Next Article in Journal
Proteomic Analysis of the Breast Cancer Brain Metastasis Microenvironment
Next Article in Special Issue
Proteolytic Rafts for Improving Intraparenchymal Migration of Minimally Invasively Administered Hydrogel-Embedded Stem Cells
Previous Article in Journal
New Materials Based on Cationic Porphyrins Conjugated to Chitosan or Titanium Dioxide: Synthesis, Characterization and Antimicrobial Efficacy
Previous Article in Special Issue
Intranasal Delivery of Mesenchymal Stromal Cells Protects against Neonatal Hypoxic–Ischemic Brain Injury
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Regenerative Capacity of Adipose Derived Stem Cells (ADSCs), Comparison with Mesenchymal Stem Cells (MSCs)

1
Laboratoire Cellules Souches et Ingénierie Tissulaire, Centre Interface Applications Médicales CIAM, Université Mohammed VI polytechnique, Ben Guérir 43150, Morocco
2
Equipe d’Accueil (EA 7460), Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Université de Bourgogne Franche Comté, Faculté des Sciences de Santé, 7 Bd Jeanne d’Arc, 21000 Dijon, France
3
Laboratoire d’Epidémiologie et de Biostatique, Centre Interface Applications Médicales CIAM, Université Mohammed VI polytechnique, Ben Guérir 43150, Morocco
4
Département de Santé Publique et de Médecine Communautaire, Faculté de Médecine et de Pharmacie, Université Cadi Ayyad, Marrakech 40000, Morocco
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2019, 20(10), 2523; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20102523
Submission received: 15 April 2019 / Revised: 3 May 2019 / Accepted: 6 May 2019 / Published: 22 May 2019
(This article belongs to the Special Issue Stem Cell-Based Therapy)

Abstract

:
Adipose tissue is now on the top one of stem cell sources regarding its accessibility, abundance, and less painful collection procedure when compared to other sources. The adipose derived stem cells (ADSCs) that it contains can be maintained and expanded in culture for long periods of time without losing their differentiation capacity, leading to large cell quantities being increasingly used in cell therapy purposes. Many reports showed that ADSCs-based cell therapy products demonstrated optimal efficacy and efficiency in some clinical indications for both autologous and allogeneic purposes, hence becoming considered as potential tools for replacing, repairing, and regenerating dead or damaged cells. In this review, we analyzed the therapeutic advancement of ADSCs in comparison to bone marrow (BM) and umbilical cord (UC)-mesenchymal stem cells (MSCs) and designed the specific requirements to their best clinical practices and safety. Our analysis was focused on the ADSCs, rather than the whole stromal vascular fraction (SVF) cell populations, to facilitate characterization that is related to their source of origins. Clinical outcomes improvement suggested that these cells hold great promise in stem cell-based therapies in neurodegenerative, cardiovascular, and auto-immunes diseases.

Graphical Abstract

1. Introduction

Present within adipose tissue (AT), multipotent mesenchymal/stromal stem cells (MSCs) have been isolated in a reproducible manner and they have changed the thought that the scientific and medical communities have on AT [1]. These cells were isolated in the stromal vascular fraction (SVF) and they were similarly identified to those from bone marrow (BM), are plastic adherent, and typical MSCs, mainly called adipose derived stem cells (ASCs or ADSCs) [1,2]. These ADSCs possess adipogenic, osteogenic, chondrogenic, myogenic, cardiogenic, and neurogenic potential in vitro [3,4], and this plasticity and multipotency have triggered much related research in recent years. ADSCs are considered as tools for replacing, repairing, and regenerating dead or damaged cells. These cells were included in clinical investigations belonging to therapeutic strategies [4,5,6,7,8].
As for hematopoietic stem cells (HSCs), the development of therapeutic strategies presenting more efficacy and safety has become a real challenge in terms of invasive collection and administration, clinical outcomes, and treatment charges. In this fact, BM-, Umbilical Cord (UC)-MSCs, and ADSCs were used as new approaches for stem cell-based therapies in regenerative medicine. The use of enriched ADSCs as cell-Assisted Lipotransfer (CAL) is now largely accepted by plastic surgeons and it was first used to overcome fat necrosis and enhance fat grafting, especially in cosmetic remodeling [9,10,11,12,13]. Currently, pre-clinical and clinical applications of fat, SVF, or enriched ADSCs to treat different diseases are very promising. These investigations have concerned wound defects, vascular ischemia, bone regeneration, neurodegenerative diseases, cartilage tissue defects, cardiovascular injuries, and graft-versus-host disease (GVHD) [10,14,15,16,17,18,19,20,21,22]. These reports have strengthened justifying whether our current knowledge meet the real therapeutic applications of these cells.
Additionally, the widespread clinical use of ADSCs depends on the manner of using them. Being purified or being within their microenvironment is critical for their therapeutic outcomes and might ensure insights regarding the induced side-effects. Another issue is the shift of ADSCs use from autologous to allogeneic setting. The development of biotechnological techniques have improved the use of highly purified ADSCs and newly performed cells [23,24], being proposed for the allogeneic setting in the absence of available autologous cells. In this way, ADSCs might play the primary role in the regenerative medicine of the 21 centuries, provided that risk factors that are related to their manipulation and cryopreservation, their concentration, and route of administration are controlled and standardized.

2. Mesenchymal Stem Cells Characteristics

Adult stem cells have been isolated from BM and identified as MSCs [25,26]. Their self-renewal and differentiation abilities raise great interest in cell-based therapy. Alternate sources of these stem cells have been characterized, such as UC tissue, blood, liver, dental pulp, and skin, according to considerations in terms of collection procedure, cell quantity, immaturity, and cell profile [1,27,28,29,30,31]. Moreover, AT and the MSCs that it contains exhibited properties making them more efficient in regenerative medicine [1,30,32,33]. Comparative analysis of these MSCs suggested that, although they share common stem cell properties for MSCs, they markedly differ regarding their population number, proliferation rates and differentiation abilities, and clinical outcomes [34,35,36]. Being defined by the Mesenchymal and Tissue Stem Cell Committee of the International Society for Cellular Therapy (ISCT), MSCs showed marked expression of stromal markers CD73 and CD105, and they were negative for hematopoietic markers CD14, CD34, and CD45 [37]. Additionally, scientific statements that were on the fact that these cells should be plastic adherent, have fibroblast-like morphology and were maintained in culture for long periods. In addition, the tri-lineage potential for osteogenic, adipogenic, and chondrogenic differentiation should be demonstrable. However, cell populations satisfying all of these criteria are likely to be still heterogeneous. Therefore, the term MSCs is currently being used to represent both mesenchymal stem cells and multipotent mesenchymal stromal cells.
The most useful characteristic reported for MSCs is being the non-immunogenic profile. MSCs are widely reported as presenting the Major Histocompatibility Complex (MHC) I, but lacking the MHC II leading to the inactivation of T cells and immunosuppressive properties [38]. Many findings reported an additional immunomodulatory effect by impairing maturation and CD80 and CD86 expression level of dendritic cells, and proliferation and differentiation of human B cells [39,40]. Interestingly, MSCs secrete different chemokines and cytokines and extracellular matrix (ECM) proteins that are involved in various biological process, including hematopoiesis, angiogenesis, leukocyte trafficking, immune, and inflammatory responses [41,42,43]. These evidences hold great promise in MSCs-based therapy allowing for allogeneic transplantation without the need for immunosuppression [44,45,46].

3. Types of Stem Cells

There are various types of stem cells that come from different places in the body or they are formed at different times in our lives. Here, this study focused on the major types of stem cells

3.1. Bone Marrow-Mesenchymal Stem Cells (BM-MSCs)

BM harvesting is a highly invasive and painful procedure implying general anesthesia and many days for hospital care. BM-MSCs constitute a rare population, with only 0.002% of total stromal cell population [25], and their isolation depend on the patient status and the volume of aspirates. These cells were first identified as accompanying the primary HSCs [26], and they have been reported as multipotent stem cells that are able to differentiate into adipogenic, chondrogenic and are more likely to osteogenic progenitors [47,48]. The most discomforting use of these cells is that their differentiation potential, their number, and maximum life span significantly decrease with age [35,49].
In expansion culture, BM-MSCs stopped growing by passage 11–12, strongly expressed the senescence-related proteins p53, p21, and p16 and they were less clonogenic when compared to those from UC blood (UCB). Their inflammatory properties that were demonstrated in different cell-based therapies were exhibited by the large secretion of Interleukin-1 α (IL-1α), IL-6, IL-8, Tumor Necrosis Factor- α (TNF-α), and Tumor growth Factor-β1 (TGF-β1), but this activity remained lower than the other MSCs sources [5]. Lee et al. have reported 24 genes that were down regulated in BM-MSCs when compared to ADSC, but these genes represent less than 1% of those that were expressed by both cells [50]. BM-MSCs also contributed to tissue repair through their ability to migrate and home to injured sites, by modulating cell surface membrane marker’s expression [51].

3.2. Umbilical Cord-Mesenchymal Stem Cells (UC-MSCs)

These cells can be isolated from different parts of UC, including Wharton’s Jelly, cord lining, and peri-vascular region. However, almost studies used MSCs that were derived from cord blood and Wharton’s jelly [52,53]. As compared to BM, MSCs frequency was reduced in UCB [31], while their multipotency was maintained for longer periods [54].
UC-MSCs presented significantly lower expression of the senescence markers p53, p21, and p16. They were slightly positive for the embryonic stem cell marker Oct4, Nanog, Sox2, and KLF4, which indicated that these cells are more primitive than the adult stem cells [5]. Interestingly, a recent finding has demonstrated that MSCs that were isolated from male-derived Wharton’s Jelly presented higher gene expression levels of Oct4 and DNA-methyltransferase1 (DNMT1), thus associating stemness profile to gender differences [55]. Their ability to differentiate into the three mesodermic lineages (adipocytes, chondrocytes, and osteocytes) was debated [31,56]; however, the greatest proliferation rates and clonogenic activity, as well as higher chondrogenic differentiation, have been shown [54,57]. As their counterparts, the UC-MSCs were known to secrete growth factors, cytokines, and chemokines, improving different cell repair mechanisms [5]. Nevertheless, their expanded use is specifically due to their low expression of MHC class II antigens, inhibition of T cell, B cell, and NK cell proliferation, and the inactivation of monocytes and dendritic cells [36,58]. Transplanted into animal model, these cells were also able to differentiate into spinal cord tissue with an increasing number of GAP43+ fibers and a higher amount of spared gray in a dose-dependent and repeated manner [59]. Parkinson’s disease, myocardial infarction, and diabetes have also benefited from the differentiation ability of these cells [60,61,62].

3.3. Adipose Derived Stem Cells (ADSCs)

Different terminologies were proposed for these cells in the literature: processed lipoaspirates, adipose-derived adult stem cells, adipose mesenchymal stem cells, adipose-derived stromal/stem cells, and adipose stromal cells. Therefore, the International Federation of Adipose Therapeutics (IFATS) adopted the ADSCs nomenclature for more uniformity.
These cells are currently isolated from the subcutaneous AT [63], which allows for them to be rapidly acquired in large numbers and with a high cellular activity [1,2,64]. Their frequency harbors nearly 2% in its stromal vascular fraction and it is considered to be the greatest one in all tissues [65,66], sometimes nearing 30% [67]. The scientific consensus, the IFATS, and the ISCT agree on the lower expression level of stromal associated markers CD13, CD29, CD44, CD73, CD90, CD105, and CD106 in the SVF population on the contrary to cultured ADSCs. These expressions were consistently pronounced by late ADSCs culture passage [47,57,68]. The specific expressions of CD10, CD36, and CD106 can distinguish them from those of BM [32,69]. ADSCs research, being predominantly carried out using culture-expanded cells, has rather led to a recent acceptance of CD34 as a marker for isolated ADSCs. Thus, there remain interesting aspects of CD34 biology to be explored and understood. A relationship between this CD34 marker and hypoxia has also been reported. CD34 might be a niche-specific marker of progenitors, and hypoxia is related to the maintenance of adult stem cells [70]. Sengenès et al. has demonstrated that the human SVF cell populations presenting the CD34 were enriched in ADSCs, on the contrary to BM-MSCs [71]. However, the expression of this antigen was only reported in the early culture passage [72]. In addition, the CD34+ subpopulation represented at least 20% of freshly isolated SVF [73]. Another surface antigen Stro-1, which is the classic BM-MSCs associated antigen, was differently reported on ADSCs within the literature [2,74]. Furthermore, one report has indicated that ADSCs might be specifically identified by the CD271 marker [75], where the expression have been maintained in elderly people and associated with high proliferative and differentiation abilities [76]. ADSCs also secrete trophic factors that regulate cell growth and display lipolytic responses to β-adrenergic agents and activated protein kinase phosphorylation in tumor necrosis [77,78].
ADSCs from younger donors exhibited a higher proliferation rate when compared to elderly ones, but the differentiation capacity was maintained with aging [79], thus having advantages on BM-MSCs. ADSCs also maintained their potential to differentiate into cells of mesodermal origin and they are commonly known for their low immunogenicity and modulatory effects [33,80]. Less than 1% of them expressed the HLADR protein on their surface, leading to immunosuppressive effects and making them suitable for clinical applications in allogeneic transplantation and in therapies for the treatment of resistant immune disorders [37,66]. Even UC-MSCs were presented as more primitive than ADSCs [5], with the latter have proven their superiority regarding availability and suitable cell quantities. As for BM, ADSCs cell growth stopped in passage 11–12, but presented the lower population doubling than BM and UCB-MSCs, hence being shorter but more highly proliferative than UC-MSCs [5]. These ADSCs features altogether seemed to promote them in tissue repair, where cell proliferation, angiogenesis, and anti-inflammatory processes were expected to occur rapidly in damaged sites (Figure 1).
From another point of view, the ADSCs were firstly identified by their ability to differentiate into mesodermic lineages. However, many findings agreed on latter, with their differentiation into ectodermic and endodermic germinal layers exhibiting a potential advantage in developing tissue engineering strategies in regenerative medicine. Even ADSCs were more susceptible to differentiate into the adipogenic lineage when compared to BM- and UC-MSCs, their multipotency was appreciated for ectodermic and endodermic tissue repair [6,30,47,81] (Figure 2). Nevertheless, their functionality was impacted by long term expansion culture and cryopreservation [32,64,82], which suggests that further studies should be performed to standardize ADSCs manipulation for clinical use.

4. Current Approaches in Adipose Derived Stem Cells (ADSCs)-Based Therapeutic Applications

Pluripotent or resident stem cells participle and induce all of the biological mechanisms that are involved in normal process of tissue regeneration and functionality during the whole life. However, MSCs seemed to be more adaptable to act in situ, thus leading to locally support degenerative diseases.
ADSCs are considered to be tools for replacing, repairing, and regenerating dead or damaged cells. These cells were included in clinical investigations that belong to therapeutic strategies and fulfill the general accepted criteria for cell-based therapies, making them the most adapted cell to play the primary role in regenerative medicine. Indeed, ADSCs are found in abundant quantities and they are harvested by a minimally invasive procedure, can differentiate into multiple cell lineages in a regulatory and reproducible manner and they are safely transplanted at the both autologous and allogeneic setting. Their possible manufacturing in accordance with current Good Manufacturing Practices (cGMP) guidelines is in progress by resolving different technical difficulties.
Clinical studies generally used the whole SVF, isolated ADSCs (alone or in combination with biomaterials), even being autologous or allogeneic. Table 1 summarizes the published results of the most reported clinical applications of ADSCs in parallel to those of BM-and UC-MSCs. To avoid misunderstandings on the use of SVF and its derived ADSCs population and their relative beneficial characteristics, it is important to distinguish between these two populations and, hence, only ADSCs applications are viewed in this table. Previous reviews have already reported the performing clinical applications of autografts that are enriched with ADSCs and or SVF and their relative benefit use in regenerative medicine [83].
Studies in Table 1 altogether reported significant differences in transplanted cells, the route of their administration and their clinical status. ADSCs represented the most benefit-to-risk in skin, onco-hematology, bone, and cartilage applications when compared to BM- and UC-MSCs. The use of these cells was related on multiple purposes, including immunomodulation effects (multiple sclerosis MS, fistula, acute and chronic GVHD, diabetes mellitus I, Crohn’s diseases), angiogenic potential (ischemia, scars, wound repair), and differentiation potential (arthritis, cardiac and spinal injury, bone regeneration). The delivery method of stem cell implantation is an important determinant for effective cell therapy.

4.1. The Effects of ADSCs on Wound Healing and Skin Regeneration

The stem cell-based therapies in wound healing were firstly performed with autologous cultured epidermal autografts (CEA) as an epidermal substitute. The addition of cellular components to these skin substitutes have led to a satisfying skin esthetic appearance and function. The use of MSCs from different sources has emerged as a unique alternative for wound management strategies to various skin-related disorders to have a near-natural skin in terms of appearance, texture, color, and metabolic properties. Regarding their accessibility, high cell number availability, and their non-invasive collection, ADSCs have been proposed to overcome CEA limitations as recurrent open wounds, long term fragility, and increased scar contractions by inducing extensive ECM protein production [7]. Their ability to restore chronic wounds was increasingly highlighted through ECM secretion, leading to proliferation and remodeling phases of wound healing [8]. These cells have been shown to promote fat tissue survival and, together with free fat, were becoming a real alternative to soft tissue augmentation surgery, as in breast augmentation and facial tissue defects [10,11,12,13] (Table 1).
ADSCs induced tissue reconstitution, thus correcting tissue defects and improving skin regeneration, helped cicatrization and modulated inflammation, thereby promoting skin healing [16,17]. The growth factors and anti-inflammatory cytokines that they secrete prevented cell apoptosis and induced neo-angiogenesis, particularly in treating critical limb ischemia [90]. Many groups have used autologous or allogenic ADSCs in the treatment of burns, their use alone, or in combination with epidermal graft have improved skin engraftment.
Some therapeutic advances have been showed when using BM- or UC-MSCs to achieve the complete remission for patients that were treated for ulcers, scars, and burns [84]. However, the ability of ADSCs to act through higher proliferation, differentiation, and paracrine manner has proven their superiority for widespread application in this field.

4.2. The Effects of ADSCs in Auto-Immune Disorders

ADSCs have been applied for many autoimmune disease’s treatments. These cells were shown safe and efficient for Systemic Lupus Erythematosus (SLE), systemic sclerosis (SS), Sjörgren’s syndrome, scleroderma, and Crohn’s diseases. Patients with Crohn’s diseases presented closed fistula after inoculation of the cells [141,142,143] (Table 1), and disease improvement, combined with patient’s safety, have been reported with no serious adverse events being estimated for short or long-term follow-up. At the other hand, a decrease in disease severity was observed after BM and UCB-MSCs treatment and this effect appeared to be up-estimated perhaps for the small sample size or the short-term follow-up [132,133,134,136]. However, many of the conducted studies reported incidences of infections and malignancies. These adverse events might not be attributable to cell infusion, but rather to the impaired immune system and the use of immune-suppressers and corticosteroids as autoimmune medications [196]. Nevertheless, these cells were reported to be the most beneficial in treating and preventing Graft-Versus-Host Disease (GVHD) resulting from HSCs transplantation [167].
ADSCs approaches appeared to be safer and more efficient in terms of cases of side effects reported. Designing MSCs transplantation protocols while using ADSCs therapeutic purposes might be predominantly selective for patients with Sjögren’s syndrome, SLE, polymyolitis, and Crohn’ disease [15,143,144] (Figure 3). The immunomodulating potential of ADSCs has also benefitted from type I diabetes mellitus, where new cells secreting insulin were regenerated [145].

4.3. The Effects of ADSCs on Hematological Disorders and Graft-Versus-Host Disease (GVHD)

The treatment of GVHD has been the most widely investigated with BM-MSCs after allogeneic HSCs transplantation and or lymphocyte infusion (Table 1). In adult patients and children with steroid-resistant acute GVHD, the infusion of allogeneic BM-MSCs resulted in the complete response and overall survival in most patients after UCB and HSCs transplantation without any side-effects during or immediately after MSCs infusion [167,168], while better outcomes were also observed with early BM-MSCs administration [169] (Figure 3). In malignancies, BM-MSCs were safe and successful in improving hematopoietic engraftment [170,197], and similar results have been reported with UCB-MSCs in pediatric and adult patients after HSC transplantation [175,176]. Accelerating hematological recovery was also found after the co-infusion of autologous blood HSC and BM-MSCs in patients presenting breast cancer that were receiving high-dose chemotherapy [198].
ADSCs, as their counterparts, were also reported to support HSCs engraftment preventing acute and chronic GVHD [14]. These cells were found to support the complete differentiation and production of mature myeloid and B lymphoid cells, but they were unable to maintain long-term survival and self-renewal of HSCs, suggesting that ADSCs could be only efficient in short term reconstitution of hematopoiesis [181]. Similar results were found in supporting the in vitro and in vivo hematopoiesis in mice [199].

4.4. The Effects of ADSCs on Bone/Cartilage Repair

Unlike other adult tissues, which generate scar tissue in response to injury, the skeleton undergoes regenerative healing, forming new bone [200]. In recent years, there has been growing interest in the use of undifferentiated progenitor cells for tissue engineering, owing to their ability to expand in culture and to differentiate into multiple cell lineages when cultured under specific growth conditions. Owing to these characteristics, adult stem cells from different tissues have been used in various cartilage and bone regenerations. ADSCs have shown significant chondrogenic potentials for use in tissue engineering approaches [201].
The widely accepted use of ADSCs is, unsurprisingly, bone and cartilage regeneration and repair. Although possessing high osteogenic differentiation potency in vitro, BM-MSCs osteogenic repair in vivo remained limited, their use for bone repair is now progressively replaced with ADSCs. As the same way, limited evidences were observed in regards to the clinical benefit of UCB-MSCs for articular cartilage repair and only one clinical trial has been performed [149] (Table 1).
Alone, or combined with scaffold, ADSCs have proven efficiency in restoring maxillary defects, accompanied with apparent bone regeneration [164]. Post-traumatic calvarial defects were also treated with autologous ADSCs and calvarial continuity with bone regeneration were obtained [165]. Their ability to secrete anti-inflammatory factors and decrease pro-inflammatory responses also makes them very attractive and efficient in the treatment of inter-vertebral disc damages, rheumatoid arthritis, osteoarthritis, and tendon damages [19,22,150,151,152,153,154]. The therapeutic benefit of ADSCs was more pronounced in cartilage defects treatment, and more case treatments were reported in the literature [18].

4.5. The Effects of ADSCs on Cardiovascular and Muscular Diseases

Adult skeletal muscle possesses extraordinary regeneration capacities. The muscle satellite cells are responsible for the postnatal growth and major regeneration capacity of adult skeletal muscle. The evidence indicates that skeletal muscle influences systemic aging. An age-dependent decline in skeletal muscle mass, strength, and endurance during the aging process is a physiological development and several factors may exacerbate this process. Capillaries are an integral part of the mechanism underlying this close matching between the blood flow and metabolism of skeletal muscle mass. In contrast with the skeletal muscle, limited regenerative capacity characterizes the myocardium. The neonatal mammalian heart can substantially regenerate after injury through cardiomyocyte proliferation until postnatal day 7. One of the factors that was shared by organisms was the ability able to regenerate the heart is the oxygenation state. It has been hypothesized that the transition to the oxygen-rich postnatal environment is the upstream signal, which results in cell-cycle arrest of cardiomyocytes [202].
Cardiac muscle has limited proliferative capacity, and regenerative therapies are in high in demand as a new treatment strategy. Fibroblasts can be directly reprogrammed to cardiomyocytes by overexpressing a combination of three cardiac-specific transcription factors (Gata4, Mef2c, Tbx5) [203]. Interestingly, it was reported that somatic cells can be directly reprogrammed to alternative differentiated fates without first becoming stem/progenitor cells. Evidence that the exposure of human skin fibroblasts to a Radio Electric Asymmetric Conveyer (REAC) provided commitment toward cardiac and skeletal muscle lineages [204].
Efficient methods to induce differentiation to cardiomyocytes that generate homogenous populations of cardiomyocytes of sufficient quality are a prerequisite for new applications. MSCs can be isolated from various human tissues, with multipotent and immunomodulatory characteristics to help in damaged tissue repair. Clinical trials have demonstrated the feasibility and efficiency for CVD therapy from diverse origins and tissue-derived MSCs [205]. In the field of the new strategies of obtaining cardiac differentiation, stem cell-based therapy has emerged as a promising therapeutic approach for treating cardiovascular diseases [206]. Evidence suggests that secreting paracrine factors prominently mediate the therapeutic effects of transplanted stem cells, importantly, microRNAs (miRNAs) are present in the secreted exosomes [207]. MSC-derived exosomal miRNAs exert cardioprotective effects through the induction of angiogenesis in ischemic heart [208].
Numerous studies employing small animals have adopted intramyocardial injections. However, these injections still show the limitation of cellular engraftment in the infarcted myocardium. Many attempts have been proposed to overcome this obstacle, like a few novel hydrogels have been introduced as potential delivery methods for ADSCs. MSCs were reliably accepted to differentiate into the cardiomyocytes in vitro [209] and they hold great promises in regenerating cardiomyocytes in the cases of myocardial infarction. Autologous and allogenic BM-MSCs that were transplanted into patients with myocardial infarcts showed no adverse side effects and increased the left ventricular function, decreased cardiac arrhythmias, and improved myocardial function [183,184,185] (Table 1). Other studies used UCB-MSCs as intravenous infusion into patients with heart failure, reduced ejection fraction, and improvement of the left ventricular function were attained in these patients [187,188].
Recently, direct cardiac reprogramming has emerged as a novel technology to regenerate damaged myocardium by directly converting endogenous cardiac fibroblasts into induced cardiomyocyte-like cells to restore cardiac function. Fibroblasts replace dead cardiomyocytes, leading to the formation of fibrosis and myocardial remodeling. There are studies regarding the interaction between ADSCs and fibroblasts. ADSCs conditioned media promotes fibroblast proliferation, which suggests the paracrine activation of fibroblasts by ADSCs. The same fibroblasts that were cultured in ADSCs conditioned media were found to secrete increased amounts of type I collagen. These findings suggest that these interactions might play a major role in myocardial protection.
In another point of view, ADSCs might be for a great cardiovascular interest and these cells were investigated in many clinical trials in last decade. ADSCs were directly administrated into myocardial tissue of patients with MI or ischemic heart failure without related events and their use was safe and efficient in improving cardiac function [191,192,193]. In animal models, ADSCs also have proven efficiency, even when administered by intracoronary, transendocardial, intramyocardial, or intravenous methods [66,210].
In skeletal muscle, the regenerating ability of MSCs is still controversial and few human studies were reported. However, clinical promising reports have shown the improvement of muscle activity in patients presenting muscle dystrophies after the infusion of UC-MSCs [189,190]. However, the paracrine activity of these MSCs was more expected to improve cardiac function by enhancing angiogenesis and anti-apoptosis, rather than direct differentiation towards cardiomyocytes [211,212].
In animal models, ADSCs can be differentiated into skeletal muscle cells and improve the ECM collagen VI deficiency in the congenital muscular dystrophy and in the mice model of Duchene Muscular Dystrophy (mdx mice) [213,214,215,216].

4.6. The Effects of ADSC on Neuro-Degenerative Diseases

Conventionally, over a long time, nervous system tissue has been considered to be problematic in regenerating, because mature neural cells do not proliferate or differentiate. It is still unclear how neural stem cells are actively maintained throughout life, and what are the cellular interactions and molecular cascades. In the adult mammalian brain, there are at least three areas that are neurogenic and contain a reservoir of neural stem cells: the subgranular zone in the hippocampal dentate gyrus, the subventricular zone around the lateral ventricles, and the hypothalamus. Recently, several components of the neural stem cell niche have been identified, which regulate neural stem cell activity by supplying various signals [217]. ADSCs could transdifferentiate into neuron-like cells that present multiple neuronal properties, such as synaptic transmission, action potential, secretion of dopamine and neurotrophic factors, and spontaneous postsynaptic current. Some recent studies indicate that miRNAs might regulate the neuronal-like differentiation of MSCs. MiR-21 has been confirmed to directly contribute to ADSCs differentiation [218]. For ADSCs to become ideal for neurological disease therapy, they must generate sufficient number of functional and high-quality neural cells. During trans-differentiation into neural cells, xenobiotics or specific factors and the corresponding partial methylation or acetylation of genomic regions and the activation of further trans-differentiation processes stimulate ADSCs.
These ADSCs and others MSCs have been proposed for novel therapy to various neurological disorders, such as Alzheimer’s and Parkinson’s diseases, stroke, amyotrophic lateral sclerosis (ALS), Huntington’s disease, spinal cord injury, traumatic brain injury, and MS. These, altogether, cells have proven their efficiency to pass through the blood brain barrier even intravenously transplanted. Neuronal morphological characteristics and markers were observed after cell infusion, either by differentiation to neuron-like cells [81] or fusion with endogenous cells [219]. However, their therapeutic benefits appeared to be induced through the secreted proteins [20,21]. Proteomic analysis have confirmed that BM- and UC-MSCs conditioned media induced neuronal differentiation, neurite outgrowth from dorsal root ganglion explants, while ADSCs showed higher axonal growth [20]. Particularly, ADSCs paracrine activity was not solely responsible for the elaboration of nerve growth factors and neurotrophic mediators that are involved in nerve regeneration [21], which adds to the complexity of microenvironment and nerve progenitors’ interactions. Used at autologous or allogenic setting, at low or high infused doses and with multiple infusions, neuroprotective and immunomodulation effects were related to MSCs from the three sources, which leads to a decrease in pathological features and an improvement of the disability of neurological disorders (Table 1).
Therapeutic properties of BM-MSCs have put profit firstly to patients suffering from Parkinson’s disease [220] and these encouraging results pave the way to others current controlled phase study. On another side, safety and clinical improvement have been shown in patients presenting ALS and stroke when using modified BM-MSCs [99,221]. A recent work investigating the BM-MSC-derived neural progenitor on patients with MS has established safety and tolerability without any serious adverse effects [100,135]. Using another alternative, conditioned media from BM-MSCs cultures, followed by BM-MSCs transplantation, potentiated clinical improvement and this ameliorative effect appeared to be dependent on IL-6, IL-8, and vascular endothelial growth factor (VEGF) conditioned the media levels [101]. At the same way, Li et al. has already demonstrated a shift from Th1 to Th2 immunity in human UC-MSCs treated patients with MS [222]. The mainly mechanisms underlying the therapeutic benefit observed for ADSCs are undeniably and irrevocably their paracrine effect after transplantation. The secreted factors might directly act on neuroprotection or through immunomodulation impacting the expression or secretion of many mediators that are involved in angiogenesis, synaptogenesis, gliogenesis, and neurogenesis [223,224,225,226,227]. Other therapeutic strategies might concern cell combination for more neurological function improvement [228] or the use of VEGF, Angiogenin (ANG), and TGF-β as predictive biomarkers for cell therapy effectiveness and for choosing patients [87].
Other ADSCs therapeutic concerns were proposed for retinal degenerative diseases and renal transplantation. These approaches, still in earlier stages of development, were respectively investigated for ADSCs differentiation potency and immunosuppressive ability in tissue repair.

4.7. The Effects of ADSCs on Radiation Injuries

The literature is rich in terms of clinical applications of ADSCs to cure radiation injury. There is an increase of radiation injuries on wounds and other organs with the widespread use of radiotherapy, interventional radiological, or cardiological procedures [229,230,231,232,233,234,235,236]. Chronic radiation wounds usually cannot be treated with conventional methods, such as flap surgery or skin grafting because of tissue ischemia and fibrosis [237]. The ischemia is due to inadequate vasculature and incompetent vessels in irradiated tissues [238,239]. The radiated skin shows erythema and abnormal pigmentation. Once a radiation wound is present, it becomes complicated with necrosis, infection, and fibrosis in various organs, such as the heart. These chronic radiation injuries could be improved by sufficient blood supply to the tissues. On the other hand, combined radiotherapy and chemotherapy have represented a major advance in the therapeutic management of cancer therapy. However, the combination of doxorubicin (DXR) and cardiac irradiation could precipitate the unexpected expression of congestive heart failure [240,241].
ADSCs therapy is promising in the treatment of chronic radiation wounds and myocardial diseases. Their administration leads to improved blood perfusion and capillary density in irradiated wounds [235]. The viability of irradiated skin flaps increased when treated with ADSCs injection in correlation with increased vascularity in the flaps injected with ADSCs. At the cellular level, the ADSCs were shown to stimulate fibroblasts proliferation and increase the expression of several cytokines, such as VEGF [236].

5. Issues Related to Autologous and Allogeneic Clinical Use of ADSCs

The stemness characteristic, plasticity, and robustness that were attributed to MSCs make them the most attractive adult stem cells in regenerative medicine. As the number, frequency, and differentiation capacity of BM-MSCs negatively correlate with age [35,49], elderly patients could not have clinically efficient autologous stem cells, suggesting that an allogeneic approach would be required. The development of allogeneic approach means that ADSCs will be isolated from a volunteer donor, expanded ex-vivo, and cryopreserved as suitable cell product until need for tissue repair. Thus, cells that were obtained from a single donor can be used to treat thousands of unrelated patients. AT from HLA identical siblings, haplo-identical relatives, or HLA-screened healthy volunteers is now considered to be a successful alternative and it might be the best choice for collection and storage until used in HLA-matched patient.
Even AT has important implications in the development of stem cell bank [242,243,244], the interest raises by ADSCs have improved two specific questions: first, their use must be practical and effective (high numbers of cells are needed); and second, the clinical outcomes should be identical to the expected use. A major advantage is that of ADSCs could be maintained for up to 24–48 h within lipoaspirates [82], cryopreserved before separation and seeding in culture with a stable and efficient ability to proliferate and differentiate [245,246], while fresh BM and UC tissues are necessary for the collection and deriving of MSCs. Consequently, the banking of allogeneic ADSCs remains full of hope for the future regenerative medicine. However, to be used on a widespread basis, efficient, simple, and especially safe methods should be provided to ensure the assurance of available cell quantities of no contaminated and functional ADSCs and it performed accordingly to cGMP. Specific insights are now focused on how these procedures should be realized with regards to their therapeutic benefits [78,247].
Many studies have been focused on this field in different diseases, but the availability of higher quantities of qualified ADSCs for banking is still debated due to the lack of best and standardized parameters of cryopreserved cells. Hence, the guidance policies covering all the processes involved in ADSCs banking (donor’s recruitment, manipulation, banking procedures, release, and defined qualification testing after thawing) would guarantee multiple ADSCs patient treatments avoiding repeated liposuctions.
We can also speculate that the allogenic model would be more attractive for cell therapy industry. However, autologous cells, rather than allogenic ones, mostly improved cell-based immunotherapy.
Despite the major interest arising concerning their use in therapy, the last decade still lacked commercial GMP-licensed products. The proposed ADSCs products appeared to be well tolerated in clinical phase I studies, provided the dosage data in phase II, and some of them were in phase III, such as in cardiovascular and autoimmune diseases. Besides academic and university communities, some biotechnological firms are now covering clinical ADSCs research for many areas. Developed laboratory ADSCs preparations or patented formulations of adult stem cells are available to undergo and evaluate the safety of different MSCs treatment for many diseases. Osiris Therapeutics Inc., Mesoblast Limited is committed to develop stem cell-based treatment to Crohn’s diseases, cardiovascular diseases, diabetes, orthopedic disorders, and radiation exposure [248]. These human BM expanded MSCs, called “Prochymal”®, appeared to be safe and efficient and improve the treatment of acute GVHD [171]. However, a major concern giving rise to ethical and justice considerations is the cost of these biotechnological products. ADSCs and derivatives might also be costly and labor-intensive, thus increasing the cost of making them available. The way that clinical trials are conducted should aim to reduce the unfair disparities in access.

6. Summary and Conclusion

ADSCs represents many therapeutic challenges in terms of origin, type, and the manner to use them, different recent investigations pave the way to their successful therapeutic use in tissue repair. More insights into standardizing technical use are warranted to evaluate the in-depth efficacy and safety of ADSCs-based therapy and evaluate the benefit-to-risk ratio in clinical applications. The beneficial effects of stem cells, and there with the paradigm of tissue regeneration, may not be restricted to cellular restoration, but may also be related to the transient paracrine actions of the cells.
These cells are phenotypically identical to BM- and UC-MSCs when using a panel of 22 surface antigens [35], the gene expression signature seems to demonstrate an up-regulation of 24 genes in ADSCs when compared with BM-MSCs [50], and several hundred expressed sequence tags [35]. The variations that were described between ADSCs and BM-MSCs regarding differentiation potential and proliferation capability may also reflect the incidence of the micro-environment of their tissue of origin. This fact might answer the differences reported in their ex-vivo expansion and promising clinical benefit. Indeed, the promising result that was observed when transplanting SVF might be refereed to its heterogeneity and the activation of the related microenvironment that was constituted by subpopulations of hematopoietic and no hematopoietic and endothelial progenitors and cells. Hence, insights into host-related factors, including local environment and optimal timing are needed to master factors governing the fate of ADSCs after infusion. Patient’s associated factors should also be considered in designing ADSCs based therapy, due to their influence on the number and functional behavior of expanded cells [249,250].
We should emphasize ADSCs purity control and identify potency assays to ensure their regenerative potency. These factors imply the identification of reproducible and consistent standardized cell preparations that are suitable for use in a large scale. Actual application may need more expanded numbers of cells and variabilities reported in clinical outcomes might be related to differences in the infused cell amounts.
All these considerations would act to reinforce their promising therapeutic benefit for tissue repair. As expected by Gimble et al., may be one day people will altruistically participate in “fat drives.” for fat donation as observed in “blood drives” [251]. Clinical grade ADSCs applications should be performed in compliance with appropriate standards and protocols relative to cellular therapies from facility and GMP compatible reagents to the clinical grade materials. Therapeutic development of HSC transplantation witnesses the success of the global standardized processes and the generalization of guidelines regarding the clinical application of these cells. ADSCs-based therapies could shortly follow the example of HSC’s therapy process.
The application of ADSCs is greater than that of BM-MSCs in regenerative medicine, because ADSCs have the need for easier technique for isolation when compared to BM-MSCs, because the technique for isolating ADSCs is easier and, consequently, they can be used in a greater number than BMSCs. The clinical use of cell-based therapies in regenerative medicine has to fulfill the minimal requirements, like any other medical treatment. Still, before ADSCs can be applied for routine clinical applications, many open questions that are related to ADSCs need to be solved. Several reports have suggested that ADSCs promote the proliferation of cancer cells that means ADSCs may stimulate the growth of pre-existing tumors. Consequently, the treatment with ADSCs in cancer diseases is not presently recommended
In conclusion, considerations should be made regarding the scientific and medical challenges for the requirement of widely use of banked ADSCs. For the medical ones, health care professionals need to be educated for the correct use and applications of ADSCs. At the other scientific side, challenges should improve the appropriate quality assurance and control in compliance with cGMP.

Author Contributions

All authors participated in the research and writing of this manuscript.

Funding

This work was supported by the University Mohammed VI polytechnic, Ben Guérir, Morocco.

Acknowledgments

We wish to think Alexandre Meloux for his assistance in references preparation.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ADSCsadipose derived stem cells
ALSamyotrophic lateral sclerosis
ATadipose tissue
b-FGFbasic Fibroblast Growth Factor
BMbone marrow
CCL-2Chemokine C-C Motif Ligand-2
cGMPcurrent good manufacturing practices
ECMextracellular matrix
EGFEndothelial Growth Factor
G-CSFGranulocyte-Colony Stimulating Factor
GDF-11, -15Growth Differentiation Factor-11, -15
GM-CSFGranulocyte Monocyte-Colony Stimulating Factor
GVHDgraft versus host disease
HSCshematopoietic stem cells
IFATSInternational Federation of Adipose Therapeutics
IGFInsulin Growth Factor
IL-1, -6, -10Interleukin-1, -6, -10
ISCTInternational Society for Cellular Therapy
KGFKeratinocyte Growth Factor
MSmultiple sclerosis
MSCsmesenchymal stem/stromal cells
NGFNerve Growth Factor
PDGFPlatelet Derived Growth Factor
PEDFPigment Epithelium Derived Factor
SLEsystemic lupus erythematosus
SSsystemic sclerosis
SVFStromal vascular fraction
TGF-βTumor growth factor-β
TLR2, 4Toll-like Receptor 2, 4
TNF-αTumor necrosis factor-α
UCumbilical cord
UCBumbilical cord blood
VEGFVascular Endothelial Growth Factor

References

  1. Zuk, P.A.; Zhu, M.; Mizuno, H.; Huang, J.; Futrell, J.W.; Katz, A.J.; Benhaim, P.; Lorenz, H.P.; Hedrick, M.H. Multilineage cells from human adipose tissue: Implications for cell-based therapies. Tissue Eng. 2001, 7, 211–228. [Google Scholar] [CrossRef]
  2. Zuk, P.A.; Zhu, M.; Ashjian, P.; De Ugarte, D.A.; Huang, J.I.; Mizuno, H.; Alfonso, Z.C.; Fraser, J.K.; Benhaim, P.; Hedrick, M.H. Human adipose tissue is a source of multipotent stem cells. Mol. Biol. Cell 2002, 13, 4279–4295. [Google Scholar] [CrossRef]
  3. Vermette, M.; Trottier, V.; Ménard, V.; Saint-Pierre, L.; Roy, A.; Fradette, J. Production of a new tissue-engineered adipose substitute from human adipose-derived stromal cells. Biomaterials 2007, 28, 2850–2860. [Google Scholar] [CrossRef] [PubMed]
  4. Choi, Y.S.; Dusting, G.J.; Stubbs, S.; Arunothayaraj, S.; Han, X.L.; Collas, P.; Morrison, W.A.; Dilley, R.J. Differentiation of human adipose-derived stem cells into beating cardiomyocytes. J. Cell. Mol. Med. 2010, 14, 878–889. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Jin, H.J.; Bae, Y.K.; Kim, M.; Kwon, S.J.; Jeon, H.B.; Choi, S.J.; Kim, S.W.; Yang, Y.S.; Oh, W.; Chang, J.W. Comparative analysis of human mesenchymal stem cells from bone marrow, adipose tissue, and umbilical cord blood as sources of cell therapy. Int. J. Mol. Sci. 2013, 14, 17986–18001. [Google Scholar] [CrossRef] [PubMed]
  6. Pittenger, M.F.; Martin, B.J. Mesenchymal stem cells and their potential as cardiac therapeutics. Circ. Res. 2004, 95, 9–20. [Google Scholar] [CrossRef]
  7. Trottier, V.; Marceau-Fortier, G.; Germain, L.; Vincent, C.; Fradette, J. IFATS collection: Using human adipose-derived stem/stromal cells for the production of new skin substitutes. Stem Cells 2008, 26, 2713–2723. [Google Scholar] [CrossRef]
  8. Hyldig, K.; Riis, S.; Pennisi, C.P.; Zachar, V.; Fink, T. Implications of Extracellular Matrix Production by Adipose Tissue-Derived Stem Cells for Development of Wound Healing Therapies. Int. J. Mol. Sci. 2017, 18, 1167. [Google Scholar] [CrossRef] [PubMed]
  9. Eto, H.; Kato, H.; Suga, H.; Aoi, N.; Doi, K.; Kuno, S.; Yoshimura, K. The fate of adipocytes after nonvascularized fat grafting: evidence of early death and replacement of adipocytes. Plast. Reconstr. Surg. 2012, 129, 1081–1092. [Google Scholar] [CrossRef] [PubMed]
  10. Koh, K.S.; Oh, T.S.; Kim, H.; Chung, I.W.; Lee, K.W.; Lee, H.B.; Park, E.J.; Jung, J.S.; Shin, I.S.; Ra, J.C.; et al. Clinical application of human adipose tissue-derived mesenchymal stem cells in progressive hemifacial atrophy (Parry-Romberg disease) with microfat grafting techniques using 3-dimensional computed tomography and 3-dimensional camera. Ann. Plast. Surg. 2012, 69, 331–337. [Google Scholar] [CrossRef] [PubMed]
  11. Wang, L.; Lu, Y.; Luo, X.; Fu, M.G.; Hu, X.; Dong, H.; Fan, Z.H. Cell-assisted lipotransfer for breast augmentation: A report of 18 patients. Chin. J. Plast. 2012, 28, 1–6. [Google Scholar]
  12. Yoshimura, K.; Sato, K.; Aoi, N.; Kurita, M.; Hirohi, T.; Harii, K. Cell-assisted lipotransfer for cosmetic breast augmentation: Supportive use of adipose-derived stem/stromal cells. Aesthetic Plast. Surg. 2008, 32, 48–55. [Google Scholar] [CrossRef] [PubMed]
  13. Yoshimura, K.; Asano, Y.; Aoi, N.; Kurita, M.; Oshima, Y.; Sato, K.; Inoue, K.; Suga, H.; Eto, H.; Kato, H.; et al. Progenitor-enriched adipose tissue transplantation as rescue for breast implant complications. Breast J. 2010, 16, 169–175. [Google Scholar] [CrossRef] [PubMed]
  14. Fang, B.; Song, Y.; Liao, L.; Zhang, Y.; Zhao, R.C. Favorable response to human adipose tissue-derived mesenchymal stem cells in steroid-refractory acute graft-versus-host disease. Transplant. Proc. 2007, 39, 3358–3362. [Google Scholar] [CrossRef] [PubMed]
  15. Stepien, A.; Dabrowska, N.L.; Maciagowska, M.; Macoch, R.P.; Zolocinska, A.; Mazur, S.; Siennicka, K.; Frankowska, E.; Kidzinski, R.; Chalimoniuk, M.; et al. Clinical Application of Autologous Adipose Stem Cells in Patients with Multiple Sclerosis: Preliminary Results. Mediat. Inflamm. 2016, 2016, 5302120. [Google Scholar] [CrossRef]
  16. Rigotti, G.; Charles-de-Sá, L.; Gontijo-de-Amorim, N.F.; Takiya, C.M.; Amable, P.R.; Borojevic, R.; Benati, D.; Bernardi, P.; Sbarbati, A. Expanded Stem Cells, Stromal-Vascular Fraction, and Platelet-Rich Plasma Enriched Fat: Comparing Results of Different Facial Rejuvenation Approaches in a Clinical Trial. Aesthetic Surg. J. 2016, 36, 261–270. [Google Scholar] [CrossRef]
  17. Charles-de-Sá, L.; Gontijo-de-Amorim, N.F.; Maeda Takiya, C.; Borojevic, R.; Benati, D.; Bernardi, P.; Sbarbati, A.; Rigotti, G. Antiaging treatment of the facial skin by fat graft and adipose-derived stem cells. Plast. Reconstr. Surg. 2015, 135, 999–1009. [Google Scholar] [CrossRef]
  18. Pak, J.; Lee, J.H.; Pak, N.; Pak, Y.; Park, K.S.; Jeon, J.H.; Jeong, B.C.; Lee, S.H. Cartilage Regeneration in Humans with Adipose Tissue-Derived Stem Cells and Adipose Stromal Vascular Fraction Cells: Updated Status. Int. J. Mol. Sci. 2018, 19, 2146. [Google Scholar] [CrossRef]
  19. Gonzalez-Rey, E.; Gonzalez, M.A.; Varela, N.; O’Valle, F.; Hernandez-Cortes, P.; Rico, L.; Büscher, D.; Delgado, M. Human adipose-derived mesenchymal stem cells reduce inflammatory and T cell responses and induce regulatory T cells in vitro in rheumatoid arthritis. Ann. Rheum. Dis. 2010, 69, 241–248. [Google Scholar] [CrossRef]
  20. Assunção-Silva, R.C.; Mendes-Pinheiro, B.; Patrício, P.; Behie, L.A.; Teixeira, F.G.; Pinto, L.; Salgado, A.J. Exploiting the impact of the secretome of MSCs isolated from different tissue sources on neuronal differentiation and axonal growth. Biochimie 2018, 155, 83–91. [Google Scholar] [CrossRef]
  21. Widgerow, A.D.; Salibian, A.A.; Lalezari, S.; Evans, G.R.D. Neuromodulatory nerve regeneration: Adipose tissue-derived stem cells and neurotrophic mediation in peripheral nerve regeneration. J. Neurosci. Res. 2013, 91, 1517–1524. [Google Scholar] [CrossRef]
  22. Im, G.I. Regeneration of articular cartilage using adipose stem cells. J. Biomed. Mater. Res. A 2016, 104, 1830–1844. [Google Scholar] [CrossRef]
  23. Duckers, H.J.; Pinkernell, K.; Milstein, A.M.; Hedrick, M.H. The Bedside Celution system for isolation of adipose derived regenerative cells. EuroIntervention 2006, 2, 395–398. [Google Scholar]
  24. Katz, A.J.; Hedrick, M.H.; Llull, R.; Futrell, J.W. A novel device for the simple and efficient refinement of liposuctioned tissue. Plast. Reconstr. Surg. 2001, 107, 595–597. [Google Scholar] [CrossRef]
  25. Caplan, A.I. Adult mesenchymal stem cells for tissue engineering versus regenerative medicine. J. Cell. Physiol. 2007, 213, 341–347. [Google Scholar] [CrossRef]
  26. Caplan, A.I. Mesenchymal stem cells. J. Orthop. Res. 1991, 9, 641–650. [Google Scholar] [CrossRef]
  27. Zomer, H.D.; Varela, G.K.D.S.; Delben, P.B.; Heck, D.; Jeremias, T.; Trentin, A.G. In vitro comparative study of human mesenchymal stromal cells from dermis and adipose tissue for application in skin wound healing. J. Tissue Eng. Regen. Med. 2019, 13. [Google Scholar] [CrossRef]
  28. Lei, M.; Li, K.; Li, B.; Gao, L.N.; Chen, F.M.; Jin, Y. Mesenchymal stem cell characteristics of dental pulp and periodontal ligament stem cells after in vivo transplantation. Biomaterials 2014, 35, 6332–6343. [Google Scholar] [CrossRef]
  29. Campagnoli, C.; Roberts, I.A.; Kumar, S.; Bennett, P.R.; Bellantuono, I.; Fisk, N.M. Identification of mesenchymal stem/progenitor cells in human first-trimester fetal blood, liver, and bone marrow. Blood 2001, 98, 2396–2402. [Google Scholar] [CrossRef]
  30. Vishnubalaji, R.; Al-Nbaheen, M.; Kadalmani, B.; Aldahmash, A.; Ramesh, T. Comparative investigation of the differentiation capability of bone-marrow- and adipose-derived mesenchymal stem cells by qualitative and quantitative analysis. Cell Tissue Res. 2012, 347, 419–427. [Google Scholar] [CrossRef]
  31. Kern, S.; Eichler, H.; Stoeve, J.; Klüter, H.; Bieback, K. Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem Cells 2006, 24, 1294–1301. [Google Scholar] [CrossRef]
  32. Bourin, P.; Bunnell, B.A.; Casteilla, L.; Dominici, M.; Katz, A.J.; March, K.L.; Redl, H.; Rubin, J.P.; Yoshimura, K.; Gimble, J.M. 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, 641–648. [Google Scholar]
  33. Puissant, B.; Barreau, C.; Bourin, P.; Clavel, C.; Corre, J.; Bousquet, C.; Taureau, C.; Cousin, B.; Abbal, M.; Laharrague, P.; et al. Immunomodulatory effect of human adipose tissue-derived adult stem cells: Comparison with bone marrow mesenchymal stem cells. Br. J. Haematol. 2005, 129, 118–129. [Google Scholar] [CrossRef]
  34. Musina, R.A.; Bekchanova, E.S.; Belyavskii, A.V.; Sukhikh, G.T. Differentiation potential of mesenchymal stem cells of different origin. Bull. Exp. Biol. Med. 2006, 141, 147–151. [Google Scholar] [CrossRef]
  35. Wagner, W.; Wein, F.; Seckinger, A.; Frankhauser, M.; Wirkner, U.; Krause, U.; Blake, J.; Schwager, C.; Eckstein, V.; Ansorge, W.; et al. Comparative characteristics of mesenchymal stem cells from human bone marrow, adipose tissue, and umbilical cord blood. Exp. Hematol. 2005, 33, 1402–1416. [Google Scholar] [CrossRef]
  36. Uccelli, A.; Moretta, L.; Pistoia, V. Mesenchymal stem cells in health and disease. Nat. Rev. Immunol. 2008, 8, 726–736. [Google Scholar] [CrossRef]
  37. Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.; Krause, D.; Deans, R.; Keating, A.; Prockop, D.; Horwitz, E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006, 8, 315–317. [Google Scholar] [CrossRef]
  38. Jacobs, S.A.; Pinxteren, J.; Roobrouck, V.D.; Luyckx, A.; van’t Hof, W.; Deans, R.; Verfaillie, C.M.; Waer, M.; Billiau, A.D.; Van Gool, S.W. Human multipotent adult progenitor cells are nonimmunogenic and exert potent immunomodulatory effects on alloreactive T-cell responses. Cell Transplant. 2013, 22, 1915–1928. [Google Scholar] [CrossRef]
  39. Spaggiari, G.M.; Abdelrazik, H.; Becchetti, F.; Moretta, L. MSCs inhibit monocyte-derived DC maturation and function by selectively interfering with the generation of immature DCs: Central role of MSC-derived prostaglandin E2. Blood 2009, 113, 6576–6583. [Google Scholar] [CrossRef]
  40. Jiang, X.X.; Zhang, Y.; Liu, B.; Zhang, S.X.; Wu, Y.; Yu, X.D.; Mao, N. Human mesenchymal stem cells inhibit differentiation and function of monocyte-derived dendritic cells. Blood 2005, 105, 4120–4126. [Google Scholar] [CrossRef] [Green Version]
  41. Jiang, R.; Han, Z.; Zhuo, G.; Qu, X.; Li, X.; Wang, X.; Shao, Y.; Yang, S.; Han, Z.C. Transplantation of placenta-derived mesenchymal stem cells in type 2 diabetes: A pilot study. Front. Med. 2011, 5, 94–100. [Google Scholar] [CrossRef]
  42. Stagg, J.; Galipeau, J. Immune plasticity of bone marrow-derived mesenchymal stromal cells. In Handbook Experimental Pharmacology; Springer: Berlin, Germany, 2007; pp. 45–66. [Google Scholar]
  43. Bernardo, M.E.; Avanzini, M.A.; Ciccocioppo, R.; Perotti, C.; Cometa, A.M.; Moretta, A.; Marconi, M.; Valli, M.; Novara, F.; Bonetti, F.; et al. Phenotypical/functional characterization of in vitro-expanded mesenchymal stromal cells from patients with Crohn’s disease. Cytotherapy 2009, 11, 825–836. [Google Scholar] [CrossRef] [PubMed]
  44. Bartholomew, A.; Sturgeon, C.; Siatskas, M.; Ferrer, K.; McIntosh, K.; Patil, S.; Hardy, W.; Devine, S.; Ucker, D.; Deans, R.; et al. Mesenchymal stem cells suppress lymphocyte proliferation in vitro and prolong skin graft survival in vivo. Exp. Hematol. 2002, 30, 42–48. [Google Scholar] [CrossRef]
  45. Meisel, R.; Zibert, A.; Laryea, M.; Göbel, U.; Däubener, W.; Dilloo, D. Human bone marrow stromal cells inhibit allogeneic T-cell responses by indoleamine 2,3-dioxygenase-mediated tryptophan degradation. Blood 2004, 103, 4619–4621. [Google Scholar] [CrossRef]
  46. Brooke, G.; Cook, M.; Blair, C.; Han, R.; Heazlewood, C.; Jones, B.; Kambouris, M.; Kollar, K.; McTaggart, S.; Pelekanos, R.; et al. Therapeutic applications of mesenchymal stromal cells. Semin. Cell Dev. Biol. 2007, 18, 846–858. [Google Scholar] [CrossRef]
  47. Pachón-Peña, G.; Yu, G.; Tucker, A.; Wu, X.; Vendrell, J.; Bunnell, B.A.; Gimble, J.M. Stromal stem cells from adipose tissue and bone marrow of age-matched female donors display distinct immunophenotypic profiles. J. Cell. Physiol. 2011, 226, 843–851. [Google Scholar] [CrossRef] [PubMed]
  48. Shafiee, A.; Soleimani, M.; Chamheidari, G.A.; Seyedjafari, E.; Dodel, M.; Atashi, A.; Gheisari, Y. Electrospun nanofiber-based regeneration of cartilage enhanced by mesenchymal stem cells. J. Biomed. Mater. Res. A 2011, 99, 467–478. [Google Scholar] [CrossRef]
  49. Barlow, S.; Brooke, G.; Chatterjee, K.; Price, G.; Pelekanos, R.; Rossetti, T.; Doody, M.; Venter, D.; Pain, S.; Gilshenan, K.; et al. Comparison of human placenta- and bone marrow-derived multipotent mesenchymal stem cells. Stem Cells Dev. 2008, 17, 1095–1107. [Google Scholar] [CrossRef]
  50. Lee, R.H.; Kim, B.; Choi, I.; Kim, H.; Choi, H.S.; Suh, K.; Bae, Y.C.; Jung, J.S. Characterization and expression analysis of mesenchymal stem cells from human bone marrow and adipose tissue. Cell. Physiol. Biochem. 2004, 14, 311–324. [Google Scholar] [CrossRef]
  51. Doorn, J.; Moll, G.; Le Blanc, K.; van Blitterswijk, C.; de Boer, J. Therapeutic applications of mesenchymal stromal cells: Paracrine effects and potential improvements. Tissue Eng. Part B Rev. 2012, 18, 101–115. [Google Scholar] [CrossRef]
  52. Chatzistamatiou, T.K.; Papassavas, A.C.; Michalopoulos, E.; Gamaloutsos, C.; Mallis, P.; Gontika, I.; Panagouli, E.; Koussoulakos, S.L.; Stavropoulos-Giokas, C. Optimizing isolation culture and freezing methods to preserve Wharton’s jelly’s mesenchymal stem cell (MSC) properties: An MSC banking protocol validation for the Hellenic Cord Blood Bank. Transfusion 2014, 54, 3108–3120. [Google Scholar] [CrossRef]
  53. Weiss, M.L.; Troyer, D.L. Stem cells in the umbilical cord. Stem Cell Rev. 2006, 2, 155–162. [Google Scholar] [CrossRef]
  54. Fong, C.Y.; Subramanian, A.; Gauthaman, K.; Venugopal, J.; Biswas, A.; Ramakrishna, S.; Bongso, A. Human umbilical cord Wharton’s jelly stem cells undergo enhanced chondrogenic differentiation when grown on nanofibrous scaffolds and in a sequential two-stage culture medium environment. Stem Cell Rev. 2012, 8, 195–209. [Google Scholar] [CrossRef]
  55. Balzano, F.; Bellu, E.; Basoli, V.; Dei Giudici, S.; Santaniello, S.; Cruciani, S.; Facchin, F.; Oggiano, A.; Capobianco, G.; Dessole, F.; et al. Lessons from human umbilical cord: Gender differences in stem cells from Wharton’s jelly. Eur. J. Obstet. Gynecol. Reprod. Biol. 2019, 234, 143–148. [Google Scholar] [CrossRef]
  56. Cheng, H.; Qiu, L.; Ma, J.; Zhang, H.; Cheng, M.; Li, W.; Zhao, X.; Liu, K. Replicative senescence of human bone marrow and umbilical cord derived mesenchymal stem cells and their differentiation to adipocytes and osteoblasts. Mol. Biol. Rep. 2011, 38, 5161–5168. [Google Scholar] [CrossRef]
  57. Zhang, X.; Hirai, M.; Cantero, S.; Ciubotariu, R.; Dobrila, L.; Hirsh, A.; Igura, K.; Satoh, H.; Yokomi, I.; Nishimura, T.; et al. Isolation and characterization of mesenchymal stem cells from human umbilical cord blood: Reevaluation of critical factors for successful isolation and high ability to proliferate and differentiate to chondrocytes as compared to mesenchymal stem cells from bone marrow and adipose tissue. J. Cell. Biochem. 2011, 112, 1206–1218. [Google Scholar]
  58. De Miguel, M.P.; Fuentes-Julián, S.; Blázquez-Martínez, A.; Pascual, C.Y.; Aller, M.A.; Arias, J.; Arnalich-Montiel, F. Immunosuppressive properties of mesenchymal stem cells: Advances and applications. Curr. Mol. Med. 2012, 12, 574–591. [Google Scholar] [CrossRef]
  59. Krupa, P.; Vackova, I.; Ruzicka, J.; Zaviskova, K.; Dubisova, J.; Koci, Z.; Turnovcova, K.; Urdzikova, L.M.; Kubinova, S.; Rehak, S.; et al. The Effect of Human Mesenchymal Stem Cells Derived from Wharton’s Jelly in Spinal Cord Injury Treatment Is Dose-Dependent and Can Be Facilitated by Repeated Application. Int. J. Mol. Sci. 2018, 19, 1503. [Google Scholar] [CrossRef]
  60. Weiss, M.L.; Medicetty, S.; Bledsoe, A.R.; Rachakatla, R.S.; Choi, M.; Merchav, S.; Luo, Y.; Rao, M.S.; Velagaleti, G.; Troyer, D. Human umbilical cord matrix stem cells: Preliminary characterization and effect of transplantation in a rodent model of Parkinson’s disease. Stem Cells 2006, 24, 781–792. [Google Scholar] [CrossRef]
  61. López, Y.; Lutjemeier, B.; Seshareddy, K.; Trevino, E.M.; Hageman, K.S.; Musch, T.I.; Borgarelli, M.; Weiss, M.L. Wharton’s jelly or bone marrow mesenchymal stromal cells improve cardiac function following myocardial infarction for more than 32 weeks in a rat model: A preliminary report. Curr. Stem Cell Res. Ther. 2013, 8, 46–59. [Google Scholar] [CrossRef]
  62. Wang, H.S.; Shyu, J.F.; Shen, W.S.; Hsu, H.C.; Chi, T.C.; Chen, C.P.; Huang, S.W.; Shyr, Y.M.; Tang, K.T.; Chen, T.H. Transplantation of insulin-producing cells derived from umbilical cord stromal mesenchymal stem cells to treat NOD mice. Cell Transplant. 2011, 20, 455–466. [Google Scholar] [CrossRef]
  63. Palumbo, P.; Miconi, G.; Cinque, B.; La Torre, C.; Lombardi, F.; Zoccali, G.; Orsini, G.; Leocata, P.; Giuliani, M.; Cifone, M.G. In vitro evaluation of different methods of handling human liposuction aspirate and their effect on adipocytes and adipose derived stem cells. J. Cell. Physiol. 2015, 230, 1974–1981. [Google Scholar] [CrossRef]
  64. Miyagi-Shiohira, C.; Kurima, K.; Kobayashi, N.; Saitoh, I.; Watanabe, M.; Noguchi, Y.; Matsushita, M.; Noguchi, H. Cryopreservation of Adipose-Derived Mesenchymal Stem Cells. Cell Med. 2015, 8, 3–7. [Google Scholar] [CrossRef] [Green Version]
  65. Vallée, M.; Côté, J.F.; Fradette, J. Adipose-tissue engineering: Taking advantage of the properties of human adipose-derived stem/stromal cells. Pathol. Biol. 2009, 57, 309–317. [Google Scholar] [CrossRef]
  66. Strem, B.M.; Hicok, K.C.; Zhu, M.; Wulur, I.; Alfonso, Z.; Schreiber, R.E.; Fraser, J.K.; Hedrick, M.H. Multipotential differentiation of adipose tissue-derived stem cells. Keio J. Med. 2005, 54, 132–141. [Google Scholar] [CrossRef] [Green Version]
  67. Lataillade, J.J.; Magne, B.; Bey, E.; Leclerc, T.; Trouillas, M. Skin engineering for severe burns. Transfus. Clin. Biol. 2017, 24, 245–250. [Google Scholar] [CrossRef]
  68. Mitchell, J.B.; McIntosh, K.; Zvonic, S.; Garrett, S.; Floyd, Z.E.; Kloster, A.; Di Halvorsen, Y.; Storms, R.W.; Goh, B.; Kilroy, G.; et al. Immunophenotype of human adipose-derived cells: Temporal changes in stromal-associated and stem cell-associated markers. Stem Cells 2006, 24, 376–385. [Google Scholar] [CrossRef]
  69. Maumus, M.; Peyrafitte, J.A.; D’Angelo, R.; Fournier-Wirth, C.; Bouloumié, A.; Casteilla, L.; Sengenès, C.; Bourin, P. Native human adipose stromal cells: Localization, morphology and phenotype. Int. J. Obes. 2011, 35, 1141–1153. [Google Scholar] [CrossRef]
  70. Zhao, D.; Liu, L.; Chen, Q.; Wang, F.; Li, Q.; Zeng, Q.; Huang, J.; Luo, M.; Li, W.; Zheng, Y.; et al. Hypoxia with Wharton’s jelly mesenchymal stem cell coculture maintains stemness of umbilical cord blood-derived CD34+ cells. Stem Cell Res. Ther. 2018, 9, 158. [Google Scholar] [CrossRef]
  71. Sengenès, C.; Lolmède, K.; Zakaroff-Girard, A.; Busse, R.; Bouloumié, A. Preadipocytes in the human subcutaneous adipose tissue display distinct features from the adult mesenchymal and hematopoietic stem cells. J. Cell. Physiol. 2005, 205, 114–122. [Google Scholar] [CrossRef]
  72. McIntosh, K.; Zvonic, S.; Garrett, S.; Mitchell, J.B.; Floyd, Z.E.; Hammill, L.; Kloster, A.; Di Halvorsen, Y.; Ting, J.P.; Storms, R.W.; et al. The immunogenicity of human adipose-derived cells: Temporal changes in vitro. Stem Cells 2006, 24, 1246–1253. [Google Scholar] [CrossRef]
  73. Scherberich, A.; Di Maggio, N.D.; McNagny, K.M. A familiar stranger: CD34 expression and putative functions in SVF cells of adipose tissue. World J. Stem Cells 2013, 5, 1–8. [Google Scholar] [CrossRef]
  74. Gronthos, S.; Franklin, D.M.; Leddy, H.A.; Robey, P.G.; Storms, R.W.; Gimble, J.M. Surface protein characterization of human adipose tissue-derived stromal cells. J. Cell. Physiol. 2001, 189, 54–63. [Google Scholar] [CrossRef]
  75. Cuevas-Diaz Duran, R.; González-Garza, M.T.; Cardenas-Lopez, A.; Chavez-Castilla, L.; Cruz-Vega, D.E.; Moreno-Cuevas, J.E. Age-related yield of adipose-derived stem cells bearing the low-affinity nerve growth factor receptor. Stem Cells Int. 2013, 2013, 372164. [Google Scholar] [CrossRef]
  76. Calabrese, G.; Giuffrida, R.; Lo Furno, D.; Parrinello, N.L.; Forte, S.; Gulino, R.; Colarossi, C.; Schinocca, L.R.; Giuffrida, R.; Cardile, V.; et al. Potential Effect of CD271 on Human Mesenchymal Stromal Cell Proliferation and Differentiation. Int. J. Mol. Sci. 2015, 16, 15609–15624. [Google Scholar] [CrossRef] [Green Version]
  77. Rydén, M.; Dicker, A.; Götherström, C.; Aström, G.; Tammik, C.; Arner, P.; Le Blanc, K. Functional characterization of human mesenchymal stem cell-derived adipocytes. Biochem. Biophys. Res. Commun. 2003, 311, 391–397. [Google Scholar] [CrossRef]
  78. Dubey, N.K.; Mishra, V.K.; Dubey, R.; Deng, Y.H.; Tsai, F.C.; Deng, W.P. Revisiting the Advances in Isolation, Characterization and Secretome of Adipose-Derived Stromal/Stem Cells. Int. J. Mol. Sci. 2018, 19, 2200. [Google Scholar] [CrossRef]
  79. Shi, Y.Y.; Nacamuli, R.P.; Salim, A.; Longaker, M.T. The osteogenic potential of adipose-derived mesenchymal cells is maintained with aging. Plast. Reconstr. Surg. 2005, 116, 1686–1696. [Google Scholar] [CrossRef]
  80. McIntosh, K.R. Evaluation of cellular and humoral immune responses to allogeneic adipose-derived stem/stromal cells. Methods Mol. Biol. 2011, 702, 133–150. [Google Scholar]
  81. Dhar, S.; Yoon, E.S.; Kachgal, S.; Evans, G.R.D. Long-term maintenance of neuronally differentiated human adipose tissue-derived stem cells. Tissue Eng. 2007, 13, 2625–2632. [Google Scholar] [CrossRef]
  82. Othmani, A.E.; Rouam, S.; Abbad, A.; Erraoui, C.; Harriba, S.; Boukind, H.; Nourlil, J.; Malka, G.; Mazini, L. Cryopreservation Impacts Cell Functionality of Long Term Expanded Adipose-Derived Stem Cells. J. Stem Cell Res. Ther. 2019, 9, 445. [Google Scholar] [CrossRef]
  83. Klar, A.S.; Zimoch, J.; Biedermann, T. Skin Tissue Engineering: Application of Adipose-Derived Stem Cells. Biomed. Res. Int. 2017, 2017, 9747010. [Google Scholar] [CrossRef]
  84. Lee, H.C.; An, S.G.; Lee, H.W.; Park, J.S.; Cha, K.S.; Hong, T.J.; Park, J.H.; Lee, S.Y.; Kim, S.P.; Kim, Y.D.; et al. Safety and effect of adipose tissue-derived stem cell implantation in patients with critical limb ischemia: A pilot study. Circ. J. 2012, 76, 1750–1760. [Google Scholar] [CrossRef]
  85. Lataillade, J.J.; Doucet, C.; Bey, E.; Carsin, H.; Huet, C.; Clairand, I.; Bottollier-Depois, J.F.; Chapel, A.; Ernou, I.; Gourven, M.; et al. New approach to radiation burn treatment by dosimetry-guided surgery combined with autologous mesenchymal stem cell therapy. Regen. Med. 2007, 2, 785–794. [Google Scholar] [CrossRef] [Green Version]
  86. García-Olmo, D.; Herreros, D.; De-La-Quintana, P.; Guadalajara, H.; Trébol, J.; Georgiev-Hristov, T.; García-Arranz, M. Adipose-derived stem cells in Crohn’s rectovaginal fistula. Case Rep. Med. 2010, 2010, 961758. [Google Scholar] [CrossRef] [PubMed]
  87. García-Arranz, M.; Herreros, M.D.; González-Gómez, C.; de la Quintana, P.; Guadalajara, H.; Georgiev-Hristov, T.; Trébol, J.; Garcia-Olmo, D. Treatment of Crohn’s-Related Rectovaginal Fistula with Allogeneic Expanded-Adipose Derived Stem Cells: A Phase I-IIa Clinical Trial. Stem Cells Transl. Med. 2016, 5, 1441–1446. [Google Scholar] [CrossRef]
  88. Panes, J. Stem Cell Therapy for Perianal Fistulas in Crohn’s Disease. Gastroenterol. Hepatol. 2016, 12, 637–640. [Google Scholar]
  89. Shi, D.; Wang, D.; Li, X.; Zhang, H.; Che, N.; Lu, Z.; Sun, L. Allogeneic transplantation of umbilical cord-derived mesenchymal stem cells for diffuse alveolar hemorrhage in systemic lupus erythematosus. Clin. Rheumatol. 2012, 31, 841–846. [Google Scholar] [CrossRef]
  90. Liang, J.; Zhang, H.; Kong, W.; Deng, W.; Wang, D.; Feng, X.; Zhao, C.; Hua, B.; Wang, H.; Sun, L. Safety analysis in patients with autoimmune disease receiving allogeneic mesenchymal stem cells infusion: A long-term retrospective study. Stem Cell Res. Ther. 2018, 9, 312. [Google Scholar] [CrossRef]
  91. Ciccocioppo, R.; Bernardo, M.E.; Sgarella, A.; Maccario, R.; Avanzini, M.A.; Ubezio, C.; Minelli, A.; Alvisi, C.; Vanoli, A.; Calliada, F.; et al. Autologous bone marrow-derived mesenchymal stromal cells in the treatment of fistulising Crohn’s disease. Gut 2011, 60, 788–798. [Google Scholar] [CrossRef]
  92. Duijvestein, M.; Vos, A.C.W.; Roelofs, H.; Wildenberg, M.E.; Wendrich, B.B.; Verspaget, H.W.; Kooy-Winkelaar, E.M.C.; Koning, F.; Zwaginga, J.J.; Fidder, H.H.; et al. Autologous bone marrow-derived mesenchymal stromal cell treatment for refractory luminal Crohn’s disease: Results of a phase I study. Gut 2010, 59, 1662–1669. [Google Scholar] [CrossRef] [PubMed]
  93. Yocum, D.E. Combination therapy: The risks of infection and tumor induction. Springer Semin. Immunopathol. 2001, 23, 63–72. [Google Scholar] [CrossRef]
  94. Le Blanc, K.; Frassoni, F.; Ball, L.; Locatelli, F.; Roelofs, H.; Lewis, I.; Lanino, E.; Sundberg, B.; Bernardo, M.E.; Remberger, M.; et al. Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: A phase II study. Lancet 2008, 371, 1579–1586. [Google Scholar] [CrossRef]
  95. Ra, J.C.; Kang, S.K.; Shin, I.S.; Park, H.G.; Joo, S.A.; Kim, J.G.; Kang, B.C.; Lee, Y.S.; Nakama, K.; Piao, M.; et al. Stem cell treatment for patients with autoimmune disease by systemic infusion of culture-expanded autologous adipose tissue derived mesenchymal stem cells. J. Transl. Med. 2011, 9, 181. [Google Scholar] [CrossRef] [PubMed]
  96. Dave, S.D.; Vanikar, A.V.; Trivedi, H.L.; Thakkar, U.G.; Gopal, S.C.; Chandra, T. Novel therapy for insulin-dependent diabetes mellitus: Infusion of in vitro-generated insulin-secreting cells. Clin. Exp. Med. 2015, 15, 41–45. [Google Scholar] [CrossRef] [PubMed]
  97. Dander, E.; Lucchini, G.; Vinci, P.; Introna, M.; Masciocchi, F.; Perseghin, P.; Balduzzi, A.; Bonanomi, S.; Longoni, D.; Gaipa, G.; et al. Mesenchymal stromal cells for the treatment of graft-versus-host disease: Understanding the in vivo biological effect through patient immune monitoring. Leukemia 2012, 26, 1681–1684. [Google Scholar] [CrossRef]
  98. Bernardo, M.E.; Cometa, A.M.; Pagliara, D.; Vinti, L.; Rossi, F.; Cristantielli, R.; Palumbo, G.; Locatelli, F. Ex vivo expansion of mesenchymal stromal cells. Best Pract. Res. Clin. Haematol. 2011, 24, 73–81. [Google Scholar] [CrossRef] [PubMed]
  99. Bernardo, M.E.; Fibbe, W.E. Mesenchymal stromal cells and hematopoietic stem cell transplantation. Immunol. Lett. 2015, 168, 215–221. [Google Scholar] [CrossRef]
  100. Ball, L.M.; Bernardo, M.E.; Roelofs, H.; Lankester, A.; Cometa, A.; Egeler, R.M.; Locatelli, F.; Fibbe, W.E. Cotransplantation of ex vivo expanded mesenchymal stem cells accelerates lymphocyte recovery and may reduce the risk of graft failure in haploidentical hematopoietic stem-cell transplantation. Blood 2007, 110, 2764–2767. [Google Scholar] [CrossRef]
  101. Wu, Y.M.; Cao, Y.B.; Li, X.H.; Xu, L.X.; Liu, Z.Y.; Liu, B.; Yan, B.; Yang, X.L.; Li, S.W.; Da, W.M.; et al. Transplantation of umbilical cord mesenchymal stem cells combined with haploidentical hematopoietic stem cells for 36 patients with refractory/relapsed myeloid leukemia. J. Exp. Hematol. 2014, 22, 1053–1057. [Google Scholar]
  102. Wu, Y.; Wang, Z.; Cao, Y.; Xu, L.; Li, X.; Liu, P.; Yan, P.; Liu, Z.; Zhao, D.; Wang, J.; et al. Cotransplantation of haploidentical hematopoietic and umbilical cord mesenchymal stem cells with a myeloablative regimen for refractory/relapsed hematologic malignancy. Ann. Hematol. 2013, 92, 1675–1684. [Google Scholar] [CrossRef] [PubMed]
  103. Koç, O.N.; Gerson, S.L.; Cooper, B.W.; Dyhouse, S.M.; Haynesworth, S.E.; Caplan, A.I.; Lazarus, H.M. Rapid hematopoietic recovery after coinfusion of autologous-blood stem cells and culture-expanded marrow mesenchymal stem cells in advanced breast cancer patients receiving high-dose chemotherapy. J. Clin. Oncol. 2000, 18, 307–316. [Google Scholar] [CrossRef] [PubMed]
  104. Corre, J.; Barreau, C.; Cousin, B.; Chavoin, J.P.; Caton, D.; Fournial, G.; Penicaud, L.; Casteilla, L.; Laharrague, P. Human subcutaneous adipose cells support complete differentiation but not self-renewal of hematopoietic progenitors. J. Cell. Physiol. 2006, 208, 282–288. [Google Scholar] [CrossRef] [PubMed]
  105. De Toni, F.; Poglio, S.; Youcef, A.B.; Cousin, B.; Pflumio, F.; Bourin, P.; Casteilla, L.; Laharrague, P. Human adipose-derived stromal cells efficiently support hematopoiesis in vitro and in vivo: A key step for therapeutic studies. Stem Cells Dev. 2011, 20, 2127–2138. [Google Scholar] [CrossRef] [PubMed]
  106. Morgan, E.F.; De Giacomo, A.; Gerstenfeld, L.C. Overview of skeletal repair (fracture healing and its assessment). Methods Mol. Biol. 2014, 1130, 13–31. [Google Scholar]
  107. Ude, C.C.; Sulaiman, S.B.; Min-Hwei, N.; Hui-Cheng, C.; Ahmad, J.; Yahaya, N.M.; Saim, A.B.; Idrus, R.B.H. Cartilage regeneration by chondrogenic induced adult stem cells in osteoarthritic sheep model. PLoS ONE 2014, 9, e98770. [Google Scholar] [CrossRef]
  108. Park, Y.B.; Ha, C.W.; Lee, C.H.; Yoon, Y.C.; Park, Y.G. Cartilage Regeneration in Osteoarthritic Patients by a Composite of Allogeneic Umbilical Cord Blood-Derived Mesenchymal Stem Cells and Hyaluronate Hydrogel: Results from a Clinical Trial for Safety and Proof-of-Concept with 7 Years of Extended Follow-Up. Stem Cells Transl. Med. 2017, 6, 613–621. [Google Scholar] [CrossRef]
  109. Mesimäki, K.; Lindroos, B.; Törnwall, J.; Mauno, J.; Lindqvist, C.; Kontio, R.; Miettinen, S.; Suuronen, R. Novel maxillary reconstruction with ectopic bone formation by GMP adipose stem cells. Int. J. Oral Maxillofac. Surg. 2009, 38, 201–209. [Google Scholar] [CrossRef]
  110. Lendeckel, S.; Jödicke, A.; Christophis, P.; Heidinger, K.; Wolff, J.; Fraser, J.K.; Hedrick, M.H.; Berthold, L.; Howaldt, H.P. Autologous stem cells (adipose) and fibrin glue used to treat widespread traumatic calvarial defects: Case report. J. Craniomaxillofac. Surg. 2004, 32, 370–373. [Google Scholar] [CrossRef]
  111. Pak, J.; Lee, J.H.; Park, K.S.; Jeong, B.C.; Lee, S.H. Regeneration of Cartilage in Human Knee Osteoarthritis with Autologous Adipose Tissue-Derived Stem Cells and Autologous Extracellular Matrix. Biores. Open Access 2016, 5, 192–200. [Google Scholar] [CrossRef]
  112. Ganey, T.; Hutton, W.C.; Moseley, T.; Hedrick, M.; Meisel, H.J. Intervertebral disc repair using adipose tissue-derived stem and regenerative cells: Experiments in a canine model. Spine 2009, 34, 2297–2304. [Google Scholar] [CrossRef]
  113. Uysal, A.C.; Mizuno, H.; Tobita, M.; Ogawa, R.; Hyakusoku, H. The effect of adipose-derived stem cells on ischemia-reperfusion injury: Immunohistochemical and ultrastructural evaluation. Plast. Reconstr. Surg. 2009, 124, 804–815. [Google Scholar] [CrossRef]
  114. Ryu, H.H.; Lim, J.H.; Byeon, Y.E.; Park, J.R.; Seo, M.S.; Lee, Y.W.; Kim, W.H.; Kang, K.S.; Kweon, O.K. Functional recovery and neural differentiation after transplantation of allogenic adipose-derived stem cells in a canine model of acute spinal cord injury. J. Vet. Sci. 2009, 10, 273–284. [Google Scholar] [CrossRef]
  115. Dragoo, J.L.; Carlson, G.; McCormick, F.; Khan-Farooqi, H.; Zhu, M.; Zuk, P.A.; Benhaim, P. Healing full-thickness cartilage defects using adipose-derived stem cells. Tissue Eng. 2007, 13, 1615–1621. [Google Scholar] [CrossRef]
  116. Puente, B.N.; Kimura, W.; Muralidhar, S.A.; Moon, J.; Amatruda, J.F.; Phelps, K.L.; Grinsfelder, D.; Rothermel, B.A.; Chen, R.; Garcia, J.A.; et al. The oxygen-rich postnatal environment induces cardiomyocyte cell-cycle arrest through DNA damage response. Cell 2014, 157, 565–579. [Google Scholar] [CrossRef]
  117. Tani, H.; Sadahiro, T.; Ieda, M. Direct Cardiac Reprogramming: A Novel Approach for Heart Regeneration. Int. J. Mol. Sci. 2018, 19, 2629. [Google Scholar] [CrossRef]
  118. Maioli, M.; Rinaldi, S.; Santaniello, S.; Castagna, A.; Pigliaru, G.; Gualini, S.; Cavallini, C.; Fontani, V.; Ventura, C. Radio electric conveyed fields directly reprogram human dermal skin fibroblasts toward cardiac, neuronal, and skeletal muscle-like lineages. Cell Transplant. 2013, 22, 1227–1235. [Google Scholar] [CrossRef]
  119. Yun, C.W.; Lee, S.H. Enhancement of Functionality and Therapeutic Efficacy of Cell-Based Therapy Using Mesenchymal Stem Cells for Cardiovascular Disease. Int. J. Mol. Sci. 2019, 20, 982. [Google Scholar] [CrossRef]
  120. Maioli, M.; Contini, G.; Santaniello, S.; Bandiera, P.; Pigliaru, G.; Sanna, R.; Rinaldi, S.; Delitala, A.P.; Montella, A.; Bagella, L.; et al. Amniotic fluid stem cells morph into a cardiovascular lineage: Analysis of a chemically induced cardiac and vascular commitment. Drug Des. Dev. Ther. 2013, 7, 1063–1073. [Google Scholar]
  121. Li, N.; Rochette, L.; Wu, Y.; Rosenblatt-Velin, N. New Insights into the Role of Exosomes in the Heart After Myocardial Infarction. J. Cardiovasc. Transl. Res. 2019, 12, 18–27. [Google Scholar] [CrossRef]
  122. Moghaddam, A.S.; Afshari, J.T.; Esmaeili, S.A.; Saburi, E.; Joneidi, Z.; Momtazi-Borojeni, A.A. Cardioprotective microRNAs: Lessons from stem cell-derived exosomal microRNAs to treat cardiovascular disease. Atherosclerosis 2019, 285, 1–9. [Google Scholar] [CrossRef] [Green Version]
  123. Makino, S.; Fukuda, K.; Miyoshi, S.; Konishi, F.; Kodama, H.; Pan, J.; Sano, M.; Takahashi, T.; Hori, S.; Abe, H.; et al. Cardiomyocytes can be generated from marrow stromal cells in vitro. J. Clin. Investig. 1999, 103, 697–705. [Google Scholar] [CrossRef] [Green Version]
  124. Assmann, A.; Heke, M.; Kröpil, P.; Ptok, L.; Hafner, D.; Ohmann, C.; Martens, A.; Karluβ, A.; Emmert, M.Y.; Kutschka, I.; et al. Laser-supported CD133+ cell therapy in patients with ischemic cardiomyopathy: Initial results from a prospective phase I multicenter trial. PLoS ONE 2014, 9, e101449. [Google Scholar] [CrossRef]
  125. Chen, S.; Fang, W.; Ye, F.; Liu, Y.H.; Qian, J.; Shan, S.; Zhang, J.; Chunhua, R.Z.; Liao, L.; Lin, S.; et al. Effect on left ventricular function of intracoronary transplantation of autologous bone marrow mesenchymal stem cell in patients with acute myocardial infarction. Am. J. Cardiol. 2004, 94, 92–95. [Google Scholar] [CrossRef]
  126. Hare, J.M.; Traverse, J.H.; Henry, T.D.; Dib, N.; Strumpf, R.K.; Schulman, S.P.; Gerstenblith, G.; DeMaria, A.N.; Denktas, A.E.; Gammon, R.S.; et al. A randomized, double-blind, placebo-controlled, dose-escalation study of intravenous adult human mesenchymal stem cells (prochymal) after acute myocardial infarction. J. Am. Coll. Cardiol. 2009, 54, 2277–2286. [Google Scholar] [CrossRef]
  127. Li, X.; Hu, Y.; Guo, Y.; Chen, Y.; Guo, D.; Zhou, H.; Zhang, F.; Zhao, Q. Safety and efficacy of intracoronary human umbilical cord-derived mesenchymal stem cell treatment for very old patients with coronary chronic total occlusion. Curr. Pharm. Des. 2015, 21, 1426–1432. [Google Scholar] [CrossRef]
  128. Bartolucci, J.; Verdugo, F.J.; González, P.L.; Larrea, R.E.; Abarzua, E.; Goset, C.; Rojo, P.; Palma, I.; Lamich, R.; Pedreros, P.A.; et al. Safety and Efficacy of the Intravenous Infusion of Umbilical Cord Mesenchymal Stem Cells in Patients with Heart Failure: A Phase 1/2 Randomized Controlled Trial (RIMECARD Trial [Randomized Clinical Trial of Intravenous Infusion Umbilical Cord Mesenchymal Stem Cells on Cardiopathy]). Circ. Res. 2017, 121, 1192–1204. [Google Scholar]
  129. Kastrup, J.; Haack-Sørensen, M.; Juhl, M.; Harary Søndergaard, R.; Follin, B.; Drozd Lund, L.; Mønsted Johansen, E.; Ali Qayyum, A.; Bruun Mathiasen, A.; Jørgensen, E.; 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 Transl. Med. 2017, 6, 1963–1971. [Google Scholar] [CrossRef]
  130. Henry, T.D.; Pepine, C.J.; Lambert, C.R.; Traverse, J.H.; Schatz, R.; Costa, M.; Povsic, T.J.; David Anderson, R.; Willerson, J.T.; Kesten, S.; et al. The Athena trials: Autologous adipose-derived regenerative cells for refractory chronic myocardial ischemia with left ventricular dysfunction. Catheter. Cardiovasc. Interv. 2017, 89, 169–177. [Google Scholar] [CrossRef]
  131. Kastrup, J.; Schou, M.; Gustafsson, I.; Nielsen, O.W.; Møgelvang, R.; Kofoed, K.F.; Kragelund, C.; Hove, J.D.; Fabricius-Bjerre, A.; Heitman, M.; et al. Rationale and Design of the First Double-Blind, Placebo-Controlled Trial with Allogeneic Adipose Tissue-Derived Stromal Cell Therapy in Patients with Ischemic Heart Failure: A Phase II Danish Multicentre Study. Stem Cells Int. 2017, 2017, 8506370. [Google Scholar] [CrossRef]
  132. Yamada, Y.; Wang, X.D.; Yokoyama, S.; Fukuda, N.; Takakura, N. Cardiac progenitor cells in brown adipose tissue repaired damaged myocardium. Biochem. Biophys. Res. Commun. 2006, 342, 662–670. [Google Scholar] [CrossRef]
  133. Li, P.; Cui, K.; Zhang, B.; Wang, Z.; Shen, Y.; Wang, X.; Zhang, J.; Tong, F.; Li, S. Transplantation of human umbilical cord-derived mesenchymal stems cells for the treatment of Becker muscular dystrophy in affected pedigree members. Int. J. Mol. Med. 2015, 35, 1051–1057. [Google Scholar] [CrossRef] [Green Version]
  134. Rajput, B.S.; Chakrabarti, S.K.; Dongare, V.S.; Ramirez, C.M.; Deb, K.D. Human Umbilical Cord Mesenchymal Stem Cells in the Treatment of Duchenne Muscular Dystrophy: Safety and Feasibility Study in India. J. Stem Cells 2015, 10, 141–156. [Google Scholar]
  135. Siegel, G.; Krause, P.; Wöhrle, S.; Nowak, P.; Ayturan, M.; Kluba, T.; Brehm, B.R.; Neumeister, B.; Köhler, D.; Rosenberger, P.; et al. Bone marrow-derived human mesenchymal stem cells express cardiomyogenic proteins but do not exhibit functional cardiomyogenic differentiation potential. Stem Cells Dev. 2012, 21, 2457–2470. [Google Scholar] [CrossRef]
  136. Sato, T.; Iso, Y.; Uyama, T.; Kawachi, K.; Wakabayashi, K.; Omori, Y.; Soda, T.; Shoji, M.; Koba, S.; Yokoyama, S.I.; et al. Coronary vein infusion of multipotent stromal cells from bone marrow preserves cardiac function in swine ischemic cardiomyopathy via enhanced neovascularization. Lab. Investig. 2011, 91, 553–564. [Google Scholar] [CrossRef] [Green Version]
  137. Vela, D.C.; Silva, G.V.; Assad, J.A.R.; Sousa, A.L.S.; Coulter, S.; Fernandes, M.R.; Perin, E.C.; Willerson, J.T.; Buja, L.M. Histopathological study of healing after allogenic mesenchymal stem cell delivery in myocardial infarction in dogs. J. Histochem. Cytochem. 2009, 57, 167–176. [Google Scholar] [CrossRef]
  138. Alexeev, V.; Arita, M.; Donahue, A.; Bonaldo, P.; Chu, M.L.; Igoucheva, O. Human adipose-derived stem cell transplantation as a potential therapy for collagen VI-related congenital muscular dystrophy. Stem Cell Res. Ther. 2014, 5, 21. [Google Scholar] [CrossRef]
  139. DiRocco, J.D.; Pavone, L.A.; Carney, D.E.; Lutz, C.J.; Gatto, L.A.; Landas, S.K.; Nieman, G.F. Dynamic alveolar mechanics in four models of lung injury. Intensive Care Med. 2006, 32, 140–148. [Google Scholar] [CrossRef]
  140. Rodriguez, A.M.; Pisani, D.; Dechesne, C.A.; Turc-Carel, C.; Kurzenne, J.Y.; Wdziekonski, B.; Villageois, A.; Bagnis, C.; Breittmayer, J.P.; Groux, H.; et al. Transplantation of a multipotent cell population from human adipose tissue induces dystrophin expression in the immunocompetent mdx mouse. J. Exp. Med. 2005, 201, 1397–1405. [Google Scholar] [CrossRef]
  141. Merkle, F.T.; Fuentealba, L.C.; Sanders, T.A.; Magno, L.; Kessaris, N.; Alvarez-Buylla, A. Adult neural stem cells in distinct microdomains generate previously unknown interneuron types. Nat. Neurosci. 2014, 17, 207–214. [Google Scholar] [CrossRef]
  142. Kim, Y.J.; Hwang, S.H.; Cho, H.H.; Shin, K.K.; Bae, Y.C.; Jung, J.S. MicroRNA 21 regulates the proliferation of human adipose tissue-derived mesenchymal stem cells and high-fat diet-induced obesity alters microRNA 21 expression in white adipose tissues. J. Cell. Physiol. 2012, 227, 183–193. [Google Scholar] [CrossRef] [PubMed]
  143. Weimann, J.M.; Charlton, C.A.; Brazelton, T.R.; Hackman, R.C.; Blau, H.M. Contribution of transplanted bone marrow cells to Purkinje neurons in human adult brains. Proc. Natl. Acad. Sci. USA 2003, 100, 2088–2093. [Google Scholar] [CrossRef] [Green Version]
  144. Canesi, M.; Giordano, R.; Lazzari, L.; Isalberti, M.; Isaias, I.U.; Benti, R.; Rampini, P.; Marotta, G.; Colombo, A.; Cereda, E.; et al. Finding a new therapeutic approach for no-option Parkinsonisms: Mesenchymal stromal cells for progressive supranuclear palsy. J. Transl. Med. 2016, 14, 127. [Google Scholar] [CrossRef] [PubMed]
  145. Steinberg, G.K.; Kondziolka, D.; Wechsler, L.R.; Lunsford, L.D.; Coburn, M.L.; Billigen, J.B.; Kim, A.S.; Johnson, J.N.; Bates, D.; King, B.; et al. Clinical Outcomes of Transplanted Modified Bone Marrow-Derived Mesenchymal Stem Cells in Stroke: A Phase 1/2a Study. Stroke 2016, 47, 1817–1824. [Google Scholar] [CrossRef] [PubMed]
  146. Petrou, P.; Gothelf, Y.; Argov, Z.; Gotkine, M.; Levy, Y.S.; Kassis, I.; Vaknin-Dembinsky, A.; Ben-Hur, T.; Offen, D.; Abramsky, O.; et al. Safety and Clinical Effects of Mesenchymal Stem Cells Secreting Neurotrophic Factor Transplantation in Patients with Amyotrophic Lateral Sclerosis: Results of Phase 1/2 and 2a Clinical Trials. JAMA Neurol. 2016, 73, 337–344. [Google Scholar] [CrossRef]
  147. Harris, V.K.; Stark, J.; Vyshkina, T.; Blackshear, L.; Joo, G.; Stefanova, V.; Sara, G.; Sadiq, S.A. Phase I Trial of Intrathecal Mesenchymal Stem Cell-derived Neural Progenitors in Progressive Multiple Sclerosis. EBioMedicine 2018, 29, 23–30. [Google Scholar] [CrossRef] [Green Version]
  148. Yamout, B.; Hourani, R.; Salti, H.; Barada, W.; El-Hajj, T.; Al-Kutoubi, A.; Herlopian, A.; Baz, E.K.; Mahfouz, R.; Khalil-Hamdan, R.; et al. Bone marrow mesenchymal stem cell transplantation in patients with multiple sclerosis: A pilot study. J. Neuroimmunol. 2010, 227, 185–189. [Google Scholar] [CrossRef]
  149. Dahbour, S.; Jamali, F.; Alhattab, D.; Al-Radaideh, A.; Ababneh, O.; Al-Ryalat, N.; Al-Bdour, M.; Hourani, B.; Msallam, M.; Rasheed, M.; et al. Mesenchymal stem cells and conditioned media in the treatment of multiple sclerosis patients: Clinical, ophthalmological and radiological assessments of safety and efficacy. CNS Neurosci. Ther. 2017, 23, 866–874. [Google Scholar] [CrossRef]
  150. Li, X.; Bai, J.; Ji, X.; Li, R.; Xuan, Y.; Wang, Y. Comprehensive characterization of four different populations of human mesenchymal stem cells as regards their immune properties, proliferation and differentiation. Int. J. Mol. Med. 2014, 34, 695–704. [Google Scholar] [CrossRef] [Green Version]
  151. Kalbermatten, D.F.; Schaakxs, D.; Kingham, P.J.; Wiberg, M. Neurotrophic activity of human adipose stem cells isolated from deep and superficial layers of abdominal fat. Cell Tissue Res. 2011, 344, 251–260. [Google Scholar] [CrossRef]
  152. Rehman, J.; Traktuev, D.; Li, J.; Merfeld-Clauss, S.; Temm-Grove, C.J.; Bovenkerk, J.E.; Pell, C.L.; Johnstone, B.H.; Considine, R.V.; March, K.L. Secretion of angiogenic and antiapoptotic factors by human adipose stromal cells. Circulation 2004, 109, 1292–1298. [Google Scholar] [CrossRef]
  153. Wei, X.; Zhao, L.; Zhong, J.; Gu, H.; Feng, D.; Johnstone, B.H.; March, K.L.; Farlow, M.R.; Du, Y. Adipose stromal cells-secreted neuroprotective media against neuronal apoptosis. Neurosci. Lett. 2009, 462, 76–79. [Google Scholar] [CrossRef]
  154. Ikegame, Y.; Yamashita, K.; Hayashi, S.I.; Mizuno, H.; Tawada, M.; You, F.; Yamada, K.; Tanaka, Y.; Egashira, Y.; Nakashima, S.; et al. Comparison of mesenchymal stem cells from adipose tissue and bone marrow for ischemic stroke therapy. Cytotherapy 2011, 13, 675–685. [Google Scholar] [CrossRef]
  155. Leu, S.; Lin, Y.C.; Yuen, C.M.; Yen, C.H.; Kao, Y.H.; Sun, C.K.; Yip, H.K. Adipose-derived mesenchymal stem cells markedly attenuate brain infarct size and improve neurological function in rats. J. Transl. Med. 2010, 8, 63. [Google Scholar] [CrossRef]
  156. Qiao, L.; Huang, F.; Zhao, M.; Xie, J.; Shi, J.; Wang, J.; Lin, X.; Zuo, H.; Wang, Y.; Geng, T. A two-year follow-up study of cotransplantation with neural stem/progenitor cells and mesenchymal stromal cells in ischemic stroke patients. Cell Transplant. 2014, 23, 65–72. [Google Scholar] [CrossRef]
  157. Kim, H.Y.; Kim, H.; Oh, K.W.; Oh, S.I.; Koh, S.H.; Baik, W.; Noh, M.Y.; Kim, K.S.; Kim, S.H. Biological markers of mesenchymal stromal cells as predictors of response to autologous stem cell transplantation in patients with amyotrophic lateral sclerosis: An investigator-initiated trial and in vivo study. Stem Cells 2014, 32, 2724–2731. [Google Scholar] [CrossRef]
  158. San-Marina, S.; Voss, S.; Crespo-Diaz, R.; Wyles, C.; Behfar, A.; Stalboeger, P.; Janus, J.R. Adipose-Derived Mesenchymal Stem Cell Features in Patients with a History of Head and Neck Radiation. Laryngoscope Investig. Otolaryngol. 2016, 1, 36–41. [Google Scholar] [CrossRef]
  159. Maria, O.M.; Shalaby, M.; Syme, A.; Eliopoulos, N.; Muanza, T. Adipose mesenchymal stromal cells minimize and repair radiation-induced oral mucositis. Cytotherapy 2016, 18, 1129–1145. [Google Scholar] [CrossRef] [Green Version]
  160. Danan, D.; Lehman, C.E.; Mendez, R.E.; Langford, B.; Koors, P.D.; Dougherty, M.I.; Peirce, S.M.; Gioeli, D.G.; Jameson, M.J. Effect of Adipose-Derived Stem Cells on Head and Neck Squamous Cell Carcinoma. Otolaryngol. Head Neck Surg. 2018, 158, 882–888. [Google Scholar] [CrossRef]
  161. Riccobono, D.; Nikovics, K.; François, S.; Favier, A.L.; Jullien, N.; Schrock, G.; Scherthan, H.; Drouet, M. First Insights into the M2 Inflammatory Response After Adipose-Tissue-Derived Stem Cell Injections in Radiation-Injured Muscles. Health Phys. 2018, 115, 37–48. [Google Scholar] [CrossRef]
  162. Van de Putte, D.; Demarquay, C.; Van Daele, E.; Moussa, L.; Vanhove, C.; Benderitter, M.; Ceelen, W.; Pattyn, P.; Mathieu, N. Adipose-Derived Mesenchymal Stromal Cells Improve the Healing of Colonic Anastomoses Following High Dose of Irradiation Through Anti-Inflammatory and Angiogenic Processes. Cell Transplant. 2017, 26, 1919–1930. [Google Scholar] [CrossRef]
  163. Grønhøj, C.; Jensen, D.H.; Vester-Glowinski, P.; Jensen, S.B.; Bardow, A.; Oliveri, R.S.; Fog, L.M.; Specht, L.; Thomsen, C.; Darkner, S.; et al. Safety and Efficacy of Mesenchymal Stem Cells for Radiation-Induced Xerostomia: A Randomized, Placebo-Controlled Phase 1/2 Trial (MESRIX). Int. J. Radiat. Oncol. Biol. Phys. 2018, 101, 581–592. [Google Scholar] [CrossRef]
  164. Ebrahimian, T.G.; Pouzoulet, F.; Squiban, C.; Buard, V.; André, M.; Cousin, B.; Gourmelon, P.; Benderitter, M.; Casteilla, L.; Tamarat, R. Cell therapy based on adipose tissue-derived stromal cells promotes physiological and pathological wound healing. Arterioscler. Thromb. Vasc. Biol. 2009, 29, 503–510. [Google Scholar] [CrossRef]
  165. Haubner, F.; Gassner, H.G. Potential of adipose-derived stem cells concerning the treatment of wound healing complications after radiotherapy. HNO 2015, 63, 111–117. [Google Scholar] [CrossRef]
  166. Singh, M.; Alavi, A.; Wong, R.; Akita, S. Radiodermatitis: A Review of Our Current Understanding. Am. J. Clin. Dermatol. 2016, 17, 277–292. [Google Scholar] [CrossRef]
  167. Mashiko, T.; Takada, H.; Wu, S.H.; Kanayama, K.; Feng, J.; Tashiro, K.; Asahi, R.; Sunaga, A.; Hoshi, K.; Kurisaki, A.; et al. Therapeutic effects of a recombinant human collagen peptide bioscaffold with human adipose-derived stem cells on impaired wound healing after radiotherapy. J. Tissue Eng. Regen. Med. 2018, 12, 1186–1194. [Google Scholar] [CrossRef]
  168. Wu, S.H.; Shirado, T.; Mashiko, T.; Feng, J.; Asahi, R.; Kanayama, K.; Mori, M.; Chi, D.; Sunaga, A.; Sarukawa, S.; et al. Therapeutic Effects of Human Adipose-Derived Products on Impaired Wound Healing in Irradiated Tissue. Plast. Reconstr. Surg. 2018, 142, 383–391. [Google Scholar] [CrossRef]
  169. Rochette, L.; Guenancia, C.; Gudjoncik, A.; Hachet, O.; Zeller, M.; Cottin, Y.; Vergely, C. Anthracyclines/trastuzumab: New aspects of cardiotoxicity and molecular mechanisms. Trends Pharmacol. Sci. 2015, 36, 326–348. [Google Scholar] [CrossRef]
  170. Dalloz, F.; Maingon, P.; Cottin, Y.; Briot, F.; Horiot, J.C.; Rochette, L. Effects of combined irradiation and doxorubicin treatment on cardiac function and antioxidant defenses in the rat. Free Radic. Biol. Med. 1999, 26, 785–800. [Google Scholar] [CrossRef]
  171. Bey, E.; Prat, M.; Duhamel, P.; Benderitter, M.; Brachet, M.; Trompier, F.; Battaglini, P.; Ernou, I.; Boutin, L.; Gourven, M.; et al. Emerging therapy for improving wound repair of severe radiation burns using local bone marrow-derived stem cell administrations. Wound Repair Regen. 2010, 18, 50–58. [Google Scholar] [CrossRef]
  172. Falanga, V.; Iwamoto, S.; Chartier, M.; Yufit, T.; Butmarc, J.; Kouttab, N.; Shrayer, D.; Carson, P. Autologous bone marrow-derived cultured mesenchymal stem cells delivered in a fibrin spray accelerate healing in murine and human cutaneous wounds. Tissue Eng. 2007, 13, 1299–1312. [Google Scholar] [CrossRef]
  173. Qin, H.L.; Zhu, X.H.; Zhang, B.; Zhou, L.; Wang, W.Y. Clinical Evaluation of Human Umbilical Cord Mesenchymal Stem Cell Transplantation After Angioplasty for Diabetic Foot. Exp. Clin. Endocrinol. Diabetes 2016, 124, 497–503. [Google Scholar] [CrossRef]
  174. Valbonesi, M.; Giannini, G.; Migliori, F.; Dalla Costa, R.; Dejana, A.M. Cord blood (CB) stem cells for wound repair. Preliminary report of 2 cases. Transfus. Apher. Sci. 2004, 30, 153–156. [Google Scholar] [CrossRef]
  175. Anderi, R.; Makdissy, N.; Azar, A.; Rizk, F.; Hamade, A. Cellular therapy with human autologous adipose-derived adult cells of stromal vascular fraction for alopecia areata. Stem Cell Res. Ther. 2018, 9, 141. [Google Scholar] [CrossRef]
  176. Trojahn Kølle, S.F.; Oliveri, R.S.; Glovinski, P.V.; Elberg, J.J.; Fischer-Nielsen, A.; Drzewiecki, K.T. Importance of mesenchymal stem cells in autologous fat grafting: A systematic review of existing studies. J. Plast. Surg. Hand Surg. 2012, 46, 59–68. [Google Scholar] [CrossRef]
  177. Shin, H.; Ryu, H.H.; Kwon, O.; Park, B.S.; Jo, S.J. Clinical use of conditioned media of adipose tissue-derived stem cells in female pattern hair loss: A retrospective case series study. Int. J. Dermatol. 2015, 54, 730–735. [Google Scholar] [CrossRef]
  178. Foubert, P.; Barillas, S.; Gonzalez, A.D.; Alfonso, Z.; Zhao, S.; Hakim, I.; Meschter, C.; Tenenhaus, M.; Fraser, J.K. Uncultured adipose-derived regenerative cells (ADRCs) seeded in collagen scaffold improves dermal regeneration, enhancing early vascularization and structural organization following thermal burns. Burns 2015, 41, 1504–1516. [Google Scholar] [CrossRef] [PubMed]
  179. Park, B.S.; Jang, K.A.; Sung, J.H.; Park, J.S.; Kwon, Y.H.; Kim, K.J.; Kim, W.S. Adipose-derived stem cells and their secretory factors as a promising therapy for skin aging. Dermatol. Surg. 2008, 34, 1323–1326. [Google Scholar] [PubMed]
  180. Rigotti, G.; Marchi, A.; Galiè, M.; Baroni, G.; Benati, D.; Krampera, M.; Pasini, A.; Sbarbati, A. Clinical treatment of radiotherapy tissue damage by lipoaspirate transplant: A healing process mediated by adipose-derived adult stem cells. Plast. Reconstr. Surg. 2007, 119, 1409–1422. [Google Scholar] [CrossRef] [PubMed]
  181. Claudio-da-Silva, C.; Baptista, L.S.; Carias, R.B.V.; Menezes Neto, H. da C.; Borojevic, R. Autologous mesenchymal stem cells culture from adipose tissue for treatment of facial rhytids. Rev. Col. Bras. Cir. 2009, 36, 288–291. [Google Scholar] [CrossRef] [PubMed]
  182. Sterodimas, A.; de Faria, J.; Nicaretta, B.; Boriani, F. Autologous fat transplantation versus adipose-derived stem cell-enriched lipografts: A study. Aesthetic Surg. J. 2011, 31, 682–693. [Google Scholar] [CrossRef]
  183. Satti, H.S.; Waheed, A.; Ahmed, P.; Ahmed, K.; Akram, Z.; Aziz, T.; Satti, T.M.; Shahbaz, N.; Khan, M.A.; Malik, S.A. Autologous mesenchymal stromal cell transplantation for spinal cord injury: A Phase I pilot study. Cytotherapy 2016, 18, 518–522. [Google Scholar] [CrossRef]
  184. Vaquero, J.; Zurita, M.; Bonilla, C.; Fernández, C.; Rubio, J.J.; Mucientes, J.; Rodriguez, B.; Blanco, E.; Donis, L. Progressive increase in brain glucose metabolism after intrathecal administration of autologous mesenchymal stromal cells in patients with diffuse axonal injury. Cytotherapy 2017, 19, 88–94. [Google Scholar] [CrossRef]
  185. Vaquero, J.; Zurita, M.; Rico, M.A.; Bonilla, C.; Aguayo, C.; Montilla, J.; Bustamante, S.; Carballido, J.; Marin, E.; Martinez, F.; et al. An approach to personalized cell therapy in chronic complete paraplegia: The Puerta de Hierro phase I/II clinical trial. Cytotherapy 2016, 18, 1025–1036. [Google Scholar] [CrossRef] [Green Version]
  186. Mendonça, M.V.P.; Larocca, T.F.; de Freitas Souza, B.S.; Villarreal, C.F.; Silva, L.F.M.; Matos, A.C.; Novaes, M.A.; Bahia, C.M.P.; de Oliveira Melo Martinez, A.C.; Kaneto, C.M.; et al. Safety and neurological assessments after autologous transplantation of bone marrow mesenchymal stem cells in subjects with chronic spinal cord injury. Stem Cell Res. Ther. 2014, 5, 126. [Google Scholar] [CrossRef]
  187. Llufriu, S.; Sepúlveda, M.; Blanco, Y.; Marín, P.; Moreno, B.; Berenguer, J.; Gabilondo, I.; Martínez-Heras, E.; Sola-Valls, N.; Arnaiz, J.A.; et al. Randomized placebo-controlled phase II trial of autologous mesenchymal stem cells in multiple sclerosis. PLoS ONE 2014, 9, e113936. [Google Scholar] [CrossRef]
  188. Shichinohe, H.; Kawabori, M.; Iijima, H.; Teramoto, T.; Abumiya, T.; Nakayama, N.; Kazumata, K.; Terasaka, S.; Arato, T.; Houkin, K. Research on advanced intervention using novel bone marrOW stem cell (RAINBOW): A study protocol for a phase I, open-label, uncontrolled, dose-response trial of autologous bone marrow stromal cell transplantation in patients with acute ischemic stroke. BMC Neurol. 2017, 17, 179. [Google Scholar] [CrossRef]
  189. Hlebokazov, F.; Dakukina, T.; Ihnatsenko, S.; Kosmacheva, S.; Potapnev, M.; Shakhbazau, A.; Goncharova, N.; Makhrov, M.; Korolevich, P.; Misyuk, N.; et al. Treatment of refractory epilepsy patients with autologous mesenchymal stem cells reduces seizure frequency: An open label study. Adv. Med. Sci. 2017, 62, 273–279. [Google Scholar] [CrossRef]
  190. Liu, X.; Fu, X.; Dai, G.; Wang, X.; Zhang, Z.; Cheng, H.; Zheng, P.; An, Y. Comparative analysis of curative effect of bone marrow mesenchymal stem cell and bone marrow mononuclear cell transplantation for spastic cerebral palsy. J. Transl. Med. 2017, 15, 48. [Google Scholar] [CrossRef]
  191. Karussis, D.; Karageorgiou, C.; Vaknin-Dembinsky, A.; Gowda-Kurkalli, B.; Gomori, J.M.; Kassis, I.; Bulte, J.W.M.; Petrou, P.; Ben-Hur, T.; Abramsky, O.; et al. Safety and immunological effects of mesenchymal stem cell transplantation in patients with multiple sclerosis and amyotrophic lateral sclerosis. Arch. Neurol. 2010, 67, 1187–1194. [Google Scholar] [CrossRef]
  192. Mazzini, L.; Vercelli, A.; Ferrero, I.; Boido, M.; Cantello, R.; Fagioli, F. Transplantation of mesenchymal stem cells in ALS. Prog. Brain Res. 2012, 201, 333–359. [Google Scholar] [PubMed]
  193. Wang, L.; Lu, M. Regulation and direction of umbilical cord blood mesenchymal stem cells to adopt neuronal fate. Int. J. Neurosci. 2014, 124, 149–159. [Google Scholar] [CrossRef]
  194. Oh, K.W.; Moon, C.; Kim, H.Y.; Oh, S.I.; Park, J.; Lee, J.H.; Chang, I.Y.; Kim, K.S.; Kim, S.H. Phase I trial of repeated intrathecal autologous bone marrow-derived mesenchymal stromal cells in amyotrophic lateral sclerosis. Stem Cells Transl. Med. 2015, 4, 590–597. [Google Scholar] [CrossRef]
  195. Moviglia, G.A.; Fernandez Viña, R.; Brizuela, J.A.; Saslavsky, J.; Vrsalovic, F.; Varela, G.; Bastos, F.; Farina, P.; Etchegaray, G.; Barbieri, M.; et al. Combined protocol of cell therapy for chronic spinal cord injury. Report on the electrical and functional recovery of two patients. Cytotherapy 2006, 8, 202–209. [Google Scholar] [CrossRef]
  196. Syková, E.; Rychmach, P.; Drahorádová, I.; Konrádová, Š.; Růžičková, K.; Voříšek, I.; Forostyak, S.; Homola, A.; Bojar, M. Transplantation of Mesenchymal Stromal Cells in Patients with Amyotrophic Lateral Sclerosis: Results of Phase I/IIa Clinical Trial. Cell Transplant. 2017, 26, 647–658. [Google Scholar] [CrossRef] [Green Version]
  197. Geffner, L.F.; Santacruz, P.; Izurieta, M.; Flor, L.; Maldonado, B.; Auad, A.H.; Montenegro, X.; Gonzalez, R.; Silva, F. Administration of autologous bone marrow stem cells into spinal cord injury patients via multiple routes is safe and improves their quality of life: Comprehensive case studies. Cell Transplant. 2008, 17, 1277–1293. [Google Scholar] [CrossRef] [PubMed]
  198. Pal, R.; Venkataramana, N.K.; Bansal, A.; Balaraju, S.; Jan, M.; Chandra, R.; Dixit, A.; Rauthan, A.; Murgod, U.; Totey, S. Ex vivo-expanded autologous bone marrow-derived mesenchymal stromal cells in human spinal cord injury/paraplegia: A pilot clinical study. Cytotherapy 2009, 11, 897–911. [Google Scholar] [CrossRef]
  199. Zhang, Z.X.; Guan, L.X.; Zhang, K.; Zhang, Q.; Dai, L.J. A combined procedure to deliver autologous mesenchymal stromal cells to patients with traumatic brain injury. Cytotherapy 2008, 10, 134–139. [Google Scholar] [CrossRef] [PubMed]
  200. Lee, M.J.; Kim, J.; Kim, M.Y.; Bae, Y.S.; Ryu, S.H.; Lee, T.G.; Kim, J.H. Proteomic analysis of tumor necrosis factor-alpha-induced secretome of human adipose tissue-derived mesenchymal stem cells. J. Proteome Res. 2010, 9, 1754–1762. [Google Scholar] [CrossRef] [PubMed]
  201. Cheng, H.; Liu, X.; Hua, R.; Dai, G.; Wang, X.; Gao, J.; An, Y. Clinical observation of umbilical cord mesenchymal stem cell transplantation in treatment for sequelae of thoracolumbar spinal cord injury. J. Transl. Med. 2014, 12, 253. [Google Scholar] [CrossRef]
  202. Jiang, Y.; Zhu, W.; Zhu, J.; Wu, L.; Xu, G.; Liu, X. Feasibility of delivering mesenchymal stem cells via catheter to the proximal end of the lesion artery in patients with stroke in the territory of the middle cerebral artery. Cell Transplant. 2013, 22, 2291–2298. [Google Scholar] [CrossRef] [PubMed]
  203. Wang, S.; Cheng, H.; Dai, G.; Wang, X.; Hua, R.; Liu, X.; Wang, P.; Chen, G.; Yue, W.; An, Y. Umbilical cord mesenchymal stem cell transplantation significantly improves neurological function in patients with sequelae of traumatic brain injury. Brain Res. 2013, 1532, 76–84. [Google Scholar] [CrossRef]
  204. Li, J.F.; Zhang, D.J.; Geng, T.; Chen, L.; Huang, H.; Yin, H.L.; Zhang, Y.; Lou, J.Y.; Cao, B.; Wang, Y.L. The potential of human umbilical cord-derived mesenchymal stem cells as a novel cellular therapy for multiple sclerosis. Cell Transplant. 2014, 23, 113–122. [Google Scholar] [CrossRef]
  205. Nakamura, K.; Mieda, T.; Suto, N.; Matsuura, S.; Hirai, H. Mesenchymal stem cells as a potential therapeutic tool for spinocerebellar ataxia. Cerebellum 2015, 14, 165–170. [Google Scholar] [CrossRef]
  206. Cordes, A.L.; Jahn, K.; Hass, R.; Schwabe, K.; Weissinger, E.M.; Ganser, A.; Götz, F.; Dengler, R.; Krauss, J.K.; Petri, S. Intramedullary spinal cord implantation of human CD34+ umbilical cord-derived cells in ALS. Amyotroph. Lateral Scler. 2011, 12, 325–330. [Google Scholar] [CrossRef] [PubMed]
  207. Hur, J.W.; Cho, T.H.; Park, D.H.; Lee, J.B.; Park, J.Y.; Chung, Y.G. Intrathecal transplantation of autologous adipose-derived mesenchymal stem cells for treating spinal cord injury: A human trial. J. Spinal Cord Med. 2016, 39, 655–664. [Google Scholar] [CrossRef]
  208. Díez-Tejedor, E.; Gutiérrez-Fernández, M.; Martínez-Sánchez, P.; Rodríguez-Frutos, B.; Ruiz-Ares, G.; Lara, M.L.; Gimeno, B.F. Reparative therapy for acute ischemic stroke with allogeneic mesenchymal stem cells from adipose tissue: A safety assessment: A phase II randomized, double-blind, placebo-controlled, single-center, pilot clinical trial. J. Stroke Cerebrovasc. Dis. 2014, 23, 2694–2700. [Google Scholar] [CrossRef]
  209. Tsai, Y.A.; Liu, R.S.; Lirng, J.F.; Yang, B.H.; Chang, C.H.; Wang, Y.C.; Wu, Y.S.; Ho, J.H.C.; Lee, O.K.; Soong, B.W. Treatment of Spinocerebellar Ataxia with Mesenchymal Stem Cells: A Phase I/IIa Clinical Study. Cell Transplant. 2017, 26, 503–512. [Google Scholar] [CrossRef] [Green Version]
  210. Fernández, O.; Izquierdo, G.; Fernández, V.; Leyva, L.; Reyes, V.; Guerrero, M.; León, A.; Arnaiz, C.; Navarro, G.; Páramo, M.D.; et al. Adipose-derived mesenchymal stem cells (AdMSC) for the treatment of secondary-progressive multiple sclerosis: A triple blinded, placebo controlled, randomized phase I/II safety and feasibility study. PLoS ONE 2018, 13, e0195891. [Google Scholar] [CrossRef]
  211. Di Summa, P.G.; Kingham, P.J.; Raffoul, W.; Wiberg, M.; Terenghi, G.; Kalbermatten, D.F. Adipose-derived stem cells enhance peripheral nerve regeneration. J. Plast. Reconstr. Aesthetic Surg. 2010, 63, 1544–1552. [Google Scholar] [CrossRef]
  212. Staff, N.P.; Madigan, N.N.; Morris, J.; Jentoft, M.; Sorenson, E.J.; Butler, G.; Gastineau, D.; Dietz, A.; Windebank, A.J. Safety of intrathecal autologous adipose-derived mesenchymal stromal cells in patients with ALS. Neurology 2016, 87, 2230–2234. [Google Scholar] [CrossRef] [Green Version]
  213. Gu, F.; Wang, D.; Zhang, H.; Feng, X.; Gilkeson, G.S.; Shi, S.; Sun, L. Allogeneic mesenchymal stem cell transplantation for lupus nephritis patients refractory to conventional therapy. Clin. Rheumatol. 2014, 33, 1611–1619. [Google Scholar] [CrossRef]
  214. Sun, L.; Wang, D.; Liang, J.; Zhang, H.; Feng, X.; Wang, H.; Hua, B.; Liu, B.; Ye, S.; Hu, X.; et al. Umbilical cord mesenchymal stem cell transplantation in severe and refractory systemic lupus erythematosus. Arthritis Rheum. 2010, 62, 2467–2475. [Google Scholar] [CrossRef]
  215. Kong, D.; Zhuang, X.; Wang, D.; Qu, H.; Jiang, Y.; Li, X.; Wu, W.; Xiao, J.; Liu, X.; Liu, J.; et al. Umbilical cord mesenchymal stem cell transfusion ameliorated hyperglycemia in patients with type 2 diabetes mellitus. Clin. Lab. 2014, 60, 1969–1976. [Google Scholar] [CrossRef] [PubMed]
  216. Cai, J.; Wu, Z.; Xu, X.; Liao, L.; Chen, J.; Huang, L.; Wu, W.; Luo, F.; Wu, C.; Pugliese, A.; et al. Umbilical Cord Mesenchymal Stromal Cell with Autologous Bone Marrow Cell Transplantation in Established Type 1 Diabetes: A Pilot Randomized Controlled Open-Label Clinical Study to Assess Safety and Impact on Insulin Secretion. Diabetes Care 2016, 39, 149–157. [Google Scholar] [CrossRef] [PubMed]
  217. Dave, S.D.; Vanikar, A.V.; Trivedi, H.L. Co-infusion of adipose tissue derived mesenchymal stem cell-differentiated insulin-making cells and haematopoietic cells with renal transplantation: A novel therapy for type 1 diabetes mellitus with end-stage renal disease. BMJ Case Rep. 2013, 2013, 009901. [Google Scholar] [CrossRef]
  218. Riordan, N.H.; Ichim, T.E.; Min, W.P.; Wang, H.; Solano, F.; Lara, F.; Alfaro, M.; Rodriguez, J.P.; Harman, R.J.; Patel, A.N.; et al. Non-expanded adipose stromal vascular fraction cell therapy for multiple sclerosis. J. Transl. Med. 2009, 7, 29. [Google Scholar] [CrossRef]
  219. Liang, J.; Zhang, H.; Hua, B.; Wang, H.; Lu, L.; Shi, S.; Hou, Y.; Zeng, X.; Gilkeson, G.S.; Sun, L. Allogenic mesenchymal stem cells transplantation in refractory systemic lupus erythematosus: A pilot clinical study. Ann. Rheum. Dis. 2010, 69, 1423–1429. [Google Scholar] [CrossRef]
  220. Fodor, P.B.; Paulseth, S.G. Adipose Derived Stromal Cell (ADSC) Injections for Pain Management of Osteoarthritis in the Human Knee Joint. Aesthetic Surg. J. 2016, 36, 229–236. [Google Scholar] [CrossRef]
  221. Song, Y.; Zhao, H.Y.; Lyu, Z.S.; Cao, X.N.; Shi, M.M.; Wen, Q.; Tang, F.F.; Wang, Y.; Xu, L.P.; Zhang, X.H.; et al. Dysfunctional Bone Marrow Mesenchymal Stem Cells in Patients with Poor Graft Function after Allogeneic Hematopoietic Stem Cell Transplantation. Biol. Blood Marrow Transplant. 2018, 24, 1981–1989. [Google Scholar] [CrossRef] [Green Version]
  222. Koh, Y.G.; Kwon, O.R.; Kim, Y.S.; Choi, Y.J.; Tak, D.H. Adipose-Derived Mesenchymal Stem Cells with Microfracture Versus Microfracture Alone: 2-Year Follow-up of a Prospective Randomized Trial. Arthroscopy 2016, 32, 97–109. [Google Scholar] [CrossRef] [PubMed]
  223. Koh, Y.G.; Choi, Y.J.; Kwon, S.K.; Kim, Y.S.; Yeo, J.E. Clinical results and second-look arthroscopic findings after treatment with adipose-derived stem cells for knee osteoarthritis. Knee Surg. Sports Traumatol. Arthrosc. 2015, 23, 1308–1316. [Google Scholar] [CrossRef] [PubMed]
  224. Marcacci, M.; Kon, E.; Moukhachev, V.; Lavroukov, A.; Kutepov, S.; Quarto, R.; Mastrogiacomo, M.; Cancedda, R. Stem cells associated with macroporous bioceramics for long bone repair: 6- to 7-year outcome of a pilot clinical study. Tissue Eng. 2007, 13, 947–955. [Google Scholar] [CrossRef]
  225. Yamada, Y.; Nakamura, S.; Ito, K.; Umemura, E.; Hara, K.; Nagasaka, T.; Abe, A.; Baba, S.; Furuichi, Y.; Izumi, Y.; et al. Injectable bone tissue engineering using expanded mesenchymal stem cells. Stem Cells 2013, 31, 572–580. [Google Scholar] [CrossRef] [PubMed]
  226. Quarto, R.; Mastrogiacomo, M.; Cancedda, R.; Kutepov, S.M.; Mukhachev, V.; Lavroukov, A.; Kon, E.; Marcacci, M. Repair of large bone defects with the use of autologous bone marrow stromal cells. N. Engl. J. Med. 2001, 344, 385–386. [Google Scholar] [CrossRef] [PubMed]
  227. Chen, C.; Qu, Z.; Yin, X.; Shang, C.; Ao, Q.; Gu, Y.; Liu, Y. Efficacy of umbilical cord-derived mesenchymal stem cell-based therapy for osteonecrosis of the femoral head: A three-year follow-up study. Mol. Med. Rep. 2016, 14, 4209–4215. [Google Scholar] [CrossRef] [PubMed]
  228. Qu, Z.; Fang, G.; Cui, Z.; Liu, Y. Cell therapy for bone nonunion: A retrospective study. Minerva Med. 2015, 106, 315–321. [Google Scholar]
  229. Castillo-Cardiel, G.; López-Echaury, A.C.; Saucedo-Ortiz, J.A.; Fuentes-Orozco, C.; Michel-Espinoza, L.R.; Irusteta-Jiménez, L.; Salazar-Parra, M.; González-Ojeda, A. Bone regeneration in mandibular fractures after the application of autologous mesenchymal stem cells, a randomized clinical trial. Dent Traumatol. 2017, 33, 38–44. [Google Scholar] [CrossRef]
  230. Kebriaei, P.; Isola, L.; Bahceci, E.; Holland, K.; Rowley, S.; McGuirk, J.; Devetten, M.; Jansen, J.; Herzig, R.; Schuster, M.; et al. Adult human mesenchymal stem cells added to corticosteroid therapy for the treatment of acute graft-versus-host disease. Biol. Blood Marrow Transplant. 2009, 15, 804–811. [Google Scholar] [CrossRef]
  231. Lazarus, H.M.; Koc, O.N.; Devine, S.M.; Curtin, P.; Maziarz, R.T.; Holland, H.K.; Shpall, E.J.; McCarthy, P.; Atkinson, K.; Cooper, B.W.; et al. Cotransplantation of HLA-identical sibling culture-expanded mesenchymal stem cells and hematopoietic stem cells in hematologic malignancy patients. Biol. Blood Marrow Transplant. 2005, 11, 389–398. [Google Scholar] [CrossRef]
  232. Ringdén, O.; Uzunel, M.; Rasmusson, I.; Remberger, M.; Sundberg, B.; Lönnies, H.; Marschall, H.U.; Dlugosz, A.; Szakos, A.; Hassan, Z.; et al. Mesenchymal stem cells for treatment of therapy-resistant graft-versus-host disease. Transplantation 2006, 81, 1390–1397. [Google Scholar] [CrossRef]
  233. Le Blanc, K.; Rasmusson, I.; Sundberg, B.; Götherström, C.; Hassan, M.; Uzunel, M.; Ringdén, O. Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cells. Lancet 2004, 363, 1439–1441. [Google Scholar] [CrossRef]
  234. Gao, L.; Zhang, Y.; Hu, B.; Liu, J.; Kong, P.; Lou, S.; Su, Y.; Yang, T.; Li, H.; Liu, Y.; et al. Phase II Multicenter, Randomized, Double-Blind Controlled Study of Efficacy and Safety of Umbilical Cord-Derived Mesenchymal Stromal Cells in the Prophylaxis of Chronic Graft-Versus-Host Disease After HLA-Haploidentical Stem-Cell Transplantation. J. Clin. Oncol. 2016, 34, 2843–2850. [Google Scholar] [CrossRef]
  235. Chen, G.; Yang, T.; Tian, H.; Qiao, M.; Liu, H.; Fu, C.; Miao, M.; Jin, Z.; Tang, X.; Han, Y.; et al. Clinical study of umbilical cord-derived mesenchymal stem cells for treatment of nineteen patients with steroid-resistant severe acute graft-versus-host disease. Chin. J. Hematol. 2012, 33, 303–306. [Google Scholar] [CrossRef]
  236. Gonzalo-Daganzo, R.; Regidor, C.; Martín-Donaire, T.; Rico, M.A.; Bautista, G.; Krsnik, I.; Forés, R.; Ojeda, E.; Sanjuán, I.; García-Marco, J.A.; et al. Results of a pilot study on the use of third-party donor mesenchymal stromal cells in cord blood transplantation in adults. Cytotherapy 2009, 11, 278–288. [Google Scholar] [CrossRef]
  237. Macmillan, M.L.; Blazar, B.R.; DeFor, T.E.; Wagner, J.E. Transplantation of ex-vivo culture-expanded parental haploidentical mesenchymal stem cells to promote engraftment in pediatric recipients of unrelated donor umbilical cord blood: Results of a phase I-II clinical trial. Bone Marrow Transplant. 2009, 43, 447–454. [Google Scholar] [CrossRef]
  238. Vanikar, A.V.; Trivedi, H.L.; Kumar, A.; Gopal, S.C.; Patel, H.V.; Gumber, M.R.; Kute, V.B.; Shah, P.R.; Dave, S.D. Co-infusion of donor adipose tissue-derived mesenchymal and hematopoietic stem cells helps safe minimization of immunosuppression in renal transplantation—Single center experience. Ren. Fail. 2014, 36, 1376–1384. [Google Scholar] [CrossRef]
  239. Nesteruk, J.; Voronina, N.; Kundt, G.; Donndorf, P.; Klopsch, C.; Kaminski, A.; Duckers, H.J.; Steinhoff, G. Stem cell registry programme for patients with ischemic cardiomyopathy undergoing coronary artery bypass grafting: What benefits does it derive? ESC Heart Fail. 2017, 4, 105–111. [Google Scholar] [CrossRef]
  240. Houtgraaf, J.H.; de Jong, R.; Kazemi, K.; de Groot, D.; van der Spoel, T.I.G.; Arslan, F.; Hoefer, I.; Pasterkamp, G.; Itescu, S.; Zijlstra, F.; et al. Intracoronary infusion of allogeneic mesenchymal precursor cells directly after experimental acute myocardial infarction reduces infarct size, abrogates adverse remodeling, and improves cardiac function. Circ. Res. 2013, 113, 153–166. [Google Scholar] [CrossRef]
  241. Panfilov, I.A.; de Jong, R.; Takashima, S.I.; Duckers, H.J. Clinical study using adipose-derived mesenchymal-like stem cells in acute myocardial infarction and heart failure. Methods Mol. Biol. 2013, 1036, 207–212. [Google Scholar]
  242. Larijani, B.; Aghayan, H.; Goodarzi, P.; Mohamadi-Jahani, F.; Norouzi-Javidan, A.; Dehpour, A.R.; Fallahzadeh, K.; Azam Sayahpour, F.; Bidaki, K.; Arjmand, B. Clinical Grade Human Adipose Tissue-Derived Mesenchymal Stem Cell Banking. Acta Med. Iran. 2015, 53, 540–546. [Google Scholar] [PubMed]
  243. Zanata, F.; Shaik, S.; Devireddy, R.V.; Wu, X.; Ferreira, L.M.; Gimble, J.M. Cryopreserved Adipose Tissue-Derived Stromal/Stem Cells: Potential for Applications in Clinic and Therapy. Adv. Exp. Med. Biol. 2016, 951, 137–146. [Google Scholar]
  244. Roato, I.; Alotto, D.; Belisario, D.C.; Casarin, S.; Fumagalli, M.; Cambieri, I.; Piana, R.; Stella, M.; Ferracini, R.; Castagnoli, C. Adipose Derived-Mesenchymal Stem Cells Viability and Differentiating Features for Orthopaedic Reparative Applications: Banking of Adipose Tissue. Stem Cells Int. 2016, 2016, 4968724. [Google Scholar] [CrossRef]
  245. MacRae, J.W.; Tholpady, S.S.; Ogle, R.C.; Morgan, R.F. Ex vivo fat graft preservation: Effects and implications of cryopreservation. Ann. Plast. Surg. 2004, 52, 281–282. [Google Scholar] [CrossRef] [PubMed]
  246. Shoshani, O.; Ullmann, Y.; Shupak, A.; Ramon, Y.; Gilhar, A.; Kehat, I.; Peled, I.J. The role of frozen storage in preserving adipose tissue obtained by suction-assisted lipectomy for repeated fat injection procedures. Dermatol. Surg. 2001, 27, 645–647. [Google Scholar]
  247. Palumbo, P.; Lombardi, F.; Siragusa, G.; Cifone, M.G.; Cinque, B.; Giuliani, M. Methods of Isolation, Characterization and Expansion of Human Adipose-Derived Stem Cells (ASCs): An Overview. Int. J. Mol. Sci. 2018, 19, 1897. [Google Scholar] [CrossRef]
  248. Kode, J.A.; Mukherjee, S.; Joglekar, M.V.; Hardikar, A.A. Mesenchymal stem cells: Immunobiology and role in immunomodulation and tissue regeneration. Cytotherapy 2009, 11, 377–391. [Google Scholar] [CrossRef]
  249. Qayyum, A.A.; Kaur, K.P.; Mathiasen, A.B.; Haack-Sørensen, M.; Ekblond, A.; Kastrup, J. Influence of patient related factors on number of mesenchymal stromal cells reached after in vitro culture expansion for clinical treatment. Scand. J. Clin. Lab. Investig. 2017, 77, 541–548. [Google Scholar] [CrossRef]
  250. Varghese, J.; Griffin, M.; Mosahebi, A.; Butler, P. Systematic review of patient factors affecting adipose stem cell viability and function: Implications for regenerative therapy. Stem Cell Res. Ther. 2017, 8, 45. [Google Scholar] [CrossRef]
  251. Gimble, J.M.; Katz, A.J.; Bunnell, B.A. Adipose-derived stem cells for regenerative medicine. Circ. Res. 2007, 100, 1249–1260. [Google Scholar] [CrossRef]
Figure 1. Immuno-modulatory effects of adipose derived stem cells (ADSCs). ADSCs stimulate macrophage change immunity and inhibit T and Dendritic cells, inducing angiogenesis, a decrease in apoptosis and fibrosis with an increase in anti-inflammation process. Interleukin-1α, -6, -10 (IL-1α, -6, -10, -13), TNF-α (Tumor Necrosis Factor-α), TGF-β (Tumor Growth Factor-β), TLR2, TLR4 (Toll Like Receptor 2, 4), VEGF (Vascular Endothelial Growth Factor), b-FGF (basic Fibroblast Growth Factor).
Figure 1. Immuno-modulatory effects of adipose derived stem cells (ADSCs). ADSCs stimulate macrophage change immunity and inhibit T and Dendritic cells, inducing angiogenesis, a decrease in apoptosis and fibrosis with an increase in anti-inflammation process. Interleukin-1α, -6, -10 (IL-1α, -6, -10, -13), TNF-α (Tumor Necrosis Factor-α), TGF-β (Tumor Growth Factor-β), TLR2, TLR4 (Toll Like Receptor 2, 4), VEGF (Vascular Endothelial Growth Factor), b-FGF (basic Fibroblast Growth Factor).
Ijms 20 02523 g001
Figure 2. Secretome of Adipose Derived Stem Cells (ADSCs) that are involved in the mechanisms related to tissue repair and regeneration. ADSCs secrete different growth factors in their microenvironment and other proteins known to induce specific cell differentiation. Interleukin-1, -6, -10 (IL-1, -6, -10), TNF-α (Tumor Necrosis Factor-α), TGF-β (Tumor Growth Factor-β), TLR2, TLR4 (Toll Like Receptor 2, 4), GDF11 (Growth Differentiation Factor 11), GDF15 (Growth Differentiation Factor 15), G-CSF (Granulocyte-Colony Stimulating Factor), GM-CSF (Granulocyte Monocyte-Colony Stimulating Factor), EGF (Endothelial Growth Factor), VEGF (Vascular Endothelial Growth Factor), IGF (Insulin Growth Factor), b-FGF (basic Fibroblast Growth Factor), PDGF (Platelet Derived Growth Factor), NGF (Nerve Growth Factor), CCL-2 (Chemokine C-C Motif Ligand 2), PEDF (Pigment Epithelium Derived Factor), KGF (Keratinocyte Growth Factor).
Figure 2. Secretome of Adipose Derived Stem Cells (ADSCs) that are involved in the mechanisms related to tissue repair and regeneration. ADSCs secrete different growth factors in their microenvironment and other proteins known to induce specific cell differentiation. Interleukin-1, -6, -10 (IL-1, -6, -10), TNF-α (Tumor Necrosis Factor-α), TGF-β (Tumor Growth Factor-β), TLR2, TLR4 (Toll Like Receptor 2, 4), GDF11 (Growth Differentiation Factor 11), GDF15 (Growth Differentiation Factor 15), G-CSF (Granulocyte-Colony Stimulating Factor), GM-CSF (Granulocyte Monocyte-Colony Stimulating Factor), EGF (Endothelial Growth Factor), VEGF (Vascular Endothelial Growth Factor), IGF (Insulin Growth Factor), b-FGF (basic Fibroblast Growth Factor), PDGF (Platelet Derived Growth Factor), NGF (Nerve Growth Factor), CCL-2 (Chemokine C-C Motif Ligand 2), PEDF (Pigment Epithelium Derived Factor), KGF (Keratinocyte Growth Factor).
Ijms 20 02523 g002
Figure 3. Therapeutic features of Adipose Derived Stem Cells (ADSCs) in auto-immunes and hematopoietic diseases compared to those of bone marrow (BM)- and umbilical cord (UC)-mesenchymal stem cells (MSCs). The size line is related to the observed effect.
Figure 3. Therapeutic features of Adipose Derived Stem Cells (ADSCs) in auto-immunes and hematopoietic diseases compared to those of bone marrow (BM)- and umbilical cord (UC)-mesenchymal stem cells (MSCs). The size line is related to the observed effect.
Ijms 20 02523 g003
Table 1. Summary of results of previous clinical studies using ADSCs as compared to those with BM-and UC-MSCs. BM: bone marrow, UC: umbilical cord, ADSCs: adipose derived stem cells, MSCs: mesenchymal stem cells, Auto: autologous, Allo: allogenic, AD: dermatitis, ALS: amyotrophic lateral sclerosis, MS: multiple sclerosis, SCA: spinocerebellar ataxia, SLE: systemic lupus erythematosus, LN: lupus nephritis, SS: systemic sclerosis, GVHD: graft versus host disease, BMD: Becker muscular disease, DMD, Duchene muscular disease.
Table 1. Summary of results of previous clinical studies using ADSCs as compared to those with BM-and UC-MSCs. BM: bone marrow, UC: umbilical cord, ADSCs: adipose derived stem cells, MSCs: mesenchymal stem cells, Auto: autologous, Allo: allogenic, AD: dermatitis, ALS: amyotrophic lateral sclerosis, MS: multiple sclerosis, SCA: spinocerebellar ataxia, SLE: systemic lupus erythematosus, LN: lupus nephritis, SS: systemic sclerosis, GVHD: graft versus host disease, BMD: Becker muscular disease, DMD, Duchene muscular disease.
Stem Cell-Based TherapyReferencesCell OriginAuto/AlloAssociated EffectsApplication
Skin[84,85,86]BM-MSCsAutoModulation of inflammation, wound repairRadiation burns, burns
[87,88,89]]UC-MSCsAlloWound healing, skin improvement, neoangiogenesis and ulcer healing, decrease in features associated with ADBurns, wound healing, severe diabetic foot, Atopic Dermatitis (AD)
[10,11,12,16,17,90,91,92,93,94,95,96,97,98]ADSCsAutoWound healing, traumatic scars, facial tissue defects, post-mastectomy radiation, breast augmentation, facial rejuvenation, hair loss and growth, breast reconstitution, neogenesis across affected arteriesSoft tissue augmentation, esthetic remodeling, anti-aging, ulcers, burns, alopecia, lipodystrophies, critical limb ischemia
Nerve system[99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119]BM-MSCsAuto
Allo
Safety and efficacy, increased functional recovery, improvement, slowdown of Amyotrophic Lateral Sclerosis (ALS) progression, safety in ALS, spinal cord repair, expanded disability scale score improvement, improvement treatment of MSIschemic stroke, traumatic brain injury, spinal cord injury, ALS, chronic spinal cord lesions, multiple sclerosis (MS), Parkinson, spastic cerebral palsy, refractory epilepsy, chronic complete paraplegia
[5,120,121,122,123,124,125]UC-MSCsAlloRecovery of neurologic function, neuroprotection from sclerosis, delay SCA progression, safety and effectiveness, feasible and safety approach in stroke, self-care in patientsALS, hereditary spinocerebellar ataxia (SCA), MS, Stroke in Middle Cerebral Artery, Traumatic Brain Injury sequelae
[126,127,128,129,130,131]ADSCsAutoRecovery from ischemia, safetySpinal cord injury, ALS, MS
Autoimmune disorders[132,133,134,135]BM-MSCsAutoimprovement the clinical condition, reduction of Crohn’s disease and perianal diseases activityCrohn’s diseases, perianal diseases, SLE
[112,132,136,137,138,139,140]UCB-MSCsAllo
Allo
Safety and improvement of insulin secretion after HSC transplantation, renal remission for LN patients, safety and increase T cell level, partial disease remission, Safety with adverse eventsDiabete I, Diabete II, Lupus Nephritis (LN), Systemic Lupus Erythematosus (SLE), systemic sclerosis (SS), Sjörgren’s syndrome
[15,141,142,143,144,145,146,147,148]ADSCsAuto
Allo
Proinflammatory cytokine decrease, improvement safety and efficacy treatment, Generation of Insulin-secreting cells, improvement in disease activity and safetyperianal and rectovaginal fistules, Crohn’s diseases, Type 1 Diabete Mellitus, refractory SLE Polymyolitis, SS
Cartilage[63,117]BM-MSCsAutoGood integration in boneknee chondral lesions, cartilage defects
[149]UC-MSCsAlloCartilage regenerationOsteoarthritis
[19,22,150,151,152,153,154,155,156,157,158]ADSCsAllo
Auto
Tendon fibers arrangement, suppressive activity and decrease inflammatory responses, Safety, Proinflammatory cytokine decrease, improvement of knee jointIntervertebral disc damage, Rheumatoid arthritis disease, Osteoarthritis
Bone[159,160,161]BM-MSCsAutoLarge bone diaphysis, good scaffold integration with host boneDurable bone regeneration, bone defects, bone segment loss, bone diaphysis defects
[162,163]UCB-MSCsAlloDecreased in healing time in bone callus formation and marrow flowBone nonunion, Necrosis of Femoral Heads
[164,165,166]ADSCsAuto
Allo
Bone regeneration, calvarial continuityMaxillary reconstitution, traumatic calvarial defects
Immune and Hematological Disorders[167,168,169,170,171,172,173,174]BM-MSCsAllo,
Auto
Improvement of HSC engraftment, Lymphocyte recovery, Hematopoietic recovery, Safety and no adverse eventsAcute and severe GVHD, Hematopoietic malignancy
[175,176,177,178,179,180]UC-MSCsAlloNeutrophil and platelet engraftment, successful engraftment, Safety, Reduced graft failure, Decrease in GVHD symptomsAcute leukemia, Hematologic malignancy, GVHD, Steroid-resistant severe and acute GVHD
[14,181,182]ADSCsAuto,Improvement of HSC engraftment, immunosuppressive activity, short term hematopoiesis, Renal transplantation immuno-suppressive minimizationAcute and chronic GVHD,
Hematopoiesis support, engraftment
Cardio-vascular and Muscle[183,184,185,186]BM-MSCsAuto, Alloimproved left ventricular function, decreased cardiac arrhythmias, improvement myocardial function, Decrease in infarct sizeHeart infarct, Ischemic cardiomyopathy
[187,188,189,190]UCB-MSCsAlloImprovement of muscle size and activity, stabilize muscle power in DMD, decrease in infarct size, improve left ventricular functionBecker muscular disease (BMD), Duchene Muscular Disease (DMD), Coronary chronic total occlusion
[191,192,193,194,195]ADSCsAuto
Allo
Reduction in myocardial scar formation, improvement of cardiac function, safe and efficientMyocardial infarction, ischemic heart disease, Heart failure

Share and Cite

MDPI and ACS Style

Mazini, L.; Rochette, L.; Amine, M.; Malka, G. Regenerative Capacity of Adipose Derived Stem Cells (ADSCs), Comparison with Mesenchymal Stem Cells (MSCs). Int. J. Mol. Sci. 2019, 20, 2523. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20102523

AMA Style

Mazini L, Rochette L, Amine M, Malka G. Regenerative Capacity of Adipose Derived Stem Cells (ADSCs), Comparison with Mesenchymal Stem Cells (MSCs). International Journal of Molecular Sciences. 2019; 20(10):2523. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20102523

Chicago/Turabian Style

Mazini, Loubna, Luc Rochette, Mohamed Amine, and Gabriel Malka. 2019. "Regenerative Capacity of Adipose Derived Stem Cells (ADSCs), Comparison with Mesenchymal Stem Cells (MSCs)" International Journal of Molecular Sciences 20, no. 10: 2523. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20102523

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop