Abstract
Background
Diabetes has evolved into a worldwide public health issue. One of the most serious complications of diabetes is diabetic foot ulcer (DFU), which frequently creates a significant financial strain on patients and lowers their quality of life. Up until now, there has been no curative therapy for DFU, only symptomatic relief or an interruption in the disease's progression. Recent studies have focused attention on mesenchymal stem cells (MSCs), which provide innovative and potential treatment candidates for several illnesses as they can differentiate into various cell types. They are mostly extracted from the placenta, adipose tissue, umbilical cord (UC), and bone marrow (BM). Regardless of their origin, they show comparable features and small deviations. Our goal is to investigate MSCs' therapeutic effects, application obstacles, and patient benefit strategies for DFU therapy.
Methodology
A comprehensive search was conducted using specific keywords relating to DFU, MSCs, and connected topics in the databases of Medline, Scopus, Web of Science, and PubMed. The main focus of the selection criteria was on English‐language literature that explored the relationship between DFU, MSCs, and related factors.
Results and Discussion
Numerous studies are being conducted and have demonstrated that MSCs can induce re‐epithelialization and angiogenesis, decrease inflammation, contribute to immunological modulation, and subsequently promote DFU healing, making them a promising approach to treating DFU. This review article provides a general snapshot of DFU (including clinical presentation, risk factors and etiopathogenesis, and conventional treatment) and discusses the clinical progress of MSCs in the management of DFU, taking into consideration the side effects and challenges during the application of MSCs and how to overcome these challenges to achieve maximum benefits.
Conclusion
The incorporation of MSCs in the management of DFU highlights their potential as a feasible therapeutic strategy. Establishing a comprehensive understanding of the complex relationship between DFU pathophysiology, MSC therapies, and related obstacles is essential for optimizing therapy outcomes and maximizing patient benefits.
Keywords: cell therapy, diabetes, diabetic foot ulcers (DFU), mesenchymal stem cell, wound healing
Key messages
DFU represents a serious threat to global health, leading to a high rate of amputations globally, with one case of an amputation occurring every second and 84% of these occurrences being linked to DFU. This emphasizes the critical need for better treatments and preventative measures.
MSCs have the ability to improve the healing process when used in concert with other treatments, showing promise as a helpful adjunct to the therapy of DFU.
Although early results point to the safety and effectiveness of MSC therapy in the treatment of DFU, there are still challenges and potential negative effects related to its application. To make MSCs more useful and efficient in preclinical and clinical settings, future research should concentrate on resolving these limitations and maximizing their utilization.
1. INTRODUCTION
Diabetes is a diverse collection of metabolic disorders caused by deficiencies in insulin production, insulin activity, or a combination of the two. Diabetes can be categorized into type 1 and type 2 diabetes. Type 1 is characterized by hyperglycemia caused by the autoimmune attack and damage to the pancreatic β‐cells, while type 2 is characterized by insulin resistance mostly caused by obesity, decreased pancreatic production of insulin, and β‐cell dysfunction. 1 , 2 , 3 By 2045, 783.2 million people worldwide are expected to have diabetes, up from 536.6 million in 2021. 4 Diabetes‐related metabolic abnormalities lead to a wide range of complications, including neuropathy, retinopathy, 5 nephropathy, cardiovascular diseases, and slow wound healing. These issues may cause death or reduce the quality of life. 6 Diabetes patients frequently have significant lower‐limb vascular problems, which can lead to DFU, as their condition gets worse and becomes more complicated. According to reports, around 19%–34% of diabetic patients will be complicated with DFU, and 20%–30% of these patients will end with limb amputation. 7 , 8
The etiology of DFU is a complex issue, and several intrinsic and extrinsic risk factors are involved in the pathogenesis. Intrinsic factors, like neuropathy and peripheral vascular disorders, on the other hand, extrinsic factors including wound infection, callus development, and increased stress on the ulcer site, all affect how well DFUs heal. 9 Given that conservative medical therapy is currently the only successful clinical strategy for diabetic foot care. Currently, conventional management strategies including wound debridement, wound dressing, treatment of wound infection, revascularization, offloading, hyperbaric oxygen therapy (HBOT), and negative pressure wound therapy (NPWT) can only help patients with their symptoms or slow the progression of the disease. Nevertheless, they are unable to restore injured blood vessels and nerves. 10 DFU seems to be a serious issue that threatens human health on a global scale. Consequently, there is an urgent need for a novel technique that accelerates the healing process in diabetic wounds.
It has been observed that a variety of stem cell types, including mesenchymal stem cells (MSCs), promote and boost the healing process in DFUs. 11 , 12 , 13 These multipotent stem cells can develop into a variety of cell types such as epithelial cells, adipocytes, osteoblasts, fibroblasts, chondrocytes, and vascular endothelial cells. 14 , 15 MSCs are an efficient method of treating DFU, as evidenced by the growing research that demonstrates their capacity to encourage re‐epithelialization and angiogenesis, decrease inflammation and the inflammatory process, take part in immunological control, and subsequently heal DFU. 16 , 17
This review article provides a general snapshot of DFU and discusses in detail MSCs and their role as a novel approach in the treatment of DFU. Moreover, it will take the challenges and general and specific limitations of MSCs into consideration and discuss how to overcome these obstacles.
2. DIABETIC FOOT ULCER
DFU is a common and serious complication of uncontrolled diabetes. 18 According to International Diabetes Federation, around 19%–34% of the approximately 540 million diabetic patients will experience a DFU over their lifetimes. 8 10% of those with DFU die within the first year of their initial diagnosis 19 , 20 and 20%–30% will need lower limb amputations, possibly minor (beneath the ankle), major (above the ankle), or both. 8 According to a study conducted in the United States, diabetes mellitus (DM) is responsible for 38% of all amputations. Morbidity and mortality may increase as a result. Neuropathy, ischemia, and infection represent the traditional triad of DFU. The danger of infection and poor wound healing are raised in DFU due to diabetes‐related metabolic dysfunction. It occurs because of several factors, including lowered peripheral blood flow, reduced angiogenesis locally, and lowered cell and growth factor responsiveness. Accordingly, peripheral nerve injury, peripheral vascular disorder, deformities, ulcerations, and gangrene all have an impact on the foot. 21
DFU is clinically presented as a cut in the epidermis and possibly a portion of the dermis. Preulcerative lesions are closed or superficial lesions that are limited to the epidermis (such as blisters, callous, erythema, or warmth), but they are more likely to develop into ulcers. 22 Repeated minor trauma mostly leads to the development of ulcers, frequently as a result of increased pressure at plantar weight‐bearing areas, friction, and shearing brought on by abnormal walking patterns, inappropriate footwear, or an undetected injury on an insensate foot (such as ingrown toenails, puncture wounds, or burns). 23 Structural deformities, such as Charcot neuroarthropathy, increase the incidence of DFU. 8 Complex and multiple pathways finally result in ulceration after a modest stressful incident. 23 , 24 The Wagner method assists in classifying the severity of the ulcer, scoring it on a range of 0–5 (Table 1). 25
Table 1.
Wanger classification for DFU. 25
| Grade | Clinical presentation |
|---|---|
| 0 | Intact skin, no ulcer but high‐risk foot |
| 1 | Superficial ulcer in the skin or subcutaneous tissue |
| 2 | Ulcer extended into joint, tendon, ligament, or capsule without abscess or osteomyelitis |
| 3 | Deep ulcer with abscess or osteomyelitis |
| 4 | Localized gangrene in the toe or forefoot |
| 5 | Extensive gangrene in the whole foot |
Abbreviation: DFU, diabetic foot ulcer.
2.1. Etiopathogenesis
DFU has a complex etiology and several risk factors involved in pathogenesis. The risk factors include peripheral neuropathy (PN), vasculopathy, immunopathy, and the spread of resistant microorganisms.
2.1.1. Peripheral neuropathy
Diabetic PN is a heterogeneous clinical manifestation that is generally described as any combination of signs or symptoms of peripheral nerve dysfunction that is thought to be a consequence of both metabolic and vascular variables associated with persistent hyperglycemia. 26 Neuropathy affects up to 66% of diabetic patients and is one of the main triggers for DFU. 27 Diabetic PN impairs the functioning of the nerve system's sensory, autonomic, and motor divisions and is responsible for poor wound healing. 28 , 29 In sensory neuropathy, sensory nerves are damaged and result in loss of pain sensation. Autonomic neuropathy reduces sebaceous and sweat functions in the lower limbs. As a result, the foot dries out, develops cracks, and becomes more susceptible to microbial infection. 30 , 31 Moreover, motor neuropathy can result in muscle atrophy and anatomical deformities of the foot. This raises the danger of ulceration by causing focally raised pressures at distinct areas of the plantar foot. 32
2.1.2. Peripheral vascular diseases
Vasculopathy is a circulatory disorder that is clinically associated with reduced blood flow to the lower extremities as a result of artery stenosis or atherosclerotic occlusion. 33 , 34 In around 50% of cases, vasculopathy is a major predisposing factor in the development of DFU. It is responsible for 70% of type 2 diabetes‐related deaths. 35 Diabetics have insufficient arterial blood flow, hence peripheral ischemia is the underlying trigger for ulceration in 35% of instances. An inadequate blood flow to the extremities results in inadequate wound healing, which aggravates the problem. 36 In turn, this increases the wound site's susceptibility to anaerobic bacterial growth, which leads to fatal side effects including gangrene and amputation. 27 , 37 Vasculopathy is not regarded as a separate risk factor; rather, it integrates with neuropathy to become the major trigger of non‐traumatic amputations. 38
2.1.3. Immunopathy and spread of resistant microorganisms
Diabetic patients’ immune systems are substantially weaker than those of healthy individuals. Therefore, the presence of infection in diabetic patients is a serious and fatal problem that can potentially result in limb loss. 39 Past research findings reveal that a variety of bacteria, including S. aureus, 40 E. coli, 41 and P. aeruginosa 42 frequently infect diabetic wounds. These microbes cause the progression of sepsis and life‐threatening infections by penetrating deeper into the fascia via irritated or poorly perfused skin. Moreover, resistant bacterial strains such as methicillin‐resistant Staphylococcus aureus commonly infect DFU, increasing the risk of amputation. 43 , 44
3. CONVENTIONAL TREATMENT OF DFU
Effective management and treatment protocol should be started soon after the development of DFU. 45 The gold standard of conventional DFU therapies includes wound debridement, dressing, treating infections, revascularization, off‐loading, HBOT, as well as NPWT. 46 Wound Debridement is the most fundamental approach for managing DFU. Debridement includes removing necrotic or contaminated tissue, blood clots, and unwanted debris from a wound bed. The most prevalent forms are ultrasonic, biological, enzyme, and surgical debridement. 47 , 48 Furthermore, debridement boosts the healing process by removing dying and infected tissue as well as bacterial biofilms. 46
Wound dressings are a conventional wound care approach consisting of natural, modified, or synthetic materials and medicinal components. 49 The wound dressing must offer a moist environment that encourages tissue regeneration, keratinocyte migration, revascularization, and granulation and interferes with bacterial growth. 50 , 51
Infection is frequently present while wounds heal, particularly in diabetic people. 52 If there are more than two of the typical signs of inflammation (redness, hotness, edema, pain, and loss of function) or purulence, it is considered to be an infection. Aerobic Gram‐positive cocci and staphylococci are the most frequently identified microorganisms that are responsible for most DFUs. 53 Narrow‐spectrum antibiotics may be used in mild or moderate wound infection, on the other hand, broad‐spectrum parenteral antibiotics may be used in severe wound infection. 54 , 55
Peripheral artery disease (PAD) is one of the best indicators for the development of chronic wounds and an elevated risk of death from cardiovascular disease. 56 , 57 According to reports, 40% of people with DFUs develop PAD. 56 DFU patients who suffer from PAD may experience slower healing, greater complication rates, as well a greater risk of amputation. Revascularization might therefore be a potential therapeutic option for individuals who suffer from DFU and chronic limb ischemia. 46 , 58
DFU healing is negatively impacted by vertical loads and shear pressure on the plantar. Consequently, Offloading aims to decrease pressure on the DFU's forefoot and plantar surfaces. 59 There are some techniques to reduce foot stress, including orthopedic walking aids and specially designed footwear for DFU therapy. 60 Total contact casting (TCC) bracing is considered an important strategy in the management of DFU. When compared to specifically made footwear, it can reduce the load by itself, and it mechanically aids in lowering and redistributing the stresses on the DFU. It can also promote ulcer healing and is thought to be a vital approach in the management of DFU. 16 , 61 , 62 TCC is proven to be significantly superior to common dressing changes and other offloading ways of boosting ulcer healing and minimizing infection. 63 , 64
HBOT is a promising approach to overcome local hypoxia. The presence of infection in diabetic ulcer lesions encourages local hypoxia, which in turn encourages the development of anaerobic bacteria, reduces local tissue perfusion, and ultimately results in necrosis and cell death. 65 , 66 , 67 , 68 , 69 HBOT is a local oxygen delivery to ulcers and systemic oxygen delivery. 16 HBOT is thought to increase local tissue perfusion, which stimulates collagen synthesis, the generation of growth factors, and neovascularization, and reduces oxidative stress, hence improving wound healing. 70 , 71 Additionally, HBOT creates a bactericidal action against anaerobic microorganisms, hence minimizing the need for antibacterial medications. 72 It's still debatable if HBOT has therapeutic significance, as determined by clinical research. 73 , 74 According to certain studies, HBOT may improve DFU's short‐term, but not permanent ulcer healing power and does not reduce DUF amputation rates. 74 Despite its apparent advantages, HBOT possesses a few adverse consequences, such as toxicity and noncompliance. 68
Recent developments in medical technology in wound dressing and treatment have led to the development of NPWT. Following debridement, negative pressure wound care involves applying a vacuum device to the ulcer region. This suction device may collect significant volumes of exudate, maintain the wound dry and clean, minimize dressing change frequency, and enhance blood flow to the ulcer region. Sustained negative pressure drainage may additionally provide a kind of irrigation to boost and accelerate the healing process. 16 , 75 NPWT causes two forms of tissue deformation: macro deformation, which can be detected by wound contraction, and micro deformation, which appears at microscopic levels. Both deformations increase blood flow and enhance the cascade of wound‐healing processes, such as tissue granulation, vessel growth, neoangiogenesis, epithelialization, and the elimination of extracellular fluid. 76 , 77
Previous studies have completely investigated additional factors such as glycemic management, use of sensitive antibiotics, vascular evaluation, and psychotherapy in DFU patients. 78 , 79 If the patient's condition deteriorates to the point that maintaining a limb is no longer an option, amputation could be a life‐saving alternative. Despite the availability of several treatment options, managing DFU remains one of the most challenging complications associated with diabetes. 80 , 81
4. STEM CELL THERAPY
4.1. Mesenchymal stem cells
MSCs are a form of multipotent stem cell that Friedenstein et al. 82 originally identified When discussing the genesis of cells, the word “mesenchymal” is used to indicate an embryonic origin. MSCs are cells that can change into different mesodermal tissues. They are also called FCUs, which stand for “fibroblast colony‐forming units,” or stromal cells, which are found in bone marrow (BM). 83 The mesoderm is the middle layer of three major layers that arise very early in embryogenesis. It is responsible for the development of a variety of connective tissues, including bones, muscles, cartilage, and fatty tissue, as well as the cells that create blood vessels, cells of the blood, and the urogenital system. 84 MSCs may also be employed to create endoderm and ectoderm‐derived cells, including hepatocytes and neural cells. 85 The capacity for MSC differentiation is influenced by the origin of these cells, the circumstances under which they are amplified, and the milieu in which they are cultured. The differentiation process may be sped up by the use of certain growth factors, hormonal substances, or particular differentiation molecules. 86
Stem cells’ self‐renewing and differentiating‐to‐many‐types abilities and wide‐ranging genetic background make them unique. Furthermore, MSCs support tissue healing through the release of growth factors and cytokines, which aid in the recruitment of additional cells to the injured location. 87 These cytokines and growth factors also encourage the development of new blood vessels, which is essential for the healing of damaged tissue. MSCs are also capable of regulating the activity of the immune system, reducing inflammation, and suppressing immunological responses, which makes MSC treatment a novel alternative for healing and rebuilding damaged tissues. 88 The majority of MSCs are found in BM, and they can self‐renew and display multilineage differentiation. 89 , 90 , 91 They might be derived from a wide range of bodily tissues and organs, including the BM, adipose tissue, blood from the peripheral circulation, Wharton's jelly, dental pulp, placenta, amniotic fluid, and the umbilical cord (UC). 92 , 93 , 94
MSCs may display a range of surface molecules and cellular cytokines according to where they originated from. CD45, CD34, CD19, CD14, CD11b, CD79, and HLA‐DR are not expressed, making CD90, CD73, and CD105 the most common markers that identify MSCs. 95 , 96 , 97 , 98 Several studies show that MSCs may be new way to treat DFU by improving angiogenesis and re‐epithelialization, helping to control the immune system, reducing inflammatory processes, and repairing DFU. 17
4.2. Types of MSCs used in DFU
MSCs can be obtained from a wide variety of sources. They can be produced from BM (BM‐MSCs), human umbilical cords (hUC‐MSCs), adipose tissue (AMSCs), and placenta (PD‐MSCs) and have been the subject of an increasing number of research investigations. 16 Table 2 summarizes the previously documented studies of MSC therapy in DFU in animal studies and preclinical human studies.
Table 2.
Documented previous studies that reported the effect of MSCs from different origins in DFU ASCs (adipose‐derived mesenchymal stem cells), HUVECs (human umbilical vein endothelial cells), PRF (Platelet‐Rich Fibrin), PD‐MSCs (placental‐derived mesenchymal stem cells), PRP (platelet rich plasma), vCPM (cryopreserved placental membrane containing viable cells), CTP (cellular‐ and tissue‐based products).
| Origin of MSCs | Authors | Study group | Method of application | Outcome |
|---|---|---|---|---|
| Bone marrow‐derived | Jain et al. 99 | Human | Topical spray and local injection | In the initial weeks of treatment, a single application of autologous bone marrow‐derived cells accelerates the pace of healing chronic lower extremities wounds. |
| Falanga et al. 100 | Human and murine model | Topical application through fibrin polymer spray system | Proportional decrease in chronic wound size with the number of cells applied, closure of full‐thickness wounds in diabetic mice, and blood vessel formation | |
| Dash et al. 101 | Human | Implant | Reduction in ulcer size from 7.26 to 2 cm and a remarkable improvement in pain‐free walking distance | |
| Amann et al. 102 | Human | Intramuscular transplantation into the ischemic leg | Significant increase in leg perfusion, sufficient to minimize major amputations and allow for long‐term limb salvage | |
| Matoba et al. 103 | Human | Intramuscular injection | A substantial decrease in the leg pain scale, ulcer size, and pain‐free walking distance, resulting in a prolongation of the amputation‐free period | |
| Lu et al. 104 | Human | Intramuscular injection | Improvement of blood circulation, pain‐free walking distance, complete healing of ulcer after 1 month in BM‐MSC group | |
| Xu et al. 105 | Rat model | Injection around the ulcer | Acceleration of wound closure, promotion of cell proliferation and angiogenesis, augmentation of cell autophagy, and reduction of cell pyroptosis in ulcers | |
| Badiavas et al. 106 | Human |
For BM‐MCS aspirate: topical application and local injection For cultured BM‐MCS for booster dose: topical application only |
Complete healing and dermal reconstruction of nonhealing chronic wounds. | |
| Human umbilical cord‐ derived | Yan et al. 107 | Mice model and in vitro cell culture | Injection for mice model and coculture for in vitro cell culture | Acceleration of diabetic cutaneous wound healing via reducing oxidative stress and increasing angiogenesis |
| Qin et al. 108 | Human | Endovascular infusion and injection around the ulcer | Complete or gradual ulcer healing, improvement in skin temperature, ankle‐brachial pressure index, transcutaneous oxygen tension, and claudication distance | |
| Zhang et al. 109 | Rat model | Tail vein injection | Acceleration of wound closure; scar reduction; improvement in regeneration of skin appendages, nerves, and arteries; and modulation of the natural distribution of collagen fibers in wound healing | |
| Adipose‐ derived | Kim et al. 110 | Mice model | Topical application | Improvement of wound healing rate, neovascularization, dermal regeneration, keratinocyte formation, and granulation tissue formation in the allogenic AMSCs group rather than the autologous group |
| Fromer et al. 111 | Murine model | Injection into the ischemic muscle | Enhancement of wound healing, proliferation, and neovascularization in HUVEC‐primed AMSCs group rather than AMSCs group. This is because, under diabetic conditions, HUVECs produce protein factors that markedly enhance the proliferation and endothelial differentiation of AMSCs. | |
| Khalil et al. 112 | Human | Topical application with spraypen | PRF enhances ulcer healing and proliferation properties of autologous AMSCs as the platelets act as a reservoir for several growth factors that stimulate healing process | |
| Cianfarani et al. 113 | Mice | Topical application | Diabetes modifies the fundamental features of AMSCs and reduces their functionality, which may impact their therapeutic efficacy when used in autologous therapy for diabetic ulcers. | |
| Rennert et al. 114 | Mice | Subcutaneous injection | Diabetic AMSCs are impaired in their capacity to form a vascular network both in vitro and in vivo. | |
| Moon et al. 115 | Human | Topical application | Significant complete wound closure in study group compared to control after 12 weeks of follow‐up | |
| Placental‐derived | Zeng et al. 116 | Human | Topical application | Reduction in wound size after 3 weeks, resulting in a shorter healing time as well as the production of dense granulation tissue that aids in wound recovery |
| Du et al. 117 | In vitro study | – | PD‐MSCs had the potential to speed up the healing process of ulcers via the generation of various cytokines and HGF, in addition to the paracrine activities that encouraged angiogenesis | |
| Meamar et al. 118 | Human | Topical application | Improvement of wound healing and pain‐free walking distance in both the PD‐MSCs and PD‐MSCs + PRP gel groups compared to control | |
| DaVanzo et al. 119 | Human | – | Significant reduction in recurrence or occurrence of ulcers and decline in mortality rate in vCPM group compared to CTP group |
Abbreviations: DFU, diabetic foot ulcer; HGF, hepatocyte growth factor; MSC, mesenchymal stem cell.
4.2.1. BM‐derived MSC therapy in DFU
BM‐derived cells such as inflammatory cell progenitors, MSCs, and multipotent stem cells represent intriguing therapeutic potential for healing chronic wounds. 120 , 121 The plasticity of BM cells raises the possibility that they may generate fresh cells in the skin. 122 BM‐MSCs are the most widely employed kind of cells in both preliminary and clinical investigations. 123 MSCs can regenerate the dermis of the skin, and it has been noted that in chronic wounds, they may exhibit phenotypical changes or become senescent. 123 Several studies have shown that the use of BM‐MSCs is effective in the healing process of chronic wounds. For example, the topical administration of BM‐MSCs has been shown to promote the regeneration of chronic wounds within 2–4 weeks after treatment. 99 Furthermore, there was a link between the number of cells delivered and the proportional reduction in lesion diameter, suggesting that bigger wounds require more MSCs to heal adequately. 100 According to the findings of Badiavas et al., 122 immediately applying BM‐MSCs led to skin regeneration and resulted in the full healing of chronic wounds in every one of the individuals who participated in the research.In a different investigation, Dash et al. 101 showed that therapies with autologous BM‐MSCs were a simple, risk‐free, and successful approach for the management of chronic wounds that were nonresolving.This treatment proved successful in reducing ulcer size, controlling pain, and increasing the distance that patients were able to walk without experiencing discomfort. After therapy, all biochemical markers were found to have remained within normal limits, showing that the medication had no adverse effects on the body. 101
MSCs, which originated in BM, are showing promise as an effective supplementary treatment that may hasten the healing of wounds and increase the likelihood of limb salvage. BM cell transplantation was shown to be safe for those with end‐stage serious limb ischemia caused by PADs. This procedure was also effective in enhancing leg perfusion, drastically decreasing the number of major amputations, and allowing for the long‐term salvage of limbs. 102 According to the findings of Matoba et al., 103 BM‐MSCs have the potential to bring about a long‐term reduction in limb ischemia, resulting in an extension of the time until amputation is required. The effectiveness of BM‐MSCs and BM‐derived mononuclear cells (BM‐MNCs) for the management of ongoing wounds among individuals with diabetic serious limb ischemia and DFU were evaluated and compared via the use of intramuscular injections of both cell types. 104 The ankle‐brachial index (ABI) and transcutaneous partial pressure of oxygen (TcO2) measurements of the study's findings show that both types of cells have the capacity to improve blood circulation and reduce pain. Results from the group that was injected with BM‐MSCs show that the wound's healing rates were much greater 6 weeks after the administration, and complete healing was achieved a month earlier compared with the BM‐MNC group. 104 Furthermore, tCO2 (total carbon dioxide), ABI, magnetic resonance angiography, and painless walking time analyses all showed significantly better outcomes in those undergoing treatment with BM‐MSCs relative to those who were administered BM‐MNCs 24 weeks after therapy. These findings suggest that, in diabetic individuals suffering from critical limb ischemia, BM‐MSCs may be well tolerated as well as more efficient than BM‐MNCs in improving limb oxygenation and speeding up the healing process of foot ulcers 104 Also, Xu et al. 105 demonstrated that BM‐MSC‐CM is an excellent therapy for DFU in rats with type 2 diabetes. Inflammation may be reduced, autophagy can be enhanced, and pyroptosis can be reduced with the use of BM‐MSC‐CM, which can aid the healing of DFUs. These results bring to light a potentially life‐saving therapeutic use of BM‐MSC‐CM for the management of DFUs that sidesteps the potential hazards of live cell therapy. 105
4.2.2. hUC‐MSC in DFU
As a part of therapeutic angiogenesis, BM‐MSC implantation has been used to treat diseases like cerebral infarction 124 , 125 heart attack 126 , 127 and ischemia of the limb. 125 , 127 The capacity to get stem cells from BM depends on the recipient's general health. 127 Hence, UC blood or peripheral blood is more commonly used as it is easier to collect. 128 hUC‐MSCs can develop into neuronal, muscle, and blood vessel tissue. High‐profile status is given to hUC‐MSCs because of their capacity for pluripotency. hUC‐MSCs have been used in a number of therapeutic contexts recently to promote wound healing. 107 In certain investigations, hUC‐MSCs have been demonstrated to improve angiogenesis and promote the rejuvenation of tissues. 107 Qin et al. 127 employed hUC‐MSCs for the management of DFU after angioplasty in two separate investigations. Better healing of wounds was seen in individuals administered hUC‐MSCs relative to those who received merely angioplasty. In patients with advanced diabetes, the combined effect of the two therapies increased blood flow, decreased the incidence of amputations, accelerated the healing of ulcers, and enhanced quality of life. This suggests that the implantation of hUC‐MSCs with angioplasty is a feasible and successful therapeutic therapy for the management of advanced DFU. 127
Studies of how hUC‐MSCs work have also shown that the exosomes made by these cells are stable and immune‐stimulating. They may also carry protein molecules and growth factors that have different roles and have different effects. 129 Exosomes made by hUC‐MSCs have been shown to control both the growth and differentiation of BM‐MSCs. 130 Yan et al. 107 investigated the impact that hUC‐MSCs and hUC‐Exos (exosomes released by hUC‐MSCs) on the healing process of diabetic wounds. They demonstrated that hUC‐MSCs regulate the expansion and activity of endothelial cells, as well as promote the healing of injuries by releasing exosomes. Through the promotion of angiogenesis and the alleviation of oxidative stresses, hUC‐Exos was able to hasten the process of wound repair. The results of this research suggested that the treatment of diabetic cutaneous wounds may be accomplished via the use of a mixed injection, which could lessen the burden that diabetes places on both the medical system and the economy. Additionally, the application of this therapeutic strategy in the management of diabetic wounds in the future may hold great promise. 107 CORLICYTE® is a novel hUC‐MSCs therapy that just passed phase I clinical trial. 131 CORLICYTE® was tested on nine patients with chronic DFU, and all patients experienced a marked reduction in wound size without any adverse effects. 132
4.2.3. Adipose‐derived MSC therapy in DFU
In comparison to BM‐MSCs, AMSCs have emerged as a viable option for cell treatment since they are easy to access and may be obtained from the subcutaneous region. They also originate from several locations and have more time to grow and change. When compared to the method of extracting BM‐MSCs, the process of harvesting AMSCs is less invasive, causes less discomfort, and requires less invasive surgery. AMSCs derived either from the patient's own body, from a donor, or from a xenograft may be used. Furthermore, the immunosuppressive effect of these cells is roughly amplified by a factor of three. 133 In addition, compared to BM‐MSCs, AMSCs have a higher ability for cell proliferation, which makes them an excellent candidate for use in cell‐based therapies intended to treat persistent diseases. 134 , 135 Minimal ethical debates are necessary due to the fact that AMSCs may be isolated from adult fat obtained from the patients themselves. 136 Numerous investigations also demonstrate that AMSCs speed up the healing process by releasing angiogenic cytokines, inhibiting inflammation and apoptosis, and boosting the production of epithelization and granulation tissue development. 137 , 138 According to research by Cianfarani et al., 139 DM inhibits the activity of AMSCs and alters their intrinsic characteristics, which in turn limits the ability of these cells to heal DFU in diabetic rat models. Also, AMSCs from diabetic mice were less able to release vascular endothelial growth factor A (VEGF‐A), insulin‐like growth factor‐1 (IGF‐1), and hepatocyte growth factor (HGF). 140 Kim et al. 140 examined the level of wound repair in mice that had been handled with either ordinary AMSCs or AMSCs produced from diabetic mice. They found that the regular AMSCs group had a higher percentage of dermal renewal, granulation tissue development, keratinocyte expansion, reepithelization, and overall wound healing. Nevertheless, the diabetic AMSCs were still able to maintain their ability to cause angiogenesis and neovascularization. 140 On the other hand, Rennert et al. 114 found that diabetic AMSCs are unable to effectively promote neovascularization and wound recovery. Based on the results of this study, there is a limited role for autologous AMSCs in cellular therapies for diabetic patients, and pretreatment steps to enhance the activity of cells are required. 114 Fromer et al. 111 investigated the potential of the secretome of human endothelial cells to counteract the detrimental impact of elevated glucose levels on AMSCs. This was achieved through a process known as priming, which enhances the regenerative and angiogenic capabilities of AMSCs in murine models. However, it is important to note that further investigation is required to simulate these findings in clinical trials. 111 In addition, research that evaluated the healing process has shown that adding platelet‐rich plasma (PRP) to AMSCs has favorable benefits. PRP particularly acts as a strong paracrine effector and cellular carrier, which increases the potency of transferred cells to be utilized in treatments. 141 Moon et al. 115 reported that the allogenic AMSC‐hydrogel complex is an effective and safe treatment option for DFU, as this complex can promote a significant complete wound closure after 12 weeks in the study group compared to a control group. Interestingly, this complex is now on phase III clinical trial and registered with ID (NCT04569409), but this trial is not completed. 142 So, it can be concluded that the use of allogeneic normal AMSCs instead of autologous diabetic and damaged AMSCs is a better way to treat wounds in people with diabetes. 140 In another study, the effects of implanting AMSCs in autologous platelet‐rich fibrin (PRF) were compared to the effects of using PRF alone to treat chronic DFU healing. They followed this regimen since it is thought to be a more effective and efficient healing method than standard persistent wound care. 112 According to Khalil et al., 112 individuals who received AMSCs in addition to PRF showed better wound healing than those who just received PRF. Their findings are in line with previous research suggesting that MSCs release significant amounts of VEGF and HGF and have a higher proportion of transforming growth factor‐3 to TGF‐1, which leads to cell growth, migration, matrix deposition, and enhancement of vascular angiogenesis. 112
4.2.4. Placental‐derived MSC therapy in DFU
PD‐MSCs have been used in a limited number of clinical trials involving humans for the management of DFU. Although BM‐MSCs are the most common source of mesenchymal cells, PD‐MSCs are a better option for several reasons. It is simple to get PD‐MSCs, and doing so does not give rise to any ethical concerns and the human placenta is less immunogenic than BM. Finally, a greater quantity of stem cells may be separated from the placenta than from BM. 143 , 144 Along with their capability to secrete substances that may speed up the healing process of wounds, it has been shown that PD‐MSCs have an impressive ability to transform into a wide variety of cell kinds. 143 , 144 , 145 , 146 In a clinical trial, Zeng et al. 116 investigated the effect of PD‐MSCs hydrogel on the healing process of DFU. According to the findings of their study, the use of PDMSC hydrogels led to a reduction in wound size after 3 weeks, resulting in a shorter healing time as well as the production of dense granulation tissue that aids in wound recovery. 116 PD‐MSCs secrete paracrine factors, stimulate vascular development, and modulate the immune system, all of which contribute to their potent healing impact in DFU. However, Zeng et al. 116 argued that larger patient samples are required for future research. Further research by Du et al. 147 revealed that PD‐MSCs had the potential to speed up the healing process of ulcers via the generation of various cytokines and HGF, in addition to the paracrine activities that encouraged angiogenesis, which is a process that is involved in wound repair.
In another study, Meamar et al. 118 investigated the efficacy of nanofibers infused with PD‐MSCs and PRP for the treatment of DFU. In this trial, PD‐MSCs coated with PRP gel, and a control group receiving routine wound care were used to treat ulcers over 12 weeks. Additionally, Meamar et al. 118 found that the size of wounds decreased by 66% and 71% in the PD‐MSCs‐treated and PD‐MSCs + PRP gel‐treated groups, respectively, but only by 36% in the control group. Both the PD‐MSCs and PD‐MSCs + PRP gel groups outperformed the control group in terms of wound healing and pain‐free walking distance. Biopsies taken from patients in both the PD‐MSCs and PD‐MSCs + PRP gel treatment groups revealed the growth of new blood vessels. 118 In a more recent investigation, DaVanzo et al. 119 compared the results of individuals with DFU who underwent therapy with a cryopreserved placental membrane containing viable cells (vCPM) to those of individuals managed with other cellular‐ and tissue‐based products (CTPs). The success of the therapy was evaluated based on two different metrics: the decrease in the incidence of after‐treatment ulcers and the decrease in mortality after 1 year. The results showed that vCPM had a significant reduction in ulcers when compared to CTP treatment, and it was able to cut mortality by 2.3 percentage points (13%–13.8% change) after 1 year when compared to other CTPs. The decline in mortality was due to the fact that vCPM was effective in reducing the number of ulcers that were present. 119
5. MECHANISMS OF MSCs IN THE TREATMENT OF DFU
The physiological mechanisms of DFU wound healing and growth include cellular expansion, differentiation, and relocation. Damage to living tissues results in wounds, and the process of coordinating wound healing begins as soon as the tissue surface is compromised. Throughout the process of repair, cytokines and growth factors work to enhance cell differentiation, expansion, movement, and protein synthesis by stimulating signal regulation and coordinating intracellular and intercellular signaling processes. Recent research has shown the critical role played by numerous growth regulators and molecular pathways in the emergence and progression of DFU. 148 , 149
5.1. MSCs induce angiogenesis by releasing several growth factors and regulators
Vascular destruction and lesions of the vessels are major causes of DFU. The regrowth and formation of new blood vessels in the ulcer zone supply nutrients for the growth of granulation tissue. Therefore, it is crucial for reducing ulcer size and encouraging healing. Extensive research has shown that MSCs release many different types of cytokines, such as epidermal growth factor (EGF), VEGF, IGF‐1, keratinocyte growth factor 2, basic fibroblast growth factor, stromal cell‐derived factor‐1 (SDF‐1), and placental growth factor‐2. Wound healing, vascular development, and improved microhemodynamics are all aided by the presence of these substances. 150 , 151 Additionally, another study showed that MSCs derived from the mouse's liver stimulate local growth factor production. These growth factors include SDF‐1, EGF, and VEGF. As a result, the formation of new blood vessels is promoted, cell recruitment to the wound is improved, and the contraction of wounds is enhanced (Figure 1). 152 , 153 , 154 Furthermore, BM‐MSCs can greatly boost the production of important growth factors like VEGF and EGF, which are necessary for the repair and regeneration of damaged tissues. They have been shown to improve the healing of wounds in diabetic rats by elevating collagen levels (types I–V). 155 Shen et al. 156 have demonstrated that BM‐MSCs can accelerate the healing of DFU in mice models by enhancing vascular endothelial cell activation and enhancing angiogenesis through paracrine VEGF and other vasoactive factors. Wan et al. 157 discovered that foot ulcers in diabetic rats healed more quickly after BM‐MSC transplantation because VEGF production in the injured area was enhanced and angiogenesis was stimulated. Diao et al. 158 demonstrated that VEGF may enhance transcription factors for controlling endothelial progenitor cells (EPCs), recruit EPCs to the BM, inhibit EPC death, and assist wound healing, in addition to directly stimulating angiogenesis. According to the findings of these investigations, MSCs can promote angiogenesis in the ulcer region directly or indirectly by releasing paracrine growth factors. This increases blood flow and accelerates DFU repair. 16
Figure 1.

Role of MSCs and their secretome in promoting wound healing in DFU. MSCs and their secretome can promote DFU healing through release of growth factors like EGF, VEGF, and SDF‐1 that promote angiogenesis, induce cell migration and regulation of wound tissue microenvironment, enhance epithelialization through differentiation and proliferation of fibroblast and keratinocytes, immunomodulatory effect through M2 macrophage polarization, increase Treg activation and inhibition of Th1 and Th17, and reduce inflammation and oxidative stress through inhibition of proinflammatory cytokines and ROS. Adapted from Yu et al., 152 El Hage et al., 153 Badillo et al. 154 DFU, diabetic foot ulcer; EGF, epidermal growth factor; MSCs, mesenchymal stem cell; ROS, reactive oxygen species; VEGF, vascular endothelial growth factor.
5.2. MSCs inhibit inflammatory T cells and activate regulatory T cells
MSCs not only have the potential to develop into a variety of different kinds of cells, but they also have a regulatory function in the immunological and inflammatory processes that the body has. Many research investigations demonstrate that following damage to a tissue or cells, MSCs can be triggered by inflammation‐related cytokines to regulate the process of tissue repair. They do this by launching several substances that can increase the growth and differentiation of progenitor cells while also playing a role in immune system regulation and preventing inflammatory reactions. This is the case after a cell or tissue has been damaged. 159 , 160 , 161
Inflammation can be mediated by both T helper 1 (Th1) and T helper 17 (Th17) cells 162 (Figure 1). T regulatory cells (Treg) are a subset of specialized immune‐suppressive T cells that express CTLA‐4 and CD25 on their surfaces and FoxP3 in their nucleus, therefore, preserving immunological self‐tolerance and homeostasis. 163 , 164 In a study by Li et al., 165 15 DFU patients who were receiving insulin treatment also received hUC‐MSC transplants. Following the transplantation, levels of blood sugar and insulin dosage fell in all 15 individuals. After 4 weeks following the transplantation, there was a considerable rise in the ratios of CD4 + CD25 (hi) FoxP3 + Treg/Th17 and CD4 + CD25 (hi) FoxP3 + Treg/Th1 cells. However, the ratios of Th17/Th1 cells remained stable, and there was a peak in blood levels of VEGF. 165
5.3. MSCs reduce proinflammatory M1 macrophages and enhance anti‐inflammatory M2
M1 macrophages are characterized by the release of proinflammatory molecules that boost immunity against infections and have significant bactericidal properties, as well as trigger tissue death and inhibit angiogenesis. 166 , 167 M1 macrophages are distinguished from other types of macrophages by their heightened capacity to release IL‐18, IL‐1b, reactive oxygen species (ROS), tumor necrosis factor (TNF), and IL‐12. 168 M2 macrophages, on the other hand, are thought to fight inflammation and aid in cell regeneration. M2 macrophages express IL‐10, Arginase 1, CD206, and chitinase 3‐like 3, resistin‐like‐α. 169 Molecular analysis suggests a role for these substances in angiogenic promotion, parasite infections, tumor immunomodulation, and tissue remodeling. 170 Proinflammatory M1 macrophages penetrate the ulcer in the early stages of healing and clear out any germs, cell debris, or unwanted objects they find. 171 As the tissue begins the healing process for an acute ulcer, the M1 macrophage pool transforms into an M2 type. M2 cells immediately begin the process of fighting inflammation and repairing damaged cells. 172 When proinflammatory macrophages with the M1 pattern fail to switch to the anti‐inflammatory M2 type, tissue healing in chronic wounds is stunted. 173 , 174 Macrophages are stimulated to generate proinflammatory mediators in response to a chronically excessive glucose situation in vivo. These cytokines include IL‐1b, ROS, IL‐6, and TNF‐α. This leads to a downward spiral of permanent M1 macrophage populations and a consistently elevated degree of inflammation in DFU. 175 Dayan et al. 176 thought that co‐culturing human BM‐MSCs and hUC‐MSCs with macrophages might reduce the number of macrophages and monocytes in the body as a whole. This reduction would include a reduction in the number of macrophages with the M1 phenotype that contribute to inflammation. In contrast, there was a considerable increase in the number of M2 anti‐inflammatory phenotypes that had undergone alternative activation. 176 To treat diabetic mouse wounds, Chen et al. 177 loaded 3D nanofiber structures with mouse BM‐MSCs. They found that there were more M2 macrophages than normally activated M1 phenotypes, which helped diabetic mice heal faster from wounds. 177 Vascular endothelial cells’ ability to function properly is dependent on the release of PGE2 by hUC‐MSC. This is accomplished by the modification of macrophage phenotypes, which then leads to an improvement in the surrounding microenvironment of the vascular endothelial cells via the production of VEGF and IL‐10. Controlling the change from M1 to M2 macrophage phenotypes in diabetic wounds boosts angiogenesis, which helps the wounds heal. 16 , 178 On the other hand, to help the body fight against infections, M1 macrophages release cytokines that trigger an inflammatory response. These macrophages are also quite effective in killing bacteria. Thus, the shift from an M1 to an M2 phenotype in mice may reduce their resistance to disease and make them more vulnerable to certain infections.
5.4. MSCs reduce inflammation and oxidative stress
ROS at high concentrations cause oxidative stress and immune system reactions that damage and impair cells, while low ROS levels are favorable for sustaining survival, proliferation, and differentiation. 179 Phagocytes digest microorganisms, cell debris, or apoptotic inflammatory cells after tissue injury. Long‐lived neutrophils produce a significant amount of ROS following phagocytosis, leading to a respiratory burst that damages tissue. MSCs may function as antioxidants by paracrine inhibition of protein oxidation and lipid peroxidation or through direct cell interaction, according to numerous publications. 16 , 180 , 181 MSCs have been shown to lower inflammation and oxidative stress in a variety of disorders. These effects include a decrease in the production of enzymes that make ROS, like inducible nitric oxide synthase, myeloperoxidase, and nitrogen oxides, as well as a decrease in the production of cytokines that cause inflammation, like IL‐6, IL‐9, IL‐1b, IL‐4, TNF‐α, and interferon‐γ. 182 , 183 MSCs have the ability to directly lower ROS as well as myeloperoxidase in activated macrophages and monocytes, which in turn inhibits the proinflammatory phenotypes of these cell types. 184 , 185 The presence of MSCs significantly reduced the formation of ROS in macrophages. This was accomplished by boosting the release and expression of stanniocalcin (STC)‐1 as well as suppressing caspase‐1 activation, the NOD‐like receptor pyrin domain containing 3 (NLRP3) inflammasome, TNF‐α, and IL‐6 transcription, and IL‐1b production. 184 Additionally, it has been demonstrated that PD‐MSC transplantation can speed up the healing of diabetic wounds by reducing cytokine levels such as TNF‐α, IL‐1, and IL‐6, and by slowing down the signaling of nuclear factor kappa B. 146 Raffaghello et al. 186 found that BM‐MSCs had the potential to inhibit ROS production without compromising neutrophil phagocytic activity. This was accomplished by inhibiting apoptosis in neutrophils, activating them, and preventing them from engaging in inappropriate or excessive oxidative metabolism. 186 Exosomes that are released by human AMSCs have the ability to slow the progress of DFU by suppressing the aging of EPCs and reducing the generation of ROS and cytokines that trigger inflammation. 187
5.5. MSCs induce cell migration and regulate the wound tissue microenvironment
Recent research has demonstrated that several different molecular pathways, such as cell signaling processes, perform essential functions in the pathogenesis and DFU healing mechanisms. 188 , 189 , 190 A protein‐serine‐threonine kinase (AKT) serves as a crucial hub for cellular signaling involved in many different processes. MSC survival, expansion, movement, and angiogenesis are all further influenced by PI13‐dependent AKT activation; this pathway has a fundamentally important regulatory function. 190 According to the findings of Hou et al., 191 the conditioned medium of BM‐MSCs prompted human umbilical vein endothelial cells to migrate and proliferate more quickly. These activities were reliant on the extracellular signal‐regulated kinases (ERK) signaling pathway but had a tight relationship with the AKT signaling system. 191 Jun et al. 192 showed that hypoxia causes an increase in the release of paracrine substances by amniotic fluid‐derived MSCs (AF‐MSCs) and that hypoxic‐conditioned medium from AF‐MSCs (AF‐MSC‐hypoCM) enhances the healing process through the promotion of the migration of cells and the stimulation of the TGF‐β/SMAD2 and PI3K/AKT pathways. This suggests that AF‐MSC‐hypoCM might be employed for the management of wound regeneration and marks left after operations. In addition, AF‐MSC‐hypoCM has the potential to provide a different pharmaceutical option for use in the medical and aesthetic industries, which would increase the efficiency of tissue repair. 192 According to the findings of Liu et al., chemokine receptor‐4 and SDF‐1 both play significant roles in the regulation of BM‐MSCs to enhance the healing of DFU. 193 It is interesting to note that combination therapy with PRP and rat ADSCs enhances angiogenesis, activates epidermal stem cell expansion and recruitment through regulation of the Notch system, and greatly speeds up the repair process of diabetic lesions induced in rats in an experimental setting. 194 These observations provide credence to the hypothesis that the Notch signaling system represents a novel and potentially promising targeted therapy for diabetic wounds. 195 , 196
MSCs have been proven to be able to discriminate into epidermal cells and function as epidermal cells in vitro investigations using a variety of different induction techniques. 197 , 198 Kato et al. 199 used BM‐MSCs to heal foot lesions in both diabetic and normal rats. They observed that the levels of phosphorylated focal adhesion kinase went back up once human keratinocytes were grown in BM‐MSC‐conditioned media with higher glucose concentrations. Also, levels of IGF‐1, EGF, and matrix metalloproteinase‐2 were found to be higher. This suggests that BM‐MSCs may be able to help diabetic foot model mice heal wounds faster by making keratinocytes work better. 199 Also, wounds treated with BM‐MSCs encourage the growth of keratinocytes and endothelial cells and the movement of macrophages, keratinocytes, and endothelial cells into the injuries of model animals, which speeds up the repair mechanisms. 200 Considering the findings of another investigation, BM‐SCs greatly increased the number of keratinocytes in the injury site, increased the production of new blood vessels, sped up the rejuvenation of epithelial cells in the injured area, and made wounds heal faster. 201 In addition, hUC‐MSCs can precisely localize to the specific wound tissues in a rat model of DFU, boost the release of cytokeratin 19, encourage the creation of keratinocytes and extracellular matrix (ECM), and enhance the renewal of epithelial cells in wounded areas. 202 Even though MSCs have been shown in a plethora of studies to be able to differentiate into endothelial cells and keratinocytes, the consequences of their engraftment are still up for debate. It has been hypothesized that MSCs have the capacity to differentiate into keratinocytes under certain conditions; however, MSCs do not possess the full complement of keratinocyte‐specific expression markers. 203
6. ROUTS OF ADMINISTRATION OF MSCs IN DFU
Stem cell therapy used for treating DFU can be given in one of two ways. Research shows that each of the local and systemic approaches is beneficial in resolving DFU. Among the several methods of cell administration, local injection is by far the most common. Clinical trials typically employ intramuscular administration. The most common routes of administration in preclinical research are intradermal and subcutaneously administered injections. 17
Systemic cell administration may be carried out endovascularly, for example, through intravenous or intraarterial injections. During angioplasty, it is possible to provide the drug systemically, where it can have an immune‐modulating function and enhance the physiological balance of glucose. However, there is a substantial potential for surgical risks, poor engraftment, and high costs associated with this mode of administration. 17
Topical uses have been more prevalent in preliminary studies than in clinical settings. Hydrogels, sprays, drops, and scaffolds are examples of topical administration methods, and ECM scaffolds have attracted scientific attention. The ECM is regarded as the primary regulator of cell regeneration, expansion, and differentiation. 17 The ability of hydrogel and collagen scaffolds to simulate the in vivo conditions for stem cells results in an increase in cell functionality, 204 engraftment, and retention. 205 , 206 , 207 , 208 In topical administration, there is minimal risk involved; it is simple, and in most circumstances, other than those involving hydrogels and scaffolds, it is believed to be quite affordable. However, before treating the wound, local cell administration might require debridement of the wound area. 17
Chiang et al. 209 conducted research in which they assessed the effectiveness of medically employing autologous stem cells in several transplantation techniques, including intramuscular injection, topical application, and intraarterial injection. The researchers found that the group that received stem cell injections through the intramuscular route had a considerably better rate of full healing compared to the group that received treatment via the intraarterial route. According to Chiang et al., 209 the findings might be connected to the fact that people who suffer from DM face microvascular difficulties or arterial occlusion, both of which result in significant ischemia of the limb, which in turn leads to peripheral perfusion. Considering that cells are carried closer to the wound area when intramuscular delivery is used, this problem might be eliminated. 209 Also, muscular tissues are able to provide infused cells with nutrients and oxygen, which contributes to the improvement of their function as well as their chances of survival. 210 According to several investigations, the intravascular infusion of stem cells may cause the cells to get lodged in the lungs, which can result in a pulmonary embolism. As a consequence of this, the injection of stem cells through the intramuscular route seems to be the method that is both safer and more successful. 209 Regarding the topically applied treatment of autologous stem cells, Chiang et al. 209 observed that it was successful in the wound repair process, as indicated in earlier research. They also reported that topically applied medication helps with cellular metabolism, cell differentiation, and migration. 209 By increasing ECM production and regeneration of tissues, topical application may also reduce the length of the therapy time, which in turn enhances the rate of survival of the transplant. 211 , 212 According to these findings, administration through intramuscular and topical routes is more efficient than treatment via intraarterial routes. 209
7. BIOMATERIALS AS MSC DELIVERY SYSTEMS
There are some drawbacks related to the topical delivery of MSCs through direct injection. Two of these drawbacks include limited cell survival and reduced cell adaptation at the wound site. As a result of this, many biomaterial conformations have evolved as vehicles for the transport of MSCs, to enhance cell survival and persistence at the site of implantation. 4 , 213 Scaffolds have been proposed as a solution to these issues because they offer a three‐dimensional framework for the movement of cells, their proliferation, and differentiation while also increasing cell survivability and retention at the wound site (Figure 2). 213 , 214
Figure 2.

Role of a scaffold‐based delivery system in delivering MSCs and promoting their wound healing action. MSC delivery can be improved by employing scaffolds and grafts that simulate or preserve the architecture of human tissue, creating a favorable milieu for MSCs to adhere, proliferate, and retain their secretome in addition to directing host cell migration. The secretome of MSCs encourages the migration and infiltration of immune cells (lymphocytes, macrophages, and neutrophils) that will control the inflammatory and immunological response in the wound area, boosting angiogenesis and enhancing wound healing. MSCs, mesenchymal stem cell.
7.1. Scaffold‐based delivery system
7.1.1. MSC distribution through hydrogel scaffolds in diabetic models
Hydrogel scaffolds evolved as vehicles for the transport of MSCs to enhance cell survival and persistence at the site of implantation. 4 Scaffolds have been proposed as a solution to these issues because they offer a three‐dimensional framework for the movement of cells, their proliferation, and differentiation while also increasing cell survivability and retention at the wound site. 214
Hydrogels are 3‐dimensional structures that can expand and retain a substantial amount of water inside their framework. These networks may be made of natural, synthetic, or mixed polymers. Because of their potential to preserve the viability of cells at the site of implantation and their versatility in terms of manufacture, hydrogels have gained a lot of attention in the discipline of treatments related to wound repair in the past few years. 215 , 216 The hydrogels that are naturally produced have shown a number of beneficial properties, including biocompatibility, biodegradability, inherent biological interactions, and structural resemblance to genuine human tissue. 217 Natural hydrogels, on the other hand, have several drawbacks, such as a restricted range of mechanical characteristics and variable results from batch to batch. 218 Composite hydrogels that blend natural and synthetic materials to achieve the desired form and function are becoming more popular. 219
Hydrogel‐mediated human MSC administration enhanced wound repair in db/db mice, with microhydrogels generated from human AD‐MSCs showing faster wound epithelial growth and thicker dermal development compared to control and hydrogel‐alone groups. 220 For treating wounds related to diabetes in mice, human AD‐MSCs have also been administered in the form of a hydrogel composed of hyaluronic acid and gellan gum. In this research, an enhanced influence on the neovascularization of diabetes wounds was seen, and the epidermis of the cured diabetic lesion was revealed to be thicker as well as more discriminated than the epidermis of the diabetic lesion that had received no therapy. 221 An additional investigation found that using a PEGDA hydrogel to encapsulate a combination of human BM‐MSCs and rat insulin‐secreting cells enhanced diabetic wound repair approximately three times quicker than in the control groups. 222
7.1.2. MSC delivery through sponge scaffolds in diabetic models
Sponge scaffolds could be prepared from synthetic or natural polymers by various techniques, such as gas foaming, porogen leaching, and freeze‐drying. They are very porous and have a consistent network of pores that are linked together. 223 , 224 Sponge scaffolds are excellent for directing cell migration to an injury site because their porous structures are identical to those of the ECM. 225 Because of their capacity for absorption and storage of water, sponge scaffolds afford a model environment for the proliferation and migration of cells in the wound site. 226 The majority of MSC carriers used for diabetic wound repair are collagen‐ and chitosan‐based sponge scaffolds. O'Loughlin et al. 227 created collagen sponge scaffolds by using the freeze‐drying technique. Seven days after delivery, allogeneic BM‐MSCs supplied topically through a collagen sponge scaffold facilitated faster wound repair and increased angiogenesis in diabetic rabbit wounds compared to the no‐treatment control group. 227 This sponge scaffold created an environment where hypoxia‐pretreated rat BM‐MSCs could survive while secreting more angiogenic factors like platelet‐derived growth factor and VEGF and upregulating the expression of essential transcription factors like HIF‐1α. Additionally, the researchers constructed a chitosan‐collagen scaffold that included simvastatin. This scaffold had great porosity, adequate mechanical strength, and elasticity comparable to that of human skin, and simvastatin was released in a regulated manner from the scaffold. 228
The delivery of MSCs derived from rat epidermis via this scaffold led to an improved wound healing rate, boosted vascularization, increased viability, and expanded MSCs in diabetic rat wounds in comparison with a control group that received no therapy and a group that received just the scaffold. 229 In an additional investigation, a sponge scaffold made of polyurethane and glycol chitosan was used to insert rat‐derived AD‐MSCs into STZ‐induced diabetic rat injuries. This method, when utilized together with acupuncture, generated complementary immune‐modulating outcomes, which led to better wound healing with full re‐epithelialization in a span of 8 days compared to what the AD‐MSC alone group experienced. 230 Additionally, sponge scaffolds may couple with growth factors to function as a means of delivery for cells. When BM‐MSCs from BALB/c mice were delivered via alginate‐chitosan‐sponge scaffolds conjugated with EGF, wound healing in diabetic rats was significantly better than in the no‐treatment control and MSC‐alone groups. This was because more granulation tissue developed, more collagen accumulated, and more blood vessels grew. 231
7.1.3. MSC delivery through fibrous scaffolds in diabetic models
Fibrous scaffolds are primarily created using an electrospinning technique to produce 3D constructions made of fibers at the micro‐ or nanoscale level to imitate the structure of normal human cells. 232 , 233 , 234 Vascular, cartilage, bone, skin, and neurological tissue engineering are a few examples of the many applications of fibrous scaffolds. 235 Because of their potential to operate as a structural model, enhance cell‐to‐cell and cellular‐matrix communications, and influence cellular behavior and function, fibrous scaffolds have been the subject of an increasing body of research in the discipline of wound healing in recent years. 235
When it comes to diabetic wound healing, fibrous scaffolds have been employed to transport MSCs for diabetic wound repair. Using gelatin, pluronic‐F‐127, and polycaprolactone, Chen et al. 177 have created a 3‐dimensional scaffold for the delivery of mouse BM‐MSC. In the wound site of diabetic mice, this fibrous scaffold‐MSC composite improved granulation tissue development, blood vessel formation, and the accumulation of collagen in comparison to no‐treatment and scaffold‐alone controls. 177 To enhance the development of new vessels and healing of wounds in diabetic mice, an integrated electrospinning nanofiber scaffold composed of 10% collagen, 10% silk, and 80% polylactic acid has been created as a cell transporter for supplying HO‐1‐overexpressing human BM‐MSCs to the injury site. 191 In addition, a polycaprolactone and aloe vera‐based fibrous scaffold was developed for injecting human UC‐MSCs or their conditioned media into the site of damage in db/db mice. After 28 days of implantation into the ulcers, both therapies showed rapid wound healing, re‐epithelialization, and a rise in the number of sebaceous glands and hair follicles, with no substantial distinction seen between the two medications. 236 After receiving both medications, the wound showed signs of healthy keratinocytes and increased production of ICAM‐1, VEGF‐A, and tissue inhibitor matrix metalloproteinase 1 at days 14 and 28. Interestingly, in a wound model using db/db mice, a silk fibroin scaffold loaded with human AD‐MSCs accelerated wound healing from 15 to 17 days to just 10 days. 237
7.1.4. MSC delivery through decellularized grafts in diabetic models
Decellularized transplants are often obtained from organs or tissues by undergoing decellularization processes that are either enzymatic (for instance, trypsin and pepsin), mechanical (e.g., force and freezing), or chemical (such as acid and Triton) to eliminate the cellular components of the donor material. 238 Organs and tissues that are frequently utilized include adipose tissue, 239 skin, 240 Wharton's jelly, 241 and in vitro cultivated cells. 242 Unlike other synthetic scaffolds, decellularized transplants still have their original ECM constituents, such as fibronectin, collagen, elastin, and laminin, and their original anatomical structure. They also do not stimulate the immune system. 239 , 242 These benefits are very necessary in the process of identifying and constructing scaffolds that may be implanted in diabetes‐related wounds. Decellularized grafts can restore the damaged ECM of diabetic ulcers by supplying ECM proteins like proteoglycans, collagen, glycoproteins, and glycosaminoglycans, thereby facilitating infiltration of host cells, modulation of the immune system reaction, promotion of new blood vessel formation, and granulation tissue development. 243 , 244
Decellularized grafts have been extensively studied as a potential MSC transport system. Among these investigations, one found that diabetic rat AD‐MSCs implanted on a decellularized transplant released cytokines (such as VEGF, TGF‐β, HGF, and bFGF) that increased the movement and development of fibroblasts, leading to better wound healing. 245 In a different investigation, a graft made from decellularized mouse skin was utilized to transfer mouse BM‐MSCs. In comparison to untreated controls, a full‐thickness cutaneous wound area in diabetic mice treated with this bio complex showed higher rates of wound closure and dramatically faster angiogenesis and re‐epithelialization. The use of a modern multiphoton microscope revealed an enhanced production of collagen type I fibers throughout diabetic wound repair, suggesting a potential pathway for wound recovery. 240 High levels of stability and robust mechanical qualities have been shown in a decellularized dermal matrix that incorporates reduced graphene oxide as a scaffold for cell administration. Mice BM‐MSCs have been effectively transferred into a wound model of a diabetic mouse using this decellularized graft. This creates an ideal environment for stem cells to stick together, move around, and multiply, as well as strong blood vessel growth and collagen buildup. 246 In addition, the delivery of human UC‐MSCs by a decellularized dermal differentiation matrix to diabetes‐related rat lesions demonstrated that the growth and discrimination of human UC‐MSCs on the decellularized dermal matrix were controlled by triggered Wnt signaling pathways. 247
7.2. MSC delivery through bionanomaterials in diabetic models
Nanomaterials are distinguished by their exceptional physicochemical and biological characteristics. When compared to other types of wound healing materials, it has been demonstrated that nanomaterials can promote quicker wound repair. They have antioxidant and antimicrobial properties, exhibit specific anti‐ and proinflammatory effects, and promote angiogenesis. They can be used by direct application to the wound or assimilated into scaffolds to generate hydrogel matrices or nanocomposites. All these properties make them useful for promoting wound healing in a variety of ways. Due to their higher surface‐to‐volume ratio, nanomaterials can be employed for more than just drug transfer. They have been used in stem cell treatments to speed up wound recovery. 248
It is possible to enhance the development of skin stem cells into fibroblasts and keratinocytes through the application of therapeutic nanomaterials during the wound‐healing process. 249 In their in vitro trials, Danková et al. 250 succeeded in promoting the growth of MSCs by mixing polycaprolactone nanofibers with integrated magnetic nanoparticles. In another investigation, it was shown that polycaprolactone nanofibers, when mixed with plant‐derived extracts from Myrtus communis, exhibited a protective effect on skin MSCs that had been aged by UV rays. Regarding the various alternatives for skin regeneration, this knowledge might be extremely valuable. 248
Başaran et al. 251 created a bionanomaterial with the potential for regeneration of the skin by encapsulating heparin in poly (lactic‐co‐glycolic acid) (PLGA) nanoparticles and incorporating it into sericin and gelatin nanofibers. This method demonstrated controlled medication release as well as a high capacity to retain water and minimal degradation rates. As a consequence of combining this system with the biopolymers gelatin and sericin, a potent medication delivery system was created for topical application in skin regeneration. 251 However, at this time, no therapeutic application of nanofibers for accelerating the healing of wounds in humans has yet been reported. To this day, it has not been able to achieve complete rejuvenation of the structural and functional qualities of the skin. Despite this, there is still a significant amount of work to be done on the research and enhancement of nanofibers for use in tissue engineering. This is because nanofibers offer a potentially effective therapeutic method. 252
8. ADVANTAGES OF MSC‑BASED THERAPY
The benefits of MSCs in comparison to those of other kinds of stem cells are outlined in the following paragraphs: unlike ESCs, which must be derived from human embryos, MSCs may be produced from adult cells and, hence, do not create ethical concerns when used for therapeutic purposes. 253 , 254 , 255 , 256 ESCs produced from embryos are allogeneic cells for the recipient and hence susceptible to immunological rejection. 257 However, MSCs display an immune privileged condition with little immunogenicity. 258 , 259 , 260 Because they express extremely low numbers of MHC class I antigens and lack the expression of MHC class II antigens or T cell costimulatory molecules. 261 , 262 MSCs are recognized for their capability to suppress immune system reactions, which may explain why they have been effective in treating graft‐versus‐host reactions and other immunological‐mediated diseases. 263 MSCs are common and may be simply isolated from a diversity of tissues, like BM, adipose tissue, Wharton's jelly, and UCs, through a simple extraction method. 264 , 265 MSCs have a tendency to migrate to locations of tissue damage or tumors, regardless of whether they are supplied locally or systemically. 266 , 267 , 268 , 269 , 270 , 271 , 272 Since MSCs are capable of being genetically modified in vitro, they have the potential to act as carrier cells for the transfer of genes. 259 Because of their capabilities for chemotrophic migration and the release of cytokines, MSCs facilitate the transport of a wide range of therapeutic medicines to cancerous, inflamed, or wounded tissues with minimal adverse consequences. 259 , 271 , 273 , 274 There was no substantial increase in toxicity associated with multiple administrations of fresh or cryopreserved MSCs. 275 , 276 Even with allogeneic MSC transplants, clinical studies haven't found any major side effects like organ system problems (such as cardiovascular and respiratory insufficiencies, etc.) or death right away or during follow‐up procedures after MSC infusions. 276 , 277 , 278 , 279 , 280 , 281 Short‐term investigations show that MSC therapy is harmless and tolerated properly, although the long‐term effects of this therapy are unclear. 282 , 283 When compared to ESCs and iPSCs, the likelihood of malignancy or teratoma development upon administration of MSCs is much lower. 284 , 285 , 286 Also, it has been revealed that MSCs can cross the blood‐brain boundary in a laboratory animal model of encephalopathy. 287
9. OBSTACLES AND CHALLENGES WITH MSCs TRANSPLANTATION THERAPY
MSC transplantation therapy is a relatively new approach that has shown substantial potential for the medical management of a number of illnesses, such as DFU. Unfortunately, there are still multiple significant challenges that need to be conquered before it can be used on patients in a clinical context.
The identification of the donors and tissues that would provide the MSCs of the greatest possible quality for usage in the treatment of certain patients is one of these challenges. The quality of MSCs might vary significantly from one donor to another. 288 , 289 Secondly, the absence of standardized protocols for dealing with MSCs. 290 There is a substantial level of variation in the quality of the cells, which is the principal obstacle in the way of standardizing the procedures for MSC. 291 , 292 , 293 In addition, aggressive separation procedures and a tedious cell culture approach. 294 MSCs have a poor rate of proliferation in vitro, which makes it difficult to scale up. For MSC therapy to be effective, it often needs a substantial number of cells to be administered. 295 There was evidence of the short‐term survival of MSCs that had been injected exogenously in vivo. 296 , 297 , 298 After receiving systematic therapy, there was either ineffective recruitment or adherence to the desired cells, as well as a decline in the efficiency of the transplantation. 278 , 296 , 299 , 300 , 301 MSC treatment has been found to be safe in several investigations, 302 , 303 but MSCs should not be used widely in healthcare settings until serious safety issues, like genetic abnormalities, unintended growth of transplanted MSCs, and the possibility of cancer formation in vivo, have been thoroughly investigated. 304 , 305 Although preclinical and clinical investigations of MSC‐based treatment have revealed promising results, these results are sometimes inconsistent and even contradictory. 306 , 307
9.1. General limitations and side effects of MSC‐based therapy
Several studies on MSCs’ therapeutic value have shown encouraging findings. However, in recent years, there have been several reports of unfavorable results and adverse effects after MSC therapy. 308 Before the use of allogeneic MSCs, patients are screened regularly to look for any signs of viruses, like the human immunodeficiency virus. However, transferred MSCs have the possibility of containing genes from other types of viruses. 309 According to research that was conducted by Sundin et al., 310 MSCs from various kinds of organs have the potential to host persistent viruses. It was discovered that the healthy participants’ MSCs carried viral genetic information from the B19 parvovirus. In addition to this, viral DNA was found in samples taken from human BM. 311 The most widespread viruses that infect MSCs are B19V, Merkel cell polyomavirus, human herpesvirus 7, Tornado tenovirus, and Epstein‐Barr virus. 308 , 311 Additionally, a number of viruses, such as the avian influenza A H5N1 strain and the respiratory syncytial virus, may infect MSCs. 312 , 313 A further dangerous concern for cultured cells is contamination with mycoplasma. Numerous things, such as interruptions in laminar flow, insufficient sterilization cycles, inappropriate lab attire, and higher antibiotic doses in culture, might lead to mycoplasma infection. 314 Although the probability of xenocontamination is very remote, it is often linked to the presence of supportive xenogenic chemicals in cell products. Cryopreservation 315 , 316 and expanding media that contain human serum albumin, the patient's plasma, or fetal bovine serum are the most common causes of contamination. 317 , 318
There is a large amount of variation in culture‐expanded MSCs concerning the shape of cells, physiological functions, and activities; this variation is a key contributor to MSC heterogeneity. 319 As a consequence of this, cell‐derived compounds and experimental models that can be duplicated under the same conditions may have affinities that are difficult to anticipate. 308 Røsland et al. 320 found that following a cultural period of a month, around 46% of human MSCs experienced a spontaneous transition into malignant cells. Several investigations have shown that extended cell culture is linked to a rise in chromosomal anomalies. Froelich et al. 321 found that the incidence of chromosomal abnormalities in AD‐MSCs increased dramatically at passage 5.
A meta‐analysis of prospective controlled studies found a substantial link between MSCs and short‐term fever after intravascular injection. 276 Within 48–72 h of receiving an MSC intra‐articular injection, patients experienced minor effusion and increased local discomfort. 322 Multiple animals and in vitro investigations have demonstrated that MSC injections promote tumor growth through a variety of mechanisms, such as the release of proangiogenic mediators and the suppression of the immune system. 323 , 324 Microthrombosis may manifest itself after MSC injections. Preliminary investigations have revealed that the majority of MSCs that are injected intravenously get stuck in the tiny capillaries of the lung parenchyma. 325 , 326 , 327 , 328 In treated individuals, this condition may cause multifocal pulmonary atelectasis and thrombus formation. 325 , 329 , 330 The glomeruli and tubules of the kidney are more likely to be damaged when an excessive amount of cell therapy is administered. 331 Interstitial cell fibrosis and tubule atrophy were seen in a patient with chronic renal disease who had received an injection of autologous MSCs derived from adipose tissue. 332 Experiments have revealed that MSCs may differentiate into cells called myofibroblasts, which may result in the development of fibrous tissue. 333 Further systemic administration of these cells triggered the onset of serious negative consequences owing to the immunosuppressive characteristics of the MSCs. For instance, after an allogeneic HSC transplant, MSC therapy is linked to a higher likelihood of mortality from pneumonia. 334 The safety and effectiveness of MSC therapy for treating COVID‐19 have been challenged due to the impact that MSCs have on blood coagulation. 335 , 336 In general, the unfavorable effects that were observed also included heart failure, allergic dermatitis, and reduced liver function, all of which are common aftereffects of life‐threatening pneumonia. 337 , 338
9.2. Specific limitations and side effects of MSCs as a treatment for DFU
MSCs have shown promising effectiveness in treating DFU in both human and animal investigations. 104 , 115 , 339 However, recent clinical investigations have shown that MSCs in DFU may cause some unwanted side effects, including diarrhea, urticaria, elevated blood creatinine levels, nausea, high body temperature, and vomiting. 340 , 341 The therapeutic impact of stem cells may decrease with repeated passage in vitro because of a loss of multidirectional differentiation capacity and paracrine function. 152 Embryonic stem cells (ESCs) have a powerful capacity for proliferation but only a limited capacity for differentiation. The insertion of these cells may provoke a rejection response from the immune system and accelerate the growth of tumors. As a result, ESCs need to be kept as far away from DFU therapy as possible. 342 , 343 , 344 Furthermore, researchers have found that increasing the dose of locally injected stem cells to enhance the effectiveness of the healing process may also raise the likelihood of tumor formation. 345
10. CONCLUSION AND FUTURE PROSPECTIVE
The complex etiology of diabetic wounds and decreased wound healing abilities in diabetic persons continue to be an obstacle for the healthcare system and all medical professionals worldwide. DFU is a recent alarm worldwide as it significantly increases the worldwide amputation rate. It was documented that around one amputation occurs each second, and 84% of these amputations are due to DFU. 17 MSCs possess a highly beneficial impact on the management of DFU, and they also have the benefit of being administrated in conjunction with other therapies to more effectively treat resistant DFU. Several studies reported a promising effect of MSCs to boost and enhance the healing process in DFU patients. Although the safety and effectiveness of MSC therapy in managing DFU not clearly provided. Given the information provided in this review, we suggest that future studies are needed to understand the treatment processes, efficacy assessments, and personal selections of stem cell sources. However, there are several challenges and side effects regarding MSCs. So future research should focus on combating these drawbacks and optimizing MSCs to be more applicable in preliminary and clinical studies.
AUTHOR CONTRIBUTIONS
Helal F Hetta: conceptualization; data curation; formal analysis; funding acquisition; validation; writing—review & editing. Alaa Elsaghir: conceptualization; data curation; investigation; methodology; resources; supervision; writing—original draft. Victor Coll Sijercic: formal analysis; investigation; visualization; writing—original draft. Mahad S Akhtar: data curation; investigation; methodology; software; writing—original draft; writing—review & editing. Sayed A Gad: data curation; formal analysis; investigation; software; validation; writing—original draft. Avinash Moses: investigation; methodology; project administration; supervision; writing—original draft. Mahlet S Zeleke: conceptualization; data curation; methodology; project administration; resources; visualization; writing—original draft. Fawaz E Alanazi: writing—review & editing. Abdulrahman K Ahmed: data curation; formal analysis; investigation; validation; writing—original draft. Yasmin N Ramadan: formal analysis; methodology; project administration; resources; software; validation; writing—original draft.
CONFLICT OF INTEREST STATEMENT
The authors declare no conflicts of interest.
TRANSPARENCY STATEMENT
The lead author Mahlet S. Zeleke affirms that this manuscript is an honest, accurate, and transparent account of the study being reported; that no important aspects of the study have been omitted; and that any discrepancies from the study as planned (and, if relevant, registered) have been explained.
ACKNOWLEDGMENTS
All figures were made by Biorender.
List of Abbreviations
1.
| ABI: Ankle Brachial Index |
|---|
| AF‐MSC‐hypoCM: hypoxic‐conditioned Medium from AF‐MSCs |
| AF‐MSCs: amniotic fluid‐derived MSCs |
| AKT: protein‐serine‐threonine kinase |
| AMSCs: adipose tissue mesenchymal stem cells |
| BM: bone marrow |
| BM‐MSCs: bone marrow mesenchymal stem cells |
| CTPs: cellular‐ and tissue‐based products |
| DFU: diabetic foot ulcer |
| EBV: Epstein‐Barr virus |
| ECM: extracellular matrix |
| EGF: epidermal growth factor |
| EPCs: endothelial progenitor cells |
| ERK: extracellular signal‐regulated kinases |
| ESCs: embryonic stem cells |
| FBS: fetal bovine syndrome |
| HA: hyaluronic acid |
| HBOT: hyperbaric oxygen therapy |
| HGF: hepatocyte growth factor |
| HHV‐7: human herpesvirus 7 |
| HIV: human immunodeficiency virus |
| HOT: hyperbaric oxygen therapy |
| hUC‐MSCs: human umbilical cord mesenchymal stem cells |
| IGF: insulin‐like growth factor |
| ISCs: insulin‐secreting cells |
| KGF: keratinocyte growth factor |
| MCPyV: Merkel cell polyomavirus |
| MRA: magnetic resonance angiography |
| MRSA: Methicillin‐Resistant Staphylococcus Aureus |
| MSC: mesenchymal stem cells |
| NLRP3: NOD‐like receptor pyrin domain containing 3 inflammasome |
| NPWT: negative pressure wound therapy |
| PAD: peripheral artery disease |
| PDGF: platelet‐derived growth factor |
| PD‐MSCs: placenta mesenchymal stem cells |
| PGF: placental growth factor |
| PLGA: poly(lactic‐co‐glycolic acid) |
| PN: peripheral neuropathy |
| PRF: platelet‐rich fibrin |
| PRP: platelet‐rich plasma |
| ROS: reactive oxygen species |
| RSV: respiratory syncytial virus |
| SDF: stromal cell‐derived factor |
| SF: silk fibroin |
| SSCs: skin stem cells |
| STC: stanniocalcin |
| TCC: total contact casting |
| TcO2: transcutaneous partial pressure of oxygen |
| TGF: transforming growth factor |
| Th: T helper |
| TIMP‐1: tissue inhibitor matrix metalloproteinase 1 |
| Treg: T regulatory cells |
| TSG‐6: TNF‐α‐stimulated Gene 6 Protein |
| TTV: tornado tenovirus |
| UC: umbilical cord |
| vCPM: cryopreserved placental membrane containing viable cells |
| VEGF: vascular endothelial growth factor |
Hetta HF, Elsaghir A, Sijercic VC, et al. Mesenchymal stem cell therapy in diabetic foot ulcer: an updated comprehensive review. Health Sci Rep. 2024;7:e2036. 10.1002/hsr2.2036
Abdulrahman K. Ahmed and Yasmin N. Ramadan contributed equally to this work.
DATA AVAILABILITY STATEMENT
All generated data are included in this manuscript.
REFERENCES
- 1. Khadem Haghighian H, Naimi AF, Gargari BP, Ali‐Asgharzadeh A, Nemati A. Effect of cinnamon supplementation on blood glucose and lipid levels in type2 diabetic patients. Diabetol Int. 2011;2:1. [Google Scholar]
- 2. Saleh A, Anwar MM, Zayed AE, et al. Impact of Ginkgo biloba extract and magnetized water on the survival rate and functional capabilities of pancreatic β‐cells in type 2 diabetic rat model. Diabetes Metab Syndr Obes. 2019;12:1339‐1347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Abd Ellah NH, Ahmed EA, Abd‐Ellatief RB, Ali MF, Zahran AM, Hetta HF. Metoclopramide nanoparticles modulate immune response in a diabetic rat model: association with regulatory T cells and proinflammatory cytokines. Int J Nanomedicine. 2019;14:2383‐2395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Du S, Zeugolis DI, O'Brien T. Scaffold‐based delivery of mesenchymal stromal cells to diabetic wounds. Stem Cell Res Ther. 2022;13(1):1‐19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Khalaf FR, Fahmy HM, Ibrahim AK, et al. Does a diabetic retinopathy educational program raise awareness among elderly diabetic patients? Diabetes Metab Syndr Obes. 2019;12:1867‐1875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Khamaisi M, Balanson SE. Stem cells for diabetes complications: a future potential cure. Rambam Maimonides Med J. 2017;8(1):e0008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Yazdanpanah L. Literature review on the management of diabetic foot ulcer. World J Diabetes. 2015;6(1):37‐53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Armstrong DG, Boulton AJM, Bus SA. Diabetic foot ulcers and their recurrence. N Engl J Med. 2017;376(24):2367‐2375. [DOI] [PubMed] [Google Scholar]
- 9. Falanga V. Wound healing and its impairment in the diabetic foot. The Lancet. 2005;366(9498):1736‐1743. [DOI] [PubMed] [Google Scholar]
- 10. Verdi J, Shirian S, Saleh M, Haghighian HK, Kavianpour M. Mesenchymal stem cells regenerate diabetic foot ulcers: a review article. World J Plast Surg. 2022;11(1):12‐22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Zhang J, Zhao B, Wei W, et al. Prospective, randomized, and controlled study of a human umbilical cord mesenchymal stem cell injection for treating diabetic foot ulcers. J Vis Exp. 2023;3(193):e65045. [DOI] [PubMed] [Google Scholar]
- 12. Hou L, Zhang X, Du H. Advances in mesenchymal stromal cells and nanomaterials for diabetic wound healing. Diabetes Metab Res Rev. 2023;39(4):e3638. [DOI] [PubMed] [Google Scholar]
- 13. Dama G, Du J, Zhu X, Liu Y, Lin J. Bone marrow‐derived mesenchymal stem cells: a promising therapeutic option for the treatment of diabetic foot ulcers. Diabetes Res Clin Pract. 2023;195:110201. [DOI] [PubMed] [Google Scholar]
- 14. Sasaki M, Abe R, Fujita Y, Ando S, Inokuma D, Shimizu H. Mesenchymal stem cells are recruited into wounded skin and contribute to wound repair by transdifferentiation into multiple skin cell type. J Immunol. 2008;180(4):2581‐2587. [DOI] [PubMed] [Google Scholar]
- 15. Liu ZJ, Zhuge Y, Velazquez OC. Trafficking and differentiation of mesenchymal stem cells. J Cell Biochem. 2009;106(6):984‐991. [DOI] [PubMed] [Google Scholar]
- 16. Yu X, Liu P, Li Z, Zhang Z. Function and mechanism of mesenchymal stem cells in the healing of diabetic foot wounds. Front Endocrinol. 2023;14:1099310. 10.3389/fendo.2023.1099310 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Lopes L, Setia O, Aurshina A, et al. Stem cell therapy for diabetic foot ulcers: a review of preclinical and clinical research. Stem Cell Res Ther. 2018;9(1):188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. The Diabetic Foot. International Diabetes Federation ; 2020.
- 19. Meloni M, Izzo V, Giurato L, Lázaro‐Martínez JL, Uccioli L. Prevalence, clinical aspects and outcomes in a large cohort of persons with diabetic foot disease: comparison between neuropathic and ischemic ulcers. J Clin Med. 2020;9(6):1780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Hoffstad O, Mitra N, Walsh J, Margolis DJ. Diabetes, lower‐extremity amputation, and death. Diabetes Care. 2015;38(10):1852‐1857. [DOI] [PubMed] [Google Scholar]
- 21. Pendsey S. Understanding diabetic foot. Int J Diabetes Dev Ctries. 2010;30(2):75‐79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. van Netten JJ, Bus SA, Apelqvist J, et al. Definitions and criteria for diabetic foot disease. Diabetes Metab Res Rev. 2020;36:e3268. [DOI] [PubMed] [Google Scholar]
- 23. Boulton AJM. The pathway to foot ulceration in diabetes. Med Clin North Am. 2013;97(5):775‐790. [DOI] [PubMed] [Google Scholar]
- 24. Yotsu RR, Pham NM, Oe M, et al. Comparison of characteristics and healing course of diabetic foot ulcers by etiological classification: neuropathic, ischemic, and neuro‐ischemic type. J Diabetes Complications. 2014;28(4):528‐535. [DOI] [PubMed] [Google Scholar]
- 25. Shah P, Inturi R, Anne D, et al. Wagner's classification as a tool for treating diabetic foot ulcers: our observations at a suburban teaching hospital. Cureus. 2022;14(1):e21501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Pop‐Busui R, Boulton AJM, Feldman EL, et al. Diabetic neuropathy: a position statement by the American diabetes association. Diabetes Care. 2016;40(1):136‐154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Chaturvedi S, Agrawal S, Garg A, Rastogi V. Potential of nanoencapsulated quercetin topical formulations in the management of diabetic foot ulcer. Revista Brasileira de Farmacognosia. 2023;33(3):484‐501. [Google Scholar]
- 28. Corbett C, Jolley J, Barson E, Wraight P, Perrin B, Fisher C. Cognition and understanding of neuropathy of inpatients admitted to a specialized tertiary diabetic foot unit with diabetes‐related foot ulcers. Int J Low Extrem Wounds. 2019;18(3):294‐300. [DOI] [PubMed] [Google Scholar]
- 29. Perez‐Favila A, Martinez‐Fierro ML, Rodriguez‐Lazalde JG, et al. Current therapeutic strategies in diabetic foot ulcers. Medicina. 2019;55(11):714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Deanfield JE, Daggett PR, Harrison MJG. The role of autonomic neuropathy in diabetic foot ulceration. J Neurol Sci. 1980;47(2):203‐210. [DOI] [PubMed] [Google Scholar]
- 31. Spallone V, Menzinger G. Autonomic neuropathy: clinical and instrumental findings. Clinical Neuroscience (New York, NY). 1997;4(6):346‐358. [PubMed] [Google Scholar]
- 32. Skopljak A, Sukalo A, BaticMujanovic O, Becirevic M, TiricCampara M, Zunic L. Assessment of diabetic polyneuropathy and plantar pressure in patients with diabetes mellitus in prevention of diabetic foot. Med Arch. 2014;68(6):389‐393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Schaper NC, Andros G, Apelqvist J, et al. Diagnosis and treatment of peripheral arterial disease in diabetic patients with a foot ulcer. A progress report of the international working group on the diabetic foot. Diabetes Metab Res Rev. 2012;28:218‐224. [DOI] [PubMed] [Google Scholar]
- 34. Association AD . Peripheral arterial disease in people with diabetes. Diabetes Care. 2003;26(12):3333‐3341. [DOI] [PubMed] [Google Scholar]
- 35. Janka HU, Standl E, Mehnert H. Peripheral vascular disease in diabetes mellitus and its relation to cardiovascular risk factors: screening with the Doppler ultrasonic technique. Diabetes Care. 1980;3(2):207‐213. [DOI] [PubMed] [Google Scholar]
- 36. Noor S, Zubair M, Ahmad J. Diabetic foot ulcer—a review on pathophysiology, classification and microbial etiology. Diabetes Metab Syndr. 2015;9(3):192‐199. [DOI] [PubMed] [Google Scholar]
- 37. Hunter P, Greco E, Cross K, Perry J. Topical oxygen therapy shifts microbiome dynamics in chronic diabetic foot ulcers. Wounds. 2020;32(3):81‐85. [PubMed] [Google Scholar]
- 38. Dinh TL, Veves A. A review of the mechanisms implicated in the pathogenesis of the diabetic foot. Int J Low Extrem Wounds. 2005;4(3):154‐159. [DOI] [PubMed] [Google Scholar]
- 39. Salah M, Badr G, Hetta HF, Khalifa WA, Shoreit AA. Fig latex inhibits the growth of pathogenic bacteria invading human diabetic wounds and accelerates wound closure in diabetic mice. Sci Rep. 2022;12(1):21852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Amjad SS, Zafar J, Shams N. Bacteriology of diabetic foot in tertiary care hospital; frequency, antibiotic susceptibility and risk factors. J Ayub Med Coll Abbottabad. 2017;29(2):234‐240. [PubMed] [Google Scholar]
- 41. Sekhar S, Vyas N, Unnikrishnan M, Rodrigues G, Mukhopadhyay C. Antimicrobial susceptibility pattern in diabetic foot ulcer: a pilot study. Ann Med Health Sci Res. 2014;4(5):742‐745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Kateel R, Augustine A, Prabhu S, Ullal S, Pai M, Adhikari P. Clinical and microbiological profile of diabetic foot ulcer patients in a tertiary care hospital. Diabetes Metab Syndr. 2018;12(1):27‐30. [DOI] [PubMed] [Google Scholar]
- 43. Bandyk DF. The diabetic foot: pathophysiology, evaluation, and treatment. Semin Vasc Surg. 2018;31(2‐4):43‐48. [DOI] [PubMed] [Google Scholar]
- 44. Abd Ellah NH, Abd El‐Aziz FEZA, Abouelmagd SA, Abd El‐Hamid BN, Hetta HF. Spidroin in carbopol‐based gel promotes wound healing in earthworm's skin model. Drug Dev Res. 2019;80(8):1051‐1061. [DOI] [PubMed] [Google Scholar]
- 45. Baltzis D, Eleftheriadou I, Veves A. Pathogenesis and treatment of impaired wound healing in diabetes mellitus: new insights. Adv Ther. 2014;31(8):817‐836. [DOI] [PubMed] [Google Scholar]
- 46. Liu M, Wei X, Zheng Z, et al. Recent advances in nano‐drug delivery systems for the treatment of diabetic wound healing. Int J Nanomedicine. 2023;18:1537‐1560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Sibbald RG, Elliott JA, Persaud‐Jaimangal R, et al. Wound bed preparation 2021. Adv Skin Wound Care. 2021;34(4):183‐195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Elraiyah T, Domecq JP, Prutsky G, et al. A systematic review and meta‐analysis of débridement methods for chronic diabetic foot ulcers. J Vasc Surg. 2016;63(2 suppl):37S‐45S.e2. [DOI] [PubMed] [Google Scholar]
- 49. Dumville JC, Deshpande S, O'Meara S, Speak K. Hydrocolloid dressings for healing diabetic foot ulcers. Cochrane Database Syst Rev. 2012;(2):Cd009099. [DOI] [PubMed]
- 50. Moura LIF, Dias AMA, Carvalho E, de Sousa HC. Recent advances on the development of wound dressings for diabetic foot ulcer treatment–a review. Acta Biomater. 2013;9(7):7093‐7114. [DOI] [PubMed] [Google Scholar]
- 51. Amirrah IN, Wee MFMR, Tabata Y, et al. Antibacterial‐integrated collagen wound dressing for diabetes‐related foot ulcers: an evidence‐based review of clinical studies. Polymers (Basel). 2020;12(9):2168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Lipsky BA, Peters EJG, Berendt AR, et al. Specific guidelines for the treatment of diabetic foot infections 2011. Diabetes Metab Res Rev. 2012;28(suppl 1):234‐235. [DOI] [PubMed] [Google Scholar]
- 53. Lipsky BA, Berendt AR, Cornia PB, et al. 2012 infectious diseases society of America clinical practice guideline for the diagnosis and treatment of diabetic foot infections. Clin Infect Dis. 2012;54(12):e132‐e173. [DOI] [PubMed] [Google Scholar]
- 54. Raja JM, Maturana MA, Kayali S, Khouzam A, Efeovbokhan N. Diabetic foot ulcer: a comprehensive review of pathophysiology and management modalities. World J Clin Cases. 2023;11(8):1684‐1693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Kwon KT, Armstrong DG. Microbiology and antimicrobial therapy for diabetic foot infections. Infect Chemother. 2018;50(1):11‐20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Albayati MA, Shearman CP. Peripheral arterial disease and bypass surgery in the diabetic lower limb. Med Clin North Am. 2013;97(5):821‐834. [DOI] [PubMed] [Google Scholar]
- 57. Carro GV, Saurral R, Witman EL, et al. Diabetic foot attack. Pathophysiological description, clinical presentation, treatment and outcomes. Medicina. 2020;80(5):523‐530. [PubMed] [Google Scholar]
- 58. Vouillarmet J, Bourron O, Gaudric J, Lermusiaux P, Millon A, Hartemann A. Lower‐extremity arterial revascularization: is there any evidence for diabetic foot ulcer‐healing? Diabetes Metab. 2016;42(1):4‐15. [DOI] [PubMed] [Google Scholar]
- 59. Okoli GN, Rabbani R, Lam OLT, et al. Offloading devices for neuropathic foot ulcers in adult persons with type 1 or type 2 diabetes: a rapid review with meta‐analysis and trial sequential analysis of randomized controlled trials. BMJ Open Diabetes Res Care. 2022;10(3):e002822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Everett E, Mathioudakis N. Update on management of diabetic foot ulcers. Ann NY Acad Sci. 2018;1411(1):153‐165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Begg L, McLaughlin P, Vicaretti M, Fletcher J, Burns J. Total contact cast wall load in patients with a plantar forefoot ulcer and diabetes. J Foot Ankle Res. 2016;9:2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Nalisa DL, Moneruzzaman M, Changwe GJ, et al. Stem cell therapy for diabetic foot ulcers: theory and practice. J Diabetes Res. 2022;2022:1‐12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Mueller MJ, Diamond JE, Sinacore DR, et al. Total contact casting in treatment of diabetic plantar ulcers. Controlled clinical trial. Diabetes Care. 1989;12(6):384‐388. [DOI] [PubMed] [Google Scholar]
- 64. Cardinal M, Eisenbud DE, Armstrong DG, et al. Serial surgical debridement: a retrospective study on clinical outcomes in chronic lower extremity wounds. Wound Repair Regen. 2009;17(3):306‐311. [DOI] [PubMed] [Google Scholar]
- 65. Faglia E, Favales F, Aldeghi A, et al. Adjunctive systemic hyperbaric oxygen therapy in treatment of severe prevalently ischemic diabetic foot ulcer. A randomized study. Diabetes Care. 1996;19(12):1338‐1343. [DOI] [PubMed] [Google Scholar]
- 66. Salama SE, Eldeeb AE, Elbarbary AH, Abdelghany SE. Adjuvant hyperbaric oxygen therapy enhances healing of nonischemic diabetic foot ulcers compared with standard wound care alone. Int J Low Extrem Wounds. 2019;18(1):75‐80. [DOI] [PubMed] [Google Scholar]
- 67. Lalieu RC, Brouwer RJ, Ubbink DT, Hoencamp R, Bol Raap R, van Hulst RA. Hyperbaric oxygen therapy for nonischemic diabetic ulcers: a systematic review. Wound Repair Regen. 2020;28(2):266‐275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Abidia A, Laden G, Kuhan G, et al. The role of hyperbaric oxygen therapy in ischaemic diabetic lower extremity ulcers: a double‐blind randomised‐controlled trial. Eur J Vasc Endovasc Surg. 2003;25(6):513‐518. [DOI] [PubMed] [Google Scholar]
- 69. Hanley ME, Manna B, Hyperbaric Treatment of Diabetic Foot UlcerStatPearls, in. 2023, StatPearls Publishing Copyright © 2023, StatPearls Publishing LLC.: Treasure Island (FL) ineligible companies. Disclosure: Biagio Manna declares no relevant financial relationships with ineligible companies.
- 70. Capó X, Monserrat‐Mesquida M, Quetglas‐Llabrés M, et al. Hyperbaric oxygen therapy reduces oxidative stress and inflammation, and increases growth factors favouring the healing process of diabetic wounds. Int J Mol Sci. 2023;24(8):7040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Rai V, Le H, Agrawal DK. Novel mediators regulating angiogenesis in diabetic foot ulcer healing. Can J Physiol Pharmacol. 2023;101:488‐501. [DOI] [PubMed] [Google Scholar]
- 72. Çimşit M, Uzun G, Yıldız Ş. Hyperbaric oxygen therapy as an anti‐infective agent. Expert Rev Anti Infect Ther. 2009;7(8):1015‐1026. [DOI] [PubMed] [Google Scholar]
- 73. Kranke P, Bennett MH, James M‐S, Schnabel A. Hyperbaric oxygen therapy for chronic wounds. Cochrane Database Syst Rev. 2015;2015(6):Cd004123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Fedorko L, Bowen JM, Jones W, et al. Hyperbaric oxygen therapy does not reduce indications for amputation in patients with diabetes with nonhealing ulcers of the lower limb: a prospective, double‐blind, randomized controlled clinical trial. Diabetes Care. 2016;39(3):392‐399. [DOI] [PubMed] [Google Scholar]
- 75. Liu S, He C, Cai Y, et al. Evaluation of negative‐pressure wound therapy for patients with diabetic foot ulcers: systematic review and meta‐analysis. Ther Clin Risk Manag. 2017;13:533‐544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Borys S, Hohendorff J, Frankfurter C, Kiec‐Wilk B, Malecki MT. Negative pressure wound therapy use in diabetic foot syndrome‐from mechanisms of action to clinical practice. Eur J Clin Invest. 2019;49(4):e13067. [DOI] [PubMed] [Google Scholar]
- 77. Chen L, Zhang S, Da J, et al. A systematic review and meta‐analysis of efficacy and safety of negative pressure wound therapy in the treatment of diabetic foot ulcer. Ann Palliat Med. 2021;10(10):10830‐10839. [DOI] [PubMed] [Google Scholar]
- 78. Bakker K, Apelqvist J, Lipsky BA, Van Netten JJ. The 2015 IWGDF guidance documents on prevention and management of foot problems in diabetes: development of an evidence‐based global consensus. Diabetes Metab Res Rev. 2016;32(suppl 1):2‐6. [DOI] [PubMed] [Google Scholar]
- 79. Doğruel H, Aydemir M, Balci MK. Management of diabetic foot ulcers and the challenging points: an endocrine view. World J Diabetes. 2022;13(1):27‐36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Zhang Z, Liu P, Yang B, et al. Necrotizing fasciitis caused by diabetic foot. Int J Infect Dis. 2021;103:3‐5. [DOI] [PubMed] [Google Scholar]
- 81. Gorden LYT, Ariel YF, Pei H, Meng L, Yi Zhen NG, Graves N. Decision‐making for early major amputation in selected diabetic foot ulcer patients with peripheral vascular disease. Health Care Science. 2022;1(2):58‐68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Friedenstein AJ, Piatetzky‐Shapiro II, Petrakova KV. Osteogenesis in transplants of bone marrow cells. Development. 1966;16(3):381‐390. [PubMed] [Google Scholar]
- 83. Caplan AI. Mesenchymal stem cells. J Orthop Res. 1991;9(5):641‐650. [DOI] [PubMed] [Google Scholar]
- 84. Ferretti E, Hadjantonakis A‐K. Mesoderm specification and diversification: from single cells to emergent tissues. Curr Opin Cell Biol. 2019;61:110‐116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Li M, Ikehara S. Bone‐marrow‐derived mesenchymal stem cells for organ repair. Stem Cells Int. 2013;2013:1‐8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Almalki SG, Agrawal DK. Key transcription factors in the differentiation of mesenchymal stem cells. Differentiation. 2016;92(1‐2):41‐51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. 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]
- 88. Ma S, Xie N, Li W, Yuan B, Shi Y, Wang Y. Immunobiology of mesenchymal stem cells. Cell Death Differ. 2014;21(2):216‐225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Zakrzewski W, Dobrzyński M, Szymonowicz M, Rybak Z. Stem cells: past, present, and future. Stem Cell Res Ther. 2019;10(1):68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Ding DC, Shyu WC, Lin SZ. Mesenchymal stem cells. Cell Transplant. 2011;20(1):5‐14. [DOI] [PubMed] [Google Scholar]
- 91. Jiang Y, Jahagirdar BN, Reinhardt RL, et al. Pluripotency of mesenchymal stem cells derived from adult marrow. Nature. 2002;418(6893):41‐49. [DOI] [PubMed] [Google Scholar]
- 92. Tang Y, Zhou Y, Li HJ. Advances in mesenchymal stem cell exosomes: a review. Stem Cell Res Ther. 2021;12(1):71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Ghorbani F, Movassaghpour AA, Talebi M, Yousefi M, Abbaszadeh H. Renoprotective effects of extracellular vesicles: a systematic review. Gene Reports. 2022;26:101491. [Google Scholar]
- 94. Meirelles LS, Chagastelles PC, Nardi NB. Mesenchymal stem cells reside in virtually all post‐natal organs and tissues. J Cell Sci. 2006;119(11):2204‐2213. [DOI] [PubMed] [Google Scholar]
- 95. Mushahary D, Spittler A, Kasper C, Weber V, Charwat V. Isolation, cultivation, and characterization of human mesenchymal stem cells. Cytometry, Part A. 2018;93(1):19‐31. [DOI] [PubMed] [Google Scholar]
- 96. Wu Y, Hoogduijn MJ, Baan CC, et al. Adipose tissue‐derived mesenchymal stem cells have a heterogenic cytokine secretion profile. Stem Cells Int. 2017;2017:1‐7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Fonseca LN, Bolívar‐Moná S, Agudelo T, et al. Cell surface markers for mesenchymal stem cells related to the skeletal system: a scoping review. Heliyon. 2023;9(2):e13464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Elsaghir A, El‐Sabaa E, Ahmed AK, Abdelwahab SF, Sayed IM, El‐Mokhtar MA. The role of cluster of differentiation 39 (CD39) and purinergic signaling pathway in viral infections. Pathogens (Basel, Switzerland). 2023;12(2):279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Jain P, Perakath B, Jesudason MR, Nayak S. The effect of autologous bone marrow‐derived cells on healing chronic lower extremity wounds: results of a randomized controlled study. Ostomy Wound Manage. 2011;57(7):38‐44. [PubMed] [Google Scholar]
- 100. Falanga V, Iwamoto S, Chartier M, et al. Autologous bone marrow–derived cultured mesenchymal stem cells delivered in a fibrin spray accelerate healing in murine and human cutaneous wounds. Tissue Eng. 2007;13(6):1299‐1312. [DOI] [PubMed] [Google Scholar]
- 101. Dash NR, Dash SN, Routray P, Mohapatra S, Mohapatra PC. Targeting nonhealing ulcers of lower extremity in human through autologous bone marrow‐derived mesenchymal stem cells. Rejuvenation Res. 2009;12(5):359‐366. [DOI] [PubMed] [Google Scholar]
- 102. Amann B, Luedemann C, Ratei R, Schmidt‐Lucke JA. Autologous bone marrow cell transplantation increases leg perfusion and reduces amputations in patients with advanced critical limb ischemia due to peripheral artery disease. Cell Transplant. 2009;18(3):371‐380. [DOI] [PubMed] [Google Scholar]
- 103. Matoba S, Tatsumi T, Murohara T, et al. Long‐term clinical outcome after intramuscular implantation of bone marrow mononuclear cells (Therapeutic Angiogenesis by Cell Transplantation [TACT] trial) in patients with chronic limb ischemia. Am Heart J. 2008;156(5):1010‐1018. [DOI] [PubMed] [Google Scholar]
- 104. Lu D, Chen B, Liang Z, et al. Comparison of bone marrow mesenchymal stem cells with bone marrow‐derived mononuclear cells for treatment of diabetic critical limb ischemia and foot ulcer: a double‐blind, randomized, controlled trial. Diabetes Res Clin Pract. 2011;92(1):26‐36. [DOI] [PubMed] [Google Scholar]
- 105. Xu YF, Wu YX, Wang HM, Gao CH, Xu YY, Yan Y. Bone marrow‐derived mesenchymal stem cell‐conditioned medium ameliorates diabetic foot ulcers in rats. Clinics. 2023;78:100181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Badiavas EV. Treatment of chronic wounds with bone marrow‐derived cells. Arch Dermatol. 2003;139(4):510‐516. [DOI] [PubMed] [Google Scholar]
- 107. Yan C, Xv Y, Lin Z, et al. Human umbilical cord mesenchymal stem cell‐derived exosomes accelerate diabetic wound healing via ameliorating oxidative stress and promoting angiogenesis. Front Bioeng Biotechnol. 2022;10:829868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Qin H, Zhu X, Zhang B, Zhou L, Wang W. Clinical evaluation of human umbilical cord mesenchymal stem cell transplantation after angioplasty for diabetic foot. Exp Clin Endocrinol Diabetes. 2016;124(8):497‐503. [DOI] [PubMed] [Google Scholar]
- 109. 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‐β receptor inhibition. Stem Cell Res Ther. 2021;12(1):434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Kim SM, Kim YH, Jun YJ, Yoo G, Rhie JW. The effect of diabetes on the wound healing potential of adipose‐tissue derived stem cells. Int Wound J. 2016;13(suppl 1):33‐41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Fromer MW, Chang S, Hagaman ALR, et al. The endothelial cell secretome as a novel treatment to prime adipose‐derived stem cells for improved wound healing in diabetes. J Vasc Surg. 2018;68(1):234‐244. [DOI] [PubMed] [Google Scholar]
- 112. Khalil C, Chaker D, Salameh R, et al. Autologous adipose‐derived mesenchymal stem cells embedded in platelet‐rich fibrin in diabetic foot ulcers. OJRM. 2021;10:19‐30. [Google Scholar]
- 113. Cianfarani F, Toietta G, Di Rocco G, Cesareo E, Zambruno G, Odorisio T. Diabetes impairs adipose tissue‐derived stem cell function and efficiency in promoting wound healing. Wound Repair Regen. 2013;21(4):545‐553. [DOI] [PubMed] [Google Scholar]
- 114. Rennert RC, Sorkin M, Januszyk M, et al. Diabetes impairs the angiogenic potential of adipose‐derived stem cells by selectively depleting cellular subpopulations. Stem Cell Res Ther. 2014;5(3):79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Moon K‐C, Suh HS, Kim KB, et al. Potential of allogeneic adipose‐derived stem cell–hydrogel complex for treating diabetic foot ulcers. Diabetes. 2019;68(4):837‐846. [DOI] [PubMed] [Google Scholar]
- 116. Zeng X, Tang Y, Hu K, et al. Three‐week topical treatment with placenta‐derived mesenchymal stem cells hydrogel in a patient with diabetic foot ulcer: a case report. Medicine. 2017;96(51):e9212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Du WJ, Chi Y, Yang ZX, et al. Heterogeneity of proangiogenic features in mesenchymal stem cells derived from bone marrow, adipose tissue, umbilical cord, and placenta. Stem Cell Res Ther. 2016;7(1):163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Meamar R, Ghasemi‐Mobarakeh L, Norouzi MR, Siavash M, Hamblin MR, Fesharaki M. Improved wound healing of diabetic foot ulcers using human placenta‐derived mesenchymal stem cells in gelatin electrospun nanofibrous scaffolds plus a platelet‐rich plasma gel: a randomized clinical trial. Int Immunopharmacol. 2021;101:108282. [DOI] [PubMed] [Google Scholar]
- 119. DaVanzo J, Hartzman A, Surfield C, Dobson A. Cryopreserved placental membrane allograft reduces the likelihood of developing a new or recurring foot ulcer and all‐cause mortality in diabetic patients when compared with other cellular‐and tissue‐based products. Adv Wound Care. 2023;12(4):169‐176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Lingen MW. Role of leukocytes and endothelial cells in the development of angiogenesis in inflammation and wound healing. Arch Pathol Lab Med. 2001;125(1):67‐71. [DOI] [PubMed] [Google Scholar]
- 121. Gillitzer R, Goebeler M. Chemokines in cutaneous wound healing. J Leukocyte Biol. 2001;69(4):513‐521. [PubMed] [Google Scholar]
- 122. Badiavas EV. Treatment of chronic wounds with bone marrow–derived cells. Arch Dermatol. 2003;139(4):510‐516. [DOI] [PubMed] [Google Scholar]
- 123. Blumberg SN, Berger A, Hwang L, Pastar I, Warren SM, Chen W. The role of stem cells in the treatment of diabetic foot ulcers. Diabetes Res Clin Pract. 2012;96(1):1‐9. [DOI] [PubMed] [Google Scholar]
- 124. Honma T, Honmou O, Iihoshi S, et al. Intravenous infusion of immortalized human mesenchymal stem cells protects against injury in a cerebral ischemia model in adult rat. Exp Neurol. 2006;199(1):56‐66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Tang J, Xie Q, Pan G, Wang J, Wang M. Mesenchymal stem cells participate in angiogenesis and improve heart function in rat model of myocardial ischemia with reperfusion. Eur J Cardiothorac Surg. 2006;30(2):353‐361. [DOI] [PubMed] [Google Scholar]
- 126. Al‐Khaldi A, Al‐Sabti H, Galipeau J, Lachapelle K. Therapeutic angiogenesis using autologous bone marrow stromal cells: improved blood flow in a chronic limb ischemia model. Ann Thorac Surg. 2003;75(1):204‐209. [DOI] [PubMed] [Google Scholar]
- 127. Qin HL, Zhu XH, Zhang B, Zhou L, Wang WY. Clinical evaluation of human umbilical cord mesenchymal stem cell transplantation after angioplasty for diabetic foot. Exp Clin Endocrinol Diabetes. 2016;124:497‐503. [DOI] [PubMed] [Google Scholar]
- 128. Rogers I, Casper RF. Umbilical cord blood stem cells. Best Pract Res Clin Obstet Gynaecol. 2004;18(6):893‐908. [DOI] [PubMed] [Google Scholar]
- 129. Zhang Y, Pan Y, Li Y, et al. Exosomes derived from human umbilical cord blood mesenchymal stem cells stimulate regenerative wound healing via transforming growth factor‐β receptor inhibition. Stem Cell Res Ther. 2021;12(1):1‐14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Wen Z, Mai M, Wu T, et al. Mesenchymal stem cell‐derived exosomes ameliorate cardiomyocyte apoptosis in hypoxic conditions through microRNA144 by targeting the PTEN/AKT pathway. Stem Cell Res Ther. 2020;11(1):1‐17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. CellResearch Corporation . Phase 1, open‐label safety study of umbilical cord lining mesenchymal stem cells (CORLICYTE®) to heal chronic diabetic foot ulcers. 2023. [DOI] [PMC free article] [PubMed]
- 132. CellResearch Corporation . CellResearch Corporation announces positive results of Phase I study for CORLICYTE®. 2023 January 2024; Available from: https://clinicaltrials.gov/study/NCT04104451?cond=Diabetic%20Foot%20Ulcer&intr=mesenchymal%20Stem%20Cell&page=2&rank=11
- 133. Qi Y, Ma J, Li S, Liu W. Applicability of adipose‐derived mesenchymal stem cells in treatment of patients with type 2 diabetes. Stem Cell Res Ther. 2019;10(1):1‐13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Hassanshahi A, Hassanshahi M, Khabbazi S, et al. Adipose‐derived stem cells for wound healing. J Cell Physiol. 2019;234(6):7903‐7914. [DOI] [PubMed] [Google Scholar]
- 135. Kokai LE, Marra K, Rubin JP. Adipose stem cells: biology and clinical applications for tissue repair and regeneration. Transl Res. 2014;163(4):399‐408. [DOI] [PubMed] [Google Scholar]
- 136. Palumbo P, Lombardi F, Siragusa G, Cifone M, Cinque B, Giuliani M. Methods of isolation, characterization and expansion of human adipose‐derived stem cells (ASCs): an overview. Int J Mol Sci. 2018;19(7):1897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Gadelkarim M, Abushouk AI, Ghanem E, Hamaad AM, Saad AM, Abdel‐Daim MM. Adipose‐derived stem cells: effectiveness and advances in delivery in diabetic wound healing. Biomed Pharmacother. 2018;107:625‐633. [DOI] [PubMed] [Google Scholar]
- 138. Álvaro‐Afonso FJ, Sanz‐Corbalán I, Lázaro‐Martínez JL, Kakagia D, Papanas N. Adipose‐derived mesenchymal stem cells in the treatment of diabetic foot ulcers: a review of preclinical and clinical studies. Angiology. 2020;71(9):853‐863. [DOI] [PubMed] [Google Scholar]
- 139. Cianfarani F, Toietta G, Di Rocco G, Cesareo E, Zambruno G, Odorisio T. Diabetes impairs adipose tissue–derived stem cell function and efficiency in promoting wound healing. Wound Repair Regen. 2013;21(4):545‐553. [DOI] [PubMed] [Google Scholar]
- 140. Kim SM, Kim YH, Jun YJ, Yoo G, Rhie JW. The effect of diabetes on the wound healing potential of adipose‐tissue derived stem cells. Int Wound J. 2016;13:33‐41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Tobita M, Tajima S, Mizuno H. Adipose tissue‐derived mesenchymal stem cells and platelet‐rich plasma: stem cell transplantation methods that enhance stemness. Stem Cell Res Ther. 2015;6(1):215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Anterogen , Phase 3 clinical study to evaluate efficacy and safety of ALLO‐ASC‐DFU in patients with diabetic wagner grade 2 foot ulcers: a randomized, comparator‐controlled, double‐blind, parallel‐group, multi‐center study. 2020.
- 143. Chang Y‐J, Shih DT, Tseng CP, Hsieh TB, Lee DC, Hwang SM. Disparate mesenchyme‐lineage tendencies in mesenchymal stem cells from human bone marrow and umbilical cord blood. Stem Cells. 2006;24(3):679‐685. [DOI] [PubMed] [Google Scholar]
- 144. Mihu CM, Mihu D, Costin N, Rus Ciucă D, Suşman S, Ciortea R. Isolation and characterization of stem cells from the placenta and the umbilical cord. Rom J Morphol Embryol. 2008;49(4):441‐446. [PubMed] [Google Scholar]
- 145. Fukuchi Y, Nakajima H, Sugiyama D, Hirose I, Kitamura T, Tsuji K. Human placenta‐derived cells have mesenchymal stem/progenitor cell potential. Stem Cells. 2004;22(5):649‐658. [DOI] [PubMed] [Google Scholar]
- 146. Wang H, Chen L, Liu Y, et al. Implantation of placenta‐derived mesenchymal stem cells accelerates murine dermal wound closure through immunomodulation. Am J Transl Res. 2016;8(11):4912‐4921. [PMC free article] [PubMed] [Google Scholar]
- 147. Du WJ, Chi Y, Yang ZX, et al. Heterogeneity of proangiogenic features in mesenchymal stem cells derived from bone marrow, adipose tissue, umbilical cord, and placenta. Stem Cell Res Ther. 2016;7:1‐11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. De Gregorio C, Contador D, Díaz D, et al. Human adipose‐derived mesenchymal stem cell‐conditioned medium ameliorates polyneuropathy and foot ulceration in diabetic BKS db/db mice. Stem Cell Res Ther. 2020;11:1‐21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Shi R, Jin Y, Cao C, et al. Localization of human adipose‐derived stem cells and their effect in repair of diabetic foot ulcers in rats. Stem Cell Res Ther. 2016;7(1):1‐13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Schlosser S, Dennler C, Schweizer R, et al. Paracrine effects of mesenchymal stem cells enhance vascular regeneration in ischemic murine skin. Microvasc Res. 2012;83(3):267‐275. [DOI] [PubMed] [Google Scholar]
- 151. Kinnaird T, Stabile E, Burnett MS, et al. Local delivery of marrow‐derived stromal cells augments collateral perfusion through paracrine mechanisms. Circulation. 2004;109(12):1543‐1549. [DOI] [PubMed] [Google Scholar]
- 152. Yu Q, Qiao G, Wang M, et al. Stem cell‐based therapy for diabetic foot ulcers. Front Cell Dev Biol. 2022;10. 10.3389/fcell.2022.812262 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. El Hage R, Knippschild U, Arnold T, Hinterseher I. Stem cell‐based therapy: a promising treatment for diabetic foot ulcer. Biomedicines. 2022;10(7):1507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Badillo AT, Redden RA, Zhang L, Doolin EJ, Liechty KW. Treatment of diabetic wounds with fetal murine mesenchymal stromal cells enhances wound closure. Cell Tissue Res. 2007;329:301‐311. [DOI] [PubMed] [Google Scholar]
- 155. Kwon DS, Gao X, Liu YB, et al. Treatment with bone marrow‐derived stromal cells accelerates wound healing in diabetic rats. Int Wound J. 2008;5(3):453‐463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Shen L, Zeng W, Wu YX, et al. Neurotrophin‐3 accelerates wound healing in diabetic mice by promoting a paracrine response in mesenchymal stem cells. Cell Transplant. 2013;22(6):1011‐1021. [DOI] [PubMed] [Google Scholar]
- 157. Wan J, Xia L, Liang W, Liu Y, Cai Q. Transplantation of bone marrow‐derived mesenchymal stem cells promotes delayed wound healing in diabetic rats. J Diabetes Res. 2013;2013:1‐11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Diao Y, Lian L, Guo L, et al. A vascular endothelial growth factor activating transcription factor increases the endothelial progenitor cells population and induces therapeutic angiogenesis in a type 1 diabetic mouse with hindlimb ischemia. Chin Med J. 2014;127(20):3623‐3629. [PubMed] [Google Scholar]
- 159. Kerkis I, Haddad MS, Valverde CW, Glosman S. Neural and mesenchymal stem cells in animal models of Huntington's disease: past experiences and future challenges. Stem Cell Res Ther. 2015;6(1):232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160. Yagi H, Soto‐Gutierrez A, Parekkadan B, et al. Mesenchymal stem cells: mechanisms of immunomodulation and homing. Cell Transplant. 2010;19(6‐7):667‐679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Pang Q‐M, Chen SY, Fu SP, et al. Regulatory role of mesenchymal stem cells on secondary inflammation in spinal cord injury. J Inflamm Res. 2022;15:573‐593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Bettelli E, Oukka M, Kuchroo VK. TH‐17 cells in the circle of immunity and autoimmunity. Nature Immunol. 2007;8(4):345‐350. [DOI] [PubMed] [Google Scholar]
- 163. Ohkura N, Sakaguchi S. Transcriptional and epigenetic basis of Treg cell development and function: its genetic anomalies or variations in autoimmune diseases. Cell Res. 2020;30(6):465‐474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164. Luz‐Crawford P, Kurte M, Bravo‐Alegría J, et al. Mesenchymal stem cells generate a CD4+CD25+Foxp3+ regulatory T cell population during the differentiation process of Th1 and Th17 cells. Stem Cell Res Ther. 2013;4:6565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Li X‐Y, Zheng ZH, Li XY, et al. Treatment of foot disease in patients with type 2 diabetes mellitus using human umbilical cord blood mesenchymal stem cells: response and correction of immunological anomalies. Curr Pharm Des. 2013;19(27):4893‐4899. [DOI] [PubMed] [Google Scholar]
- 166. Fleetwood AJ, Lawrence T, Hamilton JA, Cook AD. Granulocyte‐macrophage colony‐stimulating factor (CSF) and macrophage CSF‐dependent macrophage phenotypes display differences in cytokine profiles and transcription factor activities: implications for CSF blockade in inflammation. J Immunol. 2007;178(8):5245‐5252. [DOI] [PubMed] [Google Scholar]
- 167. Mantovani A, Biswas SK, Galdiero MR, Sica A, Locati M. Macrophage plasticity and polarization in tissue repair and remodelling. J Pathol. 2013;229(2):176‐185. [DOI] [PubMed] [Google Scholar]
- 168. Arnold CE, Whyte CS, Gordon P, Barker RN, Rees AJ, Wilson HM. A critical role for suppressor of cytokine signalling 3 in promoting M 1 macrophage activation and function in vitro and in vivo. Immunology. 2014;141(1):96‐110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169. Abdelaziz MH, Abdelwahab SF, Wan J, et al. Alternatively activated macrophages; a double‐edged sword in allergic asthma. J Transl Med. 2020;18(1):58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Al Sadoun H. Macrophage phenotypes in normal and diabetic wound healing and therapeutic interventions. Cells. 2022;11(15):2430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Wynn TA, Chawla A, Pollard JW. Macrophage biology in development, homeostasis and disease. Nature. 2013;496(7446):445‐455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Ogle ME, Segar CE, Sridhar S, Botchwey EA. Monocytes and macrophages in tissue repair: implications for immunoregenerative biomaterial design. Exp Biol Med. 2016;241(10):1084‐1097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. Kimball A, Schaller M, Joshi A, et al. Ly6CHi blood monocyte/macrophage drive chronic inflammation and impair wound healing in diabetes mellitus. Arterioscler Thromb Vasc Biol. 2018;38(5):1102‐1114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Pang J, Maienschein‐Cline M, Koh TJ. Enhanced proliferation of Ly6C+ monocytes/macrophages contributes to chronic inflammation in skin wounds of diabetic mice. J Immunol. 2021;206(3):621‐630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Huang S‐M, Wu CS, Chiu MH, et al. High glucose environment induces M1 macrophage polarization that impairs keratinocyte migration via TNF‐α: an important mechanism to delay the diabetic wound healing. J Dermatol Sci. 2019;96(3):159‐167. [DOI] [PubMed] [Google Scholar]
- 176. Dayan V, Yannarelli G, Billia F, et al. Mesenchymal stromal cells mediate a switch to alternatively activated monocytes/macrophages after acute myocardial infarction. Basic Res Cardiol. 2011;106:1299‐1310. [DOI] [PubMed] [Google Scholar]
- 177. Chen S, Wang H, Su Y, et al. Mesenchymal stem cell‐laden, personalized 3D scaffolds with controlled structure and fiber alignment promote diabetic wound healing. Acta Biomater. 2020;108:153‐167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. 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):1‐15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Liu P, Wang W, Li Z, et al. Ferroptosis: a new regulatory mechanism in osteoporosis. Oxid Med Cell Longevity. 2022;2022:1‐10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. Liu B, Ding FX, Liu Y, et al. Human umbilical cord‐derived mesenchymal stem cells conditioned medium attenuate interstitial fibrosis and stimulate the repair of tubular epithelial cells in an irreversible model of unilateral ureteral obstruction. Nephrology. 2018;23(8):728‐736. [DOI] [PubMed] [Google Scholar]
- 181. Soria B, Martin‐Montalvo A, Aguilera Y, et al. Human mesenchymal stem cells prevent neurological complications of radiotherapy. Front Cell Neurosci. 2019;13:204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182. Al‐Massri KF, Ahmed LA, El‐Abhar HS. Mesenchymal stem cells therapy enhances the efficacy of pregabalin and prevents its motor impairment in paclitaxel‐induced neuropathy in rats: role of Notch1 receptor and JAK/STAT signaling pathway. Behav Brain Res. 2019;360:303‐311. [DOI] [PubMed] [Google Scholar]
- 183. Feng J, Lu C, Dai Q, Sheng J, Xu M. SIRT3 facilitates amniotic fluid stem cells to repair diabetic nephropathy through protecting mitochondrial homeostasis by modulation of mitophagy. Cell Physiol Biochem. 2018;46(4):1508‐1524. [DOI] [PubMed] [Google Scholar]
- 184. Oh JY, Ko JH, Lee HJ, et al. Mesenchymal stem/stromal cells inhibit the NLRP3 inflammasome by decreasing mitochondrial reactive oxygen species. Stem Cells. 2014;32(6):1553‐1563. [DOI] [PubMed] [Google Scholar]
- 185. Guillén MI, Platas J, Pérez del Caz MD, Mirabet V, Alcaraz MJ. Paracrine anti‐inflammatory effects of adipose tissue‐derived mesenchymal stem cells in human monocytes. Front Physiol. 2018;9:661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Raffaghello L, Bianchi G, Bertolotto M, et al. Human mesenchymal stem cells inhibit neutrophil apoptosis: a model for neutrophil preservation in the bone marrow niche. Stem Cells. 2008;26(1):151‐162. [DOI] [PubMed] [Google Scholar]
- 187. Li X, Xie X, Lian W, et al. Exosomes from adipose‐derived stem cells overexpressing Nrf2 accelerate cutaneous wound healing by promoting vascularization in a diabetic foot ulcer rat model. Exp Mol Med. 2018;50(4):1‐14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Guo J, Hu Z, Yan F, et al. Angelica dahurica promoted angiogenesis and accelerated wound healing in db/db mice via the HIF‐1α/PDGF‐β signaling pathway. Free Radic Biol Med. 2020;160:447‐457. [DOI] [PubMed] [Google Scholar]
- 189. Zhang E, Gao B, Yang L, Wu X, Wang Z. Notoginsenoside Ft1 promotes fibroblast proliferation via PI3K/Akt/mTOR signaling pathway and benefits wound healing in genetically diabetic mice. J Pharmacol Exp Ther. 2016;356(2):324‐332. [DOI] [PubMed] [Google Scholar]
- 190. Chen J, Crawford R, Chen C, Xiao Y. The key regulatory roles of the PI3K/Akt signaling pathway in the functionalities of mesenchymal stem cells and applications in tissue regeneration. Tissue Engineering Part B: Reviews. 2013;19(6):516‐528. [DOI] [PubMed] [Google Scholar]
- 191. Hou C, Shen L, Huang Q, et al. The effect of heme oxygenase‐1 complexed with collagen on MSC performance in the treatment of diabetic ischemic ulcer. Biomaterials. 2013;34(1):112‐120. [DOI] [PubMed] [Google Scholar]
- 192. Jun E, Zhang Q, Yoon B, et al. Hypoxic conditioned medium from human amniotic fluid‐derived mesenchymal stem cells accelerates skin wound healing through TGF‐β/SMAD2 and PI3K/Akt pathways. Int J Mol Sci. 2014;15(1):605‐628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193. Lau TT, Wang D‐A. Stromal cell‐derived factor‐1 (SDF‐1): homing factor for engineered regenerative medicine. Expert Opin Biol Ther. 2011;11(2):189‐197. [DOI] [PubMed] [Google Scholar]
- 194. Ebrahim N, Dessouky AA, Mostafa O, et al. Adipose mesenchymal stem cells combined with platelet‐rich plasma accelerate diabetic wound healing by modulating the Notch pathway. Stem Cell Res Ther. 2021;12(1):392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195. Huang Y‐W, Zhu QQ, Yang XY, et al. Wound healing can be improved by (—)‐epigallocatechin gallate through targeting Notch in streptozotocin‐induced diabetic mice. FASEB J. 2019;33(1):953‐964. [DOI] [PubMed] [Google Scholar]
- 196. Yang JM, Ryu J, Kim I, Chang H, Kim IK. Dll4 blockade promotes angiogenesis in nonhealing wounds of Sox7‐deficient mice. Adv Wound Care. 2020;9(11):591‐601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197. Li B, Zheng YW, Sano Y, Taniguchi H. Evidence for mesenchymal− epithelial transition associated with mouse hepatic stem cell differentiation. PLoS One. 2011;6(2):e17092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198. Dos Santos JF, Borçari NR, da Silva Araújo M, Nunes VA. Mesenchymal stem cells differentiate into keratinocytes and express epidermal kallikreins: towards an in vitro model of human epidermis. J Cell Biochem. 2019;120(8):13141‐13155. [DOI] [PubMed] [Google Scholar]
- 199. Kato J, Kamiya H, Himeno T, et al. Mesenchymal stem cells ameliorate impaired wound healing through enhancing keratinocyte functions in diabetic foot ulcerations on the plantar skin of rats. J Diabetes Complications. 2014;28(5):588‐595. [DOI] [PubMed] [Google Scholar]
- 200. Chen L, Tredget EE, Wu PYG, Wu Y. Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing. PLoS One. 2008;3(4):e1886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201. Wu Y, Chen L, Scott PG, Tredget EE. Mesenchymal stem cells enhance wound healing through differentiation and angiogenesis. Stem Cells. 2007;25(10):2648‐2659. [DOI] [PubMed] [Google Scholar]
- 202. Zhao Q‐S, Xia N, Zhao N, et al. Localization of human mesenchymal stem cells from umbilical cord blood and their role in repair of diabetic foot ulcers in rats. Int J Biol Sci. 2014;10(1):80‐89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203. Afflerbach A‐K, Kiri MD, Detinis T, Maoz BM. Mesenchymal stem cells as a promising cell source for integration in novel in vitro models. Biomolecules. 2020;10(9):1306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204. Assi R, Foster TR, He H, et al. Delivery of mesenchymal stem cells in biomimetic engineered scaffolds promotes healing of diabetic ulcers. Regener Med. 2016;11(3):245‐260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205. Chen JS, Wong VW, Gurtner GC. Therapeutic potential of bone marrow‐derived mesenchymal stem cells for cutaneous wound healing. Front Immunol. 2012;3:192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206. Critser PJ, Kreger ST, Voytik‐Harbin SL, Yoder MC. Collagen matrix physical properties modulate endothelial colony forming cell‐derived vessels in vivo. Microvasc Res. 2010;80(1):23‐30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207. Gower RM, Shea LD. Biomaterial scaffolds for controlled, localized gene delivery of regenerative factors. Adv Wound Care. 2013;2(3):100‐106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208. Mudera V, Morgan M, Cheema U, Nazhat S, Brown R. Ultra‐rapid engineered collagen constructs tested in an in vivo nursery site. J Tissue Eng Regener Med. 2007;1(3):192‐198. [DOI] [PubMed] [Google Scholar]
- 209. Chiang K‐J, Chiu LC, Kang YN, Chen C. Autologous stem cell therapy for chronic lower extremity wounds: a meta‐analysis of randomized controlled trials. Cells. 2021;10(12):3307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210. Comerota AJ, Link A, Douville J, Burchardt ER. Upper extremity ischemia treated with tissue repair cells from adult bone marrow. J Vasc Surg. 2010;52(3):723‐729. [DOI] [PubMed] [Google Scholar]
- 211. Held M, Rahmanian‐Schwarz A, Schiefer J, et al. A novel collagen‐gelatin scaffold for the treatment of deep dermal wounds—an evaluation in a minipig model. Dermatol Surg. 2016;42(6):751‐756. [DOI] [PubMed] [Google Scholar]
- 212. Jung S‐N, Rhie JW, Kwon H, et al. In vivo cartilage formation using chondrogenic‐differentiated human adipose‐derived mesenchymal stem cells mixed with fibrin glue. J Craniofac Surg. 2010;21(2):468‐472. [DOI] [PubMed] [Google Scholar]
- 213. Keshavarz R, Olsen S, Almeida B. Using biomaterials to improve mesenchymal stem cell therapies for chronic, nonhealing wounds. Bioeng Transl Med. 2024;9(1):e10598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214. Croll TI, Gentz S, Mueller K, et al. Modelling oxygen diffusion and cell growth in a porous, vascularising scaffold for soft tissue engineering applications. Chem Eng Sci. 2005;60(17):4924‐4934. [Google Scholar]
- 215. Ahmed A, Getti G, Boateng J. Ciprofloxacin‐loaded calcium alginate wafers prepared by freeze‐drying technique for potential healing of chronic diabetic foot ulcers. Drug Delivery Transl Res. 2018;8:1751‐1768. [DOI] [PubMed] [Google Scholar]
- 216. Ahmed EM. Hydrogel: preparation, characterization, and applications: a review. J Adv Res. 2015;6(2):105‐121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217. Khayambashi P, Iyer J, Pillai S, Upadhyay A, Zhang Y, Tran S. Hydrogel encapsulation of mesenchymal stem cells and their derived exosomes for tissue engineering. Int J Mol Sci. 2021;22(2):684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218. Mardpour S, Ghanian MH, Sadeghi‐Abandansari H, et al. Hydrogel‐mediated sustained systemic delivery of mesenchymal stem cell‐derived extracellular vesicles improves hepatic regeneration in chronic liver failure. ACS Appl Mater Interfaces. 2019;11(41):37421‐37433. [DOI] [PubMed] [Google Scholar]
- 219. Catoira MC, Fusaro L, Di Francesco D, Ramella M, Boccafoschi F. Overview of natural hydrogels for regenerative medicine applications. J Mater Sci: Mater Med. 2019;30:1‐10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220. Feng J, Mineda K, Wu SH, et al. An injectable non‐cross‐linked hyaluronic‐acid gel containing therapeutic spheroids of human adipose‐derived stem cells. Sci Rep. 2017;7(1):1548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221. da Silva LP, Santos TC, Rodrigues DB, et al. Stem cell‐containing hyaluronic acid‐based spongy hydrogels for integrated diabetic wound healing. J Invest Dermatol. 2017;137(7):1541‐1551. [DOI] [PubMed] [Google Scholar]
- 222. Aijaz A, Teryek M, Goedken M, Polunas M, Olabisi RM. Coencapsulation of ISCs and MSCs enhances viability and function of both cell types for improved wound healing. Cell Mol Bioeng. 2019;12:481‐493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223. Abbasi N, Hamlet S, Love RM, Nguyen N‐T. Porous scaffolds for bone regeneration. J Sci: Adv Mater Devices. 2020;5(1):1‐9. [Google Scholar]
- 224. Lutzweiler G, Ndreu Halili A, Engin Vrana N. The overview of porous, bioactive scaffolds as instructive biomaterials for tissue regeneration and their clinical translation. Pharmaceutics. 2020;12(7):602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225. Dhandayuthapani B, Yoshida Y, Maekawa T, Kumar DS. Polymeric scaffolds in tissue engineering application: a review. Int J Polym Sci. 2011;2011:1‐19. [Google Scholar]
- 226. Udenni Gunathilake T, Ching Y, Ching K, Chuah C, Abdullah L. Biomedical and microbiological applications of bio‐based porous materials: a review. Polymers. 2017;9(5):160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227. O'Loughlin A, Kulkarni M, Creane M, et al. Topical administration of allogeneic mesenchymal stromal cells seeded in a collagen scaffold augments wound healing and increases angiogenesis in the diabetic rabbit ulcer. Diabetes. 2013;62(7):2588‐2594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228. Ní Annaidh A, Bruyère K, Destrade M, Gilchrist MD, Otténio M. Characterization of the anisotropic mechanical properties of excised human skin. J Mech Behav Biomed Mater. 2012;5(1):139‐148. [DOI] [PubMed] [Google Scholar]
- 229. Örgül D, Eroğlu H, Tiryaki M, Pınarlı FA, Hekimoglu S. In‐vivo evaluation of tissue scaffolds containing simvastatin loaded nanostructured lipid carriers and mesenchymal stem cells in diabetic wound healing. J Drug Delivery Sci Technol. 2021;61:102140. [Google Scholar]
- 230. Chen T‐Y, Wen TK, Dai NT, Hsu S. Cryogel/hydrogel biomaterials and acupuncture combined to promote diabetic skin wound healing through immunomodulation. Biomaterials. 2021;269:120608. [DOI] [PubMed] [Google Scholar]
- 231. Mohanty C, Pradhan J. A human epidermal growth factor‐curcumin bandage bioconjugate loaded with mesenchymal stem cell for in vivo diabetic wound healing. Materials Science and Engineering: C. 2020;111:110751. [DOI] [PubMed] [Google Scholar]
- 232. Yang B‐Y, Hu CH, Huang WC, Ho CY, Yao CH, Huang CH. Effects of bilayer nanofibrous scaffolds containing curcumin/lithospermi radix extract on wound healing in streptozotocin‐induced diabetic rats. Polymers. 2019;11(11):1745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233. Augustine R, Zahid AA, Hasan A, Wang M, Webster TJ. CTGF loaded electrospun dual porous core‐shell membrane for diabetic wound Healing. Int J Nanomedicine. 2019;14:8573‐8588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234. Augustine R, Rehman SRU, Ahmed R, et al. Electrospun chitosan membranes containing bioactive and therapeutic agents for enhanced wound healing. Int J Biiol Macromol. 2020;156:153‐170. [DOI] [PubMed] [Google Scholar]
- 235. Jun I, Han HS, Edwards J, Jeon H. Electrospun fibrous scaffolds for tissue engineering: viewpoints on architecture and fabrication. Int J Mol Sci. 2018;19(3):745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236. Tam K, Cheyyatraviendran S, Venugopal J, et al. A nanoscaffold impregnated with human wharton's jelly stem cells or its secretions improves healing of wounds: a nanoscaffold impregnated with human wharton's jelly stem cells. J Cell Biochem. 2014;115(4):794‐803. [DOI] [PubMed] [Google Scholar]
- 237. Navone SE, Pascucci L, Dossena M, et al. Decellularized silk fibroin scaffold primed with adipose mesenchymal stromal cells improves wound healing in diabetic mice. Stem Cell Res Ther. 2014;5:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238. Tapias LF, Ott HC. Decellularized scaffolds as a platform for bioengineered organs. Curr Opin Organ Transplant. 2014;19(2):145‐152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239. Lee YJ, Baek SE, Lee S, et al. Wound‐healing effect of adipose stem cell‐derived extracellular matrix sheet on full‐thickness skin defect rat model: histological and immunohistochemical study. Int Wound J. 2019;16(1):286‐296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240. Chu J, Shi P, Deng X, et al. Dynamic multiphoton imaging of acellular dermal matrix scaffolds seeded with mesenchymal stem cells in diabetic wound healing. J Biophotonics. 2018;11(7):e201700336. [DOI] [PubMed] [Google Scholar]
- 241. Beiki B, Zeynali B, Seyedjafari E. Fabrication of a three dimensional spongy scaffold using human Wharton's jelly derived extra cellular matrix for wound healing. Mater Sci Eng C Mater Biol Appl. 2017;78:627‐638. [DOI] [PubMed] [Google Scholar]
- 242. Han Y, Tao R, Han Y, et al. Microencapsulated VEGF gene–modified umbilical cord mesenchymal stromal cells promote the vascularization of tissue‐engineered dermis: an experimental study. Cytotherapy. 2014;16(2):160‐169. [DOI] [PubMed] [Google Scholar]
- 243. Wilshaw S‐P, Kearney J, Fisher J, Ingham E. Biocompatibility and potential of acellular human amniotic membrane to support the attachment and proliferation of allogeneic cells. Tissue Eng Part A. 2008;14(4):463‐472. [DOI] [PubMed] [Google Scholar]
- 244. Nie C, Yang D, Morris SF. Local delivery of adipose‐derived stem cells via acellular dermal matrix as a scaffold: a new promising strategy to accelerate wound healing. Med Hypotheses. 2009;72(6):679‐682. [DOI] [PubMed] [Google Scholar]
- 245. Nie C, Zhang G, Yang D, et al. Targeted delivery of adipose‐derived stem cells via acellular dermal matrix enhances wound repair in diabetic rats. J Tissue Eng Regener Med. 2015;9(3):224‐235. [DOI] [PubMed] [Google Scholar]
- 246. Fu J, Zhang Y, Chu J, et al. Reduced graphene oxide incorporated acellular dermal composite scaffold enables efficient local delivery of mesenchymal stem cells for accelerating diabetic wound healing. ACS Biomater Sci Eng. 2019;5(8):4054‐4066. [DOI] [PubMed] [Google Scholar]
- 247. Han Y, Sun T, Han Y, et al. Human umbilical cord mesenchymal stem cells implantation accelerates cutaneous wound healing in diabetic rats via the Wnt signaling pathway. Eur J Med Res. 2019;24(1):10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248. Bellu E, Medici S, Coradduzza D, Cruciani S, Amler E, Maioli M. Nanomaterials in skin regeneration and rejuvenation. Int J Mol Sci. 2021;22(13):7095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249. Kaul S, Gulati N, Verma D, Mukherjee S, Nagaich U. Role of nanotechnology in cosmeceuticals: a review of recent advances. J Pharm. 2018;2018:1‐19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250. Daňková J, Buzgo M, Vejpravová J, et al. Highly efficient mesenchymal stem cell proliferation on poly‐ε‐caprolactone nanofibers with embedded magnetic nanoparticles. Int J Nanomedicine. 2015;10:7307‐7317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251. Akolpoğlu Başaran DD, Gündüz U, Tezcaner A, Keskin D. Topical delivery of heparin from PLGA nanoparticles entrapped in nanofibers of sericin/gelatin scaffolds for wound healing. Int J Pharm. 2021;597:120207. [DOI] [PubMed] [Google Scholar]
- 252. Graça MFP, de Melo‐Diogo D, Correia IJ, Moreira AF. Electrospun asymmetric membranes as promising wound dressings: a review. Pharmaceutics. 2021;13(2):183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253. Kwon SG, Kwon YW, Lee TW, Park GT, Kim JH. Recent advances in stem cell therapeutics and tissue engineering strategies. Biomater Res. 2018;22(1):1‐8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254. Chen T, Wu Y, Gu W, Xu Q. Response of vascular mesenchymal stem/progenitor cells to hyperlipidemia. Cell Mol Life Sci. 2018;75:4079‐4091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255. Soundararajan M, Kannan S. Fibroblasts and mesenchymal stem cells: two sides of the same coin? J Cell Physiol. 2018;233(12):9099‐9109. [DOI] [PubMed] [Google Scholar]
- 256. Jackson WM, Nesti LJ, Tuan RS. Potential therapeutic applications of muscle‐derived mesenchymal stem and progenitor cells. Expert Opin Biol Ther. 2010;10(4):505‐517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257. Tomokiyo A, Yoshida S, Hamano S, Hasegawa D, Sugii H, Maeda H. Detection, characterization, and clinical application of mesenchymal stem cells in periodontal ligament tissue. Stem Cells Int. 2018;2018:1‐9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258. Ichim TE, O'Heeron P, Kesari S. Fibroblasts as a practical alternative to mesenchymal stem cells. J Transl Med. 2018;16(1):212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259. Marofi F, Vahedi G, Hasanzadeh A, et al. Mesenchymal stem cells as the game‐changing tools in the treatment of various organs disorders: mirage or reality? J Cell Physiol. 2019;234(2):1268‐1288. [DOI] [PubMed] [Google Scholar]
- 260. Beggs KJ, Lyubimov A, Borneman JN, et al. Immunologic consequences of multiple, high‐dose administration of allogeneic mesenchymal stem cells to baboons. Cell Transplant. 2006;15(8‐9):711‐721. [DOI] [PubMed] [Google Scholar]
- 261. Secunda R, Vennila R, Mohanashankar AM, Rajasundari M, Jeswanth S, Surendran R. Isolation, expansion and characterisation of mesenchymal stem cells from human bone marrow, adipose tissue, umbilical cord blood and matrix: a comparative study. Cytotechnology. 2015;67(5):793‐807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262. Klyushnenkova E, Mosca JD, Zernetkina V, et al. T cell responses to allogeneic human mesenchymal stem cells: immunogenicity, tolerance, and suppression. J Biomed Sci. 2005;12:47‐57. [DOI] [PubMed] [Google Scholar]
- 263. Jiang W, Xu J. Immune modulation by mesenchymal stem cells. Cell Proliferation. 2020;53(1):e12712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264. Scuteri A, Monfrini M. Mesenchymal stem cells as new therapeutic approach for diabetes and pancreatic disorders. Int J Mol Sci. 2018;19(9):2783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265. Ullah I, Subbarao RB, Rho GJ. Human mesenchymal stem cells‐current trends and future prospective. Biosci Rep. 2015;35(2):e00191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266. Reinders MEJ, Fibbe WE, Rabelink TJ. Multipotent mesenchymal stromal cell therapy in renal disease and kidney transplantation. Nephrol Dial Transplant. 2010;25(1):17‐24. [DOI] [PubMed] [Google Scholar]
- 267. Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease. Nat Rev Immunol. 2008;8(9):726‐736. [DOI] [PubMed] [Google Scholar]
- 268. Salem HK, Thiemermann C. Mesenchymal stromal cells: current understanding and clinical status. Stem Cells. 2010;28(3):585‐596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269. Tyndall A. Mesenchymal stem cell treatments in rheumatology—a glass half full? Nat Rev Rheumatol. 2014;10(2):117‐124. [DOI] [PubMed] [Google Scholar]
- 270. Hare JM, Traverse JH, Henry TD, et al. A randomized, double‐blind, placebo‐controlled, dose‐escalation study of intravenous adult human mesenchymal stem cells (prochymal) after acute myocardial infarction. J Am Coll Cardiol. 2009;54(24):2277‐2286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271. Park JS, Suryaprakash S, Lao YH, Leong KW. Engineering mesenchymal stem cells for regenerative medicine and drug delivery. Methods. 2015;84:3‐16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272. Macrin D, Joseph JP, Pillai AA, Devi A. Eminent sources of adult mesenchymal stem cells and their therapeutic imminence. Stem Cell Rev Rep. 2017;13:741‐756. [DOI] [PubMed] [Google Scholar]
- 273. Belmar‐Lopez C, Mendoza G, Oberg D, et al. Tissue‐derived mesenchymal stromal cells used as vehicles for anti‐tumor therapy exert different in vivo effects on migration capacity and tumor growth. BMC Med. 2013;11:13916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274. Marofi F, Vahedi G, Biglari A, Esmaeilzadeh A, Athari SS. Mesenchymal stromal/stem cells: a new era in the cell‐based targeted gene therapy of cancer. Front Immunol. 2017;8:1770. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 275. Connick P, Kolappan M, Crawley C, et al. Autologous mesenchymal stem cells for the treatment of secondary progressive multiple sclerosis: an open‐label phase 2a proof‐of‐concept study. Lancet Neurol. 2012;11(2):150‐156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276. Lalu MM, McIntyre L, Pugliese C, et al. Safety of cell therapy with mesenchymal stromal cells (safecell): a systematic review and meta‐analysis of clinical trials. PLoS One. 2012;7(10):e47559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277. Lee JS, Hong JM, Moon GJ, Lee PH, Ahn YH, Bang OY. A long‐term follow‐up study of intravenous autologous mesenchymal stem cell transplantation in patients with ischemic stroke. Stem Cells. 2010;28(6):1099‐1106. [DOI] [PubMed] [Google Scholar]
- 278. Von Bahr L, Batsis I, Moll G, et al. Analysis of tissues following mesenchymal stromal cell therapy in humans indicates limited long‐term engraftment and no ectopic tissue formation. Stem Cells. 2012;30(7):1575‐1578. [DOI] [PubMed] [Google Scholar]
- 279. Lamo‐Espinosa JM, Mora G, Blanco JF, et al. Intra‐articular injection of two different doses of autologous bone marrow mesenchymal stem cells versus hyaluronic acid in the treatment of knee osteoarthritis: long‐term follow up of a multicenter randomized controlled clinical trial (phase I/II). J Transl Med. 2018;16(1):213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 280. Akiyama K, Chen C, Wang D, et al. Mesenchymal‐stem‐cell‐induced immunoregulation involves FAS‐ligand‐/FAS‐mediated T cell apoptosis. Cell Stem Cell. 2012;10(5):544‐555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281. Skyler JS, Fonseca VA, Segal KR, Rosenstock J. Allogeneic mesenchymal precursor cells in type 2 diabetes: a randomized, placebo‐controlled, dose‐escalation safety and tolerability pilot study. Diabetes Care. 2015;38(9):1742‐1749. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 282. Si Y‐L, Zhao YL, Hao HJ, Fu XB, Han WD. MSCs: biological characteristics, clinical applications and their outstanding concerns. Ageing Res Rev. 2011;10(1):93‐103. [DOI] [PubMed] [Google Scholar]
- 283. Fujita Y, Kadota T, Araya J, Ochiya T, Kuwano K. Clinical application of mesenchymal stem cell‐derived extracellular vesicle‐based therapeutics for inflammatory lung diseases. J Clin Med. 2018;7(10):355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 284. Nunes HC, Scarano WR, Deffune E, Felisbino SL, Porreca I, Delella FK. Bisphenol a and mesenchymal stem cells: recent insights. Life Sci. 2018;206:22‐28. [DOI] [PubMed] [Google Scholar]
- 285. Gugjoo MB, Amarpal A, Fazili MR, Shah RA, Sharma GT. Mesenchymal stem cell: basic research and potential applications in cattle and buffalo. J Cell Physiol. 2019;234(6):8618‐8635. [DOI] [PubMed] [Google Scholar]
- 286. Zhuang X, Hu X, Zhang S, Li X, Yuan X, Wu Y. Mesenchymal stem cell–based therapy as a new approach for the treatment of systemic sclerosis. Clin Rev Allergy Immunol. 2022;64(3):284‐320. [DOI] [PubMed] [Google Scholar]
- 287. Guan XQ, Yu JL, Li LQ, Liu GX. Study on mesenchymal stem cells entering the brain through the blood‐brain barrier. Zhonghua Er Ke Za Zhi. 2004;42(12):920‐923. [PubMed] [Google Scholar]
- 288. Strioga M, Viswanathan S, Darinskas A, Slaby O, Michalek J. Same or not the same? Comparison of adipose tissue‐derived versus bone marrow‐derived mesenchymal stem and stromal cells. Stem Cells Dev. 2012;21(14):2724‐2752. [DOI] [PubMed] [Google Scholar]
- 289. Wagner W, Bork S, Horn P, et al. Aging and replicative senescence have related effects on human stem and progenitor cells. PLoS One. 2009;4(6):e5846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 290. Jin Y‐Z, Lee JH. Mesenchymal stem cell therapy for bone regeneration. Clin Orthop Surg. 2018;10(3):271‐278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 291. Eltoukhy HS, Sinha G, Moore CA, Gergues M, Rameshwar P. Secretome within the bone marrow microenvironment: a basis for mesenchymal stem cell treatment and role in cancer dormancy. Biochimie. 2018;155:92‐103. [DOI] [PubMed] [Google Scholar]
- 292. Gomes ED, Vieira de Castro J, Costa BM, Salgado AJ. The impact of mesenchymal stem cells and their secretome as a treatment for gliomas. Biochimie. 2018;155:59‐66. [DOI] [PubMed] [Google Scholar]
- 293. Florea V, Rieger AC, DiFede DL, et al. Dose comparison study of allogeneic mesenchymal stem cells in patients with ischemic cardiomyopathy (the TRIDENT study). Circ Res. 2017;121(11):1279‐1290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294. Kumamoto M, Nishiwaki T, Matsuo N, Kimura H, Matsushima K. Minimally cultured bone marrow mesenchymal stem cells ameliorate fibrotic lung injury. Eur Respir J. 2009;34(3):740‐748. [DOI] [PubMed] [Google Scholar]
- 295. Jakovljevic J, Harrell CR, Fellabaum C, Arsenijevic A, Jovicic N, Volarevic V. Modulation of autophagy as new approach in mesenchymal stem cell‐based therapy. Biomed Pharmacother. 2018;104:404‐410. [DOI] [PubMed] [Google Scholar]
- 296. Luo R, Lu Y, Liu J, Cheng J, Chen Y. Enhancement of the efficacy of mesenchymal stem cells in the treatment of ischemic diseases. Biomed Pharmacother. 2019;109:2022‐2034. [DOI] [PubMed] [Google Scholar]
- 297. Burst VR, Gillis M, Pütsch F, et al. Poor cell survival limits the beneficial impact of mesenchymal stem cell transplantation on acute kidney injury. Nephron Exp Nephrol. 2010;114(3):e107‐e116. [DOI] [PubMed] [Google Scholar]
- 298. Shi R‐Z, Li Q‐P. Improving outcome of transplanted mesenchymal stem cells for ischemic heart disease. Biochem Biophys Res Commun. 2008;376(2):247‐250. [DOI] [PubMed] [Google Scholar]
- 299. Karaöz E, Demircan PC, Erman G, Güngörürler E, Sarıboyacı AE. Comparative analyses of immunosuppressive characteristics of bone‐marrow, Wharton's jelly, and adipose tissue‐derived human mesenchymal stem cells. Turk J Haematol. 2017;34(3):225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 300. Song H, Cha MJ, Song BW, et al. Reactive oxygen species inhibit adhesion of mesenchymal stem cells implanted into ischemic myocardium via interference of focal adhesion complex. Stem Cells. 2010;28(3):555‐563. [DOI] [PubMed] [Google Scholar]
- 301. Becker AD, Riet IV. Homing and migration of mesenchymal stromal cells: how to improve the efficacy of cell therapy? World J Stem Cells. 2016;8(3):73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 302. Rengasamy M, Gupta PK, Kolkundkar U, et al. Preclinical safety amp; toxicity evaluation of pooled, allogeneic human bone marrow‐derived mesenchymal stromal cells. Indian J Med Res. 2016;144(6):852‐864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 303. Bernardo ME, Zaffaroni N, Novara F, et al. Human bone marrow–derived mesenchymal stem cells do not undergo transformation after long‐term in vitro culture and do not exhibit telomere maintenance mechanisms. Cancer Res. 2007;67(19):9142‐9149. [DOI] [PubMed] [Google Scholar]
- 304. Volarevic V, Markovic BS, Gazdic M, et al. Ethical and safety issues of stem cell‐based therapy. Int J Med Sci. 2018;15(1):36‐45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 305. Yong KW, Choi JR, Dolbashid AS, Wan Safwani WKZ. Biosafety and bioefficacy assessment of human mesenchymal stem cells: what do we know so far? Regener Med. 2018;13(2):219‐232. [DOI] [PubMed] [Google Scholar]
- 306. Kim N, Cho S‐G. New strategies for overcoming limitations of mesenchymal stem cell‐based immune modulation. Int J Stem Cells. 2015;8(1):54‐68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 307. Taddio A, Tommasini A, Valencic E, et al. Failure of interferon‐γ pre‐treated mesenchymal stem cell treatment in a patient with Crohn's disease. World J Gastroenterol. 2015;21(14):4379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 308. Baranovskii DS, Klabukov ID, Arguchinskaya NV, et al. Adverse events, side effects and complications in mesenchymal stromal cell‐based therapies. Stem Cell Investig. 2022;9:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 309. Thanunchai M, Hongeng S, Thitithanyanont A. Mesenchymal stromal cells and viral infection. Stem Cells Int. 2015;2015:1‐8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 310. Sundin M, Lindblom A, Örvell C, et al. Persistence of human parvovirus B19 in multipotent mesenchymal stromal cells expressing the erythrocyte P antigen: implications for transplantation. Biol Blood Marrow Transplant. 2008;14(10):1172‐1179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 311. Toppinen M, Sajantila A, Pratas D, Hedman K, Perdomo MF. The human bone marrow is host to the DNAs of several viruses. Front Cell Infect Microbiol. 2021;11:657245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 312. Cheung MB, Sampayo‐Escobar V, Green R, Moore ML, Mohapatra S, Mohapatra SS. Respiratory syncytial virus‐infected mesenchymal stem cells regulate immunity via interferon beta and indoleamine‐2, 3‐dioxygenase. PLoS One. 2016;11(10):e0163709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 313. Thanunchai M, Kanrai P, Wiboon‐ut S, Puthavathana P, Hongeng S, Thitithanyanont A. Tropism of avian influenza A (H5N1) virus to mesenchymal stem cells and CD34+ hematopoietic stem cells. PLoS One. 2013;8(12):e81805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 314. Nikfarjam L, Farzaneh P. Prevention and detection of mycoplasma contamination in cell culture. Cell journal. 2012;13(4):203‐212. [PMC free article] [PubMed] [Google Scholar]
- 315. Cottle C, Porter AP, Lipat A, et al. Impact of cryopreservation and freeze‐thawing on therapeutic properties of mesenchymal stromal/stem cells and other common cellular therapeutics. Curr Stem Cell Rep. 2022;8(2):72‐92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 316. Moll G, Geißler S, Catar R, et al. Cryopreserved or fresh mesenchymal stromal cells: only a matter of taste or key to unleash the full clinical potential of MSC therapy? Adv Exp Med Biol. 2016;951:77‐98. [DOI] [PubMed] [Google Scholar]
- 317. Sarmiento M, Ramírez P, Parody R, et al. Advantages of non‐cryopreserved autologous hematopoietic stem cell transplantation against a cryopreserved strategy. Bone Marrow Transplant. 2018;53(8):960‐966. [DOI] [PubMed] [Google Scholar]
- 318. Smagur A, Mitrus I, Ciomber A, et al. Comparison of the cryoprotective solutions based on human albumin vs. autologous plasma: its effect on cell recovery, clonogenic potential of peripheral blood hematopoietic progenitor cells and engraftment after autologous transplantation. Vox Sang. 2015;108(4):417‐424. [DOI] [PubMed] [Google Scholar]
- 319. Olmedo‐Moreno L, Aguilera Y, Baliña‐Sánchez C, Martín‐Montalvo A, Capilla‐González V. Heterogeneity of in vitro expanded mesenchymal stromal cells and strategies to improve their therapeutic actions. Pharmaceutics. 2022;14(5):1112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 320. Røsland GV, Svendsen A, Torsvik A, et al. Long‐term cultures of bone marrow–derived human mesenchymal stem cells frequently undergo spontaneous malignant transformation. Cancer Res. 2009;69(13):5331‐5339. [DOI] [PubMed] [Google Scholar]
- 321. Froelich K, Mickler J, Steusloff G, et al. Chromosomal aberrations and deoxyribonucleic acid single‐strand breaks in adipose‐derived stem cells during long‐term expansion in vitro. Cytotherapy. 2013;15(7):767‐781. [DOI] [PubMed] [Google Scholar]
- 322. Soltani SK, Forogh B, Ahmadbeigi N, et al. Safety and efficacy of allogenic placental mesenchymal stem cells for treating knee osteoarthritis: a pilot study. Cytotherapy. 2019;21(1):54‐63. [DOI] [PubMed] [Google Scholar]
- 323. Suzuki K, Sun R, Origuchi M, et al. Mesenchymal stromal cells promote tumor growth through the enhancement of neovascularization. Mol Med. 2011;17:579‐587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 324. Uccelli A, Pistoia V, Moretta L. Mesenchymal stem cells: a new strategy for immunosuppression? Trends Immunol. 2007;28(5):219‐226. [DOI] [PubMed] [Google Scholar]
- 325. Poltavtseva RA, Poltavtsev AV, Lutsenko GV, Svirshchevskaya EV. Myths, reality and future of mesenchymal stem cell therapy. Cell Tissue Res. 2019;375:563‐574. [DOI] [PubMed] [Google Scholar]
- 326. Li H, Rong P, Ma X, et al. Paracrine effect of mesenchymal stem cell as a novel therapeutic strategy for diabetic nephropathy. Life Sci. 2018;215:113‐118. [DOI] [PubMed] [Google Scholar]
- 327. Meseguer‐Olmo L, Montellano AJ, Martínez T, et al. Intraarticular and intravenous administration of 99MTc‐HMPAO‐labeled human mesenchymal stem cells (99MTC‐AH‐MSCS): in vivo imaging and biodistribution. Nucl Med Biol. 2017;46:36‐42. [DOI] [PubMed] [Google Scholar]
- 328. Leibacher J, Dauber K, Ehser S, et al. Human mesenchymal stromal cells undergo apoptosis and fragmentation after intravenous application in immune‐competent mice. Cytotherapy. 2017;19(1):61‐74. [DOI] [PubMed] [Google Scholar]
- 329. Reza‐Zaldivar EE, Hernández‐Sapiéns MA, Minjarez B, Gutiérrez‐Mercado YK, Márquez‐Aguirre AL, Canales‐Aguirre AA. Potential effects of MSC‐derived exosomes in neuroplasticity in Alzheimer's disease. Front Cell Neurosci. 2018;12:317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 330. Furlani D, Ugurlucan M, Ong L, et al. Is the intravascular administration of mesenchymal stem cells safe? Microvasc Res. 2009;77(3):370‐376. [DOI] [PubMed] [Google Scholar]
- 331. Munk A, Duvald CS, Pedersen M, et al. Dosing limitation for intra‐renal arterial infusion of mesenchymal stromal cells. Int J Mol Sci. 2022;23(15):8268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 332. Kim J‐S, Lee JH, Kwon O, et al. Rapid deterioration of preexisting renal insufficiency after autologous mesenchymal stem cell therapy. Kidney Res Clin Pract. 2017;36(2):200‐204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 333. Karthik L, Kumar G, Keswani T, Bhattacharyya A, Chandar SS, Bhaskara Rao KV. Protease inhibitors from marine actinobacteria as a potential source for antimalarial compound. PLoS One. 2014;9(3):e90972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 334. Forslöw U, Blennow O, LeBlanc K, et al. Treatment with mesenchymal stromal cells is a risk factor for pneumonia‐related death after allogeneic hematopoietic stem cell transplantation. Eur J Haematol. 2012;89(3):220‐227. [DOI] [PubMed] [Google Scholar]
- 335. Moll G, Drzeniek N, Kamhieh‐Milz J, Geissler S, Volk HD, Reinke P. MSC therapies for COVID−19: importance of patient coagulopathy, thromboprophylaxis, cell product quality and mode of delivery for treatment safety and efficacy. Front Immunol. 2020;11:1091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 336. Ahmed AK, Moustafa ER, Abd EL‐Monem AM, et al. Thromboembolism: the secret lethal weapon of coronavirus disease 2019. RRMM. 2023;34(3):167‐179. [Google Scholar]
- 337. Golchin A. Cell‐based therapy for severe COVID‐19 patients: clinical trials and cost‐utility. Stem Cell Rev Rep. 2021;17:56‐62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 338. Chen N, Zhou M, Dong X, et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. The Lancet. 2020;395(10223):507‐513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 339. Ouyang L, Qiu D, Fu X, et al. Overexpressing HPGDS in adipose‐derived mesenchymal stem cells reduces inflammatory state and improves wound healing in type 2 diabetic mice. Stem Cell Res Ther. 2022;13(1):395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 340. Rai V, Moellmer R, Agrawal DK. Stem cells and angiogenesis: implications and limitations in enhancing chronic diabetic foot ulcer healing. Cells. 2022;11(15):2287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 341. Wang Y, Yi H, Song Y. The safety of MSC therapy over the past 15 years: a meta‐analysis. Stem Cell Res Ther. 2021;12(1):1‐15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 342. Nishikawa G, Kawada K, Nakagawa J, et al. Bone marrow‐derived mesenchymal stem cells promote colorectal cancer progression via CCR5. Cell Death Dis. 2019;10(4):264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 343. Han L, He H, Yang Y, et al. Distinctive clinical and pathologic features of immature teratomas arising from induced pluripotent stem cell‐derived beta cell injection in a diabetes patient. Stem Cells Dev. 2022;31(5‐6):97‐101. [DOI] [PubMed] [Google Scholar]
- 344. Jiang X‐Y, Lu D‐B, Chen B. Progress in stem cell therapy for the diabetic foot. Diabetes Res Clin Pract. 2012;97(1):43‐50. [DOI] [PubMed] [Google Scholar]
- 345. Mathew E, Brannon AL, Del Vecchio A, et al. Mesenchymal stem cells promote pancreatic tumor growth by inducing alternative polarization of macrophages. Neoplasia. 2016;18(3):142‐151. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
All generated data are included in this manuscript.
