Skip to main content
Journal of Diabetes and Metabolic Disorders logoLink to Journal of Diabetes and Metabolic Disorders
. 2023 Aug 29;22(2):1039–1052. doi: 10.1007/s40200-023-01280-8

Transplantation of adipose derived stem cells in diabetes mellitus; limitations and achievements

Raziye Tajali 4, Akram Eidi 4, Hosein Ahmadi Tafti 5, Abdolreza Pazouki 6, Tunku Kamarul 3, Ali Mohammad Sharifi 1,2,3,
PMCID: PMC10638327  PMID: 37975135

Abstract

Objectives

Diabetes mellitus (DM) is a complex metabolic disease that results from impaired insulin secreting pancreatic β-cells or insulin resistance. Although available medications help control the disease, patients suffer from its complications. Therefore, finding effective therapeutic approaches to treat DM is a priority. Adipose Derived Stem Cells (ADSCs) based therapy is a promising strategy in various regenerative medicine applications, but its systematic translational use is still somewhat out of reach. This review is aimed at clarifying achievements as well as challenges facing the application of ADSCs for the treatment of DM, with a special focus on the mechanisms involved.

Methods

Literature searches were carried out on “Scopus”, “PubMed” and “Google Scholar” up to September 2022 to find relevant articles in the English language for the scope of this review.

Results

Recent evidence showed a significant role of ADSC therapies in DM by ameliorating insulin resistance and hyperglycemia, regulating hepatic glucose metabolism, promoting β cell function and regeneration, and functioning as a gene delivery tool. In addition, ADSCs could improve diabetic wound healing by promoting collagen deposition, inhibiting inflammation, and enhancing angiogenesis.

Conclusion

Overall, this literature review revealed the great clinical implications of ADSCs for translating into the clinical setting for the treatment of diabetes. However, further large-scale and controlled studies are needed to overcome challenges and confirm the safety and optimal therapeutic scheme before daily clinical application.

Supplementary Information

The online version contains supplementary material available at 10.1007/s40200-023-01280-8.

Keywords: Diabetes Mellitus, Pancreatic β-cells, Insulin resistance, Adipose derive stem cells (ADSCs), Regenerative medicine

Introduction

Diabetes mellitus is a non-communicable metabolic disease approaching epidemic proportions globally [1]. The most prevalent types of diabetes, type 1 and type 2, are characterized by selective autoimmune destruction of insulin-producing pancreatic β-cells with subsequent sudden and total deficiency of insulin and by a decrease in the sensitivity of target tissues to insulin [2]. Due to dietary habits and sedentary behaviors, the number of patients with diabetes is expected to rise globally [3]. According to an estimate by the International Diabetes Federation, the number of people with diabetes is about 463 million and is set to escalate to 700 million by the year 2045 [4].

Oral hypoglycemic agents and insulin therapy are the most prescribed treatments for diabetes. Although common interventions can decrease glycemic levels or temporarily ameliorate insulin sensitivity in insulin-responsive tissues, these can neither improve pancreatic β cell impairment nor insulin resistance [5]. Therefore, developing effective alternative approaches to curing diabetes is a priority. Islet cell transplantation has been proposed as a cell replacement approach for reestablishing glucose homeostasis since the 1970s [6]. However, efforts toward the routine use of this treatment have been hampered by islet source limitation, lack of oxygen and blood supply for transplanted islets, lifelong immunosuppression, and immune rejection of islets [7, 8].

Regenerative medicine, a relatively young branch of multidisciplinary research, has emerged as an important avenue that aims to treat a wide range of conditions by inducing the restoration of diseased or damaged tissue [9]. Stem cell therapies provide novel regenerative and immunomodulatory techniques for managing diabetes mellitus. Unique characteristics of mesenchymal stem cells (MSCs), including differentiation potential, immunoregulatory capacity, and paracrine effect, make them optimal for tissue regeneration [10]. Among the various sources of MSCs, adipose derived stem cells (ADSCs) have proved to be one of the most promising stem cell populations (Table 1) regarding their accessibility, abundance, efficacy, and minimally invasive collection procedure when compared to other sources [11].

Table 1.

Summary of the major clinical trials utilizing MSCs as a treatment for DM, extracted from the PubMed database, starting from the years 2017–2022

No Source of MSC in the treatment of DM Findings References
1. Pluripotent stem cell-derived pancreatic endoderm cells (PEC-01) The progressive accumulation of functional, insulin-secreting cells occurs over a period of approximately 6–9 months from the time of implant. Shapiro et al., 2021 [139]
2. Allogenic Adipose Tissue-Derived Stromal/Stem Cells ADSCs transplantation shows the short-term safety of infusions from healthy donors and daily oral cholecalciferol in patients with recent-onset T1D, as well as their potential therapeutic effect on glycemic control and pancreatic function. In most cases, mild and transient adverse events were observed. Araujo et al., 2020 [140]
3. Allogenic umbilical cord mesenchymal stromal cells Subjects in the MSC-treated group achieved insulin independence and maintained it for 3 to 12 months. Lu et al., 2021 [141]
4. Autologous bone marrow stem cell Combined therapy of intrapancreatic ASC infusion and HBOT showed increased metabolic control and reduced insulin requirements in patients with T2DM compared with standard treatment. Estrada et al., 2019 [142];
5. Allogeneic Wharton’s jelly-derived mesenchymal stem cells (WJ-MSCs) The treatment of allogeneic WJ-MSCs to evaluate long-term safety and efficacy in treating diabetic patients was well-tolerated and safe. Most of the patients reported an improvement at all time points during follow-up. The maximum improvement was reported after six months, when all participants had received two injections. Al Demour et al., 2021 [143]

In this review, we shall overview achievements as well as challenges facing the application of ADSCs for the treatment of DM, with a special focus on their role in diabetic wound healing and the underlying mechanisms involved.

Adipose derived stem cells

Adipose tissue has been widely acknowledged as a valuable origin of postnatal mesenchymal stem cells for the past four decades [12]. Zuk et al. first isolated a putative subset of adult stem cells from adipose tissue, initially called Processed Lipoaspirate or PLA cells [13]. Since then, these new multipotent stem cells have been renamed Adipose derived stem cells (ADSCs) and have made their mark as promising therapeutic cells in medicine and cell therapy [14]. In particular, adipose tissue that can be acquired as waste products of lipoaspiration or abdominoplasty offers significant therapeutic benefits, such as providing an easily accessible and ethically uncontroversial source of stem cells that yields high amounts of viable adult stem cells [15].

Adipose derived stem cells meet the criteria proposed by the International Society for Cellular Therapy (ISCT) to identify human mesenchymal stem cells (MSCs): (i) plastic adherent, [16] (ii) positive localization of CD73, CD90, and CD105 but no localization of CD45, CD34, CD14 or CD11b, CD79a or CD19, and HLA-DR surface molecules; (iii) trilineage mesenchymal differentiation potential [17]. Nevertheless, an exact immunophenotype characterization of ADSCs has been long debated due to their nonhomogeneous population [17]. Mildmay-White and Khan recommended a minimum panel of CD markers in ADSCs, including CD90+, CD44+, CD29+, CD105+, CD13+, CD34+, CD73+, CD166+, CD10+, CD49e+, CD59+, CD31-, CD45-, CD14-, CD11b-, CD19-, CD56- and CD146- [18]. Particularly, the expression of the markers varies based on the method of isolation, culture condition, passages, and donor variation [19, 20].

There have been numerous reports that ADSCs, which originate from mesodermal layers, can be differentiated into cells of the other two germ layers [21]. Actually, these cells can differentiate into adipogenic, osteogenic, chondrogenic, myogenic, angiogenic, cardiomyogenic, tenogenic, and periodontogenic lineages (11); in this respect, ADSCs and bone marrow stem cells (BMSCs) present large similarities in differentiation potential; however, several characteristics distinguish these two types of stem cells. For instance, ADSCs have an inferior potential for chondrogenic and osteogenic properties compared with BMSCs [16, 22], while being more prone to differentiate into cardiomyocytes and muscle cells [23]. In addition, ADSCs cellular activity is affected by cryopreservation [24] and long-term expansion [12]. Thus, it seems that further investigations should be carried out to optimize ADSC manipulation for clinical application.

Additionally, their expanded use is specifically due to paracrine effects through the secretion of soluble mediators, including anti-inflammatory cytokines, anti-apoptotic, neurotrophic, growth factors, and extracellular vesicles, collectively known as the secretome [25, 26]. The summary of the major clinical trials utilizing ADSCs as a treatment for various chronic diseases are listed in Table 2.

Table 2.

 A Summary of the major clinical trials utilizing ADSCs as a treatment for various chronic diseases, extracted from the PubMed database, starting from the years 2017–2022

Author, Year Disease Source Method of delivery Adverse events Main result
Lee et al., 2019 [144] Knee Osteoarthritis Autologous ADSCs

intra-articular injection

(1 × 108 cells)

No AEs at 6 months’ follow-up ADSCs provided satisfactory functional improvement and pain relief.
Zhang et al., 2022 [145] Knee Osteoarthritis Autologous SVF

intra-articular injection

(Not available)

No AEs at 12 months’ follow-up ADSCs improved the clinical symptoms through cartilage regeneration.
Freitag et al., 2019 [146] Knee Osteoarthritis Autologous ADSCs

intra-articular injection

(100 × 106 cells)

No AEs events at 12 months’ follow-up ADSCs effectively improved function and relieved pain.
Zhou et al., 2020 [134] Crohn’s fistula-in-ano Autologous ADSCs

inner orifice and around the fistula injection (− 1 × 106 cells for fistula < 1 cm

-2 × 106 cells for fistula > 1 cm)

No AEs events at 12 months’ follow-up ADSCs accelerated the healing rate.
Araujo et al., 2020 [140] Type 1 Diabetes Mellitus Allogenic ADSCs Intravenous injection (upper arm vein) (1 × 106 cells/kg) Mild transient AEs ADSCs treatment results in better glucose control and arrest β cell destruction.
Carstens et al., 2021 [147] Chronic Diabetic Foot Ulcers Autologous SVF Subcutaneous Injection (30 × 106 cells) No serious EVs ADSCs treatment enhanced wound healing, reduced amputation, and improved quality of life.
Kumar et al., 2017 [148] chronic discogenic low back pain Intradiscal implantation of ADSCs and hyaluronic acid (Low dose: 2 × 107 cells/disc. High dose: 4 × 107 cells/disc) No serious EVs in 1-year follow-up The combined implantation of ADSCs and HA is safe and tolerable and has provided significant improvement in patient outcomes.
Kastrup et al., 2017 [149] ischemic heart disease Allogeneic ADSCs Intramyocardial injection (Not available) No AEs events at 6 months’ follow-up ADSCs treatment improved cardiac function.
Huang et al., 2019 [150] Liver Cirrhosis Autologous ADSCs Intrahepatic Injection (1 × 108 cells) No AEs events at 24 weeks’ follow-up ADSCs improved liver function and quality of life.
Carstens et al., 2020 [151] Parkinson’s Disease Autologous SVF Facial injection 60 × 106 SVF No AEs events at 12 months’ follow-up in one patient and 5 years in another SVF treatment led to an improved long-term neurologic response.
Qayyum et al., 2019 [152] refractory angina Autologous ADSCs

intra-myocardial injections

72 ± 45 × 106 ASCs

3 years’ follow-up ADSCs improved cardiac symptoms and unchanged exercise capacity.
Abumoawad et al., 2020 [153] Atherosclerotic renovascular disease autologous ADSCs intra-arterial infusion of ADSCs (3 dose: 1, 2.5, and 5.0 × 105 cells/kg) Have not been reported ADSCs increase cortical and whole kidney blood flows.
Paitazoglou et al., 2019 [154] Ischaemic heart failure Allogenic ADSC Intramyocardial injection (100 × 106) No AEs events at 24 months’ follow-up Have not been reported
Squassoni et al., 2021 [155] Chronic Obstructive Pulmonary Disease autologous ADSC and BMC

Co-infusion of ADSC and BMSC

(5 × 107 ADSCs cells + 5 × 107 BMSC)

No AEs events at 12 months’ follow-up Co-infusion therapy improved gas exchange and overall quality of life.

Therapeutic properties of ADSCs in diabetes

Nowadays, stem cell therapies are hot topics in the medical world, and this unique technique is just the beginning of a long story. Stem cells such as embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), and mesenchymal stem cells (MSCs) can improve insulin production and also ameliorate insulin resistance in T2DM. The legal issues of ESCs and the tumorigenic complications of iPSCs limit their application in clinical settings. MSCs lack these disadvantages and are widely studied in T2DM. As mentioned above, adipose derived stem cells (ADSCs) are the most attractive source of MSCs for clinical cell therapy because of their self-renewal and expansion potential, easy accessibility, abundant sources, multipotency, and low immunogenicity. We tried to highlight the mechanisms and therapeutical properties of ADSCs in diabetes in Fig. 1.

Fig. 1.

Fig. 1

Mechanisms and therapeutical properties of ADSCs in diabetes. Created with BioRender.com.

Differentiation potential of ADSCs

The enhanced multidifferentiation potential of ADSCs adds a lot to their therapeutic capacity for a wide range of diseases, including DM. In fact, adipose derived stem cells have the potential to differentiate into nonmesodermal cells such as insulin producing cells (IPCs), which is the earliest therapeutic approach to curing diabetes and results in an increase in insulin levels in T2DM patients. Chandra et al. first developed a differentiation protocol for murine ADSCs to differentiate into IPCs, which could effectively bring about normoglycemia in rat models within 2 weeks [27]. In line with this study, Nam et al. differentiated human eyelid adipose tissue-derived stem cells into insulin-secreting cells. Transplantation of differentiated cells into a type 2 diabetes mouse model resulted in an increased circulating insulin level and lowered levels of IL-6, triglyceride, and free fatty acids [28]. The generation of insulin-producing cells derived from ADSCs has been successfully developed by other researchers [2933], which can effectively ameliorate hyperglycemia. However, several uncertain factors in the differentiation of ADSCs into IPCs still remain, including immune rejection and graft hypertrophy, the effect of the diabetic microenvironment on the differentiated insulin producing cells, and the tumorigenic risks associated with virus-mediated differentiation [34]. These variables are the crucial elements that affect the success of ADSC therapy. The exact dosage of cells administered has been demonstrated as another uncertain issue influencing treatment effectiveness. Moreover, problems in techniques and methodologies used in ADSC isolation, expansion, and differentiation must be resolved for promising reproducibility in the various clinical applications [34, 35].

Amelioration of insulin resistance

In addition to diminished β-cells function, insulin resistance [7] also plays a crucial role in the pathogenesis of type 2 diabetes and high-risk states [36]. Improving insulin resistance is essential for successfully alleviating hyperglycemia. Advances in research showed that MSCs lower blood sugar not only by increasing insulin production but also by reversing insulin resistance through various mechanisms. Si et al. found that the infusion of MSC into a high-fat diet/STZ-induced T2DM rat model improved insulin sensitivity by increasing glucose transporter type 4 (GLUT4) expression with increased phosphorylation of insulin receptor substrate 1 (IRS-1) and Akt (protein kinase B) in insulin target tissues [37]. Like other MSCs, ADSCs have the potential to ameliorate insulin sensitivity. The effects are due to a significant reduction of inflammatory cytokines and chemokines [38], promoting the transition of macrophages from pro-inflammatory (M1) to anti-inflammatory (M2) [39], and suppressing the expression of IL-6 [40], which is implicated in insulin resistance [41].

Inflammatory conditions and biomarkers of inflammation are major contributors to the development of IR and DM through various signaling pathways [42, 43]. Pro-inflammatory cytokines, including TNF-α (tumor necrosis factor-α) and IL-6 (interleukin-6) are involved in the activation of the NF-κB pathway leading to serine phosphorylation of IRS (insulin receptor substrate), resulting in the IR and development of DM [43, 44]. ADSC infusion in T2DM-induced Streptozotocin induced rats significantly decreased the concentration of inflammatory cytokines such as TNF-α, IL-6 and IL-1β (Interleukin-1β), which resulted in improved insulin sensitivity in insulin target tissues [45].

Accumulating evidence has revealed the key functional roles of the ADSC secretome as a cell free therapy for the treatment of diabetes and its complications [46]. Shree N. and Bhonde R. demonstrated the important role of the ADSC secretome for reversal of insulin resistance for the first time [47]. They observed an enhancement of GLUT4 and phospho Akt protein expression, a significant decrease of intramuscular triglyceride, and adipogenesis inhibition in C2C12 cells under the influence of the ADSC secretome. It has also been established that the secretome of ADSCs down regulates inflammatory markers such as IL6 and PAI1, which are significant in ameliorating IR [47]. Moreover, Sanap et al. discovered that ADSCs condition medium significantly reduced reactive oxygen species generation in insulin resistant 3T3-L1 and C2C12 cells [48]. This finding is of interest, taking into consideration the involvement of ADSC condition medium as insulin sensitizer to combat insulin resistance.

Emerging evidence has shown the therapeutic roles of EVs in modulating different singling pathways due to their delivery of bioactive molecules into target cells [46]. Extracellular vesicles participate in ameliorating insulin resistance by regulating inflammation, which is an important factor in DM pathogenesis [49]. Zhao et al. treated obese mice with ADSC-derived exosomes and found that these EVs activated M2 macrophage polarization into anti-inflammatory type 2 macrophage (M2) phenotypes and subsequently improved insulin sensitivity [50]. These results suggest that ADSC exosomes can act as a prospective insulin sensitizer for DM therapy.

Regulating hepatic glucose metabolism

The liver plays pivotal roles in the control of glucose hemostasis. An in vivo study conducted by Xie et al. confirmed that intravenous infusion of ADSCs into T2DM rats rapidly reduced blood glucose levels [51]. Xie et al. further demonstrated that this role of ADSCs is related to activation of AMP-activated protein kinase (AMPK) signaling pathway that takes place in hepatic cells, contributing to gluconeogenesis inhibition [51].

Promotion of β cell function and regeneration

ADSCs also promote the regeneration and repair of islet β cells. One of the first studies on the use of ADSCs in diabetes mellitus dates to 2010, when Abu-Abeeleh injected two million human ADSCs into Streptozotocin (STZ) induced diabetic rats. The results of their study indicated that intravenously injection of ADSCs normalized the C-peptide level in the blood. They considered the possible role of ADSCs in the restoration of pancreatic islet cells and the return of their function [52]. ADSCs infusion can restore β cell function by reducing the apoptosis of islet cells through decreasing caspase-3 activity. In addition, ADSCs facilitate regeneration by promoting islet vascularization via the release of angiogenic factors such as VEGF, HGF, and vWF [45]. Animal experiments have shown that in a mouse model of T1DM, intravenously injected ADSC can support the survival of β cell progenitors and reduce the levels of blood glucose. The positive effects of ADSCs on the protection of endogenous islet cells can be attributed to their immunoregulatory capacity and direct contact with immune cells [53]. Similarly, a study by Khatri et al. found that intrapancreatic administration of ADSCs to STZ-induced diabetic NMRI nude mice exerted a restorative effect on damaged islet cells [54].

Recent studies have revealed that β cells dedifferentiation is an important mechanism of the pathophysiology of type 2 diabetes (T2DM), which leads to the loss of β cells and dysfunctional insulin resistance [55, 56]. Several previous studies have demonstrated that mesenchymal stem cells (MSCs) can inhibit pancreatic β-cell dedifferentiation. Wang et al. demonstrated that MSC treatment can reverse β cell dedifferentiation in human T2DM islets in an IL-1Ra-mediated manner [57]. However, to date, there have been no reports specifically on the effect of adipose derived stem cells on diabetic pancreatic β cells.

As a gene delivery tool

Since MSCs are the ideal gene delivery vehicles, the combination of MSCs and gene therapy opened up a new chapter in the therapeutic strategy for the treatment of diabetes. Sun et al. used an approach involving lentivirally transduced adipose derived stem cells to deliver betatrophin to the pancreas of Streptozotocin (STZ)-induced diabetic mouse. Infusion of these ADSCs overexpressing betatrophin increased human β cell proliferation and insulin secretion [58]. Most recently, an interesting study transduced human adipocyte-derived stem cells with lentiviral vectors expressing a furin-cleavable insulin gene to treat type 1 diabetes in a mouse model [59]. These reports imply the strong potential of ADSCs as a good candidate vector for cell-based therapy.

Diabetes and chronic wound

One of the main complications of diabetes is chronic wound healing problems, which affect approximately 25% of all patients with DM [60]. In recent years, adipose derived stem cell-based therapy demonstrated efficacy in wound healing, providing a potentially therapeutic tool by addressing multifactorial etiology [61]. The impairment of self-repairing abilities in diabetic patients is due to defects in each phase of wound healing, i.e., haemostasis, inflammation, proliferation, and remodeling phase [62]. Hyperglycemia is associated with higher inflammation indices which lead to an abundance of neutrophils, altered macrophage phenotype, and excessive levels of pro-inflammatory cytokines such as IL-6, IL-8, and TNF-α [63]. Consequently, prolonged inflammatory phase in the wound creates a cascade that maintains the chronic wound state [64].

Mechanisms of ADSCs action on diabetic wound healing

Tissue repair

  • Extracellular matrix remodeling.

Several studies demonstrated that ADSC therapy can accelerate skin reconstruction by stimulating collagen synthesis, which is essential for ECM formation [65]. Liu K. et al. observed enhanced wound reepithelization in ADSC treated wounds [66].

With the treatment of ADSC, expression of collagen III/I, TGF-β3/TGF-β1, and MMP-3/tissue inhibitors of MMP-1 (TIMP-1) increased at the wound site [67]. Enhanced transforming growth factor-β1 (TGF-β1) expression is often accompanied by collagen secretion and differentiation of fibroblasts to myofibroblasts, while an elevated level of TGF-β3 leads to collagen matrix reorganization and wound reepithelization [68, 69]. A study using ADSC-derived exosomes demonstrated that ADSCs promoted the migration and proliferation of fibroblasts by modulating PI3K/Akt signaling [70].

Keratinocytes assume an urgent part in the recovery of wounds, tissue remodelling, and deposition of extracellular matrix [71]. ADSCs advance the expression of two epidermal keratinocyte marker proteins, cytokeratins CK5 and CK14 [72] enhancing the proliferation and migration of keratinocytes thus stimulating the re-epithelialization process in wound healing [73]. Other than keratinocytes, fibroblasts are vital in wound repair as well and can emit different bioactive variables to advance wound healing. ADSC transplantation can initiate the fibroblast aggregate by expanding the enrollment of endothelial cells and macrophages and advancing the development of granulation tissue [73, 74].

  • Site specific differentiation.

ADSCs are pluripotent stem cells with the ability to differentiate into various cell lineages [75]. One mechanism of ADSCs to accelerate wound healing is through differentiation into skin cells in a specific microenvironment [76]. An et al. showed improved wound healing in streptozotocin-induced diabetic mice by differentiation of green fluorescent protein-positive ddADSCs into fibroblasts and endothelial cells [77]. This finding was consistent with other studies in the mouse chronic wound models [76, 78].

ADSCs and immunomodulation

In a diabetic environment, high glucose levels often lead to elevated activity of inflammatory factors such as TNF-α, IL-6, and monocyte chemotactic protein-1 (MCP-1) [79]. ADSCs can attenuate inflammatory responses at wound sites by modulating cytokine expression. It has been reported that ADSCs can suppress the expression of IFN-γ, IL-1, TNF-α, and IL-6 [80, 81]. Particularly, it has been demonstrated that TNF- substantially increases the production of prostaglandin-E2 by ADSCs, increasing their immunosuppressive effect [82]. IFN-γ directly induces the synthesis of indoleamine-pyrrole 2,3-dioxygenase (IDO), which aids in the immunomodulatory effects of ADSCs [83]. Additionally, ADSCs can also increase the expression of anti-inflammatory factors, such as IL10 and IL4, thus controlling cell survival, apoptosis, and reducing inflammation [80, 84].

Other studies have shown that ADSCs can control the number, type, and grouping of macrophages. This means that macrophages can be controlled to help wounds heal. M1 macrophages begin to change into M2-type anti-inflammatory macrophages as wound healing progresses [85]. However, in a high glucose environment, M1 macrophages are more active, and they are more difficult to polarize into M2 macrophages. ADSCs can selectively activate M2 macrophages, thereby exerting immunomodulatory effects [86].

Prostaglandin E2 (PGE2) is a major regulatory factor of ADSC immunomodulation, having multiple functions [87]. One of these is inhibition of NK cells and T helper cells, thereby attenuating the inflammatory response [88].

Because of these findings, ADSCs are now a viable option in regenerative medicine and a powerful tool in cell-based therapy for decreasing inflammatory/immune response and promoting wound healing.

ADSCs and angiogenesis

One of the main causes of diabetic foot is damage to blood vessels and sores on the skin caused by high blood sugar and endothelial dysfunction. The new blood vessels formation results in the delivery of nutrients and oxygen to the wound sites; therefore, it is especially important for wound healing [89]. ADSCs promote angiogenesis through paracrine effects. Studies have shown that ADSCs can secrete a variety of cytokines, including VEGF, stromal cell-derived factor-1 (SDF-1), epidermal growth factor (EGF), basic fibroblast growth factor (bFGF), transforming growth factor-β (TGF-β), and platelet-derived growth factor (PDGF). These factors can promote angiogenesis and facilitate the regeneration of damaged tissues [90, 91]. One of the most important factors involved in angiogenesis is VEGF. The expression level of the VEGF protein was considerably lower in diabetic rats compared to non-diabetic rats [92]. This factor is upregulated following ADSC transplantation in rat studies [9395]. The theoretical basis for this process in diabetic foot ulcers was found by Zhou et al. illustrating that ADSCs regulate VEGF-C expression via the METTL3/IGF2BP2-m6A pathway in diabetic mice [96]. Further research revealed that the tissue around implanted ADSCs exhibits noticeably greater levels of VEGF expression [97]. In studies using human ADSCs, VEGF levels in the tissue around wounds were found to be markedly greater, regardless of whether the ADSCs came from diabetic or healthy donors [95, 98, 99]. Chen l. et al. found that ADSCs may recruit and differentiate endothelial progenitor cells (EPCs) through VEGF-PLCγ-ERK signaling pathway, thereby accelerating diabetic wound healing [100]. Furthermore, Diao et al. demonstrated that VEGF could activate transcription factors in EPCs, recruit EPCs to the bone marrow, and prevent EPC apoptosis in addition to directly promoting angiogenesis [101]. In addition, SDF-1 (CXC chemokine ligand-12) is a powerful chemokine secreted by ADSCs that plays a central role in biological functions associated with wound healing [102]. SDF-1 exerts its biological effect by activating CXC chemokine receptor (CXCR) 4 + and CXCR7 + cells [102]. The SDF-1/CXCR4 axis is perceived to be critical for cell proliferation, chemoattraction of inflammatory cells, and enhancing angiogenesis at the damage sites for wound healing [102]. The secretion of SDF-1 is reduced in diabetic patients. ADSCs have proven to overexpress SDF-1 to be recruited into the damaged site [103, 104]. Di-Rocco et al. showed that topical administration of genetically engineered ADSCs with the SDF-1 gene improved wound healing in diabetic mice (Fig. 2) [105].

Fig. 2.

Fig. 2

Mechanisms of ADSCs in the diabetic wound healing process. Created with BioRender.com.

In a diabetic foot ulcer rat model, Li et al. showed that ADSCs can potentially accelerate wound healing by promoting angiogenesis [106]. In fact, ADSCs are believed to contribute to tissue repair and regeneration by both paracrine signaling and differentiation [107]. Immunofluorescence tracing of the injected ADSCs confirmed that they could migrate to the targeted injury sites and differentiate into endothelial cells to participate in new vessel formation [98, 99, 108, 109]. Wounds treated with ADSCs generally have much higher blood vessel density compared to non-treated wounds [99, 110, 111]. Newly formed vessel density in ADSC-treated tissues is measured to be 2.5 times higher than those of non-treated groups by immunofluorescence localization [111]. Overall, the ability of ADSCs to migrate and differentiate into endothelial cells to participate in the formation of new vessels and to promote this process can be demonstrated by tracing the ADSCs that are injected into wounds’ subdermal layers [96].

Additionally, ADSCs can enhance angiogenesis through macrophage polarization. Wang X. et al. demonstrated that ADSC-Exosome-induced M2-phenotype polarization of macrophages via the JAK/STAT6 signaling pathway can promote the angiogenesis and revascularization of ischemic lower limbs in type 2 diabetic mice [112].

In diabetic ulcers, exosomes derived from MSCs may promote angiogenesis and ischemic tissue repair through the use of microRNAs. A number of microRNAs regulate angiogenesis by targeting VEGF and activating the PI3K/AKT and MAPK pathways [113]. Huang C. et al. showed that miRNA-21-5p could potently promote angiogenesis through activating serine/threonine kinase (AKT), mitogen-activated protein kinase (MAPK), and vascular endothelial growth factor receptor (VEGFR) [114]. In addition, miRNA-210-3p could enhance microcirculation and support angiogenesis by upregulating VEGF via activating a number of proangiogenic proteins, such as SRC, AKT, and ERK [115]. Exosomal miRNA-221-3p derived from MSCs enhanced the biological function of endothelial cells via the AKT/eNOS pathway, thereby facilitating the repair of diabetic wounds [116].

An Y. et al. used ADSCs overexpressing miR-21 to promote endothelial cell vascularization for regenerative purposes. They demonstrated that mir-21 secreted by engineered ADSCs could possibly be used to assist wound healing by enhancing HIF-1α and VEGF expression [117].

These studies show that paracrine growth factors, and microRNAs may help ADSCs promote angiogenesis at the injury site in a direct or indirect manner.

Safety of using adipose derived stem cells for transplantation

The emergence of ADSC therapy over the last decade has changed the therapeutic landscape for tissue engineering and organ regeneration. Numerous studies have demonstrated the significant role of ADSCs not only in DM but also in other clinical situations [118]. However, numerous uncertainties remain about ADSC therapy, which need to be addressed before daily clinical application.

The contribution of MSCs to tumor pathogenesis is widely discussed in the literature [118]. While various studies have shown that MSCs inhibit tumor progression and metastasis in multiple types of cancer [61, 63, 107, 119, 120], a large body of research has explored the capability of MSCs to contribute to tumorigenesis. It is postulated that MSCs have the capacity to differentiate into tumor-associated fibroblasts in the tumor microenvironment, providing both a functional and structural supportive environment through the production of tumor-stimulating factors and immunomodulatory mechanisms [121]. Moreover, MSCs appear to modulate immune responses and suppress both innate immune cells (neutrophils, macrophages, and NK cells) and adaptive immune system cells (DCs, B lymphocytes, and T lymphocytes) [122]. In addition, several studies have shown that MSCs can promote tumor angiogenesis by secreting proangiogenic factors (angiopoietins, EGF, galectin-1, IGF‐1, KGF, and VEGF) as well as through their potential to differentiate into pericytes, stromal fibroblasts, or endothelial-like cells [123, 124]. In addition to the abovementioned mechanisms, MSCs exert their supportive effect on tumors through inhibition of tumor cell apoptosis, stimulation of epithelial-mesenchymal transition (EMT), and promotion of tumor metastasis [125].

In fact, several discrepant results have been reported regarding the multifaceted roles of MSCs in tumor support or suppression. Thus, further studies characterizing the mechanisms of pro- or anti-tumorigenic effects of MSCs may increase the utilization of MSCs without increasing the risk of tumorigenicity. Although DSCs are typically considered stable after in vitro manipulation and expansion compared with cells derived from other sources, chromosomal instability in long-term cultures is still a major issue in the advanced application of ADSCs [102].

Another major safety issue of note is the need for 10–20% FBS during ADSC culture; actually, FBS comes with safety concerns such as transmitting unknown viral diseases and provoking immunoreactivity. Therefore, studies to develop and measure the safety of serum-free or xeno-free culture media without animal serum are necessary [71, 126].

An important issue defining any new efficient therapeutic application at a large scale is to establish the optimal dosing regimens and route of administration [72]. Further, large scale randomized placebo-controlled studies to optimize pharmacologically effective dose range, dosing schedule and rout of administration are strongly encouraged. Furthermore, another safety concern regarding using ADSCs for clinical applications is product purity.

Future Perspectives

ADSCs based therapy is a promising strategy for the clinical treatment of numerous diseases, such as diabetes mellitus, but its systematic translational use is still somewhat out of reach. In order to increase ADSC’s therapeutic efficacy and expand their application, various strategies have been examined.

Notably, priming or preconditioning of ADSCs with cytokines, hypoxia, or pharmacological drugs in vitro is one of the foremost applicable strategies to improve cell function, survival, and therapeutic potential [127]. However, several limitations, including high cost, induction of immunogenicity, and difficulty in providing good manufacturing practices (GMP) grade quality assurance for clinical application, still need to be addressed in further research [127, 128].

Genetic engineering of ADSCs is another promising approach to improve survival, migration, and immunomodulatory properties [129]. ADSCs transfected with C-X-C chemokine receptor type 4 (CXCR4) showed enhanced homing and engraftment in diabetic mice with hindlimb ischemia [130]. Sen et al. showed that ADSCs overexpressing superoxide dismutase 2 (SOD2) exhibited beneficial outcomes in reducing inflammation and improving glucose tolerance in obese diabetic mice model [131]. However, viral transduction and genetic modification are closely associated with safety concerns such as high immunogenicity, toxicity, and insertional mutagenesis [132, 133].

Recent studies have shown significant therapeutic effects of MSC-cell free therapy under a variety of experimental conditions, including metabolic disorders [134, 135]. Indeed, MSCs exert their therapeutic effects by releasing secretome, including growth factors and extracellular vesicles (EVs) [74, 136]. Recently, MSC-derived EVs have attracted significant attention as a cell free therapeutic option [137]. ADSC-derived EVs emerged as direct mediators of diabetes [134]. MiRNAs carried by adipocyte-derived EVs have been shown to have hormone-like activity, communicating with other organs and tissues to control metabolic homeostasis as well as systemic insulin resistance [104, 138]. Given the potential of adipocyte-derived EVs in regenerative therapies, future research should examine their exact therapeutic capabilities in diabetes.

Conclusions

Until now, the potential therapy effect of ADSCs in DM has been demonstrated in several studies, which increases the expectation for utilizing them in clinical application. ADSCs are able to improve insulin production and also ameliorate insulin resistance in T2DM. In addition to being able to differentiate into IPCs, ADSCs can also transport genes, increase hepatic glucose metabolism, and regenerate islet cells. Specifically, they can accelerate diabetic wound healing by synergistic effects, such as; enhancing proliferation and re-epithelialization of skin cells, site specific differentiation, immunomodulation, and promoting angiogenesis.

However, many safety challenges need to be considered, from the preparation of ADSCs to their application. Further large-scale and controlled studies are needed to overcome those issues and confirm the safety and optimal therapeutic scheme before daily clinical application.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1 (509.8KB, docx)

Author contributions

All authors have contributed and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Declarations

Conflict of interest

The authors declare that there is no conflict of interest regarding the publication of this paper.

Footnotes

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References

  • 1.Khan MAB, et al. Epidemiology of type 2 diabetes–global burden of disease and forecasted trends. J Epidemiol global health. 2020;10(1):107. doi: 10.2991/jegh.k.191028.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Ozougwu J, et al. The pathogenesis and pathophysiology of type 1 and type 2 diabetes mellitus. J Physiol Pathophysiol. 2013;4(4):46–57. doi: 10.5897/JPAP2013.0001. [DOI] [Google Scholar]
  • 3.Leitner DR, et al. Obesity and type 2 diabetes: two diseases with a need for combined treatment strategies-EASO can lead the way. Obes Facts. 2017;10(5):483–92. doi: 10.1159/000480525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Cho NH, et al. IDF Diabetes Atlas: global estimates of diabetes prevalence for 2017 and projections for 2045. Diabetes Res Clin Pract. 2018;138:271–81. doi: 10.1016/j.diabres.2018.02.023. [DOI] [PubMed] [Google Scholar]
  • 5.Huang Q, Huang Y, Liu J. Mesenchymal Stem Cells: An Excellent Candidate for the Treatment of Diabetes Mellitus International Journal of Endocrinology, 2021. 2021. DOI: 10.1155/2021/9938658. [DOI] [PMC free article] [PubMed]
  • 6.Sutherland D. Pancreas and islet transplantation. Diabetologia. 1981;20(4):435–50. doi: 10.1007/BF00253405. [DOI] [PubMed] [Google Scholar]
  • 7.Khosravi-Maharlooei M et al. Islet transplantation for type 1 diabetes: so close and yet so far away. Eur J Endocrinol, 2015. [DOI] [PubMed]
  • 8.Bruni A, et al. Islet cell transplantation for the treatment of type 1 diabetes: recent advances and future challenges. Diabetes metabolic syndrome and obesity: targets and therapy. 2014;7:211. doi: 10.2147/DMSO.S50789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.O’Brien T, Barry FP. Stem cell therapy and regenerative medicine. in Mayo Clinic Proceedings. 2009. Elsevier. DOI: 10.4065/84.10.859. [DOI] [PMC free article] [PubMed]
  • 10.Han Y, et al. Mesenchymal stem cells for regenerative medicine. Cells. 2019;8(8):886. doi: 10.3390/cells8080886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Si Z, et al. Adipose-derived stem cells: sources, potency, and implications for regenerative therapies. Biomed Pharmacother. 2019;114:108765. doi: 10.1016/j.biopha.2019.108765. [DOI] [PubMed] [Google Scholar]
  • 12.Bourin P, et al. Stromal cells from the adipose tissue-derived stromal vascular fraction and culture expanded adipose tissue-derived stromal/stem cells: a joint statement of the International Federation for Adipose therapeutics and Science (IFATS) and the International Society for Cellular Therapy (ISCT) Cytotherapy. 2013;15(6):641–8. doi: 10.1016/j.jcyt.2013.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Zuk PA, et al. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng. 2001;7(2):211–28. doi: 10.1089/107632701300062859. [DOI] [PubMed] [Google Scholar]
  • 14.Trzyna A, Banaś-Ząbczyk A. Adipose-derived stem cells secretome and its potential application in “stem cell-free therapy. Biomolecules. 2021;11(6):878. doi: 10.3390/biom11060878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Barba M, Taranto GD, Lattanzi W. Adipose-derived stem cell therapies for bone regeneration. Expert Opin Biol Ther. 2017;17(6):677–89. doi: 10.1080/14712598.2017.1315403. [DOI] [PubMed] [Google Scholar]
  • 16.Im G-I, Shin Y-W, Lee K-B. Do adipose tissue-derived mesenchymal stem cells have the same osteogenic and chondrogenic potential as bone marrow-derived cells? Osteoarthr Cartil. 2005;13(10):845–53. doi: 10.1016/j.joca.2005.05.005. [DOI] [PubMed] [Google Scholar]
  • 17.Dominici M, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8(4):315–7. doi: 10.1080/14653240600855905. [DOI] [PubMed] [Google Scholar]
  • 18.Mildmay-White A, Khan W. Cell surface markers on adipose-derived stem cells: a systematic review. Curr Stem Cell Res Therapy. 2017;12(6):484–92. doi: 10.2174/1574888X11666160429122133. [DOI] [PubMed] [Google Scholar]
  • 19.Baer PC. Adipose-derived mesenchymal stromal/stem cells: an update on their phenotype in vivo and in vitro. World J stem cells. 2014;6(3):256. doi: 10.4252/wjsc.v6.i3.256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Viswanathan S, et al. Mesenchymal stem versus stromal cells: International Society for Cell & Gene Therapy (ISCT®) mesenchymal stromal cell committee position statement on nomenclature. Cytotherapy. 2019;21(10):1019–24. doi: 10.1016/j.jcyt.2019.08.002. [DOI] [PubMed] [Google Scholar]
  • 21.Li P, Guo X. A review: therapeutic potential of adipose-derived stem cells in cutaneous wound healing and regeneration. Stem Cell Res Ther. 2018;9(1):1–7. doi: 10.1186/s13287-018-1044-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Huang JI, et al. Chondrogenic potential of progenitor cells derived from human bone marrow and adipose tissue: a patient-matched comparison. J Orthop Res. 2005;23(6):1383–9. doi: 10.1016/j.orthres.2005.03.018. [DOI] [PubMed] [Google Scholar]
  • 23.Choi YS, et al. Mechanical derivation of functional myotubes from adipose-derived stem cells. Biomaterials. 2012;33(8):2482–91. doi: 10.1016/j.biomaterials.2011.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Miyagi-Shiohira C, et al. Cryopreservation of adipose-derived mesenchymal stem cells. Cell Med. 2015;8(1–2):3–7. doi: 10.3727/215517915X689100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ceccarelli S, et al. Immunomodulatory effect of adipose-derived stem cells: the cutting edge of clinical application. Front cell Dev biology. 2020;8:236. doi: 10.3389/fcell.2020.00236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Fu Y, et al. Trophic effects of mesenchymal stem cells in tissue regeneration. Tissue Eng Part B: Reviews. 2017;23(6):515–28. doi: 10.1089/ten.teb.2016.0365. [DOI] [PubMed] [Google Scholar]
  • 27.Chandra V, et al. Generation of pancreatic hormone-expressing islet-like cell aggregates from murine adipose tissue-derived stem cells. Stem Cells. 2009;27(8):1941–53. doi: 10.1002/stem.117. [DOI] [PubMed] [Google Scholar]
  • 28.Nam JS, et al. Transplantation of insulin-secreting cells differentiated from human adipose tissue-derived stem cells into type 2 diabetes mice. Biochem Biophys Res Commun. 2014;443(2):775–81. doi: 10.1016/j.bbrc.2013.10.059. [DOI] [PubMed] [Google Scholar]
  • 29.Timper K, et al. Human adipose tissue-derived mesenchymal stem cells differentiate into insulin, somatostatin, and glucagon expressing cells. Biochem Biophys Res Commun. 2006;341(4):1135–40. doi: 10.1016/j.bbrc.2006.01.072. [DOI] [PubMed] [Google Scholar]
  • 30.Dave SD, Vanikar AV, Trivedi HL. Ex vivo generation of glucose sensitive insulin secreting mesenchymal stem cells derived from human adipose tissue. Indian J Endocrinol Metabol. 2012;16(Suppl1):S65. doi: 10.4103/2230-8210.94264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Zhang S, et al. Promoting long-term survival of insulin-producing cell grafts that differentiate from adipose tissue-derived stem cells to cure type 1 diabetes. PLoS ONE. 2011;6(12):e29706. doi: 10.1371/journal.pone.0029706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Dhanasekaran M, et al. Human omentum fat-derived mesenchymal stem cells transdifferentiates into pancreatic islet‐like cluster. Cell Biochem Funct. 2013;31(7):612–9. doi: 10.1002/cbf.2948. [DOI] [PubMed] [Google Scholar]
  • 33.Amer MG, et al. Role of adipose tissue derived stem cells differentiated into insulin producing cells in the treatment of type I diabetes mellitus. Gene. 2018;654:87–94. doi: 10.1016/j.gene.2018.02.008. [DOI] [PubMed] [Google Scholar]
  • 34.Paek HJ, Kim C, Williams SK. Adipose stem cell-based regenerative medicine for reversal of diabetic hyperglycemia. World J Diabetes. 2014;5(3):235. doi: 10.4239/wjd.v5.i3.235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Balistreri CR, et al. Stem cell therapy: old challenges and new solutions. Mol Biol Rep. 2020;47(4):3117–31. doi: 10.1007/s11033-020-05353-2. [DOI] [PubMed] [Google Scholar]
  • 36.Kahn SE, Cooper ME, Prato SD. Pathophysiology and treatment of type 2 diabetes: perspectives on the past, present, and future. The Lancet. 2014;383(9922):1068–83. doi: 10.1016/S0140-6736(13)62154-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Si Y, et al. Infusion of mesenchymal stem cells ameliorates hyperglycemia in type 2 diabetic rats: identification of a novel role in improving insulin sensitivity. Diabetes. 2012;61(6):1616–25. doi: 10.2337/db11-1141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Estrada EJ, et al. Combined treatment of intrapancreatic autologous bone marrow stem cells and hyperbaric oxygen in type 2 diabetes mellitus. Cell Transplant. 2008;17(12):1295–304. doi: 10.3727/096368908787648119. [DOI] [PubMed] [Google Scholar]
  • 39.Xie Z, et al. Human umbilical cord-derived mesenchymal stem cells elicit macrophages into an anti-inflammatory phenotype to alleviate insulin resistance in type 2 diabetic rats. Stem Cells. 2016;34(3):627–39. doi: 10.1002/stem.2238. [DOI] [PubMed] [Google Scholar]
  • 40.Blaber SP, et al. Analysis of in vitro secretion profiles from adipose-derived cell populations. J translational Med. 2012;10(1):1–16. doi: 10.1186/1479-5876-10-172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Ruan H, et al. Tumor necrosis factor-α suppresses adipocyte-specific genes and activates expression of preadipocyte genes in 3T3-L1 adipocytes: nuclear factor-κB activation by TNF-α is obligatory. Diabetes. 2002;51(5):1319–36. doi: 10.2337/diabetes.51.5.1319. [DOI] [PubMed] [Google Scholar]
  • 42.Gratas-Delamarche A, et al. Physical inactivity, insulin resistance, and the oxidative-inflammatory loop. Free Radic Res. 2014;48(1):93–108. doi: 10.3109/10715762.2013.847528. [DOI] [PubMed] [Google Scholar]
  • 43.Patel PS, Buras ED, Balasubramanyam A. The role of the immune system in obesity and insulin resistance Journal of obesity, 2013. 2013. DOI: 10.1155/2013/616193. [DOI] [PMC free article] [PubMed]
  • 44.Mahmoud F, Al-Ozairi E. Inflammatory cytokines and the risk of cardiovascular complications in type 2 diabetes. Dis Markers. 2013;35(4):235–41. doi: 10.1155/2013/931915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Hu J, et al. Effects of autologous adipose-derived stem cell infusion on type 2 diabetic rats. Endocr J. 2015;EJ14–0584. 10.1507/endocrj.EJ14-0584. [DOI] [PubMed]
  • 46.Hu W, et al. Therapeutic potentials of extracellular vesicles for the treatment of diabetes and diabetic complications. Int J Mol Sci. 2020;21(14):5163. doi: 10.3390/ijms21145163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Shree N, Bhonde RR. Conditioned media from adipose tissue derived mesenchymal stem cells reverse insulin resistance in cellular models. J Cell Biochem. 2017;118(8):2037–43. doi: 10.1002/jcb.25777. [DOI] [PubMed] [Google Scholar]
  • 48.Sanap A, Bhonde R, Joshi K. Conditioned medium of adipose derived mesenchymal stem cells reverse insulin resistance through downregulation of stress induced serine kinases. Eur J Pharmacol. 2020;881:173215. doi: 10.1016/j.ejphar.2020.173215. [DOI] [PubMed] [Google Scholar]
  • 49.Liu J, et al. Clinical potential of extracellular vesicles in type 2 diabetes. Front Endocrinol. 2021;11:596811. doi: 10.3389/fendo.2020.59681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Zhao H, et al. Exosomes from adipose-derived stem cells attenuate adipose inflammation and obesity through polarizing M2 macrophages and beiging in white adipose tissue. Diabetes. 2018;67(2):235–47. doi: 10.2337/db17-0356. [DOI] [PubMed] [Google Scholar]
  • 51.Xie M, et al. Adipose-derived mesenchymal stem cells ameliorate hyperglycemia through regulating hepatic glucose metabolism in type 2 diabetic rats. Biochem Biophys Res Commun. 2017;483(1):435–41. doi: 10.1016/j.bbrc.2016.12.125. [DOI] [PubMed] [Google Scholar]
  • 52.Abu-Abeeleh M, et al. A preliminary study of the use of human adipose tissue-derived stem cells for the treatment of streptozotocin-induced diabetes mellitus in a rat model. Comp Clin Pathol. 2010;19(1):1–4. doi: 10.1007/s00580-009-0912-x. [DOI] [Google Scholar]
  • 53.Li Y-Y, et al. Adipose-derived mesenchymal stem cells ameliorate STZ-induced pancreas damage in type 1 diabetes. Biomed Mater Eng. 2012;22(1–3):97–103. doi: 10.3233/BME-2012-0694. [DOI] [PubMed] [Google Scholar]
  • 54.Khatri R, Petry SF, Linn T. Intrapancreatic MSC transplantation facilitates pancreatic islet regeneration. Stem Cell Res Ther. 2021;12(1):1–14. doi: 10.1186/s13287-021-02173-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Saisho Y. β-cell dysfunction: its critical role in prevention and management of type 2 diabetes. World J diabetes. 2015;6(1):109. doi: 10.4239/wjd.v6.i1.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Talchai C, et al. Pancreatic β cell dedifferentiation as a mechanism of diabetic β cell failure. Cell. 2012;150(6):1223–34. doi: 10.1016/j.cell.2012.07.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Wang L, et al. Mesenchymal stem cells ameliorate β cell dysfunction of human type 2 diabetic islets by reversing β cell dedifferentiation. EBioMedicine. 2020;51:102615. doi: 10.1016/j.ebiom.2019.102615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Sun L-L, et al. Transplantation of betatrophin-expressing adipose-derived mesenchymal stem cells induces β-cell proliferation in diabetic mice. Int J Mol Med. 2017;39(4):936–48. doi: 10.3892/ijmm.2017.2914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Fang Q, et al. Adipocyte-derived stem cell-based gene therapy upon adipogenic differentiation on microcarriers attenuates type 1 diabetes in mice. Stem Cell Res Ther. 2019;10(1):1–11. doi: 10.1186/s13287-019-1135-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Burgess JL, et al. Diabetic wound-healing science. Medicina. 2021;57(10):1072. doi: 10.3390/medicina57101072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Lee DE, Ayoub N, Agrawal DK. Mesenchymal stem cells and cutaneous wound healing: novel methods to increase cell delivery and therapeutic efficacy. Stem Cell Res Ther. 2016;7(1):1–8. doi: 10.1186/s13287-016-0303-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Patel S, et al. Mechanistic insight into diabetic wounds: Pathogenesis, molecular targets and treatment strategies to pace wound healing. Biomed Pharmacother. 2019;112:108615. doi: 10.1016/j.biopha.2019.108615. [DOI] [PubMed] [Google Scholar]
  • 63.Rea IM, et al. Age and age-related diseases: role of inflammation triggers and cytokines. Front Immunol. 2018;586. 10.3389/fimmu.2018.00586. [DOI] [PMC free article] [PubMed]
  • 64.Maxson S, et al. Concise review: role of mesenchymal stem cells in wound repair. Stem cells translational medicine. 2012;1(2):142–9. doi: 10.5966/sctm.2011-0018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Nambu M, et al. Accelerated wound healing in healing-impaired db/db mice by autologous adipose tissue-derived stromal cells combined with atelocollagen matrix. Ann Plast Surg. 2009;62(3):317–21. doi: 10.1097/SAP.0b013e31817f01b6. [DOI] [PubMed] [Google Scholar]
  • 66.Liu K, et al. Adipose stem cell-derived exosomes in combination with hyaluronic acid accelerate wound healing through enhancing re‐epithelialization and vascularization. Br J Dermatol. 2019;181(4):854–6. doi: 10.1111/bjd.17984. [DOI] [PubMed] [Google Scholar]
  • 67.Wang L et al. Exosomes secreted by human adipose mesenchymal stem cells promote scarless cutaneous repair by regulating extracellular matrix remodelling (vol 7, 13321, 2017). SCIENTIFIC REPORTS, 2021. 11(1). DOI: 10.1038/s41598-017-12919-x. [DOI] [PMC free article] [PubMed]
  • 68.Shen T, et al. Accelerated healing of diabetic wound using artificial dermis constructed with adipose stem cells and poly (L-glutamic acid)/chitosan scaffold. Chin Med J. 2013;126(08):1498–503. [PubMed] [Google Scholar]
  • 69.Zografou A, et al. Autologous transplantation of adipose-derived stem cells enhances skin graft survival and wound healing in diabetic rats. Ann Plast Surg. 2013;71(2):225–32. doi: 10.1097/SAP.0b013e31826af01a. [DOI] [PubMed] [Google Scholar]
  • 70.Li C, et al. Adipose mesenchymal stem cell-derived Exosomes Promote Wound Healing through the WNT/β-catenin signaling pathway in dermal fibroblasts. Stem Cell Reviews and Reports. 2022;1–15. 10.1007/s12015-022-10378-0. [DOI] [PMC free article] [PubMed]
  • 71.Moon KM, et al. The effect of secretory factors of adipose-derived stem cells on human keratinocytes. Int J Mol Sci. 2012;13(1):1239–57. doi: 10.3390/ijms13011239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Ebrahimian T et al. Cell therapy based on adipose tissue-derived stromal cells promotes physiological and pathological wound healing Arteriosclerosis, thrombosis, and vascular biology, 2009. 29(4): p. 503–10. DOI: 10.1161/ATVBAHA.108.17896. [DOI] [PubMed]
  • 73.Tutuianu R, et al. Human mesenchymal stromal cell-derived exosomes promote in vitro wound healing by modulating the biological properties of skin keratinocytes and fibroblasts and stimulating angiogenesis. Int J Mol Sci. 2021;22(12):6239. doi: 10.3390/ijms22126239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Hong SJ, et al. Topically delivered adipose derived stem cells show an activated-fibroblast phenotype and enhance granulation tissue formation in skin wounds. PLoS ONE. 2013;8(1):e55640. doi: 10.1371/journal.pone.0055640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Poliwoda S, et al. Stem cells: a comprehensive review of origins and emerging clinical roles in medical practice. Orthop Rev. 2022;14(3). 10.52965/001c.37498. [DOI] [PMC free article] [PubMed]
  • 76.Nie C, et al. Locally administered adipose-derived stem cells accelerate wound healing through differentiation and vasculogenesis. Cell Transplant. 2011;20(2):205–16. doi: 10.3727/096368910X52006. [DOI] [PubMed] [Google Scholar]
  • 77.An R, et al. Adipose stem cells isolated from diabetic mice improve cutaneous wound healing in streptozotocin-induced diabetic mice. Stem Cell Res Ther. 2020;11(1):1–11. doi: 10.1186/s13287-020-01621-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Huang S-P, et al. Promotion of wound healing using adipose-derived stem cells in radiation ulcer of a rat model. J Biomed Sci. 2013;20(1):1–10. doi: 10.1186/1423-0127-20-51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Mulder GD, Berg JSV. Cellular senescence and matrix metalloproteinase activity in chronic wounds: relevance to debridement and new technologies. J Am Podiatr Med Assoc. 2002;92(1):34–7. doi: 10.7547/87507315-92-1-34. [DOI] [PubMed] [Google Scholar]
  • 80.Hao Z, et al. Research progress of adipose-derived stem cells in the treatment of chronic wounds. Front Chem. 2023;11. 10.3389/fchem.2023.1094693. [DOI] [PMC free article] [PubMed]
  • 81.Xiong J et al. Comparison of proangiogenic effects of adipose-derived stem cells and foreskin fibroblast exosomes on artificial dermis prefabricated flaps Stem Cells International, 2020. 2020. DOI: 10.1155/2020/5293850. [DOI] [PMC free article] [PubMed]
  • 82.Yoo KH, et al. Comparison of immunomodulatory properties of mesenchymal stem cells derived from adult human tissues. Cell Immunol. 2009;259(2):150–6. doi: 10.1016/j.cellimm.2009.06.010. [DOI] [PubMed] [Google Scholar]
  • 83.Krampera M, et al. Role for interferon-γ in the immunomodulatory activity of human bone marrow mesenchymal stem cells. Stem Cells. 2006;24(2):386–98. doi: 10.1634/stemcells.2005-0008. [DOI] [PubMed] [Google Scholar]
  • 84.Stojanović S, Najman S. The effect of conditioned media of stem cells derived from lipoma and adipose tissue on macrophages’ response and wound healing in indirect co-culture system in vitro. Int J Mol Sci. 2019;20(7):1671. doi: 10.3390/ijms20071671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Fantin A, et al. Tissue macrophages act as cellular chaperones for vascular anastomosis downstream of VEGF-mediated endothelial tip cell induction. Blood The Journal of the American Society of Hematology. 2010;116(5):829–40. doi: 10.1182/blood-2009-12-257832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Morey M, et al. Hyperglycemia acts in synergy with hypoxia to maintain the pro-inflammatory phenotype of macrophages. PLoS ONE. 2019;14(8):e0220577. doi: 10.1371/journal.pone.0220577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Gao F, et al. Mesenchymal stem cells and immunomodulation: current status and future prospects. Cell Death Dis. 2016;7(1):e2062–2. doi: 10.1038/cddis.2015.327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Eleuteri S, Fierabracci A. Insights into the secretome of mesenchymal stem cells and its potential applications. Int J Mol Sci. 2019;20(18):4597. doi: 10.3390/ijms20184597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Honnegowda TM, et al. Role of angiogenesis and angiogenic factors in acute and chronic wound healing. Plast Aesthetic Res. 2015;2:243–9. doi: 10.4103/2347-9264.165438. [DOI] [Google Scholar]
  • 90.Zhao L, Johnson T, Liu D. Therapeutic angiogenesis of adipose-derived stem cells for ischemic diseases. Stem Cell Res Ther. 2017;8(1):1–9. doi: 10.1186/s13287-017-0578-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Rehman J, et al. Secretion of angiogenic and antiapoptotic factors by human adipose stromal cells. Circulation. 2004;109(10):1292–8. doi: 10.1161/01.CIR.0000121425.42966.F1. [DOI] [PubMed] [Google Scholar]
  • 92.Schratzberger P, et al. Reversal of experimental diabetic neuropathy by VEGF gene transfer. J Clin Investig. 2001;107(9):1083–92. doi: 10.1172/JCI12188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Wang C, et al. A pilot study on ex vivo expanded autologous adipose-derived stem cells of improving fat retention in localized scleroderma patients. Stem Cells Translational Medicine. 2021;10(8):1148–56. doi: 10.1002/sctm.20-0419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Chen L, et al. Autologous nanofat transplantation accelerates foot wound healing in diabetic rats. Regen Med. 2019;14(3):231–41. doi: 10.2217/rme-2018-0169. [DOI] [PubMed] [Google Scholar]
  • 95.Seo E, et al. Exendin-4 in combination with adipose-derived stem cells promotes angiogenesis and improves diabetic wound healing. J translational Med. 2017;15(1):1–9. doi: 10.1186/s12967-017-1145-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Liu R, et al. Adipose-derived stem cells for the treatment of diabetic wound: from basic study to clinical application. Front Endocrinol. 2022;13. 10.3389/fendo.2022.882469. [DOI] [PMC free article] [PubMed]
  • 97.Kuo Y-R, et al. Adipose-derived stem cells accelerate diabetic wound healing through the induction of autocrine and paracrine effects. Cell Transplant. 2016;25(1):71–81. doi: 10.3727/096368915X687921. [DOI] [PubMed] [Google Scholar]
  • 98.Shi R, et al. Localization of human adipose-derived stem cells and their effect in repair of diabetic foot ulcers in rats. Stem Cell Res Ther. 2016;7(1):1–13. doi: 10.1186/s13287-016-0412-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Kaisang L, et al. Adipose-derived stem cells seeded in pluronic F-127 hydrogel promotes diabetic wound healing. J Surg Res. 2017;217:63–74. doi: 10.1016/j.jss.2017.04.032. [DOI] [PubMed] [Google Scholar]
  • 100.Chen L, et al. Adipose-derived stem cells promote diabetic wound healing via the recruitment and differentiation of endothelial progenitor cells into endothelial cells mediated by the VEGF-PLCγ-ERK pathway. Arch Biochem Biophys. 2020;692:108531DOI. doi: 10.1016/j.abb.2020.108531. [DOI] [PubMed] [Google Scholar]
  • 101.Diao Y, et al. A vascular endothelial growth factor activating transcription factor increases the endothelial progenitor cells population and induces therapeutic angiogenesis in a type 1 diabetic mouse with hindlimb ischemia. Chin Med J. 2014;127(20):3623–9. doi: 10.3760/cma.j.issn.0366-6999.20140883. [DOI] [PubMed] [Google Scholar]
  • 102.Li Q, et al. Stromal cell-derived factor-1 promotes human adipose tissue-derived stem cell survival and chronic wound healing. Experimental and therapeutic medicine. 2016;12(1):45–50. doi: 10.3892/etm.2016.3309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Rennert RC, et al. Stem cell recruitment after injury: lessons for regenerative medicine. Regen Med. 2012;7(6):833–50. doi: 10.2217/rme.12.82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Wu Q, et al. Stromal cell-derived factor 1 promoted migration of adipose-derived stem cells to the wounded area in traumatic rats. Biochem Biophys Res Commun. 2015;467(1):140–5. doi: 10.1016/j.bbrc.2015.09.097. [DOI] [PubMed] [Google Scholar]
  • 105.Di Rocco G et al. Enhanced healing of diabetic wounds by topical administration of adipose tissue-derived stromal cells overexpressing stromal-derived factor-1: biodistribution and engraftment analysis by bioluminescent imaging Stem cells international, 2010. 2011. DOI: 10.4061/2011/304562. [DOI] [PMC free article] [PubMed]
  • 106.Li X, et al. Exosomes from adipose-derived stem cells overexpressing Nrf2 accelerate cutaneous wound healing by promoting vascularization in a diabetic foot ulcer rat model. Exp Mol Med. 2018;50(4):1–14. doi: 10.1038/s12276-018-0058-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Mazini L, et al. Hopes and limits of adipose-derived stem cells (ADSCs) and mesenchymal stem cells (MSCs) in wound healing. Int J Mol Sci. 2020;21(4):1306. doi: 10.3390/ijms21041306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Kim EK, et al. The effect of human adipose-derived stem cells on healing of ischemic wounds in a diabetic nude mouse model. Plast Reconstr Surg. 2011;128(2):387–94. doi: 10.1097/PRS.0b013e31821e6de2. [DOI] [PubMed] [Google Scholar]
  • 109.Ding S, et al. Effect of collagen scaffold with bcl-2-modified adipose-derived stem cells on diabetic mice wound healing. Int J Low Extrem Wounds. 2020;19(2):139–47. doi: 10.1177/1534734619880055. [DOI] [PubMed] [Google Scholar]
  • 110.Kato Y, et al. Allogeneic transplantation of an adipose-derived stem cell sheet combined with artificial skin accelerates wound healing in a rat wound model of type 2 diabetes and obesity. Diabetes. 2015;64(8):2723–34. doi: 10.2337/db14-1133. [DOI] [PubMed] [Google Scholar]
  • 111.Ebrahim N, et al. Adipose mesenchymal stem cells combined with platelet-rich plasma accelerate diabetic wound healing by modulating the notch pathway. Stem Cell Res Ther. 2021;12(1):1–24. doi: 10.1186/s13287-021-02454-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Wang X, et al. Adipose-derived stem cell-secreted exosomes enhance angiogenesis by promoting macrophage M2 polarization in type 2 diabetic mice with limb ischemia via the JAK/STAT6 pathway. Heliyon. 2022;8(11):e11495. doi: 10.1016/j.heliyon.2022.e11495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Wang J, et al. Hypoxia adipose stem cell-derived exosomes promote high-quality healing of diabetic wound involves activation of PI3K/Akt pathways. J Nanobiotechnol. 2021;19(1):1–13. doi: 10.1186/s12951-021-00942-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Huang C, et al. Human mesenchymal stem cells promote ischemic repairment and angiogenesis of diabetic foot through exosome miRNA-21-5p. Stem Cell Research. 2021;52:102235. doi: 10.1016/j.scr.2021.102235. [DOI] [PubMed] [Google Scholar]
  • 115.Gangadaran P, et al. Extracellular vesicles from mesenchymal stem cells activates VEGF receptors and accelerates recovery of hindlimb ischemia. J Controlled Release. 2017;264:112–26. doi: 10.1016/j.jconrel.2017.08.022. [DOI] [PubMed] [Google Scholar]
  • 116.Yu M, et al. Exosomes derived from atorvastatin-pretreated MSC accelerate diabetic wound repair by enhancing angiogenesis via AKT/eNOS pathway. Stem Cell Res Ther. 2020;11:1–17. doi: 10.1186/s13287-020-01824-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.An Y, et al. Exosomes from adipose-derived stem cells (ADSCs) overexpressing miR-21 promote vascularization of endothelial cells. Sci Rep. 2019;9(1):12861. doi: 10.1038/s41598-019-49339-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Fadini GP, et al. Angiogenic abnormalities in diabetes mellitus: mechanistic and clinical aspects. J Clin Endocrinol Metabolism. 2019;104(11):5431–44. doi: 10.1210/jc.2019-00980. [DOI] [PubMed] [Google Scholar]
  • 119.Seo Y, Shin T-H, Kim H-S. Current strategies to enhance adipose stem cell function: an update. Int J Mol Sci. 2019;20(15):3827. doi: 10.3390/ijms20153827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Huang W, et al. Fibroblast growth factor 21 enhances angiogenesis and wound healing of human brain microvascular endothelial cells by activating PPARγ. J Pharmacol Sci. 2019;140(2):120–7. doi: 10.1016/j.jphs.2019.03.010. [DOI] [PubMed] [Google Scholar]
  • 121.Oh EJ, et al. In vivo migration of mesenchymal stem cells to burn injury sites and their therapeutic effects in a living mouse model. J Controlled Release. 2018;279:79–88. doi: 10.1016/j.jconrel.2018.04.020. [DOI] [PubMed] [Google Scholar]
  • 122.Li J, Xiao L, Rao C. Comparison of isolation and biological characteristics of adipose-derived stem cells from human and rat. J Jinan Univ (Natural Sci Med Edition) 2015;36:324–9. doi: 10.1155/2019/1609876. [DOI] [Google Scholar]
  • 123.Cha H, et al. Stem cell-derived exosomes and nanovesicles: promotion of cell proliferation, migration, and anti-senescence for treatment of wound damage and skin ageing. Pharmaceutics. 2020;12(12):1135. doi: 10.3390/pharmaceutics12121135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Niu J, et al. MCP-1-induced protein promotes endothelial-like and angiogenic properties in human bone marrow monocytic cells. J Pharmacol Exp Ther. 2013;347(2):288–97. doi: 10.1124/jpet.113.207316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Chen H, et al. Pleiotropic roles of CXCR4 in wound repair and regeneration. Front Immunol. 2021;12:668758. doi: 10.3389/fimmu.2021.668758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Jin W, et al. Modelling of the SDF-1/CXCR4 regulated in vivo homing of therapeutic mesenchymal stem/stromal cells in mice. PeerJ. 2018;6:e6072. doi: 10.7717/peerj.6072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Planat-Benard V, et al. Plasticity of human adipose lineage cells toward endothelial cells: physiological and therapeutic perspectives. Circulation. 2004;109(5):656–63. doi: 10.1161/01.CIR.0000114522.38265.61. [DOI] [PubMed] [Google Scholar]
  • 128.Kim H, Hyun MR, Kim SW. The effect of adipose-derived stem cells on wound healing: comparison of methods of application. Stem cells international, 2019. 2019. DOI: 10.1155/2019/2745640. [DOI] [PMC free article] [PubMed]
  • 129.Yuan X, et al. Strategies for improving adipose-derived stem cells for tissue regeneration. Burns & Trauma. 2022;10. 10.1093/burnst/tkac028. [DOI] [PMC free article] [PubMed]
  • 130.Dung T, et al. Autologous adipose-derived stem cell (adsc) transplantation in the management of chronic wounds. Annals of Burns and Fire Disasters. 2021;34(4):343. [PMC free article] [PubMed] [Google Scholar]
  • 131.Sen S, et al. Genetic modification of human mesenchymal stem cells helps to reduce adiposity and improve glucose tolerance in an obese diabetic mouse model. Stem Cell Res Ther. 2015;6(1):1–14. doi: 10.1186/s13287-015-0224-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Takiishi T, et al. Vitamin D and diabetes. Rheumatic Disease Clinics. 2012;38(1):179–206. doi: 10.1016/j.ecl.2013.09.010. [DOI] [PubMed] [Google Scholar]
  • 133.Zatterale F, et al. Chronic adipose tissue inflammation linking obesity to insulin resistance and type 2 diabetes. Front Physiol. 2020;1607. 10.3389/fphys.2019.01607. [DOI] [PMC free article] [PubMed]
  • 134.Zhou C, et al. Autologous adipose-derived stem cells for the treatment of Crohn’s fistula-in-ano: an open-label, controlled trial. Stem Cell Res Ther. 2020;11(1):1–13. doi: 10.1186/s13287-020-01636-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Weisberg SP, et al. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Investig. 2003;112(12):1796–808. doi: 10.1172/JCI19246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Yu J, et al. Cell sheet composed of adipose-derived stem cells demonstrates enhanced skin wound healing with reduced scar formation. Acta Biomater. 2018;77:191–200. doi: 10.1016/j.actbio.2018.07.022. [DOI] [PubMed] [Google Scholar]
  • 137.Vanderstichele S, Vranckx JJ. Anti-fibrotic effect of adipose-derived stem cells on fibrotic scars. World J Stem Cells. 2022;14(2):200. doi: 10.4252/wjsc.v14.i2.200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Zhang J, et al. Spleen-derived anti-inflammatory cytokine IL-10 stimulated by adipose tissue-derived stem cells protects against type 2 diabetes. Stem Cells Dev. 2017;26(24):1749–58. doi: 10.1089/scd.2017.0119. [DOI] [PubMed] [Google Scholar]
  • 139.Shapiro AJ, et al. Insulin expression and C-peptide in type 1 diabetes subjects implanted with stem cell-derived pancreatic endoderm cells in an encapsulation device. Cell Rep Med. 2021;2(12):100466. doi: 10.1016/j.xcrm.2021.100466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Araujo DB, et al. Allogenic adipose tissue-derived stromal/stem cells and vitamin D supplementation in patients with recent-onset type 1 diabetes mellitus: a 3-month follow-up pilot study. Front Immunol. 2020;11:993. doi: 10.3389/fimmu.2020.00993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Lu J, et al. One repeated transplantation of allogeneic umbilical cord mesenchymal stromal cells in type 1 diabetes: an open parallel controlled clinical study. Stem Cell Res Ther. 2021;12(1):1–10. doi: 10.1186/s13287-021-02417-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Estrada EJ, et al. Combination treatment of autologous bone marrow stem cell transplantation and hyperbaric oxygen therapy for type 2 diabetes mellitus: a randomized controlled trial. Cell Transplant. 2019;28(12):1632–40. doi: 10.1177/0963689719883813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Al Demour S, et al. Safety and efficacy of 2 intracavernous injections of allogeneic Wharton’s jelly-derived mesenchymal stem cells in diabetic patients with erectile dysfunction: phase 1/2 clinical trial. Urol Int. 2021;105(11–12):935–43. doi: 10.1159/000517364. [DOI] [PubMed] [Google Scholar]
  • 144.Lee W-S, et al. Intra-articular injection of autologous adipose tissue-derived mesenchymal stem cells for the treatment of knee osteoarthritis: a phase IIb, randomized, placebo-controlled clinical trial. Stem cells translational medicine. 2019;8(6):504–11. doi: 10.1002/sctm.18-0122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Zhang Y et al. The Effect of Autologous Adipose-Derived Stromal Vascular Fractions on Cartilage Regeneration Was Quantitatively Evaluated Based on the 3D-FS-SPGR Sequence: A Clinical Trial Study BioMed Research International, 2022. 2022. DOI: 10.1155/2022/2777568. [DOI] [PMC free article] [PubMed]
  • 146.Freitag J, et al. Adipose-derived mesenchymal stem cell therapy in the treatment of knee osteoarthritis: a randomized controlled trial. Regen Med. 2019;14(3):213–30. doi: 10.2217/rme-2018-0161. [DOI] [PubMed] [Google Scholar]
  • 147.Carstens MH, et al. Treatment of chronic diabetic foot ulcers with adipose-derived stromal vascular fraction cell injections: Safety and evidence of efficacy at 1 year. Stem cells translational medicine. 2021;10(8):1138–47. doi: 10.1002/sctm.20-0497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Kumar H, et al. Safety and tolerability of intradiscal implantation of combined autologous adipose-derived mesenchymal stem cells and hyaluronic acid in patients with chronic discogenic low back pain: 1-year follow-up of a phase I study. Stem Cell Res Ther. 2017;8(1):1–14. doi: 10.1186/s13287-017-0710-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Kastrup J, et al. Cryopreserved off-the-shelf allogeneic adipose-derived stromal cells for therapy in patients with ischemic heart disease and heart failure—a safety study. Stem Cells Translational Medicine. 2017;6(11):1963–71. doi: 10.1002/sctm.17-0040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Huang K-C et al. Transplantation with GXHPC1 for liver cirrhosis: phase 1 trial Cell Transplantation, 2019. 28(1_suppl): p. 100S-111S. DOI: 10.1177/0963689719884885. [DOI] [PMC free article] [PubMed]
  • 151.Carstens M, et al. Sustained clinical improvement of Parkinson’s disease in two patients with facially-transplanted adipose-derived stromal vascular fraction cells. J Clin Neurosci. 2020;81:47–51. doi: 10.1016/j.jocn.2020.09.001. [DOI] [PubMed] [Google Scholar]
  • 152.Qayyum AA, et al. Autologous adipose-derived stromal cell treatment for patients with refractory angina (MyStromalCell Trial): 3-years follow-up results. J Translational Med. 2019;17(1):1–9. doi: 10.1186/s12967-019-2110-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Abumoawad A, et al. A phase 1a escalating clinical trial, autologous mesenchymal stem cell infusion for renovascular disease increases blood flow and the glomerular filtration rate while reducing inflammatory biomarkers and blood pressure. Kidney Int. 2020;97(4):793–804. doi: 10.1016/j.kint.2019.11.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Paitazoglou C, et al. Rationale and design of the european multicentre study on Stem Cell therapy in IschEmic non-treatable cardiac diseasE (SCIENCE) Eur J Heart Fail. 2019;21(8):1032–41. doi: 10.1002/ejhf.1412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Squassoni SD, et al. Autologous infusion of bone marrow and mesenchymal stromal cells in patients with chronic obstructive Pulmonary Disease: phase I randomized clinical trial. Int J Chronic Obstr Pulm Dis. 2021;16:3561. doi: 10.2147/COPD.S332613. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

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

Supplementary Materials

Supplementary Material 1 (509.8KB, docx)

Articles from Journal of Diabetes and Metabolic Disorders are provided here courtesy of Springer

RESOURCES