induced migration of endothelial cells and fibroblasts to the wound center as well as the formation of new blood vessels and granulation tissue. The results of a study using MSCs in diabetic rats showed increased expression of CD31 and Ki67 in whole‐skin wounds. This contributed to the enhanced regenerative capacity of granulation tissue and increased expression levels of VEGF and TGFβ‐1. 181 In addition, it made wound closure significantly faster and effectively promoted proliferation of skin tissue cells. Several studies have shown that miR‐21 is highly expressed in the exosomes of adipose MSCs and can activate the PI3K/AKT signaling pathway to affect MMP‐2 and TIMP‐1 protein expression. 175 , 176 This significantly promotes accelerated wound healing. It enhances the proliferation of keratinocytes and fibroblasts while promoting the production of granulation tissue. Moreover, MSC‐Exos were reported to significantly increase the level of angiopoietin 2 in wounds and enhance angiopoietin 2 proliferation, migration, and tube‐forming abilities. 182 This mechanism suggests that stem cell exosomes play an important role in the proliferative phase of skin tissue regeneration.

The last stage of skin regeneration is the remodeling phase, which is mainly characterized by a gradual decrease in the production of new blood vessels and gradual fibrosis of the granulation tissue. During this process, fibroblasts proliferate significantly, and the newly deposited collagen molecules cross‐link, leading to an increase in the tensile strength of the tissue, which results in scar remodeling to restore the normal skin structure, a phase that can last for months to years. 183 In particular, the therapeutic effect of TSG‐6‐modified MSC‐Exos was investigated in a mouse total wound model. The results demonstrated that exosomes from TSG‐6‐modified BMSCs inhibit scar formation by reducing inflammation and inhibiting collagen deposition. 184 According to a previous report, treatment of mice with umbilical cord‐derived MSC‐Exos was found to increase the density of myofibroblasts and inhibit myofibroblast accumulation after a period of time. 185 They have also been shown to reduce the somatic inflammatory response and scarring. Stem cell exosomes reduce scarring during the remodeling phase of skin tissue regeneration, mainly by inhibiting the accumulation of fibroblasts and suppressing collagen deposition. 186 , 187 , 188

In summary, stem cell exosomes are involved in the whole process of skin tissue regeneration and effectively regulate the inflammatory response. They cause proliferative differentiation of endothelial fibroblasts, which results in neoangiogenesis and the inhibition of scarring (Figures 1 and 2).

FIGURE 1.

FIGURE 1

Stem cells, exosomes, and the role of stem cells in each step of the wound healing process.

FIGURE 2.

FIGURE 2

Hematoma tissue is produced at the beginning of the fracture in schematic diagram of bone tissue regeneration, followed by an immune response. This process involves macrophages, neutrophils, T cells, and B cells, forming new blood vessels and bone. Finally, healing tissue is formed.

3.2. Stem cell exosomes promote bone regeneration

The regeneration of bone defects caused by trauma, infection, tumors, or inherent genetic diseases is a global clinical challenge. More than 10 million bone graft procedures are performed each year, and this number is growing at a rate of 10% annually. 189 Bone has a certain ability to regenerate; however, once the critical size is exceeded, spontaneous regeneration of bone is limited. Therefore, a substance is needed to promote bone regeneration. Bone repair and regeneration are complex processes, including tissue regeneration, angiogenesis, and immune regulation, which not only involve BMSCs, osteoblasts, osteoclasts, bone precursor cells, and other bone‐related cells but also immune cells, vascular endothelial cells, and other system cells also play important roles. 190 Exosomes are tiny vesicles secreted by living cells 191 , 192 that carry relevant proteins, lipids, nucleic acids, and other substances derived from late endonucleosomal multivesicular bodies, which play an important role in intercellular information transfer. 193 , 194 , 195 Exosomes are products of the paracrine mechanism of stem cells, are more rapidly utilized than stem cells themselves, and have a wide range of clinical applications. 196 , 197 , 198 Regev‐Rudzki N et al. 194 examined the therapeutic effects of MSC‐Exos during fracture healing in a CD9−/− mouse model and found that MSC‐Exos were effective in promoting fracture healing. Zhang et al. 199 reported results of implanting human embryonic‐derived exosomes in a rat model of cartilage defect and found that exosome treatment resulted in the complete recovery of cartilage and subchondral bone by the 12th week. These two studies demonstrated the efficacy of stem cell exosomes in bone regeneration laying a foundation for subsequent studies, provided a guiding role, and showed potential of the stem cell exosomes for clinical application for bone regeneration. 200 Thus, stem cell‐derived exosomes have become a current research hotspot for tissue injury repair.

In the early stages of bone injury, the acute response to trauma produces a hematoma, preventing further serious trauma such as fractures. 201 An immune response ensues with the secretion of IL‐1, IL‐6, and TNF‐α by neutrophils and macrophages during the first 24 h after the fracture, helping to initiate the repair process through cell recruitment. 202 An important immunomodulatory property of stem cell exosomes is the promotion of M2‐type macrophage polarization and enhanced expression of anti‐inflammatory cytokines, thereby reducing the local inflammatory response. This immune response is critical for the overall repair process. Lo Sicco et al. 203 reported that adipose stem cells can secrete exosomes with anti‐inflammatory effects when cocultured with macrophages and are effectively internalized, promoting macrophage proliferation and enhancing their polarization from M1 to M2 type in a short period of time. They also showed that MSC‐Exos can effectively reduce the local inflammatory response in mice with skeletal muscle injury in in vivo experiments. 203 This study confirmed the ability of MSC‐Exos to regulate macrophage polarization, which may have potential applications in skeletal muscle tissue repair and regeneration. In an immunocompetent rat model, human embryonic MSC‐Exos promoted chondrocyte proliferation and matrix synthesis, with more M2‐type macrophage infiltration than M1‐type macrophages at cartilage defects, along with the reduced expression of inflammatory cytokines IL‐1β and TNF‐α, thereby promoting cartilage repair. 204 Zhang et al. 205 examined the role of MSC‐Exos in the regulation of the inflammatory response, nociceptive behavior, condylar cartilage and subchondral bone healing in an immunocompetent rat model of temporomandibular joint osteoarthritis. The observed exosome‐mediated repair of osteoarthritic temporomandibular joint osteoarthritis was characterized by the early inhibition of pain and inflammation‐reducing degeneration, followed by sustained proliferation. 205 The results showed progressive improvement in matrix expression and subchondral bone structure in rats, leading to overall joint recovery and regeneration. Therefore, stem cell exosomes can promote tissue regeneration by modulating immune function, providing a basis for cell‐free therapy. However, the mechanism of action of stem cell exosomes still requires thorough investigation before clinical trials can be conducted. Stem cell exosomes produce growth factors such as TGFβ, BMPs, IGF‐1, PDGF, FGF2, and VEGF and chemokines such as monocyte chemotactic protein‐1 and monocyte inflammatory protein‐1a by paracrine action, all of which contribute to the stimulation of neoangiogenesis at the fracture site. Takeuchi et al. 206 implanted human bone marrow‐derived MSC‐Exos in a rat cranial defect model. MSC‐Exos enhanced cell migration and the expression of osteogenic and angiogenic genes in MSCs, compared with that in other groups by the fourth week after treatment. Histologically, the MSC‐Exos group showed significant aggregation of osteoblast‐like cells and vascular endothelial cells. 206 This study illustrated that MSC‐Exos promote angiogenesis and bone regeneration at an early stage. Considering the tissue regeneration of transplanted cells and their secretome it was concluded that exosomes might play an important role, especially in angiogenesis. Another study showed that transplantation of UMSC‐Exos significantly enhanced angiogenesis and bone healing in a rat model of a femoral fracture. 207 Implanted UMSC‐Exos may be a key clinical strategy for accelerating fracture healing by promoting angiogenesis. Another study determined that huc‐mscs‐sev can promote osteogenesis and angiogenesis by delivering miR‐23a‐3p to activate the PTEN/AKT signaling pathway for vascularized bone regeneration. 208 This study described the key issues associated with bone regeneration and reconstruction. In mouse and rat models, hard healing tissue remodeling begins 3−4 weeks after fracture and continues for months to years. 209 As a cell‐free agent, stem cell exosomes avoid both the immune rejection of stem cell therapy and ethical controversy and reflect a better prospect for application in tissue regeneration. As nanoparticles (NPs), exosomes can cross various barriers (e.g., blood‐cerebrospinal fluid barrier, capillaries, etc.) and can be directly taken up by and act on target cells with higher action efficiency. Exosome therapy can effectively avoid safety issues associated with the direct application of stem cell therapy and is easy to prepare and transport. However, the mode, dose, and long‐term safety of its application in humans have yet to be evaluated. As the mechanisms of action of stem cell exosomes are studied further, their effects and mechanisms on tissue regeneration will become clearer.

4. NOVEL NANOFORMULATIONS LOADED WITH STEM CELLS AND EXOSOMES

With the continuous advancement in nanotechnology, combining stem cells and exosomes with nanotechnology makes treatment more precise and effective. Nanotechnology can allow stem cells to be targeted to specific locations and can also help stem cells in applying their therapeutic potential to treat, heal, and repair damaged tissues in an effective and safe manner. 210 , 211 Nanotechnology can solve the abnormal proliferation and differentiation of stem cells during the treatment process, 212 improves the possibility of reducing the limitations of stem cells in the treatment of cell regeneration in injured or degenerated organs, enhances tissue repair, and assists in cellular reconstruction. Tissue engineering combined with nanotechnology can also support the growth of stem cells. 213 To exploit the potential of stem cells for biotherapeutic applications, nanotechnology offers the ability to control the size of molecules and processes that control the fate of stem cells. 214 Many studies on exosomes have used vesicle size and biomarkers to isolate and identify exosomes, but fail to recognize exosomes properly, or make full use of them to reach the designated site. 215 Once released, exosomes are involved in various physiological activities and some of the effective substances are lost in this process. The clinical application of exosomes as drug carriers still faces some challenges, such as the effective introduction of exosomes to the target site. 216 Exosomal drug delivery strategies can be divided into two categories: endogenous loading and exogenous loading, which refers to the cargo molecule being loaded in the cell that produces the exosome and the extraction and purification of exosomes before completing the loading process, respectively. Common methods of exogenous loading include incubation, electroporation, ultrasound, and repeated freeze–thaw cycles. Limiting the possibility of exosomes binding to other components allows them to successfully reach the specified location. It is necessary to achieve a combination of nanoagents and exosomes for effective clinical application. 217 The combination of nanoagents and exosomes can solve the problems related to the exosomes delivery and their transmission to the specified location. The traditional cell culture method is susceptible to external environmental influences; the cultures require extremely specialized conditions and are unstable, leading to the wastage of resources. When exosomes were discovered in the 1980s, they were called the “trash” of cellular metabolism. Combining exosomes with nanoformulations allows resources to be repurposed, which is also an excellent material for drug delivery and tissue regeneration. 218 Using nanotechnology to adsorb or encapsulate stem cells and exosomes in nanomaterials can improve drug stability and targeting. Specific types of nanoformulations include polymeric nanoformulations, liposomes and nanoliposomes, polymeric micelles, microspheres, and hydrogels. 219 Several nanoagents that are commonly used for tissue regeneration are listed in Table 5.

TABLE 5.

Nanoformulations commonly used in tissue regeneration.

Nano preparation Material science Structure Slow and controlled release performance References
Polymeric nanoformulations

Polyester

Polycyanoacrylate (PBCA, PiBCA)

Amphiphilic block copolymer (Mal PEG, mPEG)

Dendrimer polymer(PAMAM)

Chitosan

Spherical

linear

tubular

Rod

layered

The core‐sheath structured nanoparticles loaded with KET drug with free hydrogen bonding groups detached in water exhibited a better slow release effect with a release time up to 32 h. 220 , 221
Liposome

Phospholipids:

lecithin cephalin

soybean phospholipid

other synthetic phospholipids

Cholesterol

Small monolayer liposomes

Large monolayer liposomes

Multilayer liposomes Polycystic liposomes

The results of the experiments on the slow release of liposomal pGH in depressed rats showed that GH/liposomes could exert good slow release in 3−4 d. 222 , 223
Microsphere

Natural polymers: starch, albumin, gelatin, chitosan, dextran, etc.

Synthetic polymers: polylactic acid (PLA), polylactide, polylactic acid‐hydroxyacetic acid (PLGA), polylactide glycolide (PLCG), polycaprolactone, polyhydroxybutyric acid, etc.

Solid microsphere

Hollow microsphere

Porous microsphere

Compared with PLA‐loaded microspheres, PLA/CoFE2O4‐loaded microspheres released more slowly during the test cycle, with a cumulative percentage release of 62.40% at 46 h, while PLA‐loaded microspheres already reached 74.02% at 24 h. 224 , 225
Hydrogel

Natural hydrogels: Hyaluronic acid

Collagen

Sodium alginate

Synthetic hydrogels

Polyacrylamide

Polyethylene glycol

Primary structure:

Hydrophilic groups

(carboxyl amido hydroxyl)

Hydrophobic groups: alkyl

Secondary structure:

Mildly cross‐linked three‐dimensional network structure

Three level structure:

Amorphous structure

In simulated gastric juice, the release rate was about 10% within 0.5 h due to the release of surface drug, as the low dissolution rate in simulated gastric juice makes the release very slow. 226
Polymeric micelles

Hydrophilic section material: PEG, PEO, PVP

Hydrophobic section materials: polypropylene, polystyrene, polyamino acids, polylactic acid, spermine, or short‐chain phospholipids

Block polymer micelles

Grafted polymer micelles

Polyelectrolyte micelles

Noncovalent bond micelles

Targeted polymer micelles loaded with celestin showed slow release performance, with approximately 14.87% of the cumulative release of celestin released within 1.5 h and approximately 70% of XT released and equilibrated at 20 h. 227

4.1. Polymeric nanoformulations

Polymeric NPs are efficient drug delivery carriers that can be divided into synthetic and natural polymers and have many outstanding pharmacokinetic properties. The polymeric nanoformulations bind to the stem cells and help the drug reach the designated site and exert its therapeutic effect. Researchers have identified NPs as potential candidates to regulate NSC differentiation, which currently allows stem cells to differentiate into various cell types. 228 Autologous adipose‐derived stem cells (ADSCs) can be safe and effective for the treatment of chronic myocardial ischemia and acute myocardial infarction, but neither ADSCs nor simvastatin‐coupled nanoformulations alone can achieve the desired therapeutic effect. 229 After systemic administration of both, a small number of porous polylactide‐co‐glycolide (PGLA)‐NP‐loaded ADSCs gradually release statin for tissue regeneration, 230 significantly enhancing the therapeutic effect. To overcome the resistance to chemotherapy drugs, researchers have designed a NP containing RGD 231 , 232 and increased the efficacy of adriamycin against cancer stem cells (CSCs). NPs can enhance the transplantation potential of human hematopoietic stem cells, 233 suggesting that NPs have the potential to overcome the current limitations of HSC gene editing. Chitosan medium could be used to mimic the microenvironment of CSCs, increase the expression of stem cell characterization‐related genes 220 and markers of CSCs to block differentiation and reduce the number of CSCs.

The combination of polymeric magnetic nanoformulations (MNPs) and exosomes provides new avenues for exosome‐based nanotherapies. Local release of exosomes captured under acidic pH conditions by accumulation of nanoformulations as well as cleavage of hydrazine bonds in the presence of a local magnetic field. 234 By encapsulating adriamycin (DOX) into isolated exosomes, MNPs are encapsulated to allow the accumulation of exosomes at the site of the lesion and subsequently the targeted release of exosomes. 235 In combination, the drugs can kill cancer cells. Although MNPs can deliver anti‐HIV drugs through an in vitro blood–brain barrier model, high doses of MNPs may damage cells. Correspondingly, exosomal EVs (xev) can cross the blood–brain barrier but lack long‐range site specificity. When MNPs and xevs are combined as nanocarriers, 236 they can target delivery of anti‐HIV fusion agents effectively across the blood–brain barrier. Both MNPs and exosomes from stem cells have been shown to be effective in wound healing. The preparation of exosomes using BMSCs stimulated by MNPs in conjunction with static magnetic fields 237 can further enhance wound repair.

4.2. Liposomes and nanoliposomes

Liposomes act as nanocarriers for targeted drug delivery for both hydrophilic and lipophilic drugs and improve drug solubility and stability. 222 , 223 Preparation of aspirin‐containing liposomes by a thin film dispersion method and integration into DOPA3d printed PCL scaffold 238 enhances PCL stents. It has been verified that CD146 + liposome magnetic beads could successfully separate CD146 and ADSCs, 239 which can promote the repair of articular cartilage defects. If the liposome is injected directly into the bone cavity, TGF is released over several weeks. TGF‐loaded liposome‐coupled scaffolds exhibit enhanced release and localization. The subcutaneous implantation of hydroxyphosphate composite scaffolds coupled with bisphosphate‐encapsulated liposomes in rats exhibited a strong binding effect, which increased drug retention. 240 Long‐circulating polyethylene glycolyzed poly (tritiated ester) liposomes induce CSCs apoptosis in mice and are effective therapeutic agents for further inhibition of CSCs growth. 241 Cross‐linked multilayer liposomes composed of adriamycin (Dox) and salicin (Sal) interact with breast cancer tumor cells as well as CSCs and effectively inhibit the cancer cells. 242 Biocompatible nanoprobes consisting of liposome‐indocyanine green hybrid vesicles for safe labeling of hMSCs can track cells after drug administration 243 until the drug is fully effective.

The binding of liposomes to exosomes can improve their targeting and increase their retention time, apart from the other remarkable features. A hybrid therapeutic nanovesicle in the conjugate of exosomes, drug‐loaded thermosensitive liposomes, and hGLV loaded with photothermal agents effectively targeted homologous tumors in mice resulting in excellent photothermal treatment and complete tumor elimination. 244 The efficiency of exosome encapsulation of large nucleic acids was largely enhanced by mixing the exosomes with liposomes. Subsequently, the synthesized liposome exosome mixture efficiently encapsulated large plasmids, including CRISPR‐Cas9 expression vectors. 245 The instability in the transport of exosomes to tumor cells can be resolved by fusion delivery of exosomes and liposomes, 246 which are effectively absorbed by tumor cells and then can be concentrated near the tumor to cause apoptosis.

4.3. Microspheres

Microspheres are the only effective means of introducing biological materials into real‐time differentiated ESC culture. Microspheres as nanoagents can track MSCs, deliver small molecules, and promote stem cells. 224 , 225 The encapsulation of stem cells in microlets can be used as stem cell repair units. 247 Microspheres in stirred suspension can preserve stem cells from human ADSCs (hASCs), which bind to microspheres for a period of time after higher levels of pluripotent markers expressed by hASCs, 248 and improve the proliferation, colony formation, network formation, multimesenchymal differentiation, and regenerative capacity of stem cells. Injection of umbilical cord‐derived MSCs microspheres with controlled particle size and cell encapsulation ability into the rat uterus 249 indicated their nature in promoting the repair and regeneration of the endometrium. BMSCs in the presence of open porous poly (lactic‐co‐glycolic acid) microspheres can enhance cell proliferation in vitro and in vivo promoting cartilage regeneration. 250 Sodium alginate‐based Janus microspheres for targeted delivery of MSCs to cartilage were measured and found to be less toxic to MSCs 251 with good restorative ability.

Exosomes have isolation and detection limitations during cancer diagnosis; furthermore, progress in the studies on exosome analysis is slow. Researchers have discovered a microfluidic device‐based method for exosome isolation and detection, realizing that the microsphere‐mediated dielectrophoretic separation and immunoaffinity detection 252 improve the effectiveness of exosomes in tumor marker detection and cancer diagnosis. Exosomes have significant regenerative potential for bone tissue engineering; conversely, the therapeutic potential of exosomes is limited, and there is some attrition during delivery. The high biological activity of exosomes adsorbed onto the surface of porous PLGA injectable microspheres with bioinspired polydopamine (PDA) coating enables the delivery of exosomes to effectively induce tissue regeneration, 253 thus facilitating the clinical translation of exosome‐based therapies. Integration of a PLGA microsphere delivery platform into interpenetrating network hydrogels facilitates sustained delivery of MSC‐Exos, thereby promoting endogenous AF repair, 254 , 255 illustrates the potential of exosomes for cell‐free bioactive repair.

4.4. Hydrogels

The connection between hydrogels and stem cells is complex due to the involvement of various factors, such as porosity, different polymer types, stiffness, compatibility, and degradation. As a carrier of stem cells, the right type of hydrogel must be selected for encapsulation in different situations for the purpose of transporting stem cells. 226 Hydrogel materials have good biocompatibility and biodegradability and can solve the problem of the low survival rate of stem cell transplantation after spinal cord injury (SCI). 256 Cells encapsulated in hydrogel microcapsules containing a thin oil layer ensure high cell viability before the cells reach the area of inflammation, segmental rupture of the hydrogel with reduced stiffness in the process of movement, and delivery of UCB‐MSCs to the sites with inflammation, 257 thus releasing the active substances to eliminate the inflammation. The use of acrylic hyaluronic acid (HA) hydrogel microspheres, a biodegradable hydrogel, can deliver carriers in vivo and release cells to local targets. 258 A study reported that to enhance the attachment ability and preserve bone marrow MSCs in hydrogels, the viability, density, and delivery of paracrine factors in hMSCs using agarose‐carbohydrate hydrogels must be optimized. 259 , 260

Hydrogels efficiently deliver exosomes to individual cells, maintaining the biological activity of exosomes while performing various repairs. Hydrogels can stimulate the release of MSC‐Exos and heal better than exosomes or hydrogels alone, 261 suggesting that sustained release of exosomes and hydrogels can synergistically promote wound healing. The synthesis of exosome gels by immobilizing exosomes in peptide‐modified adhesive hydrogels improved the retention and release of exosomes in injured spinal cord tissue. 262 Experiments have demonstrated that hydrogels significantly improve the residence time and stability of exosomes in vivo, and hydrogel‐bound exosomes have enhanced endothelial protection and proangiogenic capacity in vitro. 263 The combination of exosomes and hydrogels promotes wound healing, and hydrogels are effective in delivering exosomes and improving exosomal capacity. 181

4.5. Polymeric micelles

Polymeric micelles are self‐assembled from amphiphilic polymers in an easy‐to‐use manner to achieve optimal loading, stability, body circulation, and targeted delivery. 227 Dexmedetomidine micelles can be internalized by osteoblast‐like cells and rBMSCs, resulting in the release of dexmedetomidine. 264 Ibuprofen esterase‐responsive copolymer nanomicelles further enhance the adaptability of (NPPCs) in harsh environments and enhance their repair capacity. 265 , 266 Polymeric micelle drugs have higher cytotoxicity and inhibition of CSC colony formation than free drugs, and can inhibit tumor growth by CSCs. 267 Polymeric micelles assayed for their effects on stem cell proliferation and differentiation 268 showed that they promote adipogenesis, chondrogenesis, and osteogenesis of stem cells. A new micelle‐assisted method for efficiently loading anticancer drugs into exosome‐like vesicles has been discovered with improved tumor accumulation and retention. 269 Using the tumor characteristics of MSCs, a drug transfer system based on MSCs was developed, and paclitaxel (PTX)‐encapsulated HA poly (d, l‐lactate‐co‐glycolide) polymer gel (PTX/HA‐PLGA) micelles were used for treatment. 270 The results showed that the overexpression of CD44 on the surface of MSCs and tumor cells not only increased the micellar load of PTX/HA‐PLGA in MSCs but also promoted drug delivery between MSCs and adjacent cells.

In summary, both stem cells and exosomes have some defects, such as the transport and release of a single substance. Combinations with nanoformulations can compensate for the shortcomings in this area. Nanopreparations have different effects in different fields; for example, polymeric NPs enhance the stability of drugs, especially protein‐based drugs, and have better slow and controlled release properties compared with liposomes. The interactions between nanoformulations, stem cells, and exosomes allow for more efficient transport and delivery of stem cells and facilitates exosome accumulation near a specified location, better utilization of stem cells and exosomes for tissue repair and other capabilities (Figure 3).

FIGURE 3.

FIGURE 3

Nanopreparations of stem cells and their cytokines in regenerative medicine.

5. OTHER SYSTEMS LOADED WITH STEM CELLS AND EXOSOMES

In addition to nanotechnology, scaffolds, microneedles, and injectable hydrogel scaffolds are important for the delivery of stem cells and their exosomes. The primary aim of the stent is to provide primitive support, ensuring adhesion, migration, proliferation, differentiation, and long‐term survival of stem cells. Microneedling (MN) is a new type of biomaterial loaded with stem cells, allowing them to enter into damaged tissues with greater precision and effectiveness. The new hydrogel can effectively deliver stem cells and enhance their viability, enabling better attachment of stem cells to the scaffold.

5.1. Scaffolds

Due to the hostile nature of the pathological microenvironment, specific conditions must be maintained to improve their function. During the transplantation process, the scaffold acts as a bridge, a guide for axonal growth, and a carrier for the transfer of stem cells, thus changing the microenvironment. Currently, neural and MSCs have been transplanted into biomaterial scaffolds, and experiments are underway to regenerate the spinal cord. 271 , 272 Natural and synthetic biomaterials provide a controlled microenvironment for cells, thereby accelerating their growth and differentiation, and enhancing their viability in the human body. Biological scaffold materials are primarily used to repair damaged or missing tissues and to reconstruct their functions. Structural reconstitution and good clinical outcomes have been reported using biological scaffolds by the mechanism of release or production of impact factors, incorporation of endogenous stem cells into the scaffold, and modulation of innate immune responses. 273 The most common method of culturing stem cells is the two‐dimensional plastic technique. This medium does not exhibit a good stem cell niche in humans; it is a very fine microenvironment that includes supporting stromal cells, the ECM, and growth factors. Therefore, researchers and clinicians are seeking the best stem cell formulation that can be used for research and clinical applications using three‐dimensional technology. Three‐dimensional culture technology can better simulate cell–cell and cell–matrix interactions. 274 In addition to cells, different matrices and scaffolds can also be used to support complex tissues. The availability of modern technology can facilitate the expansion of cell and scaffold technology, thus providing a powerful tool for regenerative medicine. 27 In trauma or spinal fusion, scaffolding is the main bone repair method, combined with active molecules and stem cells. Cell‐based bone repair techniques have been shown to be superior to conventional techniques. A previous study showed higher osteogenic efficiency of stem cells grown in a suitable three‐dimensional scaffold support. 4 Research has shown that three‐dimensional biological scaffolds can support and mimic the in vivo environment and help stem cells differentiate into skeletal cells. Stem cells have a strong repair capacity during burn wound repair. Application of different types of stem cells can effectively improve wound healing. In human experiments, homing of stem cells to traumatized surfaces cause re‐epithelialization, angiogenesis, granulation, inhibition of apoptosis, and regeneration of skin attachments, which reduces infection rates. Animal studies have shown that stem cells are effective in accelerating wound healing. 276 The application of stem cells to burn wounds can significantly promote wound healing via scaffolding. The direct implantation of three‐dimensional bioprinted stem cells into blood vessels has great potential, especially the production of organ and tissue substitutes. 277 , 278 Promoting the differentiation of stem cells using bioprinting technology allows scaffolds to be reshaped over a period of time. 279 , 280 The ability to tune bioprinting properties, which can be used to create stem cell carriers and take full advantage of cell pluripotency, is of great importance in biomaterials and bioengineering. Optimal transplantation or implantation can be achieved by combining stem cells with an artificial scaffold. The interaction between a stem cell and its scaffold has important implications for differentiation and health. 281 Stem cells from the cystic part of rat hair were used for scanning electron microscopy, using the method of extracting collagen preparation and scaffolding. The results indicated that the growth and changes in stem cells in both scaffolded and nonscaffolded states showed clear signs of cell differentiation, displaying larger cuboidal bodies with star‐shaped nuclei. Scanning electron microscopy revealed large porosity, which facilitates cell attachment and growth. 282 Stem cells have a significant differentiation role in osteogenesis, whereas collagen scaffolds are a suitable matrix for cell growth and differentiation. There is potential for application of injectable Matrigel as a scaffold for BMP‐transduced rat dental capsule stem cells/precursor cells (rDFSCs) to promote in vitro osteogenesis and in vivo ectopic bone formation. 283 BMP9 significantly promoted osteogenic differentiation of rDFSCs, whereas Matrigel significantly promoted osteogenesis of rDFSCs induced by BMP. Biomaterial scaffolds combined with stem cell transplantation improve cell survival and differentiation efficiency. 284 The use of natural and synthetic biomaterials can mimic the ECM, which promotes the differentiation of MDSCs and thus re‐establishes the correct ecological niche of muscle stem cells, positively influencing muscle repair. 285 The binding of scaffolds with cell growth factors and ECM molecules can promote cell adhesion, proliferation, and induction of osteogenesis, thus providing signals for cell transplantation and regeneration, 286 offering tremendous opportunities for treating a wide range of diseases.

5.2. Microneedling

MN is a promising new technology that can be used to provide multiple drugs for the treatment of ischemic cardiomyopathy. By encapsulating cardiac stromal cells in fibrin gels and inoculating them on the MN matrix, polymeric MNs create “channels” between MNs and the host myocardium, allowing them to deliver regenerative factors to the damaged myocardium thereby accelerating cardiac repair. 287 , 288 The activation of hair follicle stem cells (HFSCs) can accelerate the regeneration of hair follicles, but difficulties remain in improving and managing them effectively. A keratin microneedle can be separated from the hair for the continuous delivery of HFSC‐activated substances through the microneedle tip. 289 , 290 Studies have shown that microneedle devices, in combination with MSC‐derived exosomes and small‐molecule drugs, are effective in reducing doses and improving treatment efficiency. Exosomes derived from MSCs have shown promising results for the treatment of SCI. However, conventional two‐dimensional culture methods result in stem cell loss from MSCs, which greatly limits the potential efficacy of in vitro secretions from MSCs. The three‐dimensional exosome in vitro culture method is effective. Conventional exogenous drug therapy relies on repeated local injections, which cause secondary damage and are ineffective. Researchers have developed a three‐dimensional exohydrogel hybrid microneedle array patch for the in situ repair of SCI. 291 It was shown that three‐dimensional cultured bone marrow MSCs can maintain their stem cell properties, thus three‐dimensional exosomes can effectively reduce SCI‐induced inflammation and glial scarring. A novel core–shell HA MN patch with iron‐MSC‐derived artificial nanovesicles (Fe‐MSC‐NVs) and polydopamine NPs (PDA NPs) was encapsulated in a needle tip for wound healing. Fe‐MSC‐NVs contain multiple therapeutic cytokines encapsulated in the HA nucleus at the end of the MN to promote angiogenesis. 292 In vivo experiments have shown that Fe‐MSC‐NVs/PDA MN patches are effective in wound repair in patients with diabetes. Skin MN accelerates the process of skin regeneration by creating a large amount of microdamage, which occurs when the skin is punctured by a very fine needle. To accelerate postoperative regeneration or to improve efficacy, MN is combined with drugs, vitamins, or stem cells. 293 Owing to its high efficacy, few side effects, and short recovery period, MN has been widely used in clinical practice. MSCs are injected by encapsulating them in biomaterials, thus enhancing their stability. However, because of its poor efficacy, numerous cells are required for treatment. In addition, injection to the target site achieved using conventional syringes by local injection can cause significant damage. Researchers have developed a separable hybrid microneedle library (d‐HMND) cell delivery system 294 that can effectively inject MSCs locally. Researchers have invented a detachable microneedle patch, whose main component is chitosan lactate (CL) with adipose stem cell exosomes (EXO). CL and EXO synergistically promote hair regrowth and regulate the circulation of hair follicles. 295 Animal studies have shown that drug‐free microneedle patches significantly promote hair regrowth within 7 days, compared with the typically administered minoxidil at lower doses. 296 , 297 Microneedles deliver stem cell secretomes directly to the site of injury, demonstrating the relative therapeutic benefits of live cells while avoiding potential limitations. 298 To improve the efficacy, a variety of biomaterials have been developed for the continuous and controlled delivery of stem cell secretomes. Direct delivery of stem cells and therapeutic macromolecules to the cardiac tissue and vascular smooth muscle cells 299 breaks through the nonpermeable barrier and allows the drug to go directly to the target area for maximum effect, thereby reducing the required dose. In recent years, MSCs and MN techniques have been widely used to delay aging. To investigate the effect of human umbilical cord tissue matrix (hUC‐MSC‐CM) on skin brightness and rejuvenation, researchers conducted a clinical trial in which HUC‐MSC‐CM in combination with MN was found to delay aging and could be applied to facial rejuvenation by evaluating the volunteers’ skin radiance and texture as well as their self‐satisfaction. 300

5.3. Hydrogel scaffolds

Recently, applied ADSCs have been investigated, and biologically active injectable hydrogels have been developed as cell‐transfer carriers. Hydrogel scaffolds are promising regenerative materials that can mimic the ECM and deliver signaling molecules. 301 , 302 The bionic injectable hydrogel mimics the natural microenvironment and provides the correct biochemical information for tissue regeneration. 303 Researchers have developed amniotic tissue hydrogels as a delivery system for adipose MSCs and retained the stem cells at the target site, which could serve as a potential stem cell carrier that could be used and developed as a potential therapeutic agent for osteoarthritis. 304 , 305 , 306 ADSCs have better anti‐inflammatory, chondroprotective, and paracrine functions that can be enhanced by genetic alterations. Direct cell transport without matrix support usually results in poor survival of the treated cells. Researchers have used bone grafting via genetically engineered ADSCs. 307 An injectable ECM‐mimetic hydrogel was developed as a cell‐transfer vehicle to create a favorable microenvironment for ADSC spreading and proliferation. 308 Hydrogels that mimic the ECM can reduce cell death during and after injection. Currently, self‐healing natural hydrogels suffer from poor stiffness, low healing efficiency, poor biocompatibility, and poor hydrolytic stability in the treatment of SCIs. Researchers have developed an injectable self‐healing HA gel based on multiple dynamic covalent bonds. 309 This injectable, self‐healing HA hydrogel is a promising material for nerve repair. Targeted transport of stem cells using scaffolds, microneedles, and injectable hydrogels can effectively repair lesion sites and maintain stem cells.

6. COMBINED APPLICATION OF GENETIC ENGINEERING TECHNOLOGY AND STEM CELL EXOSOMES

Stem cells have multiple functions, including migration, differentiation, and secretion of multiple therapeutic molecules such as immunomodulatory factors. 142 Therefore, stem cell therapy has been used in preclinical and clinical trials and has been shown to have great potential for application in the treatment of various diseases. Recently, stem cells have been modified to enhance their innate abilities and provide new functions that allow them to be used in different biomedical fields. There are many different types of modified stem cells; for example, engineered stem cells include genetically modified stem cells for gene transfer, NP loading and delivery, and small‐molecule drug delivery. 310 Stem cell therapy has become an effective treatment option. For example stem cell therapy has be‐come an effective treatment option for tissue regeneration. . To maximize the potential of stem cells, it is necessary to modify their properties. Genetic engineering is particularly notable in this area because of the vast array of tools available to induce gene expression in a precise and controlled manner. 311 Genetic engineering, also known as gene splicing technology and DNA recombination technology, uses the principles of molecular genetics and modern techniques from molecular biology and microbiology to construct hybrid DNA molecules in vitro from genes of different origins according to a specific structure and then transplants them into living cells to achieve the genetic characteristics of organisms, obtain new varieties, and produce new products. 312 , 313 Cell‐based bone regeneration therapy has been shown to be a better therapeutic choice because it reduces immune risk and bodily pain. Clinical studies have shown that different types of stem cells, especially MSCs, can effectively treat a variety of bone‐related diseases. 314 The clinical challenges in bone regenerative medicine have led to the development of cell and tissue engineering. 315 In particular, the development of molecular biology has laid the foundation for designing genetic strategies for bone tissue repair. Therefore, genetic engineering is an effective way to achieve continuous cell differentiation and ECM production. Gene therapy is a traditional method for improving the regenerative capacity of bone tissue. Recent studies have shown that genetic engineering techniques have become important tools for establishing effective bone, cartilage, and connective tissue. 316

Natural stem cells isolated from humans have been used for therapeutic treatments for decades. This includes the transplantation of primary cells such as hematopoietic stem cells, MSCs, and more recently, pluripotent stem cell derivatives. 317 However, with the development of cell engineering technologies, the new generation of stem cell‐based therapies will greatly expand their application. For example, stem cells improve drug and tumor lysis virus delivery to recalcitrant tumors and convert them into angiogenic, neurotrophic, and anti‐inflammatory factors, thereby accelerating the healing of injured or damaged tissue. 318 Advances in reprogramming technologies have led to the emergence of induced pluripotent stem cells (iPSCs). Thereafter, the progressive research has led to the development of various effective methods that enable iPSCs to differentiate into various cell types, thus providing new avenues for disease models. 319 In fact, cells derived from iPSCs have been widely used to study the molecular and cellular pathophysiological mechanisms underlying genetic diseases. However, the greatest difficulty in current research on iPSC cell‐based disease models is to identify the role of disease‐causing genetic variants. In the last decade, researchers have greatly improved genome editing technologies, one of which is clustering rule interval short palindromic repeat related system 9 (CRISPR/Cas9). This technique efficiently modifies a specific, discontinuous protein with a small guide RNA molecule. 320 , 321 This technology allows for the creation of isogenically regulated or mutated cell lines, thus focusing on the study of pathologies due to specific variants. 322 Combining state‐of‐the‐art tissue engineering with CRISPR/Cas9‐mediated genome engineering allows the study of hPSCs in kidney diseases with unprecedented potential. In recent years, genetically engineered mutations using CRISPR/Cas9‐mediated mutagenesis have been shown to replicate polycystic kidney disease and glomerular disease phenotypes. 323 Genetic or molecular engineering of exosomes enhances their target specificity and anticancer activity, with low toxicity. 324 The modification of cultured cells using genetic engineering techniques in vitro can play an important role in the treatment of diseases. After the isolation and purification of exosomes, ncRNAs are loaded with mimics or inhibitors to modify the exosome surface and enhance targeting ability. 325 Exosomes from stem cells have been widely used as “cell‐free” therapies to promote the regeneration and reconstruction of various tissues. However, resource constraints and a lack of effective therapeutic outcomes have limited the use of exosomes. Stem cell‐mediated gene therapy could create exosomes that promote bone regeneration. MSCs use protein‐2 gene modification to alter exosome content, promote bone repair, and make them more biocompatible and potentially clinically useful. 326 A hybrid exosome‐based nanoscale delivery vehicle consisting of engineered exosomes and liposomes fused together can enable the selective encapsulation and delivery of CRISPR/Cas9 plasmids to chondrocytes embedded in articular cartilage, thereby reducing cartilage damage. 327

7. CONCLUSIONS AND OUTLOOK

One of the main goals of tissue engineering and regenerative medicine is to restore damaged skin and bones. The issues of skin tissue and bone healing regeneration are gradually gaining attention and have become research hotspots. Recent studies have demonstrated the significant potential of stem cells in the fields of skin regeneration and bone healing. Wound healing is complex, and stem cells can simultaneously inhibit and increase inflammatory and anti‐inflammatory factors during wound repair, respectively. 302

Stem cells and their exosomes play different roles at each stage of skin and bone tissue healing to promote tissue repair. Despite the advantages of stem cell therapy, there are limitations to its use as a single substance for therapeutic purposes. Stem cells are classified as ESCs or adult stem cells (AS) according to their developmental stage. Currently, the directed differentiation of human ESCs, immune rejection caused by clinical application, and lack of sources of human ESCs have limited the rapid and widespread use of ESCs in clinical practice. 328 From the perspective of clinical application, AS cells are superior to ESCs, and self‐AS cell transplantation, such as bone marrow stem cells, is currently the ideal choice for clinical application. 329 AS cells have some disadvantages, such as extremely small content, unclear specific markers, and slow division. 330 Therefore, the isolation, purification, identification, and in vitro culture expansion of AS cells are very difficult. 331 However, the benefits of stem cell therapy remain controversial. Therefore, a deeper understanding of endogenous repair mechanisms and their interactions with stem cells is required. 6 , 332 Exosomes also have problems, such as difficulty in extraction and poor retention. Therefore, we propose the formation of stem cells and stem cell exosomes into nanopreparations to provide a new way to promote skin tissue regeneration. Although this technology is relatively new, it shows great potential in stem cell research. 333 Stem cell exosomes have anti‐inflammatory, antiaging, and wound‐healing skin tissue regeneration effects. 171 In skin regeneration, stem cells can promote cell growth and angiogenesis and reduce inflammatory responses in areas of skin damage. It is the most important link in the process of tissue regeneration, promoting new collagen and elastic fibers, inhibiting metalloproteinase activity, and anti‐UV aging. 334 The use of stem cells in the bone healing process greatly facilitates the speed and effectiveness of healing. The combination of nanotechnology and stem cells can maximize the therapeutic effects of stem cells. Nanoformulations can target and transport stem cells to specific sites and prolong the residence time, which not only improves the targeting of stem cell therapy but also reduces the adverse effects and can enhance the therapeutic effect and safety of stem cells to a great extent. 335 , 336 Despite numerous interesting studies, there is currently no uniform standard to describe the sequential manipulation of cells to obtain exosomes for clinical use. Exosomes can change the microenvironment; however, the microenvironment can also change the composition of exosomes. 337 The advantages of exosomes in tissue repair and regenerative medicine include safety and longevity. Exosomes can stimulate the proliferation and migration of senescent human dermal fibroblasts and their therapeutic potential should be explored. 338 The combination of nanoagents and exosomes can treat tissue damage caused by inflammation and prolong residence time at specific sites. 339 Nanoformulations also have some disadvantages. Polymeric NPs have disadvantages, such as a higher risk of particle aggregation and immunogenicity‐related toxicity. Therefore, very few polymeric NPs have been approved for clinical use. Liposomes also have some problems in application, such as low encapsulation rate, rapid release of water‐soluble drugs in the blood, and low storage stability. Drug‐carrying microspheres also have limitations, such as limited use, high price, and low drug loading. Hydrogels also have limitations; the mechanical properties of hydrogels with good lubrication are poor, and improved mechanical properties do not result in good lubrication properties. The practical applications of hydrogels in the field of articular cartilage replacement materials are limited. Polymeric micelles also have limitations such as low drug‐loading capacity, poor stability, and weak microenvironmental response. Currently, the study of stem cell exosomes is an active area, and advances in the development of technologies and related studies may provide valuable information for revealing the heterogeneity and biological functions of exosomes and advancing the development of stem cell exosomes in medical therapy and diagnosis. Future research is expected to provide new insights into the role of exosomes in physiological processes, such as tissue regeneration, organ degeneration, and aging. 340 , 341 The combined application of stem cells, exosomes, and nanopreparations has become a new trend in the field of skin tissue regeneration; however, many research questions remain unanswered, such as how to track the technology more accurately and consistently to explore the growth of stem cells in the body, the number of cells needed for treatment, and whether stem cells remain in the lungs after whole‐body transplantation.

Research related to the use of stem cell exosomes as drug delivery vehicles is also a hot topic, and drug delivery efficiency is another challenge. 342 Owing to the structural diversity of exosomes, their initial structural configuration can be altered using a variety of means, such as genetic engineering, chemical procedures, physical techniques, and microfluidics, thus increasing the load of exosomes and expanding their biomedical use. In targeted therapy, exosomes have great potential to overcome the limitations of conventional NP technology. 343 Future research will require the development of efficient techniques for loading sufficient doses of drugs into stem cell exosomes while maintaining their physical integrity and biological activity. 344 It is believed that in the future, researchers and medical professionals will be able to overcome these challenges as their research progresses and bring safer and more efficient stem cell transplants, giving hope to patients. With the continuous development of research and regenerative medicine, stem cells as an important new star in the field of regenerative medicine will continue to contribute to the progress of medicine.

AUTHOR CONTRIBUTION

Y. J. conceived and drafted the manuscript, drew the figures, and discussed the concepts of the manuscript. S. L. conceived and drafted the manuscript, and discussed the concepts of the manuscript. Q. Y. provided valuable discussion and funding. T. C. and D. L. discussed the concepts of the manuscript. Y. J., S. L., and Q. Y. provided valuable discussion and revised the manuscript. All authors have read and approved the final manuscript.

CONFLICT OF INTEREST STATEMENT

The authors declare no conflict of interest.

INFORMED CONSENT STATEMENT

Not applicable.

ETHICS STATEMENT

Not applicable.

ACKNOWLEDGMENTS

We thank BioRender and Figdraw for helping us in our drawing process.

Jin Y, Li S, Yu Q, Chen T, Liu D. Application of stem cells in regeneration medicine. MedComm. 2023;4:e291. 10.1002/mco2.291

Contributor Information

Tianli Chen, Email: tli_chen@163.com.

Da Liu, Email: liuda_1986@163.com.

DATA AVAILABILITY STATEMENT

Not applicable.

REFERENCES

  • 1. Klabukov ID, Krasilnikova OA, Baranovskii DS, Ivanov SA, Shegay PV, Kaprin AD. Comment on: regenerative medicine, organ bioengineering and transplantation. Br J Surg. 2021;108(11):e386. [DOI] [PubMed] [Google Scholar]
  • 2. Burns TC, Quinones‐Hinojosa A. Regenerative medicine for neurological diseases‐will regenerative neurosurgery deliver? BMJ. 2021;373:n955. [DOI] [PubMed] [Google Scholar]
  • 3. Alatyyat SM, Alasmari HM, Aleid OA, Abdel‐Maksoud MS, Elsherbiny N. Umbilical cord stem cells: background, processing and applications. Tissue Cell. 2020;65:101351. [DOI] [PubMed] [Google Scholar]
  • 4. Bacakova L, Zarubova J, Travnickova M, et al. Stem cells: their source, potency and use in regenerative therapies with focus on adipose‐derived stem cells ‐ a review. Biotechnol Adv. 2018;36(4):1111‐1126. [DOI] [PubMed] [Google Scholar]
  • 5. Dulak J, Szade K, Szade A, Nowak W, Jozkowicz A. Adult stem cells: hopes and hypes of regenerative medicine. Acta Biochim Pol. 2015;62(3):329‐337. [DOI] [PubMed] [Google Scholar]
  • 6. Yamanaka S. Pluripotent stem cell‐based cell therapy‐promise and challenges. Cell Stem Cell. 2020;27(4):523‐531. [DOI] [PubMed] [Google Scholar]
  • 7. Thomson JA, Itskovitz‐Eldor J, Shapiro SS, et al. Embryonic stem cell lines derived from human blastocysts. Science. 1998;282(5391):1145‐1147. [DOI] [PubMed] [Google Scholar]
  • 8. Franck CL, Senegaglia AC, Leite LMB, de Moura SAB, Francisco NF, Ribas Filho JM. Influence of adipose tissue‐derived stem cells on the burn wound healing process. Stem Cells Int. 2019;2019:2340725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Tyeb S, Shiekh PA, Verma V, Kumar A. Adipose‐derived stem cells (ADSCs) loaded gelatin‐sericin‐laminin cryogels for tissue regeneration in diabetic wounds. Biomacromolecules. 2020;21(2):294‐304. [DOI] [PubMed] [Google Scholar]
  • 10. Maumus M, Jorgensen C, Noel D. Mesenchymal stem cells in regenerative medicine applied to rheumatic diseases: role of secretome and exosomes. Biochimie. 2013;95(12):2229‐2234. [DOI] [PubMed] [Google Scholar]
  • 11. Platas J, Guillen MI, Perez Del Caz MD, et al. Paracrine effects of human adipose‐derived mesenchymal stem cells in inflammatory stress‐induced senescence features of osteoarthritic chondrocytes. Aging (Albany NY). 2016;8(8):1703‐1717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Koh YG, Kwon OR, Kim YS, Choi YJ, Tak DH. Adipose‐derived mesenchymal stem cells with microfracture versus microfracture alone: 2‐year follow‐up of a prospective randomized trial. Arthroscopy. 2016;32(1):97‐109. [DOI] [PubMed] [Google Scholar]
  • 13. Li Y, Huang Z, Huang X, et al. The influences of PEG‐functionalized graphdiyne on cell growth and osteogenic differentiation of bone marrow mesenchymal stem cells. J Biomed Mater Res B Appl Biomater. 2023;111(6):1309‐1317. [DOI] [PubMed] [Google Scholar]
  • 14. Sorg H, Tilkorn DJ, Hager S, Hauser J, Mirastschijski U. Skin wound healing: an update on the current knowledge and concepts. Eur Surg Res. 2017;58(1‐2):81‐94. [DOI] [PubMed] [Google Scholar]
  • 15. Bhardwaj N, Chouhan D, Mandal BB. Tissue engineered skin and wound healing: current strategies and future directions. Curr Pharm Des. 2017;23(24):3455‐3482. [DOI] [PubMed] [Google Scholar]
  • 16. Takeo M, Lee W, Ito M. Wound healing and skin regeneration. Cold Spring Harb Perspect Med. 2015;5(1):a023267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Jeschke MG, Patsouris D, Stanojcic M, et al. Pathophysiologic response to burns in the elderly. EBioMedicine. 2015;2(10):1536‐1548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Han G, Ceilley R. Chronic wound healing: a review of current management and treatments. Adv Ther. 2017;34(3):599‐610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Raghuram AC, Yu RP, Lo AY, et al. Role of stem cell therapies in treating chronic wounds: a systematic review. World J Stem Cells. 2020;12(7):659‐675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Marx C, Gardner S, Harman RM, Wagner B, Van de Walle GR. Mesenchymal stromal cell‐secreted CCL2 promotes antibacterial defense mechanisms through increased antimicrobial peptide expression in keratinocytes. Stem Cells Transl Med. 2021;10(12):1666‐1679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Chow L, Johnson V, Impastato R, Coy J, Strumpf A, Dow S. Antibacterial activity of human mesenchymal stem cells mediated directly by constitutively secreted factors and indirectly by activation of innate immune effector cells. Stem Cells Transl Med. 2020;9(2):235‐249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Rani S, Ritter T. The exosome ‐ a naturally secreted nanoparticle and its application to wound healing. Adv Mater. 2016;28(27):5542‐5552. [DOI] [PubMed] [Google Scholar]
  • 23. Domaszewska‐Szostek AP, Krzyzanowska MO, Czarnecka AM, Siemionow M. Local treatment of burns with cell‐based therapies tested in clinical studies. J Clin Med. 2021;10(3):396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Das P, Manna S, Roy S, Nandi SK, Basak P. Polymeric biomaterials‐based tissue engineering for wound healing: a systemic review. Burns Trauma. 2023;11:tkac058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Kim H, Wang SY, Kwak G, Yang Y, Kwon IC, Kim SH. Exosome‐guided phenotypic switch of M1 to M2 macrophages for cutaneous wound healing. Adv Sci (Weinh). 2019;6(20):1900513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Mead B, Ahmed Z, Tomarev S. Mesenchymal stem cell‐derived small extracellular vesicles promote neuroprotection in a genetic DBA/2J mouse model of glaucoma. Invest Ophthalmol Vis Sci. 2018;59(13):5473‐5480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Boack DH, Manegold S, Haas NP. Treatment strategy for talus fractures. Unfallchirurg. 2004;107(6):499‐514. quiz 513–4. [DOI] [PubMed] [Google Scholar]
  • 28. Van Laere AS, Nguyen M, Braunschweig M, et al. A regulatory mutation in IGF2 causes a major QTL effect on muscle growth in the pig. Nature. 2003;425(6960):832‐836. [DOI] [PubMed] [Google Scholar]
  • 29. Calvi LM, Adams GB, Weibrecht KW, et al. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature. 2003;425(6960):841‐846. [DOI] [PubMed] [Google Scholar]
  • 30. Ding L, Morrison SJ. Haematopoietic stem cells and early lymphoid progenitors occupy distinct bone marrow niches. Nature. 2013;495(7440):231‐235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Zhou BO, Yue R, Murphy MM, Peyer JG, Morrison SJ. Leptin‐receptor‐expressing mesenchymal stromal cells represent the main source of bone formed by adult bone marrow. Cell Stem Cell. 2014;15(2):154‐168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Benjamin RM. Bone health: preventing osteoporosis. Public Health Rep. 2010;125(3):368‐370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Pishavar E, Luo H, Naserifar M, et al. Advanced hydrogels as exosome delivery systems for osteogenic differentiation of MSCs: application in bone regeneration. Int J Mol Sci. 2021;22(12):6203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Liu Z, Yang S, Li X, et al. Local transplantation of GMSC‐derived exosomes to promote vascularized diabetic wound healing by regulating the Wnt/beta‐catenin pathways. Nanoscale Adv. 2023;5(3):916‐926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Peulen TO, Wilkinson KJ. Diffusion of nanoparticles in a biofilm. Environ Sci Technol. 2011;45(8):3367‐3373. [DOI] [PubMed] [Google Scholar]
  • 36. Imam RA, Rizk AA. Efficacy of erythropoietin‐pretreated mesenchymal stem cells in murine burn wound healing: possible in vivo transdifferentiation into keratinocytes. Folia Morphol (Warsz). 2019;78(4):798‐808. [DOI] [PubMed] [Google Scholar]
  • 37. Sandhiya S, Dkhar SA, Surendiran A. Emerging trends of nanomedicine–an overview. Fundam Clin Pharmacol. 2009;23(3):263‐269. [DOI] [PubMed] [Google Scholar]
  • 38. Datta S, Huntosova V, Jutkova A, et al. Influence of hydrophobic side‐chain length in amphiphilic gradient copoly(2‐oxazoline)s on the therapeutics loading, stability, cellular uptake and pharmacokinetics of nano‐formulation with curcumin. Pharmaceutics. 2022;14(12):2576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Mir M, Ali MN, Barakullah A, et al. Synthetic polymeric biomaterials for wound healing: a review. Prog Biomater. 2018;7(1):1‐21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Pinky GuptaS, Krishnakumar V, Sharma Y, Dinda AK, Mohanty S. Mesenchymal stem cell derived exosomes: a nano platform for therapeutics and drug delivery in combating COVID‐19. Stem Cell Rev Rep. 2021;17(1):33‐43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Agrawal A, Joshi A, Bhattacharya S. Recent excavation of nanoethosomes in current drug delivery. Curr Drug Deliv. 2022. [DOI] [PubMed] [Google Scholar]
  • 42. Losi P, Briganti E, Magera A, et al. Tissue response to poly(ether)urethane‐polydimethylsiloxane‐fibrin composite scaffolds for controlled delivery of pro‐angiogenic growth factors. Biomaterials. 2010;31(20):5336‐5344. [DOI] [PubMed] [Google Scholar]
  • 43. Kang M, Lee CS, Lee M. Bioactive scaffolds integrated with liposomal or extracellular vesicles for bone regeneration. Bioengineering (Basel). 2021;8(10):137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Kanda N, Anada T, Handa T, et al. Orthotopic osteogenecity enhanced by a porous gelatin sponge in a critical‐sized rat calvaria defect. Macromol Biosci. 2015;15(12):1647‐1655. [DOI] [PubMed] [Google Scholar]
  • 45. Jahani M, Rezazadeh D, Mohammadi P, Abdolmaleki A, Norooznezhad A, Mansouri K. Regenerative medicine and angiogenesis; challenges and opportunities. Adv Pharm Bull. 2020;10(4):490‐501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Shaw N, Erickson C, Bryant SJ, et al. Regenerative medicine approaches for the treatment of pediatric physeal injuries. Tissue Eng Part B Rev. 2018;24(2):85‐97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Kirana S, Stratmann B, Prante C, et al. Autologous stem cell therapy in the treatment of limb ischaemia induced chronic tissue ulcers of diabetic foot patients. Int J Clin Pract. 2012;66(4):384‐393. [DOI] [PubMed] [Google Scholar]
  • 48. Laloze J, Fievet L, Desmouliere A. Adipose‐derived mesenchymal stromal cells in regenerative medicine: state of play, current clinical trials, and future prospects. Adv Wound Care (New Rochelle). 2021;10(1):24‐48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Eraydin S. Authors' response to a letter to the editor re: the effect of foot exercises on wound healing in type 2 diabetic patients with a foot ulcer: a randomized control study. J Wound Ostomy Continence Nurs. 2018;45(2):123‐130. J Wound Ostomy Continence Nurs. 2018;45(6):495. [DOI] [PubMed] [Google Scholar]
  • 50. De D, Karmakar P, Bhattacharya D. Stem cell aging and regenerative medicine. Adv Exp Med Biol. 2021;1326:11‐37. [DOI] [PubMed] [Google Scholar]
  • 51. Suman S, Domingues A, Ratajczak J, Ratajczak MZ. Potential clinical applications of stem cells in regenerative medicine. Adv Exp Med Biol. 2019;1201:1‐22. [DOI] [PubMed] [Google Scholar]
  • 52. Liu SP, Fu RH, Huang SJ, et al. Stem cell applications in regenerative medicine for neurological disorders. Cell Transplant. 2013;22(4):631‐637. [DOI] [PubMed] [Google Scholar]
  • 53. Snyder EY. The state of the art in stem cell biology and regenerative medicine: the end of the beginning. Pediatr Res. 2018;83(1‐2):191‐204. [DOI] [PubMed] [Google Scholar]
  • 54. Zhang CP, Fu XB. Therapeutic potential of stem cells in skin repair and regeneration. Chin J Traumatol. 2008;11(4):209‐221. [DOI] [PubMed] [Google Scholar]
  • 55. Corsi KA, Pollett JB, Phillippi JA, Usas A, Li G, Huard J. Osteogenic potential of postnatal skeletal muscle‐derived stem cells is influenced by donor sex. J Bone Miner Res. 2007;22(10):1592‐1602. [DOI] [PubMed] [Google Scholar]
  • 56. Li W, Li L, Cui R, Chen X, Hu H, Qiu Y. Bone marrow mesenchymal stem cells derived exosomal Lnc TUG1 promotes bone fracture recovery via miR‐22‐5p/Anxa8 axis. Hum Cell. 2023;36(3):1041‐1053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Wang X, Yu F, Ye L. Epigenetic control of mesenchymal stem cells orchestrates bone regeneration. Front Endocrinol (Lausanne). 2023;14:1126787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Li Y, Liu D, Tan F, Yin W, Li Z. Umbilical cord derived mesenchymal stem cell‐GelMA microspheres for accelerated wound healing. Biomed Mater. 2022;18(1):015019. [DOI] [PubMed] [Google Scholar]
  • 59. Tamaki T, Hirata M, Nakajima N, et al. A long‐gap peripheral nerve injury therapy using human skeletal muscle‐derived stem cells (Sk‐SCs): an achievement of significant morphological, numerical and functional recovery. PLoS One. 2016;11(11):e0166639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Lazutkin A, Podgorny O, Enikolopov G. Modes of division and differentiation of neural stem cells. Behav Brain Res. 2019;374:112118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Ferro F, Spelat R, Baheney CS. Dental pulp stem cell (DPSC) isolation, characterization, and differentiation. Methods Mol Biol. 2014;1210:91‐115. [DOI] [PubMed] [Google Scholar]
  • 62. Chen Y, Huang H, Li G, Yu J, Fang F, Qiu W. Dental‐derived mesenchymal stem cell sheets: a prospective tissue engineering for regenerative medicine. Stem Cell Res Ther. 2022;13(1):38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Yin X, Li Q, McNutt PM, Zhang Y. Urine‐derived stem cells for epithelial tissues reconstruction and wound healing. Pharmaceutics. 2022;14(8):1669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Bento G, Shafigullina AK, Rizvanov AA, Sardao VA, Macedo MP, Oliveira PJ. Urine‐derived stem cells: applications in regenerative and predictive medicine. Cells. 2020;9(3):573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Oh EJ, Lee HW, Kalimuthu S, et al. In vivo migration of mesenchymal stem cells to burn injury sites and their therapeutic effects in a living mouse model. J Control Release. 2018;279:79‐88. [DOI] [PubMed] [Google Scholar]
  • 66. Zahorec P, Sarkozyova N, Ferancikova N, et al. Autologous mesenchymal stem cells application in post‐burn scars treatment: a preliminary study. Cell Tissue Bank. 2021;22(1):39‐46. [DOI] [PubMed] [Google Scholar]
  • 67. Nourian Dehkordi A, Mirahmadi Babaheydari F, Chehelgerdi M, Raeisi Dehkordi S. Skin tissue engineering: wound healing based on stem‐cell‐based therapeutic strategies. Stem Cell Res Ther. 2019;10(1):111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Tsai HW, Wang PH, Tsui KH. Mesenchymal stem cell in wound healing and regeneration. J Chin Med Assoc. 2018;81(3):223‐224. [DOI] [PubMed] [Google Scholar]
  • 69. Jin J, Zhu KS, Tang SM, et al. Trehalose promotes functional recovery of keratinocytes under oxidative stress and wound healing via ATG5/ATG7. Burns. 2022. [DOI] [PubMed] [Google Scholar]
  • 70. He ML, Dong X, Zuo LL, Niu YY, Wang HY. Effects of sericin and egg white on the inflammation of damaged skin in mice. Biomed Mater. 2023;18(2):025013. [DOI] [PubMed] [Google Scholar]
  • 71. Turner CT, Zeglinski MR, Boivin W, et al. Granzyme K contributes to endothelial microvascular damage and leakage during skin inflammation. Br J Dermatol. 2022. [DOI] [PubMed] [Google Scholar]
  • 72. Ueyama H, Okano T, Orita K, et al. Local transplantation of adipose‐derived stem cells has a significant therapeutic effect in a mouse model of rheumatoid arthritis. Sci Rep. 2020;10(1):3076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Ghosh T, Nandi P, Ganguly N, et al. NLGP counterbalances the immunosuppressive effect of tumor‐associated mesenchymal stem cells to restore effector T cell functions. Stem Cell Res Ther. 2019;10(1):296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Kong P, Xie X, Li F, Liu Y, Lu Y. Placenta mesenchymal stem cell accelerates wound healing by enhancing angiogenesis in diabetic Goto‐Kakizaki (GK) rats. Biochem Biophys Res Commun. 2013;438(2):410‐419. [DOI] [PubMed] [Google Scholar]
  • 75. Zhang S, Chen L, Zhang G, Zhang B. Umbilical cord‐matrix stem cells induce the functional restoration of vascular endothelial cells and enhance skin wound healing in diabetic mice via the polarized macrophages. Stem Cell Res Ther. 2020;11(1):39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Luz‐Crawford P, Djouad F, Toupet K, et al. Mesenchymal stem cell‐derived interleukin 1 receptor antagonist promotes macrophage polarization and inhibits B cell differentiation. Stem Cells. 2016;34(2):483‐492. [DOI] [PubMed] [Google Scholar]
  • 77. Francois S, Mouiseddine M, Allenet‐Lepage B, et al. Human mesenchymal stem cells provide protection against radiation‐induced liver injury by antioxidative process, vasculature protection, hepatocyte differentiation, and trophic effects. Biomed Res Int. 2013;2013:151679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Pawitan JA. Prospect of stem cell conditioned medium in regenerative medicine. Biomed Res Int. 2014;2014:965849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Li JY, Ren KK, Zhang WJ, et al. Human amniotic mesenchymal stem cells and their paracrine factors promote wound healing by inhibiting heat stress‐induced skin cell apoptosis and enhancing their proliferation through activating PI3K/AKT signaling pathway. Stem Cell Res Ther. 2019;10(1):247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Gnecchi M, Zhang Z, Ni A, Dzau VJ. Paracrine mechanisms in adult stem cell signaling and therapy. Circ Res. 2008;103(11):1204‐1219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Zhao FY, Cheng TY, Yang L, et al. G‐CSF inhibits pulmonary fibrosis by promoting BMSC homing to the lungs via SDF‐1/CXCR4 chemotaxis. Sci Rep. 2020;10(1):10515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Ellis S, Lin EJ, Tartar D. Immunology of wound healing. Curr Dermatol Rep. 2018;7(4):350‐358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Selvasandran K, Makhoul G, Jaiswal PK, et al. A tumor necrosis factor‐alpha and hypoxia‐induced secretome therapy for myocardial repair. Ann Thorac Surg. 2018;105(3):715‐723. [DOI] [PubMed] [Google Scholar]
  • 84. Smith AN, Willis E, Chan VT, et al. Mesenchymal stem cells induce dermal fibroblast responses to injury. Exp Cell Res. 2010;316(1):48‐54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. 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 Res Ther. 2021;12(1):58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Mohajer Ansari J, Ramhormozi P, Shabani R, et al. Simvastatin combined with bone marrow mesenchymal stromal cells (BMSCs) improve burn wound healing by ameliorating angiogenesis through SDF‐1alpha/CXCR4 pathway. Iran J Basic Med Sci. 2020;23(6):751‐759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Forte G, Minieri M, Cossa P, et al. Hepatocyte growth factor effects on mesenchymal stem cells: proliferation, migration, and differentiation. Stem Cells. 2006;24(1):23‐33. [DOI] [PubMed] [Google Scholar]
  • 88. Zuk PA, Zhu M, Ashjian P, et al. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 2002;13(12):4279‐4295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Kaku M, Tai M, Kawata T, et al. Mesenchymal stem cell‐induced cranial suture‐like gap in rats. Plast Reconstr Surg. 2011;127(1):69‐77. [DOI] [PubMed] [Google Scholar]
  • 90. Shen X, Yu Y, Ma P, et al. Titania nanotubes promote osteogenesis via mediating crosstalk between macrophages and MSCs under oxidative stress. Colloids Surf B Biointerfaces. 2019;180:39‐48. [DOI] [PubMed] [Google Scholar]
  • 91. Lang E, Semon JA. Mesenchymal stem cells in the treatment of osteogenesis imperfecta. Cell Regen. 2023;12(1):7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Zhang J, Chen J. Bone tissue regeneration ‐ application of mesenchymal stem cells and cellular and molecular mechanisms. Curr Stem Cell Res Ther. 2017;12(5):357‐364. [DOI] [PubMed] [Google Scholar]
  • 93. Tonnarelli B, Centola M, Barbero A, Zeller R, Martin I. Re‐engineering development to instruct tissue regeneration. Curr Top Dev Biol. 2014;108:319‐338. [DOI] [PubMed] [Google Scholar]
  • 94. Haffner‐Luntzer M, Weber B, Morioka K, et al. Altered early immune response after fracture and traumatic brain injury. Front Immunol. 2023;14:1074207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Xu QC, Wang ZG, Ji QX, et al. Systemically transplanted human gingiva‐derived mesenchymal stem cells contributing to bone tissue regeneration. Int J Clin Exp Pathol. 2014;7(8):4922‐4929. [PMC free article] [PubMed] [Google Scholar]
  • 96. Walmsley GG, Ransom RC, Zielins ER, et al. Stem cells in bone regeneration. Stem Cell Rev Rep. 2016;12(5):524‐529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Adamiak M, Sahoo S. Exosomes in myocardial repair: advances and challenges in the development of next‐generation therapeutics. Mol Ther. 2018;26(7):1635‐1643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Muthu S, Patil SC, Jeyaraman N, et al. Comparative effectiveness of adipose‐derived mesenchymal stromal cells in the management of knee osteoarthritis: a meta‐analysis. World J Orthop. 2023;14(1):23‐41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Huang B, Qian J, Ma J, et al. Myocardial transfection of hypoxia‐inducible factor‐1alpha and co‐transplantation of mesenchymal stem cells enhance cardiac repair in rats with experimental myocardial infarction. Stem Cell Res Ther. 2014;5(1):22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Folestad E, Kunath A, Wagsater D. PDGF‐C and PDGF‐D signaling in vascular diseases and animal models. Mol Aspects Med. 2018;62:1‐11. [DOI] [PubMed] [Google Scholar]
  • 101. Zhang SJ, Song XY, He M, Yu SB. Effect of TGF‐beta1/SDF‐1/CXCR4 signal on BM‐MSCs homing in rat heart of ischemia/perfusion injury. Eur Rev Med Pharmacol Sci. 2016;20(5):899‐905. [PubMed] [Google Scholar]
  • 102. Gnecchi M, Danieli P, Malpasso G, Ciuffreda MC. Paracrine mechanisms of mesenchymal stem cells in tissue repair. Methods Mol Biol. 2016;1416:123‐146. [DOI] [PubMed] [Google Scholar]
  • 103. Ma M, Cui G, Liu Y, et al. Mesenchymal stem cell‐derived extracellular vesicles, osteoimmunology and orthopedic diseases. PeerJ. 2023;11:e14677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Peng J, Xiao S, Xie J, Fu W. Bulleyaconitine A reduces fracture‐induced pain and promotes fracture healing in mice. Front Pharmacol. 2023;14:1046514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Martinez FO, Gordon S. The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep. 2014;6:13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Kim HK, Lee SG, Lee SW, et al. A subset of paracrine factors as efficient biomarkers for predicting vascular regenerative efficacy of mesenchymal stromal/stem cells. Stem Cells. 2019;37(1):77‐88. [DOI] [PubMed] [Google Scholar]
  • 107. Nakamura Y, Ishikawa H, Kawai K, Tabata Y, Suzuki S. Enhanced wound healing by topical administration of mesenchymal stem cells transfected with stromal cell‐derived factor‐1. Biomaterials. 2013;34(37):9393‐9400. [DOI] [PubMed] [Google Scholar]
  • 108. Granero‐Molto F, Weis JA, Miga MI, et al. Regenerative effects of transplanted mesenchymal stem cells in fracture healing. Stem Cells. 2009;27(8):1887‐1898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Batoon L, Millard SM, Raggatt LJ, Pettit AR. Osteomacs and bone regeneration. Curr Osteoporos Rep. 2017;15(4):385‐395. [DOI] [PubMed] [Google Scholar]
  • 110. Bastian O, Pillay J, Alblas J, Leenen L, Koenderman L, Blokhuis T. Systemic inflammation and fracture healing. J Leukoc Biol. 2011;89(5):669‐673. [DOI] [PubMed] [Google Scholar]
  • 111. Wu AC, Raggatt LJ, Alexander KA, Pettit AR. Unraveling macrophage contributions to bone repair. Bonekey Rep. 2013;2:373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat Rev Immunol. 2008;8(12):958‐969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Mahon OR, Browe DC, Gonzalez‐Fernandez T, et al. Nano‐particle mediated M2 macrophage polarization enhances bone formation and MSC osteogenesis in an IL‐10 dependent manner. Biomaterials. 2020;239:119833. [DOI] [PubMed] [Google Scholar]
  • 114. Li T, Liu ZL, Xiao M, et al. Impact of bone marrow mesenchymal stem cell immunomodulation on the osteogenic effects of laponite. Stem Cell Res Ther. 2018;9(1):100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Liu C, Sun J. Osteogenically differentiated mesenchymal stem cells induced by hydrolyzed fish collagen maintain their immunomodulatory effects. Life Sci. 2019;238:116970. [DOI] [PubMed] [Google Scholar]
  • 116. Zhang HJ, Li FS, Wang F, et al. Transgenic PDGF‐BB sericin hydrogel potentiates bone regeneration of BMP9‐stimulated mesenchymal stem cells through a crosstalk of the Smad‐STAT pathways. Regen Biomater. 2023;10:rbac095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Chappell AG, Ramsey MD, Dabestani PJ, Ko JH. Vascularized bone graft reconstruction for upper extremity defects: a review. Arch Plast Surg. 2023;50(1):82‐95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Rui YF, Lui PP, Lee YW, Chan KM. Higher BMP receptor expression and BMP‐2‐induced osteogenic differentiation in tendon‐derived stem cells compared with bone‐marrow‐derived mesenchymal stem cells. Int Orthop. 2012;36(5):1099‐1107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Wang X, Wang Y, Gou W, Lu Q, Peng J, Lu S. Role of mesenchymal stem cells in bone regeneration and fracture repair: a review. Int Orthop. 2013;37(12):2491‐2498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Liao S, Yue W, Cai S, et al. Improvement of gold nanorods in photothermal therapy: recent progress and perspective. Front Pharmacol. 2021;12:664123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Pearson HB, Mason DE, Kegelman CD, et al. Effects of bone morphogenetic protein‐2 on neovascularization during large bone defect regeneration. Tissue Eng Part A. 2019;25(23‐24):1623‐1634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Kitaura H, Kimura K, Ishida M, Kohara H, Yoshimatsu M, Takano‐Yamamoto T. Immunological reaction in TNF‐alpha‐mediated osteoclast formation and bone resorption in vitro and in vivo. Clin Dev Immunol. 2013;2013:181849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Yu X, Sun H, Yang J, et al. Evaluation of bone‐regeneration effects and ectopic osteogenesis of collagen membrane chemically conjugated with stromal cell‐derived factor‐1 in vivo. Biomed Mater. 2019;15(1):015009. [DOI] [PubMed] [Google Scholar]
  • 124. Wang Y, Pan J, Wang D, Liu J. The use of stem cells in neural regeneration: a review of current opinion. Curr Stem Cell Res Ther. 2018;13(7):608‐617. [DOI] [PubMed] [Google Scholar]
  • 125. Li Y, Kamei Y, Kambe M, Ebisawa K, Oishi M, Takanari K. Peripheral nerve regeneration using different germ layer‐derived adult stem cells in the past decade. Behav Neurol. 2021;2021:5586523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Maki D, Tamaki T, Fukuzawa T, et al. Peripheral nerve regeneration using a cytokine cocktail secreted by skeletal muscle‐derived stem cells in a mouse model. J Clin Med. 2021;10(4):824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Yang C, Li X, Sun L, Guo W, Tian W. Potential of human dental stem cells in repairing the complete transection of rat spinal cord. J Neural Eng. 2017;14(2):026005. [DOI] [PubMed] [Google Scholar]
  • 128. Yoon Y, Kim HS, Jeon I, et al. Implantation of the clinical‐grade human neural stem cell line, CTX0E03, rescues the behavioral and pathological deficits in the quinolinic acid‐lesioned rodent model of Huntington's disease. Stem Cells. 2020;38(8):936‐947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Yang Z, Yang Y, Xu Y, et al. Biomimetic nerve guidance conduit containing engineered exosomes of adipose‐derived stem cells promotes peripheral nerve regeneration. Stem Cell Res Ther. 2021;12(1):442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Sinden JD, Hicks C, Stroemer P, Vishnubhatla I, Corteling R. Human neural stem cell therapy for chronic ischemic stroke: charting progress from laboratory to patients. Stem Cells Dev. 2017;26(13):933‐947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Wang D, Wang Y, Tian W, Pan J. Advances of tooth‐derived stem cells in neural diseases treatments and nerve tissue regeneration. Cell Prolif. 2019;52(3):e12572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Vojnits K, Pan H, Dai X, et al. Functional neuronal differentiation of injury‐induced muscle‐derived stem cell‐like cells with therapeutic implications. Sci Rep. 2017;7(1):1177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Wang F, Jia Y, Liu J, et al. Dental pulp stem cells promote regeneration of damaged neuron cells on the cellular model of Alzheimer's disease. Cell Biol Int. 2017;41(6):639‐650. [DOI] [PubMed] [Google Scholar]
  • 134. Li B, Hu W, Ma K, Zhang C, Fu X. Are hair follicle stem cells promising candidates for wound healing? Expert Opin Biol Ther. 2019;19(2):119‐128. [DOI] [PubMed] [Google Scholar]
  • 135. Yuan AR, Bian Q, Gao JQ. Current advances in stem cell‐based therapies for hair regeneration. Eur J Pharmacol. 2020;881:173197. [DOI] [PubMed] [Google Scholar]
  • 136. Wang X, Chen H, Tian R, et al. Macrophages induce AKT/beta‐catenin‐dependent Lgr5(+) stem cell activation and hair follicle regeneration through TNF. Nat Commun. 2017;8:14091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Lee SA, Li KN, Tumbar T. Stem cell‐intrinsic mechanisms regulating adult hair follicle homeostasis. Exp Dermatol. 2021;30(4):430‐447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Chueh SC, Lin SJ, Chen CC, et al. Therapeutic strategy for hair regeneration: hair cycle activation, niche environment modulation, wound‐induced follicle neogenesis, and stem cell engineering. Expert Opin Biol Ther. 2013;13(3):377‐391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Bu ZY, Wu LM, Yu XH, Zhong JB, Yang P, Chen J. Isolation and characterization of in vitro culture of hair follicle cells differentiated from umbilical cord blood mesenchymal stem cells. Exp Ther Med. 2017;14(1):303‐307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Fukuoka H, Narita K, Suga H. Hair regeneration therapy: application of adipose‐derived stem cells. Curr Stem Cell Res Ther. 2017;12(7):531‐534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Lee J, Böscke R, Tang PC, Hartman BH, Heller S, Koehler KR. Hair follicle development in mouse pluripotent stem cell‐derived skin organoids. Cell Rep. 2018;22(1):242‐254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Fu X, Liu G, Halim A, Ju Y, Luo Q, Song AG. Mesenchymal stem cell migration and tissue repair. Cells. 2019;8(8):784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Hirsch T, Rothoeft T, Teig N, et al. Regeneration of the entire human epidermis using transgenic stem cells. Nature. 2017;551(7680):327‐332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Bahraminasab M, Janmohammadi M, Arab S, et al. Bone scaffolds: an incorporation of biomaterials, cells, and biofactors. ACS Biomater Sci Eng. 2021;7(12):5397‐5431. [DOI] [PubMed] [Google Scholar]
  • 145. Liang X, Ding Y, Zhang Y, Tse HF, Lian Q. Paracrine mechanisms of mesenchymal stem cell‐based therapy: current status and perspectives. Cell Transplant. 2014;23(9):1045‐1059. [DOI] [PubMed] [Google Scholar]
  • 146. Sarasua JG, Lopez SP, Viejo MA, et al. Treatment of pressure ulcers with autologous bone marrow nuclear cells in patients with spinal cord injury. J Spinal Cord Med. 2011;34(3):301‐307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. An T, Chen Y, Tu Y, Lin P. Mesenchymal stromal cell‐derived extracellular vesicles in the treatment of diabetic foot ulcers: application and challenges. Stem Cell Rev Rep. 2021;17(2):369‐378. [DOI] [PubMed] [Google Scholar]
  • 148. Anderson S, Das H. Cutaneous wound generation in diabetic NOD/SCID mice and the use of nanofiber‐expanded hematopoietic stem cell therapy. Methods Mol Biol. 2021;2193:41‐48. [DOI] [PubMed] [Google Scholar]
  • 149. Zhang H, Xue F, Jun Xiao H. Ilizarov method in combination with autologous mesenchymal stem cells from iliac crest shows improved outcome in tibial non‐union. Saudi J Biol Sci. 2018;25(4):819‐825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Zhao SJ, Kong FQ, Jie J, et al. Macrophage MSR1 promotes BMSC osteogenic differentiation and M2‐like polarization by activating PI3K/AKT/GSK3beta/beta‐catenin pathway. Theranostics. 2020;10(1):17‐35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Sansone V, Branes M, Romeo P. A novel bimodal approach for treating atrophic bone non‐unions with extracorporeal shockwaves and autologous mesenchymal stem cell transplant. Med Hypotheses. 2018;111:4‐7. [DOI] [PubMed] [Google Scholar]
  • 152. Schlundt C, Bucher CH, Tsitsilonis S, Schell H, Duda GN, Schmidt‐Bleek K. Clinical and research approaches to treat non‐union fracture. Curr Osteoporos Rep. 2018;16(2):155‐168. [DOI] [PubMed] [Google Scholar]
  • 153. Pan BT, Johnstone RM. Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: selective externalization of the receptor. Cell. 1983;33(3):967‐978. [DOI] [PubMed] [Google Scholar]
  • 154. Johnstone RM, Adam M, Hammond JR, Orr L, Turbide C. Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J Biol Chem. 1987;262(19):9412‐9420. [PubMed] [Google Scholar]
  • 155. Kalluri R. The biology and function of exosomes in cancer. J Clin Invest. 2016;126(4):1208‐1215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013;200(4):373‐383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Borges FT, Melo SA, Ozdemir BC, et al. TGF‐beta1‐containing exosomes from injured epithelial cells activate fibroblasts to initiate tissue regenerative responses and fibrosis. J Am Soc Nephrol. 2013;24(3):385‐392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Skokos D, Botros HG, Demeure C, et al. Mast cell‐derived exosomes induce phenotypic and functional maturation of dendritic cells and elicit specific immune responses in vivo. J Immunol. 2003;170(6):3037‐3045. [DOI] [PubMed] [Google Scholar]
  • 159. Lamichhane TN, Sokic S, Schardt JS, Raiker RS, Lin JW, Jay SM. Emerging roles for extracellular vesicles in tissue engineering and regenerative medicine. Tissue Eng Part B Rev. 2015;21(1):45‐54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Hade MD, Suire CN, Suo Z. Mesenchymal stem cell‐derived exosomes: applications in regenerative medicine. Cells. 2021;10(8):1959. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. Shabbir A, Cox A, Rodriguez‐Menocal L, Salgado M, Van Badiavas E. Mesenchymal stem cell exosomes induce proliferation and migration of normal and chronic wound fibroblasts, and enhance angiogenesis in vitro. Stem Cells Dev. 2015;24(14):1635‐1647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. de Souza W, Gemini‐Piperni S, Grenho L, et al. Titanium dioxide nanoparticles affect osteoblast‐derived exosome cargos and impair osteogenic differentiation of human mesenchymal stem cells. Biomater Sci. 2023;11(7):2427‐2444. [DOI] [PubMed] [Google Scholar]
  • 163. Harrell CR, Jovicic N, Djonov V, Arsenijevic N, Volarevic V. Mesenchymal stem cell‐derived exosomes and other extracellular vesicles as new remedies in the therapy of inflammatory diseases. Cells. 2019;8(12):1605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. Zhu B, Zhang L, Liang C, et al. Stem cell‐derived exosomes prevent aging‐induced cardiac dysfunction through a novel exosome/lncRNA MALAT1/NF‐kappaB/TNF‐alpha signaling pathway. Oxid Med Cell Longev. 2019;2019:9739258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Yu S, Zhou Y, Niu L, Qiao Y, Yan Y. Mesenchymal stem cell‐derived exosome mir‐342‐3p inhibits metastasis and chemo‐resistance of breast cancer through regulating ID4. Genes Genomics. 2022;44(5):539‐550. [DOI] [PubMed] [Google Scholar]
  • 166. Hong P, Yang H, Wu Y, Li K, Tang Z. The functions and clinical application potential of exosomes derived from adipose mesenchymal stem cells: a comprehensive review. Stem Cell Res Ther. 2019;10(1):242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167. Toh WS, Lai RC, Zhang B, Lim SK. MSC exosome works through a protein‐based mechanism of action. Biochem Soc Trans. 2018;46(4):843‐853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Nikfarjam S, Rezaie J, Zolbanin NM, Jafari R. Mesenchymal stem cell derived‐exosomes: a modern approach in translational medicine. J Transl Med. 2020;18(1):449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Ding J, Wang X, Chen B, Zhang J, Xu J. Exosomes derived from human bone marrow mesenchymal stem cells stimulated by deferoxamine accelerate cutaneous wound healing by promoting angiogenesis. Biomed Res Int. 2019;2019:9742765. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 170. Casado‐Diaz A, Quesada‐Gomez JM, Dorado G. Extracellular vesicles derived from mesenchymal stem cells (MSC) in regenerative medicine: applications in skin wound healing. Front Bioeng Biotechnol. 2020;8:146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171. Ha DH, Kim HK, Lee J, et al. Mesenchymal stem/stromal cell‐derived exosomes for immunomodulatory therapeutics and skin regeneration. Cells. 2020;9(5):1157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172. Kose O, Botsali A, Caliskan E. Role of exosomes in skin diseases. J Cosmet Dermatol. 2022;21(8):3219‐3225. [DOI] [PubMed] [Google Scholar]
  • 173. Liu W, Yu M, Xie D, et al. Melatonin‐stimulated MSC‐derived exosomes improve diabetic wound healing through regulating macrophage M1 and M2 polarization by targeting the PTEN/AKT pathway. Stem Cell Res Ther. 2020;11(1):259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174. Li X, Liu L, Yang J, et al. Exosome derived from human umbilical cord mesenchymal stem cell mediates mir‐181c attenuating burn‐induced excessive inflammation. EBioMedicine. 2016;8:72‐82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Yang C, Luo L, Bai X, et al. Highly‐expressed micoRNA‐21 in adipose derived stem cell exosomes can enhance the migration and proliferation of the HaCaT cells by increasing the MMP‐9 expression through the PI3K/AKT pathway. Arch Biochem Biophys. 2020;681:108259. [DOI] [PubMed] [Google Scholar]
  • 176. He L, Zhu C, Jia J, et al. ADSC‐Exos containing MALAT1 promotes wound healing by targeting miR‐124 through activating Wnt/beta‐catenin pathway. Biosci Rep. 2020;40(5):BSR20192549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Medhat D, Rodriguez CI, Infante A. Immunomodulatory effects of MSCs in Bone Healing. Int J Mol Sci. 2019;20(21):5467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Su N, Hao Y, Wang F, Hou W, Chen H, Luo Y. Mesenchymal stromal exosome‐functionalized scaffolds induce innate and adaptive immunomodulatory responses toward tissue repair. Sci Adv. 2021;7(20):eabf7207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179. Guillamat‐Prats R. The role of MSC in wound healing, scarring and regeneration. Cells. 2021;10(7):1729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. An Y, Lin S, Tan X, et al. Exosomes from adipose‐derived stem cells and application to skin wound healing. Cell Prolif. 2021;54(3):e12993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181. 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. Int J Nanomedicine. 2020;15:5911‐5926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182. Liu J, Yan Z, Yang F, et al. Exosomes derived from human umbilical cord mesenchymal stem cells accelerate cutaneous wound healing by enhancing angiogenesis through delivering angiopoietin‐2. Stem Cell Rev Rep. 2021;17(2):305‐317. [DOI] [PubMed] [Google Scholar]
  • 183. Wu M, Ben Amar M. Growth and remodelling for profound circular wounds in skin. Biomech Model Mechanobiol. 2015;14(2):357‐370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184. Jiang L, Zhang Y, Liu T, et al. Exosomes derived from TSG‐6 modified mesenchymal stromal cells attenuate scar formation during wound healing. Biochimie. 2020;177:40‐49. [DOI] [PubMed] [Google Scholar]
  • 185. Shi Y, Yang Y, Guo Q, et al. Exosomes derived from human umbilical cord mesenchymal stem cells promote fibroblast‐to‐myofibroblast differentiation in inflammatory environments and benefit cardioprotective effects. Stem Cells Dev. 2019;28(12):799‐811. [DOI] [PubMed] [Google Scholar]
  • 186. Vu NB, Nguyen HT, Palumbo R, Pellicano R, Fagoonee S, Pham PV. Stem cell‐derived exosomes for wound healing: current status and promising directions. Minerva Med. 2021;112(3):384‐400. [DOI] [PubMed] [Google Scholar]
  • 187. Qian L, Pi L, Fang BR, Meng XX. Adipose mesenchymal stem cell‐derived exosomes accelerate skin wound healing via the lncRNA H19/miR‐19b/SOX9 axis. Lab Invest. 2021;101(9):1254‐1266. [DOI] [PubMed] [Google Scholar]
  • 188. Zhang Y, Pan Y, Liu Y, et al. Exosomes derived from human umbilical cord blood mesenchymal stem cells stimulate regenerative wound healing via transforming growth factor‐beta receptor inhibition. Stem Cell Res Ther. 2021;12(1):434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189. Diseases GBD, Injuries C. Global burden of 369 diseases and injuries in 204 countries and territories, 1990–2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet. 2020;396(10258):1204‐1222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190. Paiva KBS, Granjeiro JM. Matrix metalloproteinases in bone resorption, remodeling, and repair. Prog Mol Biol Transl Sci. 2017;148:203‐303. [DOI] [PubMed] [Google Scholar]
  • 191. Zeng ZL, Xie H. Mesenchymal stem cell‐derived extracellular vesicles: a possible therapeutic strategy for orthopaedic diseases: a narrative review. Biomater Transl. 2022;3(3):175‐187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192. Wu C, He Y, Yao Y, Yang H, Lu F. Exosomes treating osteoarthritis: hope with challenge. Heliyon. 2023;9(1):e13152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193. Mittelbrunn M, Gutierrez‐Vazquez C, Villarroya‐Beltri C, et al. Unidirectional transfer of microRNA‐loaded exosomes from T cells to antigen‐presenting cells. Nat Commun. 2011;2:282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194. Regev‐Rudzki N, Wilson DW, Carvalho TG, et al. Cell‐cell communication between malaria‐infected red blood cells via exosome‐like vesicles. Cell. 2013;153(5):1120‐1133. [DOI] [PubMed] [Google Scholar]
  • 195. Couzin J. Cell biology: the ins and outs of exosomes. Science. 2005;308(5730):1862‐1863. [DOI] [PubMed] [Google Scholar]
  • 196. Lei F, Li M, Lin T, Zhou H, Wang F, Su X. Treatment of inflammatory bone loss in periodontitis by stem cell‐derived exosomes. Acta Biomater. 2022;141:333‐343. [DOI] [PubMed] [Google Scholar]
  • 197. Aldoghachi AF, Loh JK, Wang ML, et al. Current developments and therapeutic potentials of exosomes from induced pluripotent stem cells (iPSCs)‐derived mesenchymal stem cells (iMSCs). J Chin Med Assoc. 2023;86(4):356‐365. [DOI] [PubMed] [Google Scholar]
  • 198. Shan SK, Lin X, Li F, et al. Exosomes and bone disease. Curr Pharm Des. 2019;25(42):4536‐4549. [DOI] [PubMed] [Google Scholar]
  • 199. Zhang S, Chu WC, Lai RC, Lim SK, Hui JH, Toh WS. Exosomes derived from human embryonic mesenchymal stem cells promote osteochondral regeneration. Osteoarthritis Cartilage. 2016;24(12):2135‐2140. [DOI] [PubMed] [Google Scholar]
  • 200. Wang L, Wang J, Zhou X, et al. A new self‐healing hydrogel containing hucMSC‐derived exosomes promotes bone regeneration. Front Bioeng Biotechnol. 2020;8:564731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201. Dittmer KE, Firth EC. Mechanisms of bone response to injury. J Vet Diagn Invest. 2017;29(4):385‐395. [DOI] [PubMed] [Google Scholar]
  • 202. Einhorn TA, Gerstenfeld LC. Fracture healing: mechanisms and interventions. Nat Rev Rheumatol. 2015;11(1):45‐54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203. Lo Sicco C, Reverberi D, Balbi C, et al. Mesenchymal stem cell‐derived extracellular vesicles as mediators of anti‐inflammatory effects: endorsement of macrophage polarization. Stem Cells Transl Med. 2017;6(3):1018‐1028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204. Zhang S, Chuah SJ, Lai RC, Hui JHP, Lim SK, Toh WS. MSC exosomes mediate cartilage repair by enhancing proliferation, attenuating apoptosis and modulating immune reactivity. Biomaterials. 2018;156:16‐27. [DOI] [PubMed] [Google Scholar]
  • 205. Guo H, Zhang Y, Liang W, et al. An inorganic magnetic fluorescent nanoprobe with favorable biocompatibility for dual‐modality bioimaging and drug delivery. J Inorg Biochem. 2019;192:72‐81. [DOI] [PubMed] [Google Scholar]
  • 206. Takeuchi R, Katagiri W, Endo S, Kobayashi T. Exosomes from conditioned media of bone marrow‐derived mesenchymal stem cells promote bone regeneration by enhancing angiogenesis. PLoS One. 2019;14(11):e0225472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207. Li W, Yang J, Luo L, et al. Targeting photodynamic and photothermal therapy to the endoplasmic reticulum enhances immunogenic cancer cell death. Nat Commun. 2019;10(1):3349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208. Hu H, Zhang H, Bu Z, et al. Small extracellular vesicles released from bioglass/hydrogel scaffold promote vascularized bone regeneration by transferring miR‐23a‐3p. Int J Nanomedicine. 2022;17:6201‐6220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209. Marsell R, Einhorn TA. The biology of fracture healing. Injury. 2011;42(6):551‐555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210. Mazzeo A, Santos EJC. Nanotechnology and multipotent adult progenitor cells in reparative medicine: therapeutic perspectives. Einstein (Sao Paulo). 2018;16(4):eRB4587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211. Wei M, Yang Z, Li S, Le W. Nanotherapeutic and Stem cell therapeutic strategies in neurodegenerative diseases: a promising therapeutic approach. Int J Nanomedicine. 2023;18:611‐626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212. Alexander A, Saraf S, Saraf S, et al. Amalgamation of stem cells with nanotechnology: a unique therapeutic approach. Curr Stem Cell Res Ther. 2019;14(2):83‐92. [DOI] [PubMed] [Google Scholar]
  • 213. Dubey SK, Alexander A, Sivaram M, et al. Uncovering the diversification of tissue engineering on the emergent areas of stem cells, nanotechnology and biomaterials. Curr Stem Cell Res Ther. 2020;15(3):187‐201. [DOI] [PubMed] [Google Scholar]
  • 214. Deb KD, Griffith M, Muinck ED, Rafat M. Nanotechnology in stem cells research: advances and applications. Front Biosci (Landmark Ed). 2012;17(5):1747‐1760. [DOI] [PubMed] [Google Scholar]
  • 215. Nagarajah S. Exosome secretion ‐ more than simple waste disposal? Implications for physiology, diagnostics and therapeutics. J Circ Biomark. 2016;5:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216. Xi XM, Xia SJ, Lu R. Drug loading techniques for exosome‐based drug delivery systems. Pharmazie. 2021;76(2):61‐67. [DOI] [PubMed] [Google Scholar]
  • 217. Familtseva A, Jeremic N, Tyagi SC. Exosomes: cell‐created drug delivery systems. Mol Cell Biochem. 2019;459(1‐2):1‐6. [DOI] [PubMed] [Google Scholar]
  • 218. Hessvik NP, Llorente A. Current knowledge on exosome biogenesis and release. Cell Mol Life Sci. 2018;75(2):193‐208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219. Tang Z, Yin L, Zhang Y, Yu W, Wang Q, Zhan Z. Preparation and study of two kinds of ophthalmic nano‐preparations of everolimus. Drug Deliv. 2019;26(1):1235‐1242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220. Sun Y, Li B, Cao Q, Liu T, Li J. Targeting cancer stem cells with polymer nanoparticles for gastrointestinal cancer treatment. Stem Cell Res Ther. 2022;13(1):489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221. Jayaraman P, Gandhimathi C, Venugopal JR, Becker DL, Ramakrishna S, Srinivasan DK. Controlled release of drugs in electrosprayed nanoparticles for bone tissue engineering. Adv Drug Deliv Rev. 2015;94:77‐95. [DOI] [PubMed] [Google Scholar]
  • 222.Abu Lila AS, Ishida T. Liposomal delivery systems: design optimization and current applications. Biol Pharm Bull. 2017;40(1):1‐10. [DOI] [PubMed] [Google Scholar]
  • 223. Li M, Du C, Guo N, et al. Composition design and medical application of liposomes. Eur J Med Chem. 2019;164:640‐653. [DOI] [PubMed] [Google Scholar]
  • 224. Sharma R, Benwood C, Willerth SM. Drug‐releasing microspheres for stem cell differentiation. Curr Protoc. 2021;1(12):e331. [DOI] [PubMed] [Google Scholar]
  • 225. Li J, Lee WY, Wu T, et al. Near‐infrared light‐triggered release of small molecules for controlled differentiation and long‐term tracking of stem cells in vivo using upconversion nanoparticles. Biomaterials. 2016;110:1‐10. [DOI] [PubMed] [Google Scholar]
  • 226. Tsou YH, Khoneisser J, Huang PC, Xu X. Hydrogel as a bioactive material to regulate stem cell fate. Bioact Mater. 2016;1(1):39‐55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227. Ghosh B, Biswas S. Polymeric micelles in cancer therapy: state of the art. J Control Release. 2021;332:127‐147. [DOI] [PubMed] [Google Scholar]
  • 228. Asgari V, Landarani‐Isfahani A, Salehi H, et al. The story of nanoparticles in differentiation of stem cells into neural cells. Neurochem Res. 2019;44(12):2695‐2707. [DOI] [PubMed] [Google Scholar]
  • 229. Scioli MG, Storti G, D'Amico F, et al. Adipose‐derived stem cells in cancer progression: new perspectives and opportunities. Int J Mol Sci. 2019;20(13):3296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230. Yokoyama R, Ii M, Masuda M, et al. Cardiac regeneration by statin‐polymer nanoparticle‐loaded adipose‐derived stem cell therapy in myocardial infarction. Stem Cells Transl Med. 2019;8(10):1055‐1067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231. Wang W, Lei B, Li L, et al. Single‐cell proteomic profiling identifies nanoparticle enhanced therapy for triple negative breast cancer stem cells. Cells. 2021;10(11):2842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232. Gao Y, Wang K, Zhang J, Duan X, Sun Q, Men K. Multifunctional nanoparticle for cancer therapy. MedComm. 2023;4(1):e187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233. El‐Kharrag R, Berckmueller KE, Madhu R, et al. Efficient polymer nanoparticle‐mediated delivery of gene editing reagents into human hematopoietic stem and progenitor cells. Mol Ther. 2022;30(6):2186‐2198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234. Liu S, Chen X, Bao L, et al. Treatment of infarcted heart tissue via the capture and local delivery of circulating exosomes through antibody‐conjugated magnetic nanoparticles. Nat Biomed Eng. 2020;4(11):1063‐1075. [DOI] [PubMed] [Google Scholar]
  • 235. Wang J, Chen P, Dong Y, et al. Designer exosomes enabling tumor targeted efficient chemo/gene/photothermal therapy. Biomaterials. 2021;276:121056. [DOI] [PubMed] [Google Scholar]
  • 236. Caobi A, Andre M, Miles J, et al. Magnetic nanoparticle and exosomal therapeutic (M‐NEXT) effects on HIV‐associated neurotoxicity. Crit Rev Biomed Eng. 2020;48(3):189‐198. [DOI] [PubMed] [Google Scholar]
  • 237. Wu D, Kang L, Tian J, et al. Exosomes derived from bone mesenchymal stem cells with the stimulation of Fe(3)O(4) nanoparticles and static magnetic field enhance wound healing through upregulated miR‐21‐5p. Int J Nanomedicine. 2020;15:7979‐7993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238. Li Y, Bai Y, Pan J, et al. A hybrid 3D‐printed aspirin‐laden liposome composite scaffold for bone tissue engineering. J Mater Chem B. 2019;7(4):619‐629. [DOI] [PubMed] [Google Scholar]
  • 239. Xie A, Peng Y, Yao Z, Lu L, Ni T. Effect of a subset of adipose‐derived stem cells isolated with liposome magnetic beads to promote cartilage repair. J Cell Mol Med. 2021;25(9):4204‐4215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240. Mandpe P, Prabhakar B, Shende P. Role of liposomes‐based stem cell for multimodal cancer therapy. Stem Cell Rev Rep. 2020;16(1):103‐117. [DOI] [PubMed] [Google Scholar]
  • 241. Li M, Shi F, Fei X, et al. PEGylated long‐circulating liposomes deliver homoharringtonine to suppress multiple myeloma cancer stem cells. Exp Biol Med (Maywood). 2017;242(9):996‐1004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242. Kim YJ, Liu Y, Li S, et al. Co‐eradication of breast cancer cells and cancer stem cells by cross‐linked multilamellar liposomes enhances tumor treatment. Mol Pharm. 2015;12(8):2811‐2822. [DOI] [PubMed] [Google Scholar]
  • 243. Mazza M, Lozano N, Vieira DB, Buggio M, Kielty C, Kostarelos K. Liposome‐indocyanine green nanoprobes for optical labeling and tracking of human mesenchymal stem cells post‐transplantation in vivo. Adv Healthc Mater. 2017;6(21):1700374. [DOI] [PubMed] [Google Scholar]
  • 244. Cheng L, Zhang X, Tang J, Lv Q, Liu J. Gene‐engineered exosomes‐thermosensitive liposomes hybrid nanovesicles by the blockade of CD47 signal for combined photothermal therapy and cancer immunotherapy. Biomaterials. 2021;275:120964. [DOI] [PubMed] [Google Scholar]
  • 245. Lin Y, Wu J, Gu W, et al. Exosome‐liposome hybrid nanoparticles deliver CRISPR/Cas9 system in MSCs. Adv Sci (Weinh). 2018;5(4):1700611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246. Li L, He D, Guo Q, et al. Exosome‐liposome hybrid nanoparticle codelivery of TP and miR497 conspicuously overcomes chemoresistant ovarian cancer. J Nanobiotechnology. 2022;20(1):50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247. Yang S, Xiao X, Huang Z, et al. Human adipose‐derived mesenchymal stem cells‐based microspheres ameliorate atherosclerosis progression in vitro. Stem Cells Dev. 2023. [DOI] [PubMed] [Google Scholar]
  • 248. Mashiko T, Kanayama K, Saito N, et al. Selective proliferation of highly functional adipose‐derived stem cells in microgravity culture with stirred microspheres. Cells. 2021;10(3):560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249. Xu B, Cao Y, Zheng Z, et al. Injectable mesenchymal stem cell‐laden matrigel microspheres for endometrium repair and regeneration. Adv Biol (Weinh). 2021;5(8):e2000202. [DOI] [PubMed] [Google Scholar]
  • 250. Qu M, Liao X, Jiang N, et al. Injectable open‐porous PLGA microspheres as cell carriers for cartilage regeneration. J Biomed Mater Res A. 2021;109(11):2091‐2100. [DOI] [PubMed] [Google Scholar]
  • 251. Thomas RG, Unnithan AR, Moon MJ, et al. Electromagnetic manipulation enabled calcium alginate Janus microsphere for targeted delivery of mesenchymal stem cells. Int J Biol Macromol. 2018;110:465‐471. [DOI] [PubMed] [Google Scholar]
  • 252. Zhao W, Zhang L, Ye Y, et al. Microsphere mediated exosome isolation and ultra‐sensitive detection on a dielectrophoresis integrated microfluidic device. Analyst. 2021;146(19):5962‐5972. [DOI] [PubMed] [Google Scholar]
  • 253. Gao Y, Yuan Z, Yuan X, et al. Bioinspired porous microspheres for sustained hypoxic exosomes release and vascularized bone regeneration. Bioact Mater. 2022;14:377‐388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254. DiStefano TJ, Vaso K, Panebianco CJ, et al. Hydrogel‐embedded poly(lactic‐co‐glycolic acid) microspheres for the delivery of hMSC‐derived exosomes to promote bioactive annulus fibrosus repair. cartil. 2022;13(3):19476035221113959. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255. Swanson WB, Zhang Z, Xiu K, et al. Scaffolds with controlled release of pro‐mineralization exosomes to promote craniofacial bone healing without cell transplantation. Acta Biomater. 2020;118:215‐232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256. Yuan X, Ding L, Deng DYB. Research progress of hydrogel combined with mesenchymal stem cells in the treatment of spinal cord injury. Sheng Wu Yi Xue Gong Cheng Xue Za Zhi. 2021;38(4):805‐811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257. Kim DW, Jeong HS, Kim E, Lee H, Choi CH, Lee SJ. Oral delivery of stem‐cell‐loaded hydrogel microcapsules restores gut inflammation and microbiota. J Control Release. 2022;347:508‐520. [DOI] [PubMed] [Google Scholar]
  • 258. Hamilton M, Harrington S, Dhar P, Stehno‐Bittel L. Hyaluronic acid hydrogel microspheres for slow release stem cell delivery. ACS Biomater Sci Eng. 2021;7(8):3754‐3763. [DOI] [PubMed] [Google Scholar]
  • 259. Caron I, Rossi F, Papa S, et al. A new three dimensional biomimetic hydrogel to deliver factors secreted by human mesenchymal stem cells in spinal cord injury. Biomaterials. 2016;75:135‐147. [DOI] [PubMed] [Google Scholar]
  • 260. Ramzan A, Mehmood A, Ashfaq R, et al. Freeze gelation (chitosan‐elastin‑sodium alginate)/zinc oxide nanocomposite gel with enhanced adipose stem cell proliferation and antibacterial properties. Int J Biol Macromol. 2023;233:123519. [DOI] [PubMed] [Google Scholar]
  • 261. Wang C, Wang M, Xu T, et al. Engineering bioactive self‐healing antibacterial exosomes hydrogel for promoting chronic diabetic wound healing and complete skin regeneration. Theranostics. 2019;9(1):65‐76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262. Li L, Zhang Y, Mu J, et al. Transplantation of human mesenchymal stem‐cell‐derived exosomes immobilized in an adhesive hydrogel for effective treatment of spinal cord injury. Nano Lett. 2020;20(6):4298‐4305. [DOI] [PubMed] [Google Scholar]
  • 263. Zhang K, Zhao X, Chen X, et al. Enhanced therapeutic effects of mesenchymal stem cell‐derived exosomes with an injectable hydrogel for hindlimb ischemia treatment. ACS Appl Mater Interfaces. 2018;10(36):30081‐30091. [DOI] [PubMed] [Google Scholar]
  • 264. Santo VE, Ratanavaraporn J, Sato K, et al. Cell engineering by the internalization of bioinstructive micelles for enhanced bone regeneration. Nanomedicine (Lond). 2015;10(11):1707‐1721. [DOI] [PubMed] [Google Scholar]
  • 265. Xia KS, Li DD, Wang CG, et al. An esterase‐responsive ibuprofen nano‐micelle pre‐modified embryo derived nucleus pulposus progenitor cells promote the regeneration of intervertebral disc degeneration. Bioact Mater. 2023;21:69‐85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266. Quek JY, Bright R, Dabare P, Vasilev K. ROS‐responsive copolymer micelles for inflammation triggered delivery of ibuprofen. Colloids Surf B Biointerfaces. 2022;217:112590. [DOI] [PubMed] [Google Scholar]
  • 267. Zhao Y, Alakhova DY, Zhao X, Band V, Batrakova EV, Kabanov AV. Eradication of cancer stem cells in triple negative breast cancer using doxorubicin/pluronic polymeric micelles. Nanomedicine. 2020;24:102124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268. Zhang J, Peng CA. Enhanced proliferation and differentiation of mesenchymal stem cells by astaxanthin‐encapsulated polymeric micelles. PLoS One. 2019;14(5):e0216755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269. Lin Q, Qu M, Zhou B, et al. Exosome‐like nanoplatform modified with targeting ligand improves anti‐cancer and anti‐inflammation effects of imperialine. J Control Release. 2019;311‐312:104‐116. [DOI] [PubMed] [Google Scholar]
  • 270. Wang XL, Zhao WZ, Fan JZ, et al. Tumor tropic delivery of hyaluronic acid‐poly (D,L‐lactide‐co‐glycolide) polymeric micelles using mesenchymal stem cells for glioma therapy. Molecules. 2022;27(8):2419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271. Shrestha B, Coykendall K, Li Y, Moon A, Priyadarshani P, Yao L. Repair of injured spinal cord using biomaterial scaffolds and stem cells. Stem Cell Res Ther. 2014;5(4):91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272. Hashimoto S, Nagoshi N, Shinozaki M, et al. Microenvironmental modulation in tandem with human stem cell transplantation enhances functional recovery after chronic complete spinal cord injury. Biomaterials. 2023;295:122002. [DOI] [PubMed] [Google Scholar]
  • 273. Costa A, Naranjo JD, Londono R, Badylak SF. Biologic scaffolds. Cold Spring Harb Perspect Med. 2017;7(9):a025676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274. Giuliani C. The flavonoid quercetin induces AP‐1 activation in FRTL‐5 thyroid cells. Antioxidants (Basel). 2019;8(5):112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275. !!! INVALID CITATION !!! 265;.
  • 276. Ullah S, Mansoor S, Ayub A, et al. An update on stem cells applications in burn wound healing. Tissue Cell. 2021;72:101527. [DOI] [PubMed] [Google Scholar]
  • 277. Kang Y, Xu J, Meng L, et al. 3D bioprinting of dECM/Gel/QCS/nHAp hybrid scaffolds laden with mesenchymal stem cell‐derived exosomes to improve angiogenesis and osteogenesis. Biofabrication. 2023;15(2):024103. [DOI] [PubMed] [Google Scholar]
  • 278. Yan Z, Yin H, Wu J, et al. Engineering exosomes by three‐dimensional porous scaffold culture of human umbilical cord mesenchymal stem cells promote osteochondral repair. Mater Today Bio. 2023;19:100549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279. Irvine SA, Venkatraman SS. Bioprinting and differentiation of stem cells. Molecules. 2016;21(9):1188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280. Ahn SH, Lee J, Park SA, Kim WD. Three‐dimensional bio‐printing equipment technologies for tissue engineering and regenerative medicine. Tissue Eng Regen Med. 2016;13(6):663‐676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281. Duska LR, Scalici JM, Temkin SM, et al. Results of an early safety analysis of a study of the combination of pembrolizumab and pelvic chemoradiation in locally advanced cervical cancer. Cancer. 2020;126(22):4948‐4956. [DOI] [PubMed] [Google Scholar]
  • 282. Aran S, Zahri S, Asadi A, Khaksar F, Abdolmaleki A. Hair follicle stem cells differentiation into bone cells on collagen scaffold. Cell Tissue Bank. 2020;21(2):181‐188. [DOI] [PubMed] [Google Scholar]
  • 283. Fu T, Liang P, Song J, et al. Matrigel scaffolding enhances BMP9‐induced bone formation in dental follicle stem/precursor cells. Int J Med Sci. 2019;16(4):567‐575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 284. Agbay A, Edgar JM, Robinson M, et al. Biomaterial strategies for delivering stem cells as a treatment for spinal cord injury. Cells Tissues Organs. 2016;202(1‐2):42‐51. [DOI] [PubMed] [Google Scholar]
  • 285. Fuoco C, Petrilli LL, Cannata S, Gargioli C. Matrix scaffolding for stem cell guidance toward skeletal muscle tissue engineering. J Orthop Surg Res. 2016;11(1):86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286. Vadaye Kheiry E, Fazly Bazzaz BS, Kerachian MA. Implantation of stem cells on synthetic or biological scaffolds: an overview of bone regeneration. Biotechnol Genet Eng Rev. 2021;37(2):238‐268. [DOI] [PubMed] [Google Scholar]
  • 287. Fang J, Li JJ, Zhong X, et al. Engineering stem cell therapeutics for cardiac repair. J Mol Cell Cardiol. 2022;171:56‐68. [DOI] [PubMed] [Google Scholar]
  • 288. Yuan J, Yang H, Liu C, et al. Microneedle patch loaded with exosomes containing microRNA‐29b prevents cardiac fibrosis after myocardial infarction. Adv Healthc Mater. 2023:e2202959. [DOI] [PubMed] [Google Scholar]
  • 289. Yang G, Chen Q, Wen D, et al. A therapeutic microneedle patch made from hair‐derived keratin for promoting hair regrowth. ACS Nano. 2019;13(4):4354‐4360. [DOI] [PubMed] [Google Scholar]
  • 290. Yuan A, Gu Y, Bian Q, et al. Conditioned media‐integrated microneedles for hair regeneration through perifollicular angiogenesis. J Control Release. 2022;350:204‐214. [DOI] [PubMed] [Google Scholar]
  • 291. Han M, Yang H, Lu X, et al. Three‐dimensional‐cultured MSC‐derived exosome‐hydrogel hybrid microneedle array patch for spinal cord repair. Nano Lett. 2022;22(15):6391‐6401. [DOI] [PubMed] [Google Scholar]
  • 292. Ma W, Zhang X, Liu Y, et al. Polydopamine decorated microneedles with Fe‐MSC‐derived nanovesicles encapsulation for wound healing. Adv Sci (Weinh). 2022;9(13):e2103317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 293. Zdunska K, Kolodziejczak A, Rotsztejn H. Is skin microneedling a good alternative method of various skin defects removal. Dermatol Ther. 2018;31(6):e12714. [DOI] [PubMed] [Google Scholar]
  • 294. Lee K, Xue Y, Lee J, et al. A patch of detachable hybrid microneedle depot for localized delivery of mesenchymal stem cells in regeneration therapy. Adv Funct Mater. 2020;30(23):2070143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295. Zhang Z, Li W, Chang D, et al. A combination therapy for androgenic alopecia based on quercetin and zinc/copper dual‐doped mesoporous silica nanocomposite microneedle patch. Bioact Mater. 2023;24:81‐95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296. Shi Y, Zhao J, Li H, et al. A drug‐free, hair follicle cycling regulatable, separable, antibacterial microneedle patch for hair regeneration therapy. Adv Healthc Mater. 2022;11(19):e2200908. [DOI] [PubMed] [Google Scholar]
  • 297. Kittigul L, Meephansan J, Sirithanabadeekul P, et al. The efficacy of LED microneedle patch on hair growth in mice. Arch Dermatol Res. 2023;315(4):971‐982. [DOI] [PubMed] [Google Scholar]
  • 298. Li F, Zhang J, Yi K, et al. Delivery of stem cell secretome for therapeutic applications. ACS Appl Bio Mater. 2022;5(5):2009‐2030. [DOI] [PubMed] [Google Scholar]
  • 299. Panda A, Matadh VA, Suresh S, Shivakumar HN, Murthy SN. Non‐dermal applications of microneedle drug delivery systems. Drug Deliv Transl Res. 2022;12(1):67‐78. [DOI] [PubMed] [Google Scholar]
  • 300. Liang X, Li J, Yan Y, et al. Efficacy of microneedling combined with local application of human umbilical cord‐derived mesenchymal stem cells conditioned media in skin brightness and rejuvenation: a randomized controlled split‐face study. Front Med (Lausanne). 2022;9:837332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 301. Han B, Cao C, Wang A, et al. Injectable double‐network hydrogel‐based three‐dimensional cell culture systems for regenerating dental pulp. ACS Appl Mater Interfaces. 2023;15(6):7821‐7832. [DOI] [PubMed] [Google Scholar]
  • 302. Luo L, Zhu Q, Li Y, et al. Application of thermosensitive‐hydrogel combined with dental pulp stem cells on the injured fallopian tube mucosa in an animal model. Front Bioeng Biotechnol. 2022;10:1062646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 303. Bhattacharjee M, Escobar Ivirico JL, Kan HM, et al. Injectable amnion hydrogel‐mediated delivery of adipose‐derived stem cells for osteoarthritis treatment. Proc Natl Acad Sci U S A. 2022;119(4):e2120968119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 304. Bhattacharjee M, Ivirico JLE, Kan HM, et al. Preparation and characterization of amnion hydrogel and its synergistic effect with adipose derived stem cells towards IL1beta activated chondrocytes. Sci Rep. 2020;10(1):18751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 305. Li H, Xiang D, Gong C, Wang X, Liu L. Naturally derived injectable hydrogels with ROS‐scavenging property to protect transplanted stem cell bioactivity for osteoarthritic cartilage repair. Front Bioeng Biotechnol. 2022;10:1109074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 306. Manferdini C, Gabusi E, Saleh Y, et al. Mesenchymal stromal cells laden in hydrogels for osteoarthritis cartilage regeneration: a systematic review from in vitro studies to clinical applications. Cells. 2022;11(24):3969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 307. van de Looij SM, de Jong OG, Vermonden T, Lorenowicz MJ. Injectable hydrogels for sustained delivery of extracellular vesicles in cartilage regeneration. J Control Release. 2023;355:685‐708. [DOI] [PubMed] [Google Scholar]
  • 308. Yu W, Hu B, Boakye‐Yiadom KO, et al. Injectable hydrogel mediated delivery of gene‐engineered adipose‐derived stem cells for enhanced osteoarthritis treatment. Biomater Sci. 2021;9(22):7603‐7616. [DOI] [PubMed] [Google Scholar]
  • 309. Li S, Ke Z, Peng X, et al. Injectable and fast gelling hyaluronate hydrogels with rapid self‐healing ability for spinal cord injury repair. Carbohydr Polym. 2022;298:120081. [DOI] [PubMed] [Google Scholar]
  • 310. Yin PT, Han E, Lee KB. Engineering stem cells for biomedical applications. Adv Healthc Mater. 2016;5(1):10‐55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 311. Nowakowski A, Andrzejewska A, Janowski M, Walczak P, Lukomska B. Genetic engineering of stem cells for enhanced therapy. Acta Neurobiol Exp (Wars). 2013;73(1):1‐18. [DOI] [PubMed] [Google Scholar]
  • 312. Lanigan TM, Kopera HC, Saunders TL. Principles of genetic engineering. Genes (Basel). 2020;11(3):291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 313. Kitada T, DiAndreth B, Teague B, Weiss R. Programming gene and engineered‐cell therapies with synthetic biology. Science. 2018;359(6376):eaad1067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 314. Kirankumar S, Gurusamy N, Rajasingh S, et al. Modern approaches on stem cells and scaffolding technology for osteogenic differentiation and regeneration. Exp Biol Med (Maywood). 2022;247(5):433‐445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 315. Zhang Q, Nettleship I, Schmelzer E, et al. Tissue engineering and regenerative medicine therapies for cell senescence in bone and cartilage. Tissue Eng Part B Rev. 2020;26(1):64‐78. [DOI] [PubMed] [Google Scholar]
  • 316. Gersbach CA, Phillips JE, Garcia AJ. Genetic engineering for skeletal regenerative medicine. Annu Rev Biomed Eng. 2007;9:87‐119. [DOI] [PubMed] [Google Scholar]
  • 317. Okae H, Toh H, Sato T, et al. Derivation of human trophoblast stem cells. Cell Stem Cell. 2018;22(1):50‐63. .e6. [DOI] [PubMed] [Google Scholar]
  • 318. Kimbrel EA, Lanza R. Next‐generation stem cells ‐ ushering in a new era of cell‐based therapies. Nat Rev Drug Discov. 2020;19(7):463‐479. [DOI] [PubMed] [Google Scholar]
  • 319. Ohnuki M, Takahashi K. Present and future challenges of induced pluripotent stem cells. Philos Trans R Soc Lond B Biol Sci. 2015;370(1680):20140367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 320. Paquet D, Kwart D, Chen A, et al. Efficient introduction of specific homozygous and heterozygous mutations using CRISPR/Cas9. Nature. 2016;533(7601):125‐129. [DOI] [PubMed] [Google Scholar]
  • 321. Itoh M, Kawagoe S, Tamai K, Nakagawa H, Asahina A, Okano HJ. Footprint‐free gene mutation correction in induced pluripotent stem cell (iPSC) derived from recessive dystrophic epidermolysis bullosa (RDEB) using the CRISPR/Cas9 and piggyBac transposon system. J Dermatol Sci. 2020;98(3):163‐172. [DOI] [PubMed] [Google Scholar]
  • 322.Ben Jehuda R, Shemer Y, Binah O. Genome editing in induced pluripotent stem cells using CRISPR/Cas9. Stem Cell Rev Rep. 2018;14(3):323‐336. [DOI] [PubMed] [Google Scholar]
  • 323. Garreta E, Gonzalez F, Montserrat N. Studying kidney disease using tissue and genome engineering in human pluripotent stem cells. Nephron. 2018;138(1):48‐59. [DOI] [PubMed] [Google Scholar]
  • 324. Lim W, Kim HS. Exosomes as therapeutic vehicles for cancer. Tissue Eng Regen Med. 2019;16(3):213‐223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 325. Li C, Ni YQ, Xu H, et al. Roles and mechanisms of exosomal non‐coding RNAs in human health and diseases. Signal Transduct Target Ther. 2021;6(1):383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 326. Li F, Wu J, Li D, et al. Engineering stem cells to produce exosomes with enhanced bone regeneration effects: an alternative strategy for gene therapy. J Nanobiotechnology. 2022;20(1):135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 327. Liang Y, Xu X, Xu L, et al. Chondrocyte‐specific genomic editing enabled by hybrid exosomes for osteoarthritis treatment. Theranostics. 2022;12(11):4866‐4878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 328. Grogan S, Kopcow J, D'Lima D. Challenges facing the translation of embryonic stem cell therapy for the treatment of cartilage lesions. Stem Cells Transl Med. 2022;11(12):1186‐1195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 329. Parisi S, Piscitelli S, Passaro F, Russo T. HMGA proteins in stemness and differentiation of embryonic and adult stem cells. Int J Mol Sci. 2020;21(1):362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 330. Gorecka J, Kostiuk V, Fereydooni A, et al. The potential and limitations of induced pluripotent stem cells to achieve wound healing. Stem Cell Res Ther. 2019;10(1):87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 331. Tan S, Yao Y, Yang Q, Yuan XL, Cen LP, Ng TK. Diversified treatment options of adult stem cells for optic neuropathies. Cell Transplant. 2022;31:9636897221123512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 332. Sano T, Ishigami S, Ito T, Sano S. Stem cell therapy in heart disease: limitations and future possibilities. Acta Med Okayama. 2020;74(3):185‐190. [DOI] [PubMed] [Google Scholar]
  • 333. Vakhshiteh F, Atyabi F, Ostad SN. Mesenchymal stem cell exosomes: a two‐edged sword in cancer therapy. Int J Nanomedicine. 2019;14:2847‐2859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 334. Jo H, Brito S, Kwak BM, Park S, Lee MG, Bin BH. Applications of mesenchymal stem cells in skin regeneration and rejuvenation. Int J Mol Sci. 2021;22(5):2410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 335. Askari E, Naghib SM. Detection and monitoring of stem cell differentiation using nanotechnology. Methods Mol Biol. 2020;2125:197‐204. [DOI] [PubMed] [Google Scholar]
  • 336. Alzate‐Correa D, Lawrence WR, Salazar‐Puerta A, Higuita‐Castro N, Gallego‐Perez D. Nanotechnology‐driven cell‐based therapies in regenerative medicine. AAPS J. 2022;24(2):43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 337. Skuratovskaia D, Vulf M, Khaziakhmatova O, et al. Exosome limitations in the treatment of inflammatory diseases. Curr Pharm Des. 2021;27(28):3105‐3121. [DOI] [PubMed] [Google Scholar]
  • 338. Oh M, Lee J, Kim YJ, Rhee WJ, Park JH. Exosomes derived from human induced pluripotent stem cells ameliorate the aging of skin fibroblasts. Int J Mol Sci. 2018;19(6):1715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 339. Kim J, Li S, Zhang S, Wang J. Plant‐derived exosome‐like nanoparticles and their therapeutic activities. Asian J Pharm Sci. 2022;17(1):53‐69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 340. Chen MY, Wang QG, Fan YJ. Research progress in promotion of tissue regeneration and reconstruction with exosomes derived from mesenchymal stem cells. Sichuan Da Xue Xue Bao Yi Xue Ban. 2021;52(3):380‐386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 341. Zhang S, Hou Y, Yang J, et al. Application of mesenchymal stem cell exosomes and their drug‐loading systems in acute liver failure. J Cell Mol Med. 2020;24(13):7082‐7093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 342. Sun Y, Liu G, Zhang K, Cao Q, Liu T, Li J. Mesenchymal stem cells‐derived exosomes for drug delivery. Stem Cell Res Ther. 2021;12(1):561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 343. Mondal J, Pillarisetti S, Junnuthula V, et al. Hybrid exosomes, exosome‐like nanovesicles and engineered exosomes for therapeutic applications. J Control Release. 2022;353:1127‐1149. [DOI] [PubMed] [Google Scholar]
  • 344. Beck B, Blanpain C. Unravelling cancer stem cell potential. Nat Rev Cancer. 2013;13(10):727‐738. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Not applicable.


Articles from MedComm are provided here courtesy of Wiley

RESOURCES