Review Open Access
Copyright ©The Author(s) 2023. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Diabetes. Nov 15, 2023; 14(11): 1585-1602
Published online Nov 15, 2023. doi: 10.4239/wjd.v14.i11.1585
Mesenchymal stem cells-based drug delivery systems for diabetic foot ulcer: A review
Hong-Min Zhang, Department of Endocrinology, People’s Hospital of Chongqing Liangjiang New Area, Chongqing 400030, China
Meng-Liu Yang, Gang-Yi Yang, Department of Endocrinology, The Second Affiliated Hospital of The Chongqing Medical University, Chongqing 400030, China
Jia-Zhuang Xi, Qi-Nan Wu, Department of Endocrinology, Dazu Hospital of Chongqing Medical University, The People’s Hospital of Dazu, Chongqing 406230, China
ORCID number: Hong-Min Zhang (0000-0002-1726-5721); Qi-Nan Wu (0000-0002-8733-9191).
Author contributions: Zhang HM, Yang ML, Xi JZ, Yang GY and Wu QN contributed to writing the manuscript and participated in helpful discussions; Wu QN is the guarantor of this work.
Supported by Science and Health Joint Medical Research Project of Chongqing, No. 2022MSXM133; Natural Science Foundation of Chongqing, No. CSTB2022NSCQ-MSX1522, No. CSTB2023NSCQ-MSX0246, and No. CSTB2022NSCQ-MSX1271; The First Batch of Key Disciplines on Public Health in Chongqing and Science; and Health Joint Project of Dazu District Science and Technology Bureau, No. DZKJ,2022CCC1001.
Conflict-of-interest statement: The authors have no conflicts of interest and all agreed to publish the manuscript.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Qi-Nan Wu, Doctor, MD, Director, Professor, Department of Endocrinology, Dazu Hospital of Chongqing Medical University, The People’s Hospital of Dazu, No. 1073 Second Ring South Road, Hongxing Community, Tangxiang Street, Dazu District, Chongqing 406230, China. wqn11@126.com
Received: July 1, 2023
Peer-review started: July 1, 2023
First decision: July 6, 2023
Revised: July 16, 2023
Accepted: September 11, 2023
Article in press: September 11, 2023
Published online: November 15, 2023

Abstract

The complication of diabetes, which is known as diabetic foot ulcer (DFU), is a significant concern due to its association with high rates of disability and mortality. It not only severely affects patients’ quality of life, but also imposes a substantial burden on the healthcare system. In spite of efforts made in clinical practice, treating DFU remains a challenging task. While mesenchymal stem cell (MSC) therapy has been extensively studied in treating DFU, the current efficacy of DFU healing using this method is still inadequate. However, in recent years, several MSCs-based drug delivery systems have emerged, which have shown to increase the efficacy of MSC therapy, especially in treating DFU. This review summarized the application of diverse MSCs-based drug delivery systems in treating DFU and suggested potential prospects for the future research.

Key Words: Diabetic foot ulcer, Mesenchymal stem cells, Drug delivery systems, Diabetes, Wound healing

Core Tip: Diabetic foot ulcer (DFU) is a significant concern due to its association with high rates of disability and mortality. Mesenchymal stem cell (MSC) therapy has been extensively studied in treating DFU, the current efficacy of DFU healing using this method is still inadequate. However, in recent years, several MSCs-based drug delivery systems have emerged, which have shown to increase the efficacy of MSC therapy, especially in treating DFU.



INTRODUCTION

Diabetes, along with its associated metabolic complications, has emerged as one of the most rapidly growing global health emergencies in the 21st century[1-3]. Diabetic foot ulcer (DFU) is a multifaceted and troubling complication of diabetes that poses significant challenges to healthcare providers and patients. DFU is characterized by the development of chronic wounds on the feet, which can lead to severe morbidity and mortality rates if left untreated[4-6]. Approximately 19%-34% of diabetics experience DFU, a condition with a recurrence rate of 40% within one year. This poses a significant threat to patients’ physical and mental well-being and places a substantial financial strain on both patients and their families[7]. The 10th edition of the International Diabetes Federation (IDF) Diabetes Atlas has revealed a sobering reality concerning the global burden of diabetes. With an estimated 536.6 million diabetic cases diagnosed worldwide in 2021, diabetes has become a significant public health crisis that demands immediate attention and action. Even more alarming is the prediction that this number will surge to a staggering 783 million by 2045 if current trends continue. Furthermore, it is anticipated that the healthcare costs associated with diabetes and its complications could globally reach a staggering 966 billion USD in 2021[8-10]. The reported statistics revealed that DFU, a condition that affects the skin tissue of the feet and can cause serious complications, has a prevalence of 6.3% among the global population. It is noteworthy that this condition is more frequently observed in men than in women, indicating a gender-based disparity in its incidence rate. Moreover, the data suggested that patients with type 2 diabetes (T2D), a chronic metabolic disorder, are at a higher risk of developing DFU, with an estimated prevalence rate of 6.4%, as opposed to those with type 1 diabetes, who have a lower rate of approximately 5.5%. The prevalence of DFU varies significantly across different regions of the world, as per available data. Specifically, North America has the highest recorded incidence rate of 13%, followed by Africa at 7.2%. Meanwhile, relatively lower rates of 5.1% and 5.5% were reported in Europe and Asia, respectively. The prevalence of DFU in Oceania is the lowest among the regions mentioned above, with a rate of 3.0%[11-13]. Previous studies have also demonstrated that DFU increases the risk of death by 2.5-fold in diabetics[14,15].

The synthesis of iodine (I)-polyvinyl alcohol (PVA)@polydopamine (PDA) microspheres was documented in Yang et al’s study[16]. The aim was to attain computed tomography images, drug loading and controlled release capabilities, as well as improved embolization of the liver portal vein. The in vivo embolization findings demonstrated the presence of focal necrosis in hepatocytes, along with necrotic cell fragments and infiltration of inflammatory cells in liver tissue. These observations provided evidence that the I-PVA@PDA microspheres exhibit a more potent embolization effect compared to PVA particles. Additionally, the I-PVA@PDA microspheres were utilized for the delivery and controlled release of 5-fluorouracil, a chemotherapeutic drug. The results showed an initial rapid release (29.74% released) within the first 24 h, followed by sustained release (34.48%) over a period of 72 h. In Ouyang et al’s research[17], a multifunctional bio-hemostatic hydrogel (CODM) was prepared based on hydrogen bonding and Schiff base bonding by using modified alginate, polyvinylpyrrolidone (PVP), and carboxymethyl chitosan. The amino group-modified montmorillonite was uniformly dispersed in the hydrogel through amido bond formation with the carboxyl groups of carboxymethyl chitosan and oxidized alginate. The catechol group (-CHO) and PVP were able to form hydrogen bonds with the tissue surface, resulting in firm tissue adhesion and wound hemostasis. The addition of montmorillonite-NH2 further improved the hemostatic ability, surpassing that of commercial hemostatic materials. Furthermore, the photothermal conversion capability (derived from polydopamine) was synergized with the phenolic hydroxyl group, quinone group, and protonated amino group, effectively eliminating bacteria both in vitro and in vivo. In a recent review[18], it was reported that a multifunctional CH hyaluronic acid three-dimensional (3D) hydrogel possesses a notable capacity for water absorption. This property holds potential for its application in managing inflammatory bowel diseases, with a concentration on various aspects, such as adhesion, synergistic therapy, pH sensitivity, particle size, and temperature sensitivity. A desirable polymer hydrogel for hemostasis is expected to possess the following characteristics[18]: (1) It should exhibit a rapid gelation rate to promptly stop bleeding and promote active wound healing; (2) in dynamic and humid environments, the hemostatic hydrogel should demonstrate adequate adhesion and exceptional mechanical properties to effectively seal the wound and prevent the displacement of the hemostatic hydrogel from the bleeding site; and (3) it should exhibit favorable biocompatibility. Furthermore, it is important for the hydrogel to exhibit controllable swelling behavior as overly swollen hydrogels may exert pressure on the surrounding tissue.

The physiological mechanism underlying wound healing encompasses a series of coordinated events, primarily involving inflammation, angiogenesis, and extracellular matrix (ECM) remodeling. However, DFU is associated with an abnormal microenvironment with prolonged inflammatory cell infiltration and slow angiogenesis and ECM remodeling, leading to impaired wound repair via the vascular and neurotrophic pathways. This in turn impedes local tissue regeneration and greatly reduces wound healing[19-22]. In addition, due to decreased granulocyte functions and chemotaxis, DFU cases are prone to infections[23-25]. The challenge of healing wounds in DFU is a multifaceted issue that arises from a combination of different factors. These factors include peripheral arterial disorders, which can impair blood flow and delivery of essential nutrients to the wound site, peripheral neuropathy, which can affect nerve function and lead to reduced sensation and poor healing response, foot deformities that can create pressure points and limit mobility, and also bacterial infections that can further complicate the healing process[26-29]. Wound debridement, which is the standard treatment for DFU and involves surgical removal of thickened, necrotic, damaged or infected tissues, has been widely used in clinical practice; the wound is then covered with dressing and/or treated with antibiotics to prevent infection[30-32]. A wound dressing that meets the ideal standards should facilitate a moist wound environment, shield against secondary infections, eliminate wound exudates, regulate biofilm formation, and stimulate tissue regeneration[33,34]. However, none of the existing dressings are able to meet all of these requirements, and due to the influence of various factors, traditional treatments can no longer achieve satisfactory outcomes[35,36]. DFU patients continue to face significant clinical hurdles when it comes to wound healing. Despite numerous chronic wound management techniques and treatments having been developed, including gene therapy, growth factor therapy, stem cell therapy, and biomaterial application, successfully repairing these wounds remains a formidable task[37,38]. Because of their diverse characteristics that involve producing numerous growth factors, cytokines, and chemokines, as well as regulating immune responses, supporting the development of new blood vessels, and restructuring tissue, mesenchymal stem cells (MSCs) have demonstrated significant therapeutic capabilities in improving wound healing for cases with DFU[39-41]. Most of the current cell-based therapies are administered via systemic or subcutaneous injection of cells[42,43]. However, MSCs cannot be delivered to the wound via the systemic route, as cells are mainly retained in the lung or liver[44,45]. Although intradermal injection of MSCs into the wound was reported to significantly improve healing[46-48], despite the promising potential of MSC therapy, the effectiveness of this approach is still hampered by challenges, such as inadequate cell localization and compromised cell viability at the injury site[49]. To eliminate these problems, researchers have utilized delivery systems to deliver stem cells to the site of injury, and these delivery systems have been suggested to significantly improve stem cell viability and wound implantation rates. Furthermore, the delivery system scaffold also provides a 3D structure for stem cell migration, proliferation, and differentiation[50]. Figure 1 illustrates the summary of the main content of this review.

Figure 1
Figure 1 The summary of the main content of this review. MSC: Mesenchymal stem cell; DFU: Diabetic foot ulcer.

In the context of MSCs-based treatment of DFU, we reviewed the advancements made in preclinical and clinical application of various delivery systems, as depicted in Figure 2.

Figure 2
Figure 2 Themesenchymal stem cells-based drug delivery system for the treatment of diabetic foot ulcer.
POTENTIAL MECHANISM OF ACTION OF MSC THERAPY FOR DFU

Stem cells exhibited diverse developmental potentials, enabling them to be classified into three categories: Totipotent stem cells (comprising embryonic stem cells), pluripotent stem cells, and specialized stem cells (e.g., hematopoietic stem cells and neural stem cells). Furthermore, stem cells can be distinguished from embryonic and somatic stem cells based on their developmental stage. Embryonic stem cells are derived from embryonic and fetal tissues, while somatic stem cells are extracted from the organs or tissues of postnatal individuals. The two types of stem cells each have advantages. However, the utilization of embryonic stem cells is subject to restrictions for several reasons, including the ethical controversy surrounding their use for medical purposes and the presence of legal constraints limiting their application. In addition, the sources of embryonic stem cells are limited. In vitro technologies for amplification and purification are still in an early stage of development. The utilization of allogeneic transplantation of embryonic stem cells in individuals of different genetic backgrounds is accompanied by the potential hazards of immune rejection and teratoma formation. In contrast, somatic stem cells, including hematopoietic stem cells, neural stem cells, liver stem cells, and MSCs, possess relatively lower immunogenicity and reduced risk of tumorigenesis, rendering them more appropriate for various clinical applications. The choice of stem cell type for a particular therapeutic intervention should be based on several important factors, including their safety profiles, efficacy, and compatibility with the recipient’s immune system[51]. Clinical and animal experiments have indicated that there are two striking biological features of somatic stem cells. First, once transplanted, somatic stem cells undergo chemotaxis and are recruited to the site of damage in massive numbers. Second, once they reach the site of damage, somatic stem cells undergo induced differentiation into cells essential for damaged tissue repair in the local microenvironment. Site-specific differentiation is one of the mechanisms by which somatic stem cells promote damaged tissue repair, and researchers have revealed that MSCs promote reconstruction of the local microcirculation by releasing cytokines and growth factors through paracrine and endocrine effects; this is the main mechanism by which these cells accelerate wound healing[52,53]. These findings may expand the indications for clinical treatment using somatic stem cells and facilitate the development of somatic cell and tissue engineering approaches. MSCs are nonhematopoietic stem cells resulting from mesoderm differentiation. They are adherent cells in vitro and can be massively expanded and differentiated into mesenchymal cells, also known as mesenchymal progenitor cells. The latter can be further differentiated into various connective tissue cells, including adipose cells, osteocytes, chondrocytes, vascular endothelial cells, osteoblasts, myoblasts, and nerve cells[54]. MSCs are a versatile cell population that can be obtained from various sources, including bone marrow, umbilical cord and blood, peripheral blood, fat, liver, gingiva, oral mucosa, amniotic fluid, as well as interstitial and connective tissues of organs. Due to the abundance of MSCs in these sources and their ability to differentiate into multiple cell types, they are valuable tools in regenerative medicine and tissue engineering research. One of the major advantages of MSCs is that their isolation does not pose ethical issues, unlike some other stem cells. Furthermore, studies have demonstrated that treatment with MSCs is safe and can lead to few side effects, providing another level of confidence in their use. The versatility and safety of MSCs make them an ideal candidate for utilization in tissue engineering investigations and clinical trials[55]. MSCs are easily isolated from different sources and have high proliferative potential and genetic stability. They migrate to damaged tissues, where they exhibit resistance to inflammation, influence the microenvironment, promote angiogenesis, and exert antifibrotic and antiapoptotic effects. Additionally, they release cytokines involved in damage repair and tissue regeneration, contributing to the healing process[56].

The intricate process of wound healing can be divided into four distinct overlapping phases, including homeostasis, inflammation, proliferation, and maturation. During the homeostasis stage, blood vessels in the affected area quickly constrict to minimize bleeding, while platelets form a plug to seal the injured site. During this stage, coagulation factors are activated, coordinating their efforts to form a fibrin clot that plays a vital role in stabilizing the wound. The inflammation phase is characterized by the influx of immune cells, such as neutrophils and macrophages, into the wound area. These cells are responsible for the removal of debris, pathogens, and damaged tissue. Additionally, they release growth factors and cytokines that initiate the subsequent phases of the healing process. In the proliferation stage, fibroblasts and endothelial cells start to proliferate and migrate into the wound bed, leading to the formation of new blood vessels and ECM. This stage also involves the deposition of collagen fibers, which provide structural support to the healing tissue. Finally, in the maturation phase, the newly formed tissue undergoes remodeling and maturation, resulting in a stronger and more organized scar. This process can take several months to complete, during which time the collagen fibers realign and cross-link to increase the tensile strength of the tissue[57]. MSCs exert therapeutic effects on DFU via several mechanisms. First, a key cause of failure of DFU healing is poor blood supply to the site of the ulcer and disrupted angiogenesis. MSCs, through their ability to secrete various growth factors, have been shown to enhance angiogenesis, referring to the process of forming new blood vessels from pre-existing ones. This effect is mediated via both autocrine and endocrine signaling pathways, leading to the upregulation of several growth factors that are crucial for promoting angiogenesis. Vascular endothelial growth factor (VEGF) is a critical growth factor that stands out among others because of its ability to stimulate the proliferation and differentiation of endothelial cells, leading to the formation of new blood vessels. Basic fibroblast growth factor (bFGF), placental growth factor (PIGF), insulin-like growth factor 1 (IGF-1), and angiopoietin-1 (Ang-1) are also key players in promoting angiogenesis by inducing endothelial cell migration and proliferation. In addition to these growth factors, MSCs can also secrete stromal cell-derived factor-1 (SDF-1), erythropoietin, inducible nitric oxide synthase (iNOS), epidermal growth factor (EGF), and keratinocyte growth factor 2. These growth factors work synergistically to further enhance angiogenesis and promote wound healing. The ability of MSCs to increase the levels of these growth factors is particularly relevant in DFUs wherein impaired angiogenesis is a significant contributing factor to poor wound healing. By improving blood flow in the affected area, MSCs can significantly accelerate the repair of DFUs, thus offering a promising therapeutic option for this debilitating condition[58]. Second, MSCs are involved in immunoregulation via different pathways, and they can improve the microenvironment, reduce the inflammatory response and alleviate tissue injury[59]. (1) MSCs exert immunomodulatory effects by inhibiting T-cell activation[60]. T cells usually secrete a variety of proinflammatory factors after skin damage, delaying wound healing. MSCs secrete cytokines, including interferon-γ (IFN-γ), tumour necrosis factor(TNF)-α, interleukin (IL)-1α or IL-1β, and NOS, which inhibit T-cell activation. In addition, MSCs can block antigen-presenting cell maturation, thereby inhibiting the ability of T cells to respond and exert immunomodulatory effects; (2) MSCs inhibit proinflammatory T cells, and immunomodulatory effects are mainly mediated by Th17 cells and Treg cells[61]. One study showed that after the injection of bone-derived mesenchymal stem cells (BMSCs) into a mouse model of experimental allergic encephalomyelitis, Th17 cells were inhibited, accompanied by increases in the percentages of CD4+CD25+Foxp3+ Treg cells and IL-10-producing cells[62]. According to another report, MSCs modulate cytokine secretion by different T-cell subsets. Specifically, in experimental studies, it has been observed that the administration of MSCs results in a noticeable decrease in the secretion of certain proinflammatory cytokines, specifically IFN-γ and TNF-α. On the other hand, there is a concomitant increase in the secretion of anti-inflammatory cytokines, such as IL-4 and IL-10. Moreover, the percentage of Treg cells was reported to increase after MSC treatment[63]; (3) MSCs exert immunomodulatory effects by reducing the number of classically activated macrophages (M1-type, proinflammatory) and increasing the number of selectively activated macrophages (M2-type, anti-inflammatory)[64]. It has been shown that the coculture of MSCs and macrophages reduces the overall number of macrophages/monocytes, including M1 macrophages, but increases the percentage of M2 macrophages. In addition, MSCs induce M2 polarization of macrophages to exert immunomodulatory effects, enhancing wound repair; and (4) MSCs exert immunomodulatory effects by reducing reactive oxygen species (ROS) levels[65]. In damaged tissues, macrophages engulf bacteria, apoptotic inflammatory cells or cell fragments, thereby killing pathogens and eliminating other harmful factors. However, the prolonged presence of neutrophils after phagocytosis usually results in massive production of ROS, which ultimately causes a respiratory burst and tissue injury. MSCs prevent excessive or improper activation of oxidative metabolism in neutrophils, while preserving the phagocytic ability of neutrophils. MSCs also inhibit neutrophil apoptosis, reducing ROS generation and the intensity of the respiratory burst. In summary, MSCs have exhibited to exert immunomodulatory effects, leading to the alleviation of inflammatory responses and tissue injury, as well as the promotion of wound healing. Another important characteristic of MSCs is their ability to self-replicate and differentiate into different types of mature cells that possess distinct morphology, specific molecular markers, and specialized functions. This multidirectional differentiation potential allows for the generation of a diverse range of cell types, which has significant implications for regenerative medicine and other therapeutic applications involving tissue repair and regeneration[66]. MSCs can be divided into endothelial cells and keratinocytes that are involved in injury repair. Following transplantation of MSCs, there is a notable rise in the levels of angiogenic factors such as IGF-1, EGF, and IL-8. Moreover, the expression of keratinocyte-specific proteins and cytokeratin in wounds leads to the significant proliferation of various cell types, including epithelial cells and keratinocytes. These proteins expressed in wounds facilitate angiogenesis, epithelial cell regeneration, and wound healing. According to another study, in a rat model of DFU, MSCs were specifically localized to target ulcers, where keratin 19 secretion, formation of keratinocytes and ECM, and epithelial cell regeneration were promoted[67]. MSCs show promise for the treatment of DFU, and some encouraging results have been obtained from clinical trials. Further optimization is needed in terms of the following aspects of treatment with MSCs: The feasibility of treatment using autologous and allogeneic MSC transplantation in patients with DFU, factors related to transplantation efficiency, the standardization of MSC quality detection methods and assessment criteria, MSC delivery systems, and methods to determine the survival rate of transplanted MSCs and the effectiveness and long-term efficacy of MSC transplantation. MSC therapy has potential for promoting tissue regeneration and healing in DFU through the differentiation of MSCs into various cell types and the release of growth factors and cytokines. Furthermore, MSC therapy can enhance angiogenesis and blood vessel formation, increasing blood flow to the ulcerated area and promoting healing, while also preventing infections and reducing the need for antibiotics through its antimicrobial effects. Overall, these mechanisms suggest that MSC therapy may be a promising approach for treating DFU, and MSCs can provide neuroprotection by promoting nerve regeneration and reducing neuropathic pain associated with DFU. MSCs possess the capacity to produce and release neurotrophic factors, such as nerve growth factor and brain-derived neurotrophic factor, which are potent mediators of nerve growth and survival. The secretion of these factors by MSCs can contribute to the repair and regeneration of damaged nerve tissues by promoting neuronal cell proliferation and differentiation, enhancing axonal sprouting and myelination, and reducing neuronal apoptosis.

ROLES OF MSCS-DERIVED EXOSOMES IN DFUHEALING

Medical professionals have been perplexed by the limited efficacy of MSCs in promoting wound healing. Low survival and proliferation rates of MSCs due to micro-environmental factors, such as ischemia, hypoxia, and inflammation, further affect the efficacy of MSCs-based treatment. An increasing body of research suggests that the transplantation of MSCs facilitates wound healing through two distinct mechanisms. One way is through direct differentiation, where MSCs differentiate into specific cell types such as fibroblasts, myofibroblasts, and endothelial cells, all of which contribute to tissue repair and angiogenesis. The other mechanism involves a paracrine effect, in which MSCs release various bioactive molecules, such as growth factors and cytokines, promoting the proliferation and migration of nearby cells involved in wound healing[68]. MSCs-derived exosomes act as mediators that deliver membrane receptors, proteins, mRNAs and microRNAs to receptor cells. Due to modulatory effects, gene expression and protein translation undergo changes in receptor cells, thereby influencing the biological activity of target cells. Exosome-based treatment provides a promising approach to overcome various limitations associated with stem cell-based treatment. These limitations include the challenge of large cell volume impeding capillary flow, the low dose and potency of stem cells, the potential presence of mutations or damaged DNA in stem cells, their potential to impair immunocompetence and the immune response, as well as their tendency to exhibit poor differentiation. Exosomes are paracrine products of stem cells and exert similar effects as stem cells. Exosomes are involved in a series of important processes during wound healing, including inflammatory regulation, angiogenesis, epithelial regeneration, and collagen deposition. Bone marrow is the most common site for harvesting MSCs. The utilization of BMSCs that exert their therapeutic effects through paracrine exosomes has been the subject of extensive investigation in the treatment of DFU. For instance, Wang et al[69] conducted a comprehensive analysis of the effects of exosomes originating from BMSCs on the tube-forming capabilities of endothelial progenitor cells (EPCs). Their results highlighted that BMSCs-derived exosomes exerted a significant modulatory influence on Nrf2, which ultimately led to reduced wound inflammation. Consequently, the exosomes played a vital role in promoting wound healing, re-epithelialization, collagen deposition, and angiogenesis in diabetic rats. Ding et al[70] carried out subcutaneous injection of exosomes into skin wounds on the backs of diabetic rats. Following the transplantation of exosomes, the wound healing rate was notably higher in the group after 7 and 14 dvs that in the control group. The findings suggest that exosomes derived from BMSCs triggered the PI3K/AKT signaling pathway via miRNA-126-mediated PTEN downregulation, leading to proangiogenic characteristics both in vivo and in vitro. Furthermore, adipose-derived stem cells (ADSCs) also facilitate the repair of diabetic ulcers. ADSC-derived exosome transplantation may be a new method for treating DFU. A group of researchers conducted a study in which they extracted exosomes from ADSC-conditioned medium and combined them with EPC cells. Their findings indicated that the exosomes derived from ADSCs had a regulatory impact on LINC00511. This, in turn, impeded Twist1 ubiquitination and degradation induced by PAQR3. Ultimately, this process encouraged the proliferation, migration, and angiogenesis of EPCs, thereby accelerating the healing of DFUs[71]. Li et al[72]revealed that ADSC-derived exosomes inhibited ROS and inflammatory cytokine production, thereby inhibiting EPC aging induced by high glucose and an oxidative microenvironment. Other effects included enhancing EPC viability, proliferation, and angiogenesis capacity and improving vascularization. Moreover, it was suggested that Nrf2-overexpressing ADSC-derived exosomes facilitated diabetic wound healing by enhancing collagen deposition, tissue fibrosis, and micro-angiogenesis. The mechanism may involve a promoting effect on vascularization and growth factor release and mitigation of the oxidative stress response. In addition, researchers have demonstrated the influences of human umbilical cord MSCs-derived exosomes on DFU healing. For instance, Hu et al’s research involved investigating the effects of exosomes derived from human umbilical cord MSCs on promoting angiogenesis and fibroblast functions, leading to improved skin wound healing. This was achieved through PTEN inhibition by miR-21-3p and SPRY1, which resulted in enhanced healing of skin wounds[73]. Liu et al[74] conducted a study to determine the efficacy of MSCs derived from human umbilical cords in improving wound healing and angiogenesis in a rat model of deep second-degree burn wounds. They observed a higher rate of wound closure and increased expression of CD31 in vivo. Furthermore, they found evidence suggesting that these MSCs facilitated the proliferation, migration, and tube formation abilities of human umbilical vein endothelial cells via exosome-mediated secretion of Ang-2. Despite these findings, there is a dearth of studies comparing the therapeutic potential of exosomes derived from MSCs of varied tissues in treating DFU, highlighting the need for further research in this area. There are still many problems to be resolved, such as the source and type of cells, isolation technique, dosage, transplantation method, and amplification method for MSCs-derived exosomes. Preclinical and clinical studies with large sample sizes are still needed in the future. Exosomes derived from MSCs have emerged as critical players in the process of wound healing acceleration and promotion for individuals afflicted with DFU. These minute vesicles are laden with a plethora of bioactive molecules, including growth factors, cytokines, and microRNAs, that intricately regulate multiple cellular processes crucial to the wound healing cascade. Their multifaceted mechanisms of action make them an attractive therapeutic avenue for DFUs, as they not only promote angiogenesis and cell proliferation, but also modulate inflammation and ECM remodeling. Overall, MSCs-derived exosomes offer a promising approach to treat DFU, as they provide a safe and effective alternative to the use of whole cells. They can be easily obtained from MSCs through noninvasive techniques, and their administration has minimal risks compared with the use of whole cells (Figure 3)[75].

Figure 3
Figure 3 Illustration of mesenchymal stem cells-derived exosomes in the healing of diabetic foot ulcer[75].
MSC DELIVERY VIA HYDROGEL SCAFFOLDS FOR DFU TREATMENT

Hydrogels are complex structures made up of a mesh-like network of polymer chains that are chemically linked together. This unique arrangement allows hydrogels to absorb vast amounts of water, up to hundreds of times their own weight, while still maintaining their structural integrity. The implantation of hydrogels has been demonstrated to enhance surface cytocompatibility, antibacterial properties, and the preservation of cell viability at the targeted site, reflecting their capability in alleviating wound healing[76,77]. Naturally derived hydrogels are biocompatible and biodegradable, interact with innate immune cells, and they are structurally similar to natural human tissues[78-80]. However, there has been a growing interest in the development of hydrogels with tailored properties and performance. However, studies have demonstrated that natural hydrogels possess a limited set of mechanical characteristics and tend to have significant fluctuations in properties between batches[81,82]. This limitation has led to a shift towards the use of synthetic hydrogels, which offer numerous advantages over their natural counterparts. Synthetic hydrogels have significantly longer lifetimes, higher water absorption capacities, and greater gel strengths compared to natural hydrogels[83,84]. Furthermore, they are known to be stable even under severe temperature fluctuations, making them ideal for a wide range of applications[85,86]. To capitalize on the unique properties of both types of hydrogels, composite hydrogels that combine natural and synthetic components have emerged as a promising alternative. These composite hydrogels offer the possibility of creating materials with controllable and customizable structures and functions, thereby expanding the range of potential applications[87,88]. Composite hydrogels are emerging as promising materials for tissue engineering due to their ability to be engineered with specific properties such as size, shape, surface activity, biodegradability, and biocompatibility. By carefully tailoring these characteristics, hydrogel scaffolds can provide a precise mechanical and biological environment to support cell growth and tissue regeneration[89]. New research has revealed that the use of type I rat tail collagen hydrogel to deliver murine BMSCs and adipose-derived MSCs (ADMSCs) could noticeably modulate immune and inflammatory responses at wound sites. This could be attained via upregulating the expression levels of growth factors, including VEGF, as well as by attracting macrophages, which play a crucial role in tissue repair. This approach has been particularly successful in promoting wound healing in diabetic mice, where impaired immune function and chronic inflammation mainly hinder the natural healing process[90-93]. It has been demonstrated that collagen hydrogels can successfully deliver MSCs to the wound site and improve healing. Murine ADMSCs delivered via a hyperbranched polyethylene glycol diacrylate-cross-linked gelatin hydrogel exhibit superior cell adhesion and are viable and metabolically active for 3 wk[94,95]. Db/db diabetic mice that are injected with an ADMSC-loaded hydrogel at the wound surface have a significantly improved cell retention rate in the wound, accelerated wound closure, enhanced angiogenesis, and attenuated inflammation[96]. In an effort to promote effective wound healing in the context of diabetes-induced impaired healing, innovative methods for delivering beneficial cells and substances have been investigated. Specifically, studies have revealed that the utilization of a biodegradable n-isopropylacrylamide thermo-sensitive hydrogel to deliver mouse BMSCs could remarkably improve wound healing in db/db mice. This delivery method has been demonstrated to enhance ECM deposition, angiogenesis, re-epithelialization, and granulation tissue formation within wounds. Additionally, it has exhibited the ability to regulate polarization of M1 and M2 macrophages at the wound site[97,98]. Additionally, the utilization of Pluronic F-127, a synthetic biocompatible hydrogel with unique thermo-sensitivity, offers an effective means of encapsulating and delivering numerous rat ADMSCs to the wound site. Such delivery has been found to stimulate angiogenesis and cell proliferation, ultimately leading to expedited wound healing in diabetic rats. These findings suggest the potential for novel therapeutic approaches utilizing these delivery methods to improve healing outcomes in individuals with diabetes-induced impairment[99,100]. Moreover, umbilical MSC implantation withPF-127 and sodium phosphate promotes wound healing and angiogenesis as well as improves dermal regeneration and collagen deposition in diabetic rats[101]. Diabetic rats that received ADMSCs encapsulated in silk fibroin/chitosan hydrogel exhibited significantly increased re-epithelialization, granulosa tissue formation and capillary formation at the wound site 7 d after treatment[102]. Furthermore, the expression levels of epithermal growth factor (EGF), TGF-β, and VEGF were also upregulated in the wound tissues on day 14 post-treatment. Utilizing hydrogels composed of hyaluronic acid and N-carboxyethyl chitosan cross-linked by adipic acid dihydrazide as a delivery mechanism, rat BMSCs have been found to effectively inhibit chronic inflammation, promote granulosa tissue formation, collagen deposition, nucleated cell proliferation, and stimulate angiogenesis in diabetic rats. As a result, these hydrogels can significantly enhance wound healing outcomes in this population[103]. Efforts to improve wound healing outcomes in individuals with diabetes-induced impairment have spurred innovative research in the field of cell delivery mechanisms. Researchers have discovered that delivering rabbit BMSCs via a nitric oxide-releasing S-nitroso-N-acetyl-penicillamine-loaded chitosan/polyvinyl-alcohol hydrogel can significantly enhance wound healing rates, re-epithelialization, and collagen deposition in diabetic rabbits, as described in literature sources[104-106]. Jin et al[107] developed an injectable hydrogel with unique properties such as suitable electrical conductivity and sustained hypoxia that can upregulate HIF-1α and connexin-43 expression in loaded ADMSCs, ultimately facilitating wound closure in diabetic rats. This hydrogel has been found to enhance angiogenesis, promoting the reconstruction of blood vessels, hair follicles, and dermal collagen matrix, further contributing to improved wound healing outcomes[107]. Srifa et al[108] conducted a study wherein they administered VEGFA-overexpressing human BMSCs to wounds in db/db mice either through direct injection or embedding them within a HyStem HP hydrogel. The researchers discovered that both methods of cell delivery enhanced the rate of wound healing; however, between days 7-9 after treatment, the hydrogel group exhibited significantly better wound healing compared to the direct injection group[108]. In a phase II clinical trial of MSC delivery via a hydrogen scaffold (NTC02619877), the authors developed an allogeneic ADMSC hydrogel sheet that can maintain long-term stability under cryopreservation and has been approved for marketing by the Ministry of Food and Drug Safety of South Korea (Approval No. ALL-ASC-DFU-201). This trial showed that 82% of diabetic patients had complete wound closure at week 12 after receiving the allogeneic ADMSC hydrogel sheet compared with 53% of controls, and adverse reactions were not observed after treatment, demonstrating that ADMSC delivery via the hydrogel is effective and safe for diabetic wound healing[109]. An in-depth case study was undertaken to explore the implications of utilizing sodium alginate hydrogel-encapsulated placenta-derived MSCs as a topical treatment for foot ulcers in patients with T2DM. The findings were exceptionally promising, with complete wound healing observed three weeks post-treatment, along with marked improvements in foot pain and minimal toxicity. Furthermore, no recurrence was noted during the six-month follow-up period[110]. However, as it was a case study, further investigation is required.

At present, MSC delivery systems supported by hydrogels meet the need for local controlled release and create a 3D bionic environment. Hydrogel scaffolds not only increase bioavailability and antibacterial capacity by improving transport dynamics, but also provide a moist and stable wound repair environment for damaged ulcers, promoting synergy between skin cells and cytokines. Functional materials should be optimized in terms of the degree of crosslinking, porosity, swelling property, mechanical performance, cell adhesion, permeability, toxicity and cost efficiency. They should be synthesized with optimal healing strength to best mimic the ECM microenvironment to maintain the features and activity of each component. The development of hydrogel materials as MSC delivery systems or scaffolds holds significant potential for the future advancements in this field. In recent years, increasing research on the complex dynamics of biological systems has improved hydrogel material properties and hydrogel preparation methods and led to continual optimization of the interactions between organisms and scaffold materials. It can be concluded that the use of hydrogel scaffolds for the delivery of MSCs can significantly improve the therapeutic potential of MSCs-based treatments for DFU. Hydrogels offer a 3D framework that maintains the viability, proliferation, and specialization of cells, and simultaneously enables regulated discharge of growth factors that stimulate recovery of wounds and regeneration of tissues. Additionally, the mechanical properties of hydrogels can be modified to match the specific needs of DFU treatment, such as flexibility to accommodate weight-bearing or stiffness to support tissue regeneration. Overall, MSC-hydrogel therapies can effectively reduce inflammation, stimulate angiogenesis, and improve wound closure rates in human/animal models of DFU (Figure 4)[111]. However, there is an expectation for the development and application of a wider range of functional hydrogels that can perform distinct functions to facilitate DFU repair.

Figure 4
Figure 4 Delivery of mesenchymal stem cells and adipose-derived stem cells by hydrogel scaffolds for diabetic foot ulcer[111]. MSC: Mesenchymal stem cells; ASC: Adipose-derived stem cells.
MSC DELIVERY VIA FIBER SCAFFOLDS FOR DFU TREATMENT

Fiber scaffolds are 3D structures primarily composed of micro- or nanoscale fibers prepared by electrospinning to simulate the structure of natural human tissues[112]. The utilization of fiber scaffolds has been observed in multiple domains of tissue engineering, such as bone, cartilage, skin, vascular, and neural tissue engineering[113-115]. The notable surface-to-volume ratio of fiber scaffolds provides an ideal setting for cell adhesion, although the limited pore size may pose a challenge to cell migration. Consequently, the properties of fiber scaffolds should be tailored based on the specific cell type being cultured[116,117]. The employment of fiber scaffolds for the purpose of wound healing has recently garnered significant attention, as they have demonstrated a remarkable potential in promoting cell-cell and cell-ECM interactions, while also directing the functions and behaviors (e.g., cell morphology, proliferation, and differentiation) of diverse cells, including MSCs[118-121]. In the realm of diabetes wound healing, the utilization of fiber scaffolds for MSC transportation has been extensively employed. Chen et al[122] have devised a 3D scaffold that is comprised of vertically or radially aligned nanofibers that can be customized to fit the size, depth, and configuration of different T2D wounds. The scaffold itself possesses an impressive ability to regain its shape both in water and the atmosphere, even after undergoing compression. When infused with BMSCs, this 3D fiber scaffold has the potential to stimulate the development of granulosa tissue, encourage angiogenesis, and facilitate collagen deposition within the T2D wound[122,123]. Furthermore, Hou et al[124] developed a novel electrospun nanofibrous scaffold, which was composed of 80% polylactic acid, 10% silk, and 10% collagen. The scaffold was designed to deliver HO-1-overexpressing human BMSCs to wounds in diabetic mice. This hybrid scaffold has exhibited promising results in improving wound healing in diabetic mice. The authors found that this approach significantly improved angiogenesis and wound healing via the Akt signaling pathway[124]. He et al[125] delivered human BMSCs that overexpressed neurotrophic factors to wounds in diabetic mice via an electrospun biomaterial, and this method was found to significantly accelerate wound closure and increase angiogenesis[125]. In addition, delivery of human ADMSCs to wounds via silk fibroin scaffolds led to faster complete wound closure in db/db mice (d10) than in control mice (d15-17)[126-128]. It is easy to manipulate fiber scaffolds, but their preparation is very complicated. The dimensions and morphology of fiber scaffolds are affected by many factors, including solution viscosity, voltage, temperature, humidity, the distance between receiver and nozzle, and the loading flow rate of the solution. Ideal nanoscale fiber scaffolds can be fabricated only by systematically optimizing the above parameters. Fiber scaffolds offer a favorable setting for the growth and differentiation of MSCs, facilitate the regulated release of growth factors, and assist in the process of wound healing. Thus, MSC delivery via fiber scaffolds can improve wound closure rates, boost angiogenesis, reduce inflammation, and potentially offer better outcomes than other platforms for MSC delivery. Additional research is essential to enhance the design and production of fiber scaffolds for delivering MSCs, develop universally recognized procedures, and assess the enduring safety and efficacy of this technique.

MSC DELIVERY VIA SPONGE SCAFFOLDS FOR DFU TREATMENT

Scaffold sponges, which are widely used in tissue engineering and regenerative medicine, can be fabricated using a diverse range of techniques, involving porogen leaching, gas foaming, and freeze-drying. These methods enable the creation of scaffolds made from natural or synthetic polymers that possess a high degree of porosity and a uniform network of interconnected pores[129-131]. Despite the longstanding use of sponge scaffolds in the biomedical field, researchers have long been striving to generate an environment that can provide support for the ECM in autologous cells and tissues. A 3D system with the ability to modulate cell viability and customize the structural and architectural properties, such as porosity, pore size, and interconnected dimension offers a significant degree of freedom. These features synergistically contribute to the regulation of cell-material interactions and consequently promote tissue growth within the scaffold gap[132,133]. Sponge and hydrogel scaffolds mainly differ in their method of fabrication, which results in differences in the water content of the scaffold. In contrast to hydrogels, the creation of sponge scaffolds is a time-consuming process that necessitates surface and structural modifications based on the type of cell and host tissue being used, as stated in the original citation. However, sponge scaffolds offer several potential benefits for skin wound healing. First, their highly porous structure closely resembles that of the ECM, which aids in supporting cell migration to the site of injury[134-136]. Second, because of their water absorption and retention capabilities, sponge scaffolds can absorb exudates from the wound site, providing a favorable environment for cell proliferation and migration[137-139]. The utilization of MSC scaffolds for diabetic wound healing frequently involves the use of sponge scaffolds made with collagen and chitosan. To create a collagen sponge scaffold, O’Loughlin et al[140] utilized freeze-drying techniques. Delivery of allogeneic BMSCs via topical application of the collagen sponge scaffold resulted in superior wound closure and angiogenesis on day 7 following implantation in diabetic rabbits when compared to the no treatment control group[140,141]. Tong et al[142] developed a collagen-chitosan sponge scaffold that is suitable for BMSC delivery by employing cross-linking and freeze-drying techniques, as mentioned in the original citation. This sponge scaffold has a 100 μm pore network and appropriate biodegradability and swelling ratio[142]. This type of scaffold creates an environment that is favorable for cell growth and stimulates hypoxia-pretreated rat BMSCs to produce higher levels of VEGF and platelet-derived growth factor, as well as upregulate expression of key transcription factors, including HIF-1α, while retaining cell viability. In STZ-induced diabetic rats, the BMSC-sponge scaffold group exhibited significantly improved wound closure, increased angiogenesis, and reduced inflammation (upregulated IL-10 gene and protein expression on days 7 and 14 post-implantation) vs the control group. Furthermore, Ní Annaidh et al[143] fabricated a sponge scaffold made of collagen and chitosan that was infused with simvastatin. The scaffold had high porosity, with pore sizes ranging from 20-200 μm, and possessed sufficient mechanical strength while maintaining elasticity similar to human skin. Additionally, the release of simvastatin from the scaffold could be controlled[143-145]. It was previously shown that delivery of rat ADMSCs by a sponge scaffold made of glycol chitosan and polyurethane combined with acupuncture had a synergistic immunomodulatory effect on wounds in mice with STZ-induced diabetes. This combination therapy improved wound closure and promoted complete re-epithelialization within 8 d, in contrast with ADMSCs alone[146-150]. In addition, on day 8 after treatment, the wound displayed an increase in secretion of SDF-1 and TGFβ-1, while production of TNF-α and IL-1β was reduced. Additionally, sponge scaffolds have the potential to serve as a cell delivery system in conjunction with growth factors. Delivery of Balb/c mouse BMSCs by chitosan-alginate sponge scaffolds combined with EGF can enhance cell viability and transcription factor expression, maintain MSC pluripotency and self-renewal capability, and promote collagen deposition and angiogenesis by increasing granulosa tissue formation in the wounds of diabetic rats[151-154]. De Francesco et al[155] conducted a study to assess how effective autologous dermal micro-grafts, similar to MSCs, could be in treating DFUs by delivering them through collagen sponge scaffolds. The dermal micro-grafts were obtained through mechanical dissociation of small pieces of skin tissues and express MSC markers (e.g., CD34, CD73, CD90, and CD105) in vitro. The results showed that the micro-grafts remained viable and proliferative in the collagen scaffold, indicating that MSC-loaded sponge scaffolds could remarkably improve ulcer wound closure and enhance patients’ quality of life[155-157].

A pore size of a few hundred microns is usually considered most suitable to ensure that cells obtain the needed nutrients. In addition, the porosity is generally above 70%, which provides enough space for cell penetration and mass transfer. Initial cell attachment is also guaranteed by the adequacy of the materials. The size of interconnections is a crucial factor that affects the transport characteristics of the entire porous structure. There seems to be a consensus that the minimal interconnection size is approximately 50 μm to allow angiogenesis and cell migration. The regulation of cell differentiation and function can be influenced by physical parameters, involving material hardness, viscoelasticity, and pore curvature. However, it is important to acknowledge the significant role that endogenous factors play in promoting complete cell maturation. Sponge scaffolds have demonstrated their effectiveness as delivery vehicles for MSCs, providing a supportive environment for cell growth and promoting tissue regeneration at the site of the ulcer. The utilization of sponge scaffolds has the potential to regulate the immune response and alleviate inflammation at the site of injury, thereby facilitating the process of wound healing. MSCs delivered by sponge scaffolds can improve angiogenesis and blood vessel formation, increasing blood flow to the ulcerated area and promoting healing. The controlled release of MSCs from sponge scaffolds can provide sustained therapeutic effects over time, reducing the need for frequent treatments. Compared to other methods of MSC delivery, the use of sponge scaffolds may improve patient compliance and reduce the risk of infection. The potential of MSCs delivered via sponge scaffolds to promote DFU wound healing is notable. These natural polymer-based scaffolds provide a 3D environment that supports MSC survival, proliferation, and differentiation, facilitating interactions with surrounding tissues. Overall, compared with the delivery of MSCs via a silk fibroin sponge scaffold, utilizing chitosan-based sponge scaffolds for MSC delivery could potentially lead to faster healing rates and increased collagen deposition. Moreover, significant reduction in ulcer size and improvement in wound closure time could also be noted.

MSC DELIVERY BY ACELLULAR BIOSCAFFOLDS FOR DFU REPAIR

Acellular bioscaffolds refer to biological substances that are derived from human or animal organs or tissues, which undergo decellularization techniques for the removal of immunogenic cellular components[158-161]. Acellular bioscaffolds have exhibited favorable results in diverse tissues and organs and have garnered noticeable interest in the domain of tissue engineering. Mechanical (freezing or force), chemical (acid or Triton), and enzymatic (trypsin or pepsin) methods are decellularization techniques that can be employed. While combining multiple techniques is often more effective than using a single method, it is crucial to choose the appropriate decellularization approach based on the distinctive features of each tissue type[162]. Acellular bioscaffolds are composed mainly of ECM and other extracellular macromolecules (e.g., collagen, elastin, fibronectin, laminin, and stromal cell proteins). They possess the unique characteristic of being nonimmunogenic 3D structures, distinguishing them from other synthetic scaffolds[160]. These characteristics are critical for the identification and development of implantable scaffolds for diabetic wounds. The utilization of acellular bioscaffolds has numerous benefits in treating diabetic wounds, involving the ability to replace damaged ECM with a variety of proteins (e.g., collagen, glycosaminoglycans, proteoglycans, and glycoproteins). Additionally, acellular bioscaffolds facilitate the infiltration of host cells and regulation of immune responses. They also promote angiogenesis and granulation tissue formation[163,164]. There are currently few commercially available acellular bioscaffolds for wound healing[165,166]. These scaffolds are manufactured differently and hence have differentmechanical properties and varying abilities to support skin regeneration[167]. Several studies have examined the delivery of MSCs by acellular bioscaffolds for the treatment of diabetic wounds. Shi et al[168] developed a decellularized dermal matrix scaffold, called book-shaped decellularized dermal matrix (BDDM), closely resembling native dermal tissues in terms of histology, microstructure, and composition. This noncytotoxic scaffold exhibited low immunogenicity and supported the attachment and proliferation of ADMSCs. The researchers also synthesized a recombinant growth factor, collagen-binding domain (CBD)-bFGF, by fusing a CBD-bFGF, and tethered it to the collagen fibers of the BDDM scaffold. This was resulted in the creation of a functional scaffold (CBD-bFF/BDDM), promoting endothelial cell inducibility more effectively. In vitro tests revealed that CBD-bFGF/BDDM scaffold can gradually release tethered bFGF and facilitate ADMSC interactions until endothelial differentiation is achieved. To evaluate the effectiveness of this scaffold, ADMSCs were cultured to create a cell sheet which was placed between PIGF, CBD-bFGF and BDDM before being transplanted into diabetic rats. Results from in vivo experiments showed that the implantation of ADMSC-loaded CBD-bFGF/BDDM scaffold promoted the formation of granulation tissue and angiogenesis. It also facilitated collagen deposition and remodeling. Zhang et al[169] developed a novel delivery system for exogenous cells using nanoparticles encapsulating IL-8 along with polylactic-co-glycolic acid (PLGA) loaded onto acellular matrix insulin-like growth factor 1. This efficient delivery medium, termed PLGA@IL-8/ADM, was found to promote significant proliferation and endothelial differentiation of the MSCs while increasing their survival rate. Moreover, PLGA@IL-8/ADM scaffold loaded with MSCs facilitated capillary construction, collagen deposition, and Ang-1 wound healing in skin wounds of mice with STZ-induced diabetes, thereby demonstrating its effectiveness as a therapeutic intervention for diabetic wounds. These findings highlight the promise of the PLGA@IL-18/ADM scaffold as a novel delivery system for exogenous cells that can aid in tissue regeneration. Chu et al[170] conducted a study, in which they loaded mouse BMSCs onto a decellularized dermal matrix scaffold obtained from normal mouse skin and applied it at full-thickness cutaneous wound sites in diabetic mice. The use of MSC-ADM for treating these wounds resulted in a noteworthy increase in the percentage of wound closure, a boost in type I collagen fiber synthesis, and an acceleration of both angiogenesis and re-epithelization[170-172]. Moreover, a cell delivery platform comprised of acellular dermal matrix and reduced graphene oxide has high stability and promising mechanical properties. Upon delivery of murine BMSCs to wounds in diabetic mice, this acellular bioscaffold provides a favorable milieu for BMSC adhesion and proliferation, promotes angiogenesis and collagen deposition, and accelerates wound healing[173,174]. A study using acellular dermal matrix to deliver human umbilical cord MSCs showed that MSC proliferation and differentiation were regulated by activation of Wnt signaling, which ultimately promoted wound healing in diabetic rats[175].

Matrix formed by the removal of cells from dermal tissues should be used to deliver MSCs to wounds, thereby resolving immune rejection of allografts. Research priorities related to the use of decellularized vascular bioscaffolds for MSC delivery and wound repair include cell growth promotion, vascularization, and appendage regeneration, which may represent the future of DFU treatment.

Acellular bioscaffolds have demonstrated their efficacy as a successful delivery platform for MSCs, facilitating a supportive environment for cell growth and driving tissue regeneration at the site of the ulcer. The use of acellular bioscaffolds can modulate the immune response and reduce inflammation at the wound site, aiding in the healing process. MSCs delivered by acellular bioscaffolds can improve angiogenesis and blood vessel formation, increasing blood flow to the ulcerated area and promoting healing. The controlled release of MSCs from acellular bioscaffolds can provide sustained therapeutic effects over time, reducing the need for frequent treatments. The use of acellular bioscaffolds may improve patient compliance and reduce the risk of infection compared with other methods of MSC delivery. Overall, MSCs can promote wound healing by secreting cytokines and growth factors, reducing inflammation, and stimulating angiogenesis. However, delivering MSCs directly to the site of DFU wounds can be challenging due to low survival rates and poor engraftment. Using an acellular bioscaffold as a delivery vehicle could potentially improve the viability and functionality of transplanted MSCs and enhance their therapeutic effects for DFUs. The bioscaffold provides a 3D microenvironment similar to the natural ECM, allowing for cell attachment and migration, promoting angiogenesis, and increasing nutrient and oxygen availability.

CONCLUSION

In conclusion, the increasing incidence of nontraumatic amputation and the poor therapeutic effect of currently available treatments make DFU one of the most important clinical challenges[176]. MSCs are widely utilized in the treatment of DFU, however, their efficacy needs to be improved. The application of different MSCs-based drug delivery systems for DFU and the relevant mechanisms were discussed (Table 1). Several preclinical investigations have exhibited impressive results, indicating that diverse MSCs-based drug delivery mechanisms can expedite wound healing and stimulate skin regeneration in DFU. However, there are still limited clinical data regarding the utilization of MSCs-based drug delivery systems for treating DFU. There is no consistent correlation between the results obtained in animal and human models. The safety, efficacy, and cost of different MSCs-based drug delivery systems should be deeply investigated in the future research. An interdisciplinary approach is required to develop cells-based drug delivery systems for the clinical treatment of DFU.

Table 1 Summary of four mesenchymal stem cells-based drug delivery systems for diabetic foot ulcer.
Scaffold
Advantage
Disadvantage
HydrogelExcellent biocompatibility and biodegradability[33-36], low cytotoxicity[74-79], with similar structure as human tissues[77-82], longer lifetime, higher water absorption capacity[90-95], and greater gel strength[107-111]Weak mechanical strength and high batch-to-batch variability[161-165]
FiberWell-mimicking the human tissues and excellent cell attachment[112-118]Small pore size limiting cell migration[46-48]
SpongeAppropriate microenvironment for cell attachment, migration, and nutrient transition[59-63]; Exceptional ability of loading, retaining and releasing of fluids[103-107]Longer time for fabrication procedure[79-83];Requirement to adjust according to cell type and host tissues[137-142]
DecellularizationMimicking an optimal non-immune environment with native three-dimensional structures and various bioactive components[121-124]; Flexible mechanical properties[127-133]; Satisfactory mechanical strength[139-142]Complete decellularization is complex and time consuming[152-156]
Footnotes

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

Peer-review model: Single blind

Specialty type: Endocrinology and metabolism

Country/Territory of origin: China

Peer-review report’s scientific quality classification

Grade A (Excellent): A, A

Grade B (Very good): B, B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Sikiric P, Croatia; Wang S, China; Cai L, United States S-Editor: Fan JR L-Editor: A P-Editor: Yuan YY

References
1.  International Hypoglycaemia Study Group. Hypoglycaemia, cardiovascular disease, and mortality in diabetes: epidemiology, pathogenesis, and management. Lancet Diabetes Endocrinol. 2019;7:385-396.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 190]  [Cited by in F6Publishing: 236]  [Article Influence: 47.2]  [Reference Citation Analysis (1)]
2.  Lei X, Qiu S, Yang G, Wu Q. Adiponectin and metabolic cardiovascular diseases: Therapeutic opportunities and challenges. Genes Dis. 2023;10:1525-1536.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
3.  Yang L, Rong GC, Wu QN. Diabetic foot ulcer: Challenges and future. World J Diabetes. 2022;13:1014-1034.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 2]  [Cited by in F6Publishing: 8]  [Article Influence: 4.0]  [Reference Citation Analysis (1)]
4.  Pu D, Lei X, Leng W, Zheng Y, Chen L, Liang Z, Chen B, Wu Q. Lower limb arterial intervention or autologous platelet-rich gel treatment of diabetic lower extremity arterial disease patients with foot ulcers. Ann Transl Med. 2019;7:485.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
5.  Schaper NC, van Netten JJ, Apelqvist J, Bus SA, Hinchliffe RJ, Lipsky BA; IWGDF Editorial Board. Practical Guidelines on the prevention and management of diabetic foot disease (IWGDF 2019 update). Diabetes Metab Res Rev. 2020;36 Suppl 1:e3266.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 195]  [Cited by in F6Publishing: 282]  [Article Influence: 70.5]  [Reference Citation Analysis (0)]
6.  Sloan G, Selvarajah D, Tesfaye S. Pathogenesis, diagnosis and clinical management of diabetic sensorimotor peripheral neuropathy. Nat Rev Endocrinol. 2021;17:400-420.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 126]  [Article Influence: 42.0]  [Reference Citation Analysis (0)]
7.  Armstrong DG, Boulton AJM, Bus SA. Diabetic Foot Ulcers and Their Recurrence. N Engl J Med. 2017;376:2367-2375.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1953]  [Cited by in F6Publishing: 1745]  [Article Influence: 249.3]  [Reference Citation Analysis (1)]
8.  Ogurtsova K, Guariguata L, Barengo NC, Ruiz PL, Sacre JW, Karuranga S, Sun H, Boyko EJ, Magliano DJ. IDF diabetes Atlas: Global estimates of undiagnosed diabetes in adults for 2021. Diabetes Res Clin Pract. 2022;183:109118.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 202]  [Article Influence: 101.0]  [Reference Citation Analysis (0)]
9.  Saeedi P, Salpea P, Karuranga S, Petersohn I, Malanda B, Gregg EW, Unwin N, Wild SH, Williams R. Mortality attributable to diabetes in 20-79 years old adults, 2019 estimates: Results from the International Diabetes Federation Diabetes Atlas, 9(th) edition. Diabetes Res Clin Pract. 2020;162:108086.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 262]  [Cited by in F6Publishing: 281]  [Article Influence: 70.3]  [Reference Citation Analysis (1)]
10.  Williams R, Karuranga S, Malanda B, Saeedi P, Basit A, Besançon S, Bommer C, Esteghamati A, Ogurtsova K, Zhang P, Colagiuri S. Global and regional estimates and projections of diabetes-related health expenditure: Results from the International Diabetes Federation Diabetes Atlas, 9th edition. Diabetes Res Clin Pract. 2020;162:108072.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 352]  [Cited by in F6Publishing: 384]  [Article Influence: 96.0]  [Reference Citation Analysis (0)]
11.  Zhang P, Lu J, Jing Y, Tang S, Zhu D, Bi Y. Global epidemiology of diabetic foot ulceration: a systematic review and meta-analysis (†). Ann Med. 2017;49:106-116.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 666]  [Cited by in F6Publishing: 783]  [Article Influence: 111.9]  [Reference Citation Analysis (1)]
12.  Fu XL, Ding H, Miao WW, Mao CX, Zhan MQ, Chen HL. Global recurrence rates in diabetic foot ulcers: A systematic review and meta-analysis. Diabetes Metab Res Rev. 2019;35:e3160.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 60]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
13.  Schreml S, Berneburg M. The global burden of diabetic wounds. Br J Dermatol. 2017;176:845-846.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 15]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
14.  Walsh JW, Hoffstad OJ, Sullivan MO, Margolis DJ. Association of diabetic foot ulcer and death in a population-based cohort from the United Kingdom. Diabet Med. 2016;33:1493-1498.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 238]  [Cited by in F6Publishing: 268]  [Article Influence: 33.5]  [Reference Citation Analysis (0)]
15.  Vadiveloo T, Jeffcoate W, Donnan PT, Colhoun HC, McGurnaghan S, Wild S, McCrimmon R, Leese GP; Scottish Diabetes Research Network Epidemiology Group. Amputation-free survival in 17,353 people at high risk for foot ulceration in diabetes: a national observational study. Diabetologia. 2018;61:2590-2597.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 29]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
16.  Yang X, Wang S, Zhang X, Ye C, An X. Development of PVA-based microsphere as a potential embolization agent. Mater Sci Eng C Mater Biol Appl. 2022;135:112677.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
17.  Ouyang Y, Zhao Y, Zheng X, Zhang Y, Zhao J, Wang S, Gu Y. Rapidly degrading and mussel-inspired multifunctional carboxymethyl chitosan/montmorillonite hydrogel for wound hemostasis. Int J Biol Macromol. 2023;242:124960.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 19]  [Reference Citation Analysis (0)]
18.  Ouyang Y, Zhao J, Wang S. Multifunctional hydrogels based on chitosan, hyaluronic acid and other biological macromolecules for the treatment of inflammatory bowel disease: A review. Int J Biol Macromol. 2023;227:505-523.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 32]  [Article Influence: 32.0]  [Reference Citation Analysis (0)]
19.  Liu Y, Liu Y, He W, Mu X, Wu X, Deng J, Nie X. Fibroblasts: Immunomodulatory factors in refractory diabetic wound healing. Front Immunol. 2022;13:918223.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 22]  [Article Influence: 11.0]  [Reference Citation Analysis (1)]
20.  Louiselle AE, Niemiec SM, Zgheib C, Liechty KW. Macrophage polarization and diabetic wound healing. Transl Res. 2021;236:109-116.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 171]  [Article Influence: 57.0]  [Reference Citation Analysis (0)]
21.  Wan R, Weissman JP, Grundman K, Lang L, Grybowski DJ, Galiano RD. Diabetic wound healing: The impact of diabetes on myofibroblast activity and its potential therapeutic treatments. Wound Repair Regen. 2021;29:573-581.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 38]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
22.  Lin CW, Hung CM, Chen WJ, Chen JC, Huang WY, Lu CS, Kuo ML, Chen SG. New Horizons of Macrophage Immunomodulation in the Healing of Diabetic Foot Ulcers. Pharmaceutics. 2022;14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 18]  [Reference Citation Analysis (0)]
23.  Chakraborty R, Borah P, Dutta PP, Sen S. Evolving spectrum of diabetic wound: Mechanistic insights and therapeutic targets. World J Diabetes. 2022;13:696-716.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 5]  [Cited by in F6Publishing: 11]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
24.  Aitcheson SM, Frentiu FD, Hurn SE, Edwards K, Murray RZ. Skin Wound Healing: Normal Macrophage Function and Macrophage Dysfunction in Diabetic Wounds. Molecules. 2021;26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 106]  [Article Influence: 35.3]  [Reference Citation Analysis (0)]
25.  Pawar KB, Desai S, Bhonde RR, Bhole RP, Deshmukh AA. Wound with Diabetes: Present Scenario and Future. Curr Diabetes Rev. 2021;17:136-142.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 11]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
26.  Bus SA, Lavery LA, Monteiro-Soares M, Rasmussen A, Raspovic A, Sacco ICN, van Netten JJ; International Working Group on the Diabetic Foot. Guidelines on the prevention of foot ulcers in persons with diabetes (IWGDF 2019 update). Diabetes Metab Res Rev. 2020;36 Suppl 1:e3269.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 144]  [Article Influence: 36.0]  [Reference Citation Analysis (0)]
27.  Liu C, Ponsero AJ, Armstrong DG, Lipsky BA, Hurwitz BL. The dynamic wound microbiome. BMC Med. 2020;18:358.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 31]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
28.  Nickerson DS, Barrett SL. Comment on IWGDF ulcer prevention guidelines. Endocrinol Diabetes Metab. 2021;4:e00169.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
29.  Jnana A, Muthuraman V, Varghese VK, Chakrabarty S, Murali TS, Ramachandra L, Shenoy KR, Rodrigues GS, Prasad SS, Dendukuri D, Morschhauser A, Nestler J, Peter H, Bier FF, Satyamoorthy K. Microbial Community Distribution and Core Microbiome in Successive Wound Grades of Individuals with Diabetic Foot Ulcers. Appl Environ Microbiol. 2020;86.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 16]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
30.  Patton D, Avsar P, Wilson P, Mairghani M, O'Connor T, Nugent L, Moore Z. Treatment of diabetic foot ulcers: review of the literature with regard to the TIME clinical decision support tool. J Wound Care. 2022;31:771-779.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
31.  Durand BARN, Pouget C, Magnan C, Molle V, Lavigne JP, Dunyach-Remy C. Bacterial Interactions in the Context of Chronic Wound Biofilm: A Review. Microorganisms. 2022;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 15]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
32.  Smythe P, Wilkinson HN. The Skin Microbiome: Current Landscape and Future Opportunities. Int J Mol Sci. 2023;24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 7]  [Reference Citation Analysis (0)]
33.  Wang H, Xu Z, Zhao M, Liu G, Wu J. Advances of hydrogel dressings in diabetic wounds. Biomater Sci. 2021;9:1530-1546.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 64]  [Article Influence: 21.3]  [Reference Citation Analysis (0)]
34.  Wu H, Ni R, Shi Y, Hu Y, Shen Z, Pang Q, Zhu Y. The Promising Hydrogel Candidates for Preclinically Treating Diabetic Foot Ulcer: A Systematic Review and Meta-Analysis. Adv Wound Care (New Rochelle). 2023;12:28-37.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 9]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
35.  Chang M, Nguyen TT. Strategy for Treatment of Infected Diabetic Foot Ulcers. Acc Chem Res. 2021;54:1080-1093.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 191]  [Article Influence: 63.7]  [Reference Citation Analysis (0)]
36.  Perez-Favila A, Martinez-Fierro ML, Rodriguez-Lazalde JG, Cid-Baez MA, Zamudio-Osuna MJ, Martinez-Blanco MDR, Mollinedo-Montaño FE, Rodriguez-Sanchez IP, Castañeda-Miranda R, Garza-Veloz I. Current Therapeutic Strategies in Diabetic Foot Ulcers. Medicina (Kaunas). 2019;55.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 78]  [Article Influence: 15.6]  [Reference Citation Analysis (0)]
37.  Golledge J, Thanigaimani S. Novel therapeutic targets for diabetes-related wounds or ulcers: an update on preclinical and clinical research. Expert Opin Ther Targets. 2021;25:1061-1075.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
38.  Stratman S, Schneider C, Kirsner RS. New Therapies for the Treatment of Diabetic Foot Ulcers: Updated Review of Clinical Trials. Surg Technol Int. 2020;37:37-47.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Yu Q, Qiao GH, Wang M, Yu L, Sun Y, Shi H, Ma TL. Stem Cell-Based Therapy for Diabetic Foot Ulcers. Front Cell Dev Biol. 2022;10:812262.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 22]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
40.  El Hage R, Knippschild U, Arnold T, Hinterseher I. Stem Cell-Based Therapy: A Promising Treatment for Diabetic Foot Ulcer. Biomedicines. 2022;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 7]  [Reference Citation Analysis (0)]
41.  Hsieh MW, Wang WT, Lin CY, Kuo YR, Lee SS, Hou MF, Wu YC. Stem Cell-Based Therapeutic Strategies in Diabetic Wound Healing. Biomedicines. 2022;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
42.  Lopes L, Setia O, Aurshina A, Liu S, Hu H, Isaji T, Liu H, Wang T, Ono S, Guo X, Yatsula B, Guo J, Gu Y, Navarro T, Dardik A. Stem cell therapy for diabetic foot ulcers: a review of preclinical and clinical research. Stem Cell Res Ther. 2018;9:188.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 71]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
43.  Rai V, Moellmer R, Agrawal DK. Stem Cells and Angiogenesis: Implications and Limitations in Enhancing Chronic Diabetic Foot Ulcer Healing. Cells. 2022;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 22]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
44.  Wu Q, Lei X, Chen L, Zheng Y, Huang H, Qian C, Liang Z. Autologous platelet-rich gel combined with in vitro amplification of bone marrow mesenchymal stem cell transplantation to treat the diabetic foot ulcer: a case report. Ann Transl Med. 2018;6:307.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 20]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
45.  Huang YZ, Gou M, Da LC, Zhang WQ, Xie HQ. Mesenchymal Stem Cells for Chronic Wound Healing: Current Status of Preclinical and Clinical Studies. Tissue Eng Part B Rev. 2020;26:555-570.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 91]  [Article Influence: 22.8]  [Reference Citation Analysis (0)]
46.  El Sadik AO, El Ghamrawy TA, Abd El-Galil TI. The Effect of Mesenchymal Stem Cells and Chitosan Gel on Full Thickness Skin Wound Healing in Albino Rats: Histological, Immunohistochemical and Fluorescent Study. PLoS One. 2015;10:e0137544.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 21]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
47.  Rajabian MH, Ghorabi GH, Geramizadeh B, Sameni S, Ayatollahi M. Evaluation of bone marrow derived mesenchymal stem cells for full-thickness wound healing in comparison to tissue engineered chitosan scaffold in rabbit. Tissue Cell. 2017;49:112-121.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 12]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
48.  de Mayo T, Conget P, Becerra-Bayona S, Sossa CL, Galvis V, Arango-Rodríguez ML. The role of bone marrow mesenchymal stromal cell derivatives in skin wound healing in diabetic mice. PLoS One. 2017;12:e0177533.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 55]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
49.  Marquardt LM, Heilshorn SC. Design of Injectable Materials to Improve Stem Cell Transplantation. Curr Stem Cell Rep. 2016;2:207-220.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 110]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
50.  Santra M, Liu YC, Jhanji V, Yam GH. Human SMILE-Derived Stromal Lenticule Scaffold for Regenerative Therapy: Review and Perspectives. Int J Mol Sci. 2022;23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
51.  Rammal H, Harmouch C, Lataillade JJ, Laurent-Maquin D, Labrude P, Menu P, Kerdjoudj H. Stem cells: a promising source for vascular regenerative medicine. Stem Cells Dev. 2014;23:2931-2949.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
52.  Dulak J, Szade K, Szade A, Nowak W, Józkowicz A. Adult stem cells: hopes and hypes of regenerative medicine. Acta Biochim Pol. 2015;62:329-337.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 95]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
53.  Merkert S, Martin U. Site-Specific Genome Engineering in Human Pluripotent Stem Cells. Int J Mol Sci. 2016;17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 13]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
54.  Jacobs SA, Roobrouck VD, Verfaillie CM, Van Gool SW. Immunological characteristics of human mesenchymal stem cells and multipotent adult progenitor cells. Immunol Cell Biol. 2013;91:32-39.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 160]  [Cited by in F6Publishing: 164]  [Article Influence: 14.9]  [Reference Citation Analysis (0)]
55.  Lee CC, Hirasawa N, Garcia KG, Ramanathan D, Kim KD. Stem and progenitor cell microenvironment for bone regeneration and repair. Regen Med. 2019;14:693-702.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
56.  Čamernik K, Barlič A, Drobnič M, Marc J, Jeras M, Zupan J. Mesenchymal Stem Cells in the Musculoskeletal System: From Animal Models to Human Tissue Regeneration? Stem Cell Rev Rep. 2018;14:346-369.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 27]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
57.  Sorg H, Tilkorn DJ, Hager S, Hauser J, Mirastschijski U. Skin Wound Healing: An Update on the Current Knowledge and Concepts. Eur Surg Res. 2017;58:81-94.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 473]  [Cited by in F6Publishing: 578]  [Article Influence: 72.3]  [Reference Citation Analysis (0)]
58.  Azari Z, Nazarnezhad S, Webster TJ, Hoseini SJ, Brouki Milan P, Baino F, Kargozar S. Stem cell-mediated angiogenesis in skin tissue engineering and wound healing. Wound Repair Regen. 2022;30:421-435.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 23]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
59.  Wang Y, Chen X, Cao W, Shi Y. Plasticity of mesenchymal stem cells in immunomodulation: pathological and therapeutic implications. Nat Immunol. 2014;15:1009-1016.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 830]  [Cited by in F6Publishing: 949]  [Article Influence: 105.4]  [Reference Citation Analysis (0)]
60.  Kovach TK, Dighe AS, Lobo PI, Cui Q. Interactions between MSCs and immune cells: implications for bone healing. J Immunol Res. 2015;2015:752510.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 98]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
61.  Tumangelova-Yuzeir K, Naydenov E, Ivanova-Todorova E, Krasimirova E, Vasilev G, Nachev S, Kyurkchiev D. Mesenchymal Stem Cells Derived and Cultured from Glioblastoma Multiforme Increase Tregs, Downregulate Th17, and Induce the Tolerogenic Phenotype of Monocyte-Derived Cells. Stem Cells Int. 2019;2019:6904638.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 13]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
62.  Luz-Crawford P, Kurte M, Bravo-Alegría J, Contreras R, Nova-Lamperti E, Tejedor G, Noël D, Jorgensen C, Figueroa F, Djouad F, Carrión F. 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;4:65.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 286]  [Cited by in F6Publishing: 321]  [Article Influence: 29.2]  [Reference Citation Analysis (0)]
63.  Mohammadpour H, Pourfathollah AA, Nikougoftar Zarif M, Hashemi SM. Increasing proliferation of murine adipose tissue-derived mesenchymal stem cells by TNF-α plus IFN-γ. Immunopharmacol Immunotoxicol. 2016;38:68-76.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
64.  Pajarinen J, Lin T, Gibon E, Kohno Y, Maruyama M, Nathan K, Lu L, Yao Z, Goodman SB. Mesenchymal stem cell-macrophage crosstalk and bone healing. Biomaterials. 2019;196:80-89.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 273]  [Cited by in F6Publishing: 430]  [Article Influence: 71.7]  [Reference Citation Analysis (0)]
65.  Muinos-López E, Ripalda-Cemboráin P, López-Martínez T, González-Gil AB, Lamo-Espinosa JM, Valentí A, Mortlock DP, Valentí JR, Prósper F, Granero-Moltó F. Hypoxia and Reactive Oxygen Species Homeostasis in Mesenchymal Progenitor Cells Define a Molecular Mechanism for Fracture Nonunion. Stem Cells. 2016;34:2342-2353.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 19]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
66.  Ong HT, Dilley RJ. Novel non-angiogenic role for mesenchymal stem cell-derived vascular endothelial growth factor on keratinocytes during wound healing. Cytokine Growth Factor Rev. 2018;44:69-79.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 33]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
67.  Paulsen JD, Zeck B, Sun K, Simoes C, Theise ND, Chiriboga L. Keratin 19 and mesenchymal markers for evaluation of epithelial-mesenchymal transition and stem cell niche components in primary biliary cholangitis by sequential elution-stripping multiplex immunohistochemistry. J Histotechnol. 2020;43:163-173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
68.  Hade MD, Suire CN, Suo Z. Mesenchymal Stem Cell-Derived Exosomes: Applications in Regenerative Medicine. Cells. 2021;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 153]  [Article Influence: 51.0]  [Reference Citation Analysis (0)]
69.  Wang L, Cai Y, Zhang Q, Zhang Y. Pharmaceutical Activation of Nrf2 Accelerates Diabetic Wound Healing by Exosomes from Bone Marrow Mesenchymal Stem Cells. Int J Stem Cells. 2022;15:164-172.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 20]  [Reference Citation Analysis (0)]
70.  Ding J, Wang X, Chen B, Zhang J, Xu J. Exosomes Derived from Human Bone Marrow Mesenchymal Stem Cells Stimulated by Deferoxamine Accelerate Cutaneous Wound Healing by Promoting Angiogenesis. Biomed Res Int. 2019;2019:9742765.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 120]  [Article Influence: 24.0]  [Reference Citation Analysis (0)]
71.  Qiu J, Shu C, Li X, Ye C, Zhang WC. Exosomes from linc00511-overexpressing ADSCs accelerates angiogenesis in diabetic foot ulcers healing by suppressing PAQR3-induced Twist1 degradation. Diabetes Res Clin Pract. 2021;180:109032.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 25]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
72.  Li X, Xie X, Lian W, Shi R, Han S, Zhang H, Lu L, Li M. 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;50:1-14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 137]  [Cited by in F6Publishing: 229]  [Article Influence: 38.2]  [Reference Citation Analysis (0)]
73.  Hu Y, Rao SS, Wang ZX, Cao J, Tan YJ, Luo J, Li HM, Zhang WS, Chen CY, Xie H. Exosomes from human umbilical cord blood accelerate cutaneous wound healing through miR-21-3p-mediated promotion of angiogenesis and fibroblast function. Theranostics. 2018;8:169-184.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 243]  [Cited by in F6Publishing: 330]  [Article Influence: 55.0]  [Reference Citation Analysis (0)]
74.  Liu J, Yan Z, Yang F, Huang Y, Yu Y, Zhou L, Sun Z, Cui D, Yan Y. Exosomes Derived from Human Umbilical Cord Mesenchymal Stem Cells Accelerate Cutaneous Wound Healing by Enhancing Angiogenesis through Delivering Angiopoietin-2. Stem Cell Rev Rep. 2021;17:305-317.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 49]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
75.  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;96:1-9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 60]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
76.  Ahmed EM. Hydrogel: Preparation, characterization, and applications: A review. J Adv Res. 2015;6:105-121.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2699]  [Cited by in F6Publishing: 2331]  [Article Influence: 211.9]  [Reference Citation Analysis (0)]
77.  Liu Y, Wang J, Chen H, Cheng D. Environmentally friendly hydrogel: A review of classification, preparation and application in agriculture. Sci Total Environ. 2022;846:157303.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 27]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
78.  Wu Q, Chen B, Liang Z. Mesenchymal Stem Cells as a Prospective Therapy for the Diabetic Foot. Stem Cells Int. 2016;2016:4612167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 24]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
79.  Akbari A, Jabbari N, Sharifi R, Ahmadi M, Vahhabi A, Seyedzadeh SJ, Nawaz M, Szafert S, Mahmoodi M, Jabbari E, Asghari R, Rezaie J. Free and hydrogel encapsulated exosome-based therapies in regenerative medicine. Life Sci. 2020;249:117447.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 91]  [Article Influence: 22.8]  [Reference Citation Analysis (0)]
80.  Pishavar E, Luo H, Naserifar M, Hashemi M, Toosi S, Atala A, Ramakrishna S, Behravan J. Advanced Hydrogels as Exosome Delivery Systems for Osteogenic Differentiation of MSCs: Application in Bone Regeneration. Int J Mol Sci. 2021;22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 40]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
81.  Mardpour S, Ghanian MH, Sadeghi-Abandansari H, Mardpour S, Nazari A, Shekari F, Baharvand H. Hydrogel-Mediated Sustained Systemic Delivery of Mesenchymal Stem Cell-Derived Extracellular Vesicles Improves Hepatic Regeneration in Chronic Liver Failure. ACS Appl Mater Interfaces. 2019;11:37421-37433.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 104]  [Article Influence: 20.8]  [Reference Citation Analysis (0)]
82.  Maqsood M, Kang M, Wu X, Chen J, Teng L, Qiu L. Adult mesenchymal stem cells and their exosomes: Sources, characteristics, and application in regenerative medicine. Life Sci. 2020;256:118002.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 86]  [Article Influence: 21.5]  [Reference Citation Analysis (0)]
83.  Mahmood A, Patel D, Hickson B, DesRochers J, Hu X. Recent Progress in Biopolymer-Based Hydrogel Materials for Biomedical Applications. Int J Mol Sci. 2022;23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 47]  [Article Influence: 23.5]  [Reference Citation Analysis (0)]
84.  Li Y, Wang X, Han Y, Sun HY, Hilborn J, Shi L. Click chemistry-based biopolymeric hydrogels for regenerative medicine. Biomed Mater. 2021;16:022003.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 20]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
85.  Lu L, Yuan S, Wang J, Shen Y, Deng S, Xie L, Yang Q. The Formation Mechanism of Hydrogels. Curr Stem Cell Res Ther. 2018;13:490-496.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 56]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
86.  Braun GA, Ary BE, Dear AJ, Rohn MCH, Payson AM, Lee DSM, Parry RC, Friedman C, Knowles TPJ, Linse S, Åkerfeldt KS. On the Mechanism of Self-Assembly by a Hydrogel-Forming Peptide. Biomacromolecules. 2020;21:4781-4794.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 21]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
87.  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;30:115.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 361]  [Cited by in F6Publishing: 331]  [Article Influence: 66.2]  [Reference Citation Analysis (0)]
88.  Cassuto D, Bellia G, Schiraldi C. An Overview of Soft Tissue Fillers for Cosmetic Dermatology: From Filling to Regenerative Medicine. Clin Cosmet Investig Dermatol. 2021;14:1857-1866.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 12]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
89.  Eivazzadeh-Keihan R, Moghim Aliabadi HA, Radinekiyan F, Sobhani M, Farzane Khalili, Maleki A, Madanchi H, Mahdavi M, Shalan AE. Investigation of the biological activity, mechanical properties and wound healing application of a novel scaffold based on lignin-agarose hydrogel and silk fibroin embedded zinc chromite nanoparticles. RSC Adv. 2021;11:17914-17923.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 45]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
90.  Geng X, Qi Y, Liu X, Shi Y, Li H, Zhao L. A multifunctional antibacterial and self-healing hydrogel laden with bone marrow mesenchymal stem cell-derived exosomes for accelerating diabetic wound healing. Biomater Adv. 2022;133:112613.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 32]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
91.  Guo J, Hu H, Gorecka J, Bai H, He H, Assi R, Isaji T, Wang T, Setia O, Lopes L, Gu Y, Dardik A. Adipose-derived mesenchymal stem cells accelerate diabetic wound healing in a similar fashion as bone marrow-derived cells. Am J Physiol Cell Physiol. 2018;315:C885-C896.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 56]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
92.  Wang C, Wang M, Xu T, Zhang X, Lin C, Gao W, Xu H, Lei B, Mao C. Erratum: Engineering Bioactive Self-Healing Antibacterial Exosomes Hydrogel for Promoting Chronic Diabetic Wound Healing and Complete Skin Regeneration: Erratum. Theranostics. 2021;11:10174-10175.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
93.  Pomatto M, Gai C, Negro F, Cedrino M, Grange C, Ceccotti E, Togliatto G, Collino F, Tapparo M, Figliolini F, Lopatina T, Brizzi MF, Camussi G. Differential Therapeutic Effect of Extracellular Vesicles Derived by Bone Marrow and Adipose Mesenchymal Stem Cells on Wound Healing of Diabetic Ulcers and Correlation to Their Cargoes. Int J Mol Sci. 2021;22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 97]  [Article Influence: 32.3]  [Reference Citation Analysis (0)]
94.  Wei M, Hsu YI, Asoh TA, Sung MH, Uyama H. Injectable poly(γ-glutamic acid)-based biodegradable hydrogels with tunable gelation rate and mechanical strength. J Mater Chem B. 2021;9:3584-3594.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 8]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
95.  Ma X, Xu T, Chen W, Qin H, Chi B, Ye Z. Injectable hydrogels based on the hyaluronic acid and poly (γ-glutamic acid) for controlled protein delivery. Carbohydr Polym. 2018;179:100-109.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 71]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
96.  Dong Y, Rodrigues M, Kwon SH, Li X, A S, Brett EA, Elvassore N, Wang W, Gurtner GC. Acceleration of Diabetic Wound Regeneration using an In Situ-Formed Stem-Cell-Based Skin Substitute. Adv Healthc Mater. 2018;7:e1800432.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 41]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
97.  Chen S, Shi J, Zhang M, Chen Y, Wang X, Zhang L, Tian Z, Yan Y, Li Q, Zhong W, Xing M. Mesenchymal stem cell-laden anti-inflammatory hydrogel enhances diabetic wound healing. Sci Rep. 2015;5:18104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 107]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
98.  Lei Z, Singh G, Min Z, Shixuan C, Xu K, Pengcheng X, Xueer W, Yinghua C, Lu Z, Lin Z. Bone marrow-derived mesenchymal stem cells laden novel thermo-sensitive hydrogel for the management of severe skin wound healing. Mater Sci Eng C Mater Biol Appl. 2018;90:159-167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 36]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
99.  Kaisang L, Siyu W, Lijun F, Daoyan P, Xian CJ, Jie S. Adipose-derived stem cells seeded in Pluronic F-127 hydrogel promotes diabetic wound healing. J Surg Res. 2017;217:63-74.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 63]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
100.  Yang J, Chen Z, Pan D, Li H, Shen J. Umbilical Cord-Derived Mesenchymal Stem Cell-Derived Exosomes Combined Pluronic F127 Hydrogel Promote Chronic Diabetic Wound Healing and Complete Skin Regeneration. Int J Nanomedicine. 2020;15:5911-5926.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 203]  [Article Influence: 50.8]  [Reference Citation Analysis (0)]
101.  Jiao Y, Chen X, Niu Y, Huang S, Wang J, Luo M, Shi G, Huang J. Wharton's jelly mesenchymal stem cells embedded in PF-127 hydrogel plus sodium ascorbyl phosphate combination promote diabetic wound healing in type 2 diabetic rat. Stem Cell Res Ther. 2021;12:559.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 13]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
102.  Wu YY, Jiao YP, Xiao LL, Li MM, Liu HW, Li SH, Liao X, Chen YT, Li JX, Zhang Y. Experimental Study on Effects of Adipose-Derived Stem Cell-Seeded Silk Fibroin Chitosan Film on Wound Healing of a Diabetic Rat Model. Ann Plast Surg. 2018;80:572-580.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 37]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
103.  Bai H, Kyu-Cheol N, Wang Z, Cui Y, Liu H, Feng Y, Zhao Y, Lin Q, Li Z. Regulation of inflammatory microenvironment using a self-healing hydrogel loaded with BM-MSCs for advanced wound healing in rat diabetic foot ulcers. J Tissue Eng. 2020;11:2041731420947242.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 59]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
104.  Ahmed R, Afreen A, Tariq M, Zahid AA, Masoud MS, Ahmed M, Ali I, Akram Z, Hasan A. Bone marrow mesenchymal stem cells preconditioned with nitric-oxide-releasing chitosan/PVA hydrogel accelerate diabetic wound healing in rabbits. Biomed Mater. 2021;16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
105.  Zahid AA, Augustine R, Dalvi YB, Reshma K, Ahmed R, Raza Ur Rehman S, Marei HE, Alfkey R, Hasan A. Development of nitric oxide releasing visible light crosslinked gelatin methacrylate hydrogel for rapid closure of diabetic wounds. Biomed Pharmacother. 2021;140:111747.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 22]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
106.  Zahid AA, Ahmed R, Raza Ur Rehman S, Augustine R, Tariq M, Hasan A. Nitric oxide releasing chitosan-poly (vinyl alcohol) hydrogel promotes angiogenesis in chick embryo model. Int J Biol Macromol. 2019;136:901-910.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 46]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
107.  Jin X, Shang Y, Zou Y, Xiao M, Huang H, Zhu S, Liu N, Li J, Wang W, Zhu P. Injectable Hypoxia-Induced Conductive Hydrogel to Promote Diabetic Wound Healing. ACS Appl Mater Interfaces. 2020;12:56681-56691.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 47]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
108.  Srifa W, Kosaric N, Amorin A, Jadi O, Park Y, Mantri S, Camarena J, Gurtner GC, Porteus M. Cas9-AAV6-engineered human mesenchymal stromal cells improved cutaneous wound healing in diabetic mice. Nat Commun. 2020;11:2470.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 39]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
109.  Moon KC, Suh HS, Kim KB, Han SK, Young KW, Lee JW, Kim MH. Potential of Allogeneic Adipose-Derived Stem Cell-Hydrogel Complex for Treating Diabetic Foot Ulcers. Diabetes. 2019;68:837-846.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 92]  [Article Influence: 18.4]  [Reference Citation Analysis (0)]
110.  Zeng X, Tang Y, Hu K, Jiao W, Ying L, Zhu L, Liu J, Xu J. Three-week topical treatment with placenta-derived mesenchymal stem cells hydrogel in a patient with diabetic foot ulcer: A case report. Medicine (Baltimore). 2017;96:e9212.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 32]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
111.  Sivaraj D, Chen K, Chattopadhyay A, Henn D, Wu W, Noishiki C, Magbual NJ, Mittal S, Mermin-Bunnell AM, Bonham CA, Trotsyuk AA, Barrera JA, Padmanabhan J, Januszyk M, Gurtner GC. Hydrogel Scaffolds to Deliver Cell Therapies for Wound Healing. Front Bioeng Biotechnol. 2021;9:660145.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 57]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
112.  Yao T, Baker MB, Moroni L. Strategies to Improve Nanofibrous Scaffolds for Vascular Tissue Engineering. Nanomaterials (Basel). 2020;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 23]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
113.  Jun I, Han HS, Edwards JR, Jeon H. Electrospun Fibrous Scaffolds for Tissue Engineering: Viewpoints on Architecture and Fabrication. Int J Mol Sci. 2018;19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 318]  [Cited by in F6Publishing: 242]  [Article Influence: 40.3]  [Reference Citation Analysis (0)]
114.  Ding X, Huang Y, Li X, Liu S, Tian F, Niu X, Chu Z, Chen D, Liu H, Fan Y. Three-dimensional silk fibroin scaffolds incorporated with graphene for bone regeneration. J Biomed Mater Res A. 2021;109:515-523.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 15]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
115.  Carvalho MS, Silva JC, Udangawa RN, Cabral JMS, Ferreira FC, da Silva CL, Linhardt RJ, Vashishth D. Co-culture cell-derived extracellular matrix loaded electrospun microfibrous scaffolds for bone tissue engineering. Mater Sci Eng C Mater Biol Appl. 2019;99:479-490.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 59]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
116.  Bružauskaitė I, Bironaitė D, Bagdonas E, Bernotienė E. Scaffolds and cells for tissue regeneration: different scaffold pore sizes-different cell effects. Cytotechnology. 2016;68:355-369.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 375]  [Cited by in F6Publishing: 382]  [Article Influence: 42.4]  [Reference Citation Analysis (0)]
117.  Song L, Wang L, Hou Y, Zhou J, Chen C, Ye X, Dong W, Gao H, Liu Y, Qiao G, Pan T, Chen Q, Cao Y, Hu F, Rao Z, Chen Y, Han Y, Zheng M, Luo Y, Li X, Huang Z. FGF4 protects the liver from nonalcoholic fatty liver disease by activating the AMP-activated protein kinase-Caspase 6 signal axis. Hepatology. 2022;76:1105-1120.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 18]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
118.  Li P, Ruan L, Jiang G, Sun Y, Wang R, Gao X, Yunusov KE, Aharodnikau UE, Solomevich SO. Design of 3D polycaprolactone/ε-polylysine-modified chitosan fibrous scaffolds with incorporation of bioactive factors for accelerating wound healing. Acta Biomater. 2022;152:197-209.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
119.  Pandey VK, Ajmal G, Upadhyay SN, Mishra PK. Nano-fibrous scaffold with curcumin for anti-scar wound healing. Int J Pharm. 2020;589:119858.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 11]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
120.  Su N, Gao PL, Wang K, Wang JY, Zhong Y, Luo Y. Fibrous scaffolds potentiate the paracrine function of mesenchymal stem cells: A new dimension in cell-material interaction. Biomaterials. 2017;141:74-85.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 136]  [Cited by in F6Publishing: 128]  [Article Influence: 18.3]  [Reference Citation Analysis (0)]
121.  Liu C, Zhu Y, Lun X, Sheng H, Yan A. Effects of wound dressing based on the combination of silver@curcumin nanoparticles and electrospun chitosan nanofibers on wound healing. Bioengineered. 2022;13:4328-4339.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 22]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
122.  Chen S, Wang H, Su Y, John JV, McCarthy A, Wong SL, Xie J. Mesenchymal stem cell-laden, personalized 3D scaffolds with controlled structure and fiber alignment promote diabetic wound healing. Acta Biomater. 2020;108:153-167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 60]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
123.  Li J, Zhang T, Pan M, Xue F, Lv F, Ke Q, Xu H. Nanofiber/hydrogel core-shell scaffolds with three-dimensional multilayer patterned structure for accelerating diabetic wound healing. J Nanobiotechnology. 2022;20:28.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 15]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
124.  Hou C, Shen L, Huang Q, Mi J, Wu Y, Yang M, Zeng W, Li L, Chen W, Zhu C. The effect of heme oxygenase-1 complexed with collagen on MSC performance in the treatment of diabetic ischemic ulcer. Biomaterials. 2013;34:112-120.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 36]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
125.  He S, Shen L, Wu Y, Li L, Chen W, Hou C, Yang M, Zeng W, Zhu C. Effect of brain-derived neurotrophic factor on mesenchymal stem cell-seeded electrospinning biomaterial for treating ischemic diabetic ulcers via milieu-dependent differentiation mechanism. Tissue Eng Part A. 2015;21:928-938.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 15]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
126.  Navone SE, Pascucci L, Dossena M, Ferri A, Invernici G, Acerbi F, Cristini S, Bedini G, Tosetti V, Ceserani V, Bonomi A, Pessina A, Freddi G, Alessandrino A, Ceccarelli P, Campanella R, Marfia G, Alessandri G, Parati EA. Decellularized silk fibroin scaffold primed with adipose mesenchymal stromal cells improves wound healing in diabetic mice. Stem Cell Res Ther. 2014;5:7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 85]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
127.  Millán-Rivero JE, Martínez CM, Romecín PA, Aznar-Cervantes SD, Carpes-Ruiz M, Cenis JL, Moraleda JM, Atucha NM, García-Bernal D. Silk fibroin scaffolds seeded with Wharton's jelly mesenchymal stem cells enhance re-epithelialization and reduce formation of scar tissue after cutaneous wound healing. Stem Cell Res Ther. 2019;10:126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 41]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
128.  Kolimi P, Narala S, Nyavanandi D, Youssef AAA, Dudhipala N. Innovative Treatment Strategies to Accelerate Wound Healing: Trajectory and Recent Advancements. Cells. 2022;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 37]  [Reference Citation Analysis (0)]
129.  Chen G, Kawazoe N. Porous Scaffolds for Regeneration of Cartilage, Bone and Osteochondral Tissue. Adv Exp Med Biol. 2018;1058:171-191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 31]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
130.  Chen Y, Kawazoe N, Chen G. Preparation of dexamethasone-loaded biphasic calcium phosphate nanoparticles/collagen porous composite scaffolds for bone tissue engineering. Acta Biomater. 2018;67:341-353.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 80]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
131.  Wang C, Yue H, Huang W, Lin X, Xie X, He Z, He X, Liu S, Bai L, Lu B, Wei Y, Wang M. Cryogenic 3D printing of heterogeneous scaffolds with gradient mechanical strengths and spatial delivery of osteogenic peptide/TGF-β1 for osteochondral tissue regeneration. Biofabrication. 2020;12:025030.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 34]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
132.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 52]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
133.  Zhang L, Yang G, Johnson BN, Jia X. Three-dimensional (3D) printed scaffold and material selection for bone repair. Acta Biomater. 2019;84:16-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 313]  [Cited by in F6Publishing: 359]  [Article Influence: 71.8]  [Reference Citation Analysis (0)]
134.  Han F, Dong Y, Su Z, Yin R, Song A, Li S. Preparation, characteristics and assessment of a novel gelatin-chitosan sponge scaffold as skin tissue engineering material. Int J Pharm. 2014;476:124-133.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 95]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
135.  Oryan A, Sahvieh S. Effectiveness of chitosan scaffold in skin, bone and cartilage healing. Int J Biol Macromol. 2017;104:1003-1011.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 119]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
136.  Agarwal T, Narayan R, Maji S, Behera S, Kulanthaivel S, Maiti TK, Banerjee I, Pal K, Giri S. Gelatin/Carboxymethyl chitosan based scaffolds for dermal tissue engineering applications. Int J Biol Macromol. 2016;93:1499-1506.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 55]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
137.  Udenni Gunathilake TMS, Ching YC, Ching KY, Chuah CH, Abdullah LC. Biomedical and Microbiological Applications of Bio-Based Porous Materials: A Review. Polymers (Basel). 2017;9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 48]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
138.  Sampath UGTM, Ching YC, Chuah CH, Sabariah JJ, Lin PC. Fabrication of Porous Materials from Natural/Synthetic Biopolymers and Their Composites. Materials (Basel). 2016;9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 61]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
139.  Abdul Khalil HPS, Yahya EB, Tajarudin HA, Balakrishnan V, Nasution H. Insights into the Role of Biopolymer-Based Xerogels in Biomedical Applications. Gels. 2022;8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 13]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
140.  O'Loughlin A, Kulkarni M, Creane M, Vaughan EE, Mooney E, Shaw G, Murphy M, Dockery P, Pandit A, O'Brien T. 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;62:2588-2594.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 92]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
141.  Kerstan A, Dieter K, Niebergall-Roth E, Klingele S, Jünger M, Hasslacher C, Daeschlein G, Stemler L, Meyer-Pannwitt U, Schubert K, Klausmann G, Raab T, Goebeler M, Kraft K, Esterlechner J, Schröder HM, Sadeghi S, Ballikaya S, Gasser M, Waaga-Gasser AM, Murphy GF, Orgill DP, Frank NY, Ganss C, Scharffetter-Kochanek K, Frank MH, Kluth MA. Translational development of ABCB5(+) dermal mesenchymal stem cells for therapeutic induction of angiogenesis in non-healing diabetic foot ulcers. Stem Cell Res Ther. 2022;13:455.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 15]  [Reference Citation Analysis (0)]
142.  Tong C, Hao H, Xia L, Liu J, Ti D, Dong L, Hou Q, Song H, Liu H, Zhao Y, Fu X, Han W. Hypoxia pretreatment of bone marrow-derived mesenchymal stem cells seeded in a collagen-chitosan sponge scaffold promotes skin wound healing in diabetic rats with hindlimb ischemia. Wound Repair Regen. 2016;24:45-56.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 61]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
143.  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;5:139-148.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 389]  [Cited by in F6Publishing: 362]  [Article Influence: 27.8]  [Reference Citation Analysis (0)]
144.  Shah RG, Pierce MC, Silver FH. Morphomechanics of dermis-A method for non-destructive testing of collagenous tissues. Skin Res Technol. 2017;23:399-406.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
145.  Joodaki H, Panzer MB. Skin mechanical properties and modeling: A review. Proc Inst Mech Eng H. 2018;232:323-343.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 180]  [Cited by in F6Publishing: 184]  [Article Influence: 30.7]  [Reference Citation Analysis (0)]
146.  Chen TY, Wen TK, Dai NT, Hsu SH. Cryogel/hydrogel biomaterials and acupuncture combined to promote diabetic skin wound healing through immunomodulation. Biomaterials. 2021;269:120608.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 79]  [Article Influence: 19.8]  [Reference Citation Analysis (0)]
147.  Akin B, Ozmen MM. Antimicrobial cryogel dressings towards effective wound healing. Prog Biomater. 2022;11:331-346.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
148.  Fu CY, Chuang WT, Hsu SH. A Biodegradable Chitosan-Polyurethane Cryogel with Switchable Shape Memory. ACS Appl Mater Interfaces. 2021;13:9702-9713.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 13]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
149.  Shiekh PA, Singh A, Kumar A. Exosome laden oxygen releasing antioxidant and antibacterial cryogel wound dressing OxOBand alleviate diabetic and infectious wound healing. Biomaterials. 2020;249:120020.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 185]  [Article Influence: 46.3]  [Reference Citation Analysis (0)]
150.  Wang M, Hu J, Ou Y, He X, Wang Y, Zou C, Jiang Y, Luo F, Lu D, Li Z, Li J, Tan H. Shape-Recoverable Hyaluronic Acid-Waterborne Polyurethane Hybrid Cryogel Accelerates Hemostasis and Wound Healing. ACS Appl Mater Interfaces. 2022;14:17093-17108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 21]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
151.  Mohanty C, Pradhan J. A human epidermal growth factor-curcumin bandage bioconjugate loaded with mesenchymal stem cell for in vivo diabetic wound healing. Mater Sci Eng C Mater Biol Appl. 2020;111:110751.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 39]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
152.  Lee HJ, Jeong M, Na YG, Kim SJ, Lee HK, Cho CW. An EGF- and Curcumin-Co-Encapsulated Nanostructured Lipid Carrier Accelerates Chronic-Wound Healing in Diabetic Rats. Molecules. 2020;25.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 28]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
153.  Kamar SS, Abdel-Kader DH, Rashed LA. Beneficial effect of Curcumin Nanoparticles-Hydrogel on excisional skin wound healing in type-I diabetic rat: Histological and immunohistochemical studies. Ann Anat. 2019;222:94-102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 41]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
154.  Ghufran H, Mehmood A, Azam M, Butt H, Ramzan A, Yousaf MA, Ejaz A, Tarar MN, Riazuddin S. Curcumin preconditioned human adipose derived stem cells co-transplanted with platelet rich plasma improve wound healing in diabetic rats. Life Sci. 2020;257:118091.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 5]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
155.  De Francesco F, Graziano A, Trovato L, Ceccarelli G, Romano M, Marcarelli M, Cusella De Angelis GM, Cillo U, Riccio M, Ferraro GA. A Regenerative Approach with Dermal Micrografts in the Treatment of Chronic Ulcers. Stem Cell Rev Rep. 2017;13:139-148.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 18]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
156.  Niimi Y, Baba K, Tsuchida M, Takeda A. A Histological Evaluation of Artificial Dermal Scaffold Used in Micrograft Treatment: A Case Study of Micrograft and NPWT Performed on a Postoperative Ulcer Formation after Tumor Resection. Medicina (Kaunas). 2022;58.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
157.  Li Y, Sakamoto M, Sawaragi E, Nakano T, Katayama Y, Yamanaka H, Tsuge I, Morimoto N. Comparison of Wound Healing Effect of Skin Micrograft Impregnated into Two Kinds of Artificial Dermis in a Murine Wound Model. Plast Reconstr Surg Glob Open. 2022;10:e4636.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
158.  Amirazad H, Dadashpour M, Zarghami N. Application of decellularized bone matrix as a bioscaffold in bone tissue engineering. J Biol Eng. 2022;16:1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 43]  [Article Influence: 21.5]  [Reference Citation Analysis (0)]
159.  Safdari M, Bibak B, Soltani H, Hashemi J. Recent advancements in decellularized matrix technology for bone tissue engineering. Differentiation. 2021;121:25-34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 12]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
160.  Zhang X, Chen X, Hong H, Hu R, Liu J, Liu C. Decellularized extracellular matrix scaffolds: Recent trends and emerging strategies in tissue engineering. Bioact Mater. 2022;10:15-31.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 178]  [Cited by in F6Publishing: 192]  [Article Influence: 96.0]  [Reference Citation Analysis (0)]
161.  Sahranavard M, Sarkari S, Safavi S, Ghorbani F. Three-dimensional bio-printing of decellularized extracellular matrix-based bio-inks for cartilage regeneration: a systematic review. Biomater Transl. 2022;3:105-115.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
162.  Rajab TK, O'Malley TJ, Tchantchaleishvili V. Decellularized scaffolds for tissue engineering: Current status and future perspective. Artif Organs. 2020;44:1031-1043.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 32]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
163.  Cao Y, Shi X, Zhao X, Chen B, Li X, Li Y, Chen Y, Chen C, Lu H, Liu J. Acellular dermal matrix decorated with collagen-affinity peptide accelerate diabetic wound healing through sustained releasing Histatin-1 mediated promotion of angiogenesis. Int J Pharm. 2022;624:122017.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
164.  Kim MJ, Lee WB, Park BY. Effect of Morphologically Transformed Acellular Dermal Matrix on Chronic Diabetic Wounds: An Experimental Study in a Calvarial Bone Exposure Diabetic Rat Model. J Surg Res. 2022;272:153-165.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
165.  Cazzell S, Vayser D, Pham H, Walters J, Reyzelman A, Samsell B, Dorsch K, Moore M. A randomized clinical trial of a human acellular dermal matrix demonstrated superior healing rates for chronic diabetic foot ulcers over conventional care and an active acellular dermal matrix comparator. Wound Repair Regen. 2017;25:483-497.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 40]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
166.  Kavros SJ, Dutra T, Gonzalez-Cruz R, Liden B, Marcus B, McGuire J, Nazario-Guirau L. The use of PriMatrix, a fetal bovine acellular dermal matrix, in healing chronic diabetic foot ulcers: a prospective multicenter study. Adv Skin Wound Care. 2014;27:356-362.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 26]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
167.  Rennert RC, Sorkin M, Garg RK, Januszyk M, Gurtner GC. Cellular response to a novel fetal acellular collagen matrix: implications for tissue regeneration. Int J Biomater. 2013;2013:527957.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 23]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
168.  Shi X, Jiang L, Zhao X, Chen B, Shi W, Cao Y, Chen Y, Li X, He Y, Li C, Liu X, Lu H, Chen C, Liu J. Adipose-Derived Stromal Cell-Sheets Sandwiched, Book-Shaped Acellular Dermal Matrix Capable of Sustained Release of Basic Fibroblast Growth Factor Promote Diabetic Wound Healing. Front Cell Dev Biol. 2021;9:646967.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 9]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
169.  Zhang Y, Jiang W, Kong L, Fu J, Zhang Q, Liu H. PLGA@IL-8 nanoparticles-loaded acellular dermal matrix as a delivery system for exogenous MSCs in diabetic wound healing. Int J Biol Macromol. 2023;224:688-698.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
170.  Chu J, Shi P, Deng X, Jin Y, Liu H, Chen M, Han X. Dynamic multiphoton imaging of acellular dermal matrix scaffolds seeded with mesenchymal stem cells in diabetic wound healing. J Biophotonics. 2018;11:e201700336.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 16]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
171.  Yan W, Liu H, Deng X, Jin Y, Wang N, Chu J. Acellular dermal matrix scaffolds coated with connective tissue growth factor accelerate diabetic wound healing by increasing fibronectin through PKC signalling pathway. J Tissue Eng Regen Med. 2018;12:e1461-e1473.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 14]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
172.  Zhou W, Zhao X, Shi X, Chen C, Cao Y, Liu J. Constructing Tissue-Engineered Dressing Membranes with Adipose-Derived Stem Cells and Acellular Dermal Matrix for Diabetic Wound Healing: A Comparative Study of Hypoxia- or Normoxia-Culture Modes. Stem Cells Int. 2022;2022:2976185.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
173.  Fu J, Zhang Y, Chu J, Wang X, Yan W, Zhang Q, Liu H. 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;5:4054-4066.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 27]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
174.  Chu J, Shi P, Yan W, Fu J, Yang Z, He C, Deng X, Liu H. PEGylated graphene oxide-mediated quercetin-modified collagen hybrid scaffold for enhancement of MSCs differentiation potential and diabetic wound healing. Nanoscale. 2018;10:9547-9560.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 65]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
175.  Han Y, Sun T, Han Y, Lin L, Liu C, Liu J, Yan G, Tao R. Human umbilical cord mesenchymal stem cells implantation accelerates cutaneous wound healing in diabetic rats via the Wnt signaling pathway. Eur J Med Res. 2019;24:10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 25]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
176.  Chu DT, Phuong TNT, Tien NLB, Tran DK, Thanh VV, Quang TL, Truong DT, Pham VH, Ngoc VTN, Chu-Dinh T, Kushekhar K. An Update on the Progress of Isolation, Culture, Storage, and Clinical Application of Human Bone Marrow Mesenchymal Stem/Stromal Cells. Int J Mol Sci. 2020;21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 78]  [Article Influence: 19.5]  [Reference Citation Analysis (0)]