Next Article in Journal
Modulation of the Unfolded Protein Response by Tauroursodeoxycholic Acid Counteracts Apoptotic Cell Death and Fibrosis in a Mouse Model for Secondary Biliary Liver Fibrosis
Next Article in Special Issue
Micro-Computed Tomography Detection of Gold Nanoparticle-Labelled Mesenchymal Stem Cells in the Rat Subretinal Layer
Previous Article in Journal
The Potential of Targeting Ribosome Biogenesis in High-Grade Serous Ovarian Cancer
Previous Article in Special Issue
Cannabidiol Modulates the Expression of Alzheimer’s Disease-Related Genes in Mesenchymal Stem Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mesenchymal Stem Cells from Adipose Tissue in Clinical Applications for Dermatological Indications and Skin Aging

1
Aelan Cell Technology, San Francisco, CA 94107, USA
2
Division of Plastic Surgery, University of California, San Diego, La Jolla, CA 92121, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2017, 18(1), 208; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18010208
Submission received: 25 October 2016 / Revised: 5 January 2017 / Accepted: 5 January 2017 / Published: 20 January 2017

Abstract

:
Operating at multiple levels of control, mesenchymal stem cells from adipose tissue (ADSCs) communicate with organ systems to adjust immune response, provide signals for differentiation, migration, enzymatic reactions, and to equilibrate the regenerative demands of balanced tissue homeostasis. The identification of the mechanisms by which ADSCs accomplish these functions for dermatological rejuvenation and wound healing has great potential to identify novel targets for the treatment of disorders and combat aging. Herein, we review new insights into the role of adipose-derived stem cells in the maintenance of dermal and epidermal homeostasis, and recent advances in clinical applications of ADSCs related to dermatology.

Graphical Abstract

1. Introduction

The skin is a vital organ that functions not only as a protective barrier against environmental factors, but also in the synthesis, processing and metabolism of structural biomolecules such as lipids, proteins and glycans, and in the production and secretion of hormones. Skin aging is a complex biological process that becomes evident through the individual’s appearance and is caused by intrinsic factors such as structural and hormonal, and by environmental/extrinsic factors such as UV (longwavelength ultraviolet radiations (UVA) and medium wavelength ultraviolet radiations (UVB) exposure) radiations and chemicals [1,2]. Dermatological aging is driven by deficiencies in (a) important intrinsic cellular processes such as DNA repair and stability; mitochondrial function; cellular control over cell cycle and apoptosis; cellular metabolism; as well as (b) progressive loss of epidermal homeostasis such as the integrity of the extracellular matrix; changes in hormonal levels and the ability of complex cellular systems to provide communication between all of the counterparts. The physiological changes associated with aging of the skin are manifested in xerosis, dramatic loss of skin elasticity due to damage to collagen and elastin fibers; as well as barrier function, striae, modification of rhytides and deficiencies in the regenerative property of the skin (efficient turnover of epidermal cells) all of which ultimately result in thinning of the skin, malar fat atrophy and pigmentary changes [2,3].
The regenerative property of skin is essential for maintenance of the skin barrier during normal developmental process and for wound healing. Impaired skin regeneration results in debilitating effects on the organism in general and is a leading cause of a number of pathological conditions. The regenerative ability of skin is maintained by the stem cells (SCs) that are present in the skin to provide for cellular turn over during skin homeostasis and repair upon injury [4,5]. Whereas the morphogenetic changes that are associated with SC-driven regeneration and orchestrated cell stratification are intensively investigated, the underlying molecular mechanisms which execute such complex decision-making processes such as cell proliferation, differentiation and migration within the epidermis remain poorly understood.
Interplay between multiple signaling pathways on genetic and epigenetic levels seems to be important for regenerative properties of the skin SCs [6,7,8,9,10]. During adult skin homeostasis, the resident SCs from the different stem cell niches, including the hair follicle (HF), interfollicular epidermis [11] and sebaceous glands (SGs) have been proven to be important in the maintenance of healthy dermis and epidermis. In addition to SCs, other types of epidermal progenitors have come to the light in recent investigations, suggesting no less complex mesenchymal-epithelial crosstalk that governs skin regeneration through the secreted stimulatory factors: cytokines, chemotactic factors extracellular matrix (ECM) remodelers, growth factors and their inhibitors [12,13].
In parallel, recent clinical research points to the fact that adult stem cells of mesenchymal origin from the perivascular adipose depots can be successfully deployed for the treatment of several skin conditions. They can boost skin regeneration after injuries and those due to natural aging-related decline [14]. In this review, we will discuss the applications of adipose–derived stem cells ADSCs aimed for more efficient re-epithelialization and highlight the current view on the cascade of the factors essential for skin regeneration, many of which are secreted by these cells. We will touch upon the complex and the hierarchical relations between ADSCs and numerous reservoirs of long-term SCs and progenitors found to reside in the skin epithelium, as well as review the recent conceptual paradigms aimed to explain how these interactions modulate SC niches in a remarkably orchestrated fashion to allow different resident niches to be appropriately fueled during skin homeostasis and how interruptions in the spatial and temporal order of these events result in pathological conditions. Current research has raised many new fascinating questions and has unraveled new and exciting paradigms through which one can seek an understanding of how aging influences a number of the functional relationships important for skin regeneration and repair. We will attempt to render these new ideas in this manuscript.

2. Skin Structure and Cellular Composition as It Relates to Regenerative Medicine

Before we dive into the complexity of biological pathways governing skin regeneration and repair, let us take a look into the structure and composition of the skin. The human skin is the largest organ in the body that performs numerous indispensible physiological functions, such as: protection from exogenous chemical, physical, immune and pathogen insults; defense against free radical and UV radiation; major participation in thermoregulation and thermoisolation; and, importantly, is the organ entrusted with significant endocrine function burden (vitamin D synthesis, peripheral conversion of pro-hormones) [15]. Its thickness varies from 0.05 to 2 mm and is composed of four main layers shown in Figure 1.
The external, highly organized structure, stratum corneum, 10–20 μm thick, is highly hydrophobic and contains 10–15 layers of interdigitated dead cells called corneocytes. It is a major barrier to the penetration of environmental factors, as well as “active ingredients” included in cosmetic and dermatological products. Often defined as a “brick and mortar” structure, the corneocytes of hydrated keratin comprise the “bricks”; and multiple lipid bilayers semicrystalline composed of fatty acids, ceramides, cholesterol and cholesterol esters are viewed as a “mortar” holding the structure [16,17]. Skin penetration by topical agents occurs via this intercellular microroute and numerous techniques exist to provide for temporary disruption of this highly organized structure [18] shown in Figure 1A.
The second outermost layer is viable epidermis, which originates from the ectoderm during embryonic development (Figure 1) [19,20]. It is (~100–150 μm thick) and is often divided into stratum lucidium, stratum granulosm, stratum spinosum, and stratum germinatevum (basale). All these sub-layers are composed of keratinocytes at various stages of differentiation. Keratinocyte is the predominant cell type in the epidermis, constituting 95% of the cells. The keratinocytes found in the basal layer (stratum germinatevum) of the epidermis are sometimes referred to as “basal cells” or “basal keratinocytes”. In addition to the keratinocytes, numerous other cell types can be found in the epidermis (dendritic T cells, adipose cells, melanocytes, Merkel cells, Langerhans cells, and epidermotropic lymphocytes). Numerous active catabolic enzymes including, but not limited to, lipases, phosphatases, esterases, nucleotidases and proteases are present in the intercellular space as shown in Figure 2 and reviewed in [19]. As mesenchymal cells populate the skin, they transmit signals that instruct the stratification of the epidermis during development and dictate the positioning of downgrowths that mark the initiation of hair follicle (HF) morphogenesis [21]. The epidermis is subjected to homeostatic regulation during maturation as the stratum basale cells undergo mitotic proliferation during which the daughter cells move outwards up from basal to suprabasal to spinosum. The migrating cells change their morphology as they undergo multiple stages of cell differentiation due to deployment of dedicated transcriptional programs. Upon migration and entry of the spinous layer, the cellular genome undergoes changes in its transcriptional programs. For instance, the expression of genes encoding keratin 5 and 14 (KRT5/K5 and KRT14) is turned off, and the transcriptional upregulation of genes encoding KRT1 and KRT10 is turned on. This switch in the keratin production is viewed to be instrumental to forging a robust, interlinked with desmosomes an intermediate filament (IF) proteins network [22]. This IF network is viewed as a resilient structural scaffold aimed to reinforce cell–cell junctions and provide a resistance against mechanical stresses. At the late stage of terminal differentiation, granular cells express structural proteins that, when cross-linked, form structural 3-D support for lipid bilayers that are extruded from intracellular lamellar granules into the extra-cellular space between squamous (dead stratum corneum cells). This process may be viewed as “waterproofing” or sealing the skin surface. Eventually terminally differentiated cells shed from the stratum corneum surface (desquamation).
The innermost basement membrane layer is rich in extracellular matrix (ECM) proteins and growth factors. The formation of this layer takes its humble beginnings as a collaboration between the mesenchyme and stratifying epidermis, both of which participate in the production and organization of the basement membrane. The basement membrane that is attached to the epidermis provides a physical boundary between the epithelium and the third layer of the skin, dermis. It also serves as “a provision support center” providing growth-promoting factors, enzymes and structural components. The structural composition of the dermis is saturated with nerves, blood vessels, hair follicles, and sebaceous and sweat glands (shown in Figure 1). The presence of various lineages of dermal fibroblasts is important for manufacturing and supporting the polymerization of collagen and elastin fibers comprising ECM, a structure that provides for the elasticity of the dermis (Figure 2). Dermal adipose cells, together with mast cells and infiltrating leukocytes, are also present in this skin layer and shown in Figure 1B. Skin homeostasis and remarkable resilience of the skin is provided by SCs, which reside in different niches of the skin. Recent studies have begun to explain the mysteries behind these special “fountains of youth” (for extensive review [4]).
The last layer, the hypodermis or subcutaneous layer, is encompassed by adipose cells, mesenchymal stem (stromal) cells (MSCs), blood and lymph vessels (Figure 1). Numerous studies demonstrate that adipose tissue provides a remarkable influence on the microenvironment by the secretion of a wide-range of bioactive factors with a diverse set of functions. This includes lipid metabolism, energy balance and insulin sensitivity, which also have proven to be important in the regulation of angiogenesis, immunomodulation and inflammatory response. The secretory repertoire of adipose tissue can influence tissue and organ homeostasis at the autocrine, paracrine and/or endocrine, and gene expression level [23]. In addition, it has been reported that mesenchymal stem cells contain 500 times higher per gram of subcutaneous adipose tissue than bone marrow [24,25,26,27]. A complex and interconnected relationship has been shown between hematopoietic cells, fibroblasts and keratinocytes during skin homeostasis; and yet the contribution of the SC from subcutaneous and intradermal adipose tissue depots and intradermal adipocytes themselves have just recently been investigated despite the intensive use of these cells in plastic and reconstructive surgery [28,29,30,31]. Functional analysis of rodent models demonstrate that adipose depots are involved in mediation of the communication among multiple cell types, as well as in coordination and management of keratinocyte and fibroblast proliferation and migration in order to ensure epidermal and dermal repair [12,32,33]. Consequently, the beneficial therapeutic effects in the intradermal and subcutaneous adipose compartments could be attributed to the capacity of resident mesenchymal SCs, not only to heal sites of inflammation and injury, but also for their ability to secrete numerous pro-regenerative, anti-fibrotic and anti-apoptotic signaling molecular and growth factors integral to endogenous repair processes [34,35]. In other words, adipose tissue-resident mesenchymal stem cells could well be viewed as “endogenous factories” producing trophic mediators able to support all of the functional skin layers to ensure skin homeostasis, regeneration and repair.

3. Dermatological Aging and Practical Use and Significance of Non-Invasive Methods for Its Assessment

Although dermatological aging is a natural process that is caused and modified by intrinsic and external factors, it inevitably leads to changes that are physical (appearance), structural, and physiological, affecting barrier function and neurosensory perception. The key structural changes are caused by proteins, glycosaminoglycan, water, and lipids; and the structural modifications in these biological molecules have been reviewed previously [36,37,38]. Structurally, aging skin undergoes thinning of the epidermis that is caused by the reduction of vascularity and hydration without change in the number of epidermal layers [39,40]. On average, the thickness of the epidermis is reduced by about 6.4% during each decade of aging and there is a decreased number of mast cells and fibroblasts [40,41,42]. The thinning may also be caused by the decrease in the keratinocyte proliferation rate and by the reduction of the glycoprotein levels; for example, epidermal transmembrane keratinocyte glycoprotein. CD44 positive cells have been reported to have a regulatory role in proliferation of keratinocytes and in the maintenance of hyaluronic acid homeostasis decreases with aging [43,44]. Decreased production of collagen, elastin, glycosaminoglycans and hyaluronic acid due to the reduction in total fibroblasts leads to the thinning of dermis [1]. The extracellular matrix (ECM) changes associated with aging have also been attributed to circulating sugars and glycation end-products (AGEs). AGEs are generated as a result of non-enzymatic glycation of amino groups on proteins by reducing sugars, following the Maillard reaction, impacting on the changes in ECM structures [45,46]. AGEs are believed to activate the downstream signaling pathways responsible for increased oxidative stress and inflammation ultimately leading to tissue damage and a number of the diseases associated with aging that have been reviewed in detail [47]. Structural changes in the dermis occur because of a higher degree of calcification in the aged skin, degradation of elastin fibers, and changes in the organization of collagen bundles as they become disorganized with aging [48]. These ECM molecules are critical for providing structural support and in the regulation of signaling pathways for normal growth and development of skin cells, as well as during wound healing.
Skin aging effects are external as well as intrinsic, but they can be monitored/measured based on the skin appearance: texture and roughness, fine lines and wrinkles, structure, elasticity, hydration and barrier function. Many new non-invasive or minimally invasive bioengineering advances in recent years have enabled the quantitative analysis of skin properties during the aging process [49,50,51]. Trans-epidermal water loss (TEWL) is widely used for measuring the changes in the skin barrier integrity [51,52]. TEWL is defined as the measurement of the water that passes through the epidermis to the surrounding area by diffusion and evaporation. Skin corneometer measures electrical capacitance of skin, thereby measuring the extent of stratum corneum hydration [52]. Confocal Raman spectroscopy has been used extensively to measure the in vivo skin penetration of biological molecules, such as retinol, as it allows for the quantitation of water and other molecules in the skin [53,54]. This sophisticated technique relies on the Raman (inelastic) of monochromatic light and presents a highly detailed spatial resolution. Videomicroscopy and Fringe Projection are non-contact quantitative techniques for skin surface changes, while the Dermal Torque meter, Ballistometer and Cutometer are used for measuring skin firmness and elasticity [52,55]. Recently, Hahn et al. 2016, measured skin wrinkles by PRIMOS Lite 3D Face and Skin Scanner Analyzing System and used the DermaLab USB elasticity probe (Cortex Technology ApS, Hadsund, Denmark) for skin elasticity measurement [50].
The latest non-invasive non-contact techniques such as phase-shift rapid in vivo measurement of skin (PRIMOS) and surface evaluation of living skin (SELS) using Visioscan VC 98 rely on the use of optical systems for directly measuring skin roughness [56,57]. These systems are easy and time efficient as compared to the techniques that needed skin replica to be generated. Recent studies compared the data obtained using PRIMOS and Visioscan VC98 and concluded that Visioscan VC98 measurements generated highly reliable and consistent results for the parameters measured [56,57].
In addition, developments of novel devices made it possible to evaluate not only skin morphology but also molecular processes within the skin by noninvasive instrumental measuring techniques [26,40,51,52,55,58,59]. Raman spectroscopy is a non-destructive analytical method for analysis of molecular compounds [60]. Using this technique, Gniadecka, et al. [60] compared changes in the collagen properties such as fragmentation, thickening, solubility and arrangement of collagen fibers in photoaged vs. chronologically aged skin. These techniques can be successfully applied to quantify and objectively assess the efficacy of the products or procedures, as well as to decipher important signaling pathways important for skin homeostasis.
Next, we will discuss the signaling pathways that outline the complexity of the balanced interactions within skin infrastructure and its impact on derma aging.

4. Signaling Pathways of Cellular Interactions within Skin

All major cells such as keratinocytes, fibroblasts, macrophages, adipocytes and SCs within the skin layers have a capacity to communicate between themselves and interact with their environment. The internal and external stimuli can trigger the complex changes in these cell-to-cell communication pathways to orchestrate the appropriate responses [61]. The skin cell systems use several clearly defined signaling pathways that will transfer information from the cell surface to the genome to facilitate gene activities.
Mouse genetics studies have uncovered multiple signaling pathways of great importance for different events related to skin homeostasis. The complexity of skin differentiation and epidermal stratification, wound healing and regeneration in development and disease settings, is orchestrated by multiple spatially and temporarily coordinated pathways including Wnt, Notch, mitogen-activated protein kinase (MAPK), nuclear factor-κB (NF-κB) and integrin-linked kinases. Numerous transcriptional factors that drive developmental cascades and cellular viability transcriptional programs seem to be enlisted to cross-communicate the instructions of the signaling pathways to the gene targets, such as transcriptional regulators p63 (which is related to p53), the AP2 family, the CCAAT/enhancer-binding protein (C/EBP), Kruppel-like factor 4 (KLF4), transforming growth factor (TGF-β) activation and numerous others [10,21,62,63,64,65,66].
The interplay between critical signaling pathways, gene transcription and the production of growth factors imposes very specific influences (instructions) on heterogeneous cell compositions in normal and pathological settings. The full complexity of these elaborate cascades is just beginning to emerge. In particular, increased understanding of the role of growth factors, cytokines and matrikines in skin rejuvenation and regeneration, wound repair, and reversal of photo-aging has provoked a great deal of interest, resulting in numerous studies aimed to evaluate their role in repair, regeneration and remodeling of dermal infrastructure. A large number of cosmeceutical products include in their composition either a single or combination of multiple human growth factors. Cytokines and matrikines are currently marketed for skin repair and rejuvenation. The results of the clinical assessment of some of these topical products show beneficial effects in reducing the signs of facial skin aging thus supporting skin homeostasis [67,68,69,70,71,72,73,74].
We have summarized some, but not all, of the most interesting factors in Table 1. Our hope is to give the reader a high-level view of an ever-expanding realm of the inter-connective pathways and their immediate read-outs by specific and/or cohort of the targeted cells leading to important functional outcomes for skin homeostasis, regeneration and repair functions. This is essential for the critical evaluation of a plethora of emerging claims regarding regenerative dermatological therapies.
Several studies emphasized the role of interleukins and interleukin receptors in the skin. These factors and their receptors have an essential role in regulation of the inflammatory stage of wound healing and regeneration [116,117,118]. In addition, low doses of interleukins are found to have an anti-aging effect (see Table 1) and [119,120].
An intriguing example worthy of discussion in the context of this review is insulin growth factor (IGF-1) and its binding proteins (IGFBP1, IGFBP2, IGFBP3). The ratio between IGF-1 and IGFBP3, have been associated with facial aging and skin wrinkling [103]. IGF binding protein 1 (IGFBP1) also regulates the signals generated by adhesion receptors and growth receptors, thereby regulating the cellular processes. In addition, the cross talk between insulin-like growth receptor and β1-integrin receptor has been reported to play an important role in the prolidase activity and collagen biosynthesis [102], which together with IGFBP1 and 2 and their proteolytic fragments, have been shown to improve tissue repair process under inflammatory conditions, by acting upon the pathways that control proliferation and migration of HDFs [121].
Human dermal growth factors and cytokines are important for collagen, elastin and hyaluronan production [122]. Hyaluronan (HA) synthesis and regulation of its degradation is essential for maintenance of extracellular matrix homeostasis. Alteration in the expression of hyaluronic acid (HA) and its metabolizing enzymes is associated with dermatological aging of the human skin [123]. Notably, TGF-β1, bFGF, EGF, and PDGF-BB are commonly linked to enhancement of the total amount of HA production in skin fibroblasts through the targeted up-regulation of HA synthase expression [124]. It has been reported that fibroblasts treated with growth factors are capable of triggering healing and regenerative processes such as proliferation, angiogenesis and extracellular matrix component (ECM) synthesis. This results in the release of VEGF and HGF by fibroblasts and it has been reported that enhanced synthesis of type I collagen and hyaluronic acid (HA) can be accomplished in these experimental conditions [125]. In addition, recently it has been shown that VEGFR are involved in cell migration [101,126]. PDGF signaling and its receptor is involved in hair follicle morphogenesis [127].
Intriguingly, recently it has been postulated that dermal adipocytes secrete adipocytokines, such as adiponectin and leptin. Sensing these adipokines by human dermal fibroblasts expressing receptors for adiponectin and leptin results in the significant increased production of hyaluronic acid (HA) and collagen, a major extracellular matrix component (ECM) of dermis [128]. In accordance with these findings, it has been shown that leptin promotes wound healing in the skin [129]. Among these findings then another member of the TGF-β superfamily, Activin A, have shown to be involved in physiological and pathological processes associated with tissue homeostasis, cell differentiation and, ultimately, with wound healing [130].
Some of the listed protein factors, especially the transforming growth factor-β (TGF-β), α-granule-derived factors such as platelet-derived growth factor (PDGF), fibroblast growth factor (FGF-2/bFGF), epidermal and hepatocyte growth factors EGF and HGF, insulin-like growth factor (IGF), and angiogenic vascular endothelial growth factor (VEGF) have been commonly called skin matrikines and assigned a functional significance in wound-healing and/or in skin remodeling and regeneration [131,132]. Matrikines themselves or their hydrolytic peptides can link a matching cell-surface receptor to a transducer in order to relay information into the cell(s) using various signaling pathways. For example, TGF-β-PI3K axis that governs collagen-1 production (shown in Figure 3)—to be discussed below. In simplistic terms, each matrikine works like a key to a keyhole. Different matrikines can “unlock different doors”, relay a different messages to cell systems and trigger different functional outcomes.
Next we will tease apart some mechanistic aspects of these complex cascades by taking a closer look into skin wound healing process.

5. Wound Healing Paradigms

Skin is a self-repairing organ and maintains homeostasis through very organized yet complex mechanisms. Any injury or damage to skin results in cutaneous wounds that either heal itself or require an intervention based on the etiology. Understanding of this process of wound healing is important to tease apart critical signaling circuitry in communication of multiple cell types that coordinates proliferation, migration and factors secretion during epidermal and dermal homeostasis. While it is important to acknowledge that wound healing process is different from skin regeneration in its core, we believe that knowledge gained from its building blocks will shed light into the regenerative process itself and will empower many therapeutic approaches in medical practice for treating pathological conditions and aging.
Effective wound healing leads to the restoration of skin tissue integrity and occurs through a highly organized multistage and truly dynamic process that involves distinct stages such as (1) inflammation; (2) proliferation and granularization and; finally, (3) re-epithelialization (shown in Figure 3).
Wound healing process is initiated following changes in fibrin assembly at the wound site and recruitment of SCs in the skin, where functional activities will proceed through all of three distinct stages for successful wound healing [109,133,134].
The first inflammatory stage is triggered by fibrin changes and is immediately followed by hemostasis and migration of inflammatory cells and mediators of inflammatory response to the wound. This includes monocytes, neutrophils, macrophages as well as SCs from different niches, including cells from dermal and subcutaneous adipose depots. All these cells secrete growth factors and cytokines that regulate the signaling cascade and immunological pathways for wound healing [33,135,136,137]. Several studies show the role of interleukin receptors in the skin. These receptors have an essential role in the regulation of the inflammatory stage of wound healing and regeneration (see Table 1 and [116]). The natural course of wound healing at this stage involves remodeling of dermal collagen and other matrix molecules at the initial inflammatory phase, which has been reported to manifest massively high levels of matrix metalloproteinases (MMPs) that degrade the fragmented collagenous matrix [138,139,140]. Growth factors and cytokines such as TGF-β, TNF-α, PDGF, IL-1, IL-6, and IL-10 participate in the step-wise balancing act between development of inflammation and its rapid resolution [141].
Similar to above, the spatial-temporal process involving numerous growth factors and cytokines marks the transition from inflammatory phase of wound healing to the second (proliferative) granulation phase. This process engages FGFs, TGF-α, TGF-β, PDGF, HGF, EGF, IGF-1, as well as CSF, IL2, IL6, IL8, and TNF-α [29,33,79,95,106,109,110,111,142,143,144,145,146,147]. Several transmembrane adhesion proteins have been shown to be expressed at this stage. For example, CXCL16 is a transmembrane adhesion protein and is expressed by epidermal keratinocytes of normal skin upon wound injury or photodamage [148] and CXCL10 was demonstrated to have a role in the production of type I collagen, elastin and hyaluronan in dermal fibroblasts [122]. Granularization stage/proliferative stage is initiated by the proliferation, accumulation and migration of fibroblasts and epidermal keratinocytes, which are guided by the presence of environment growth factors and cytokines. The fibroblasts and keratinocytes themselves also secrete intricate cascades of matrikines. Multiple metabolic pathways lead to the formation of new collagen and to the repair of extracellular matrix during the granulation phase. Many cell adhesion molecules have recently been proven to be important modulators of fibroblast, keratinocytes and immune cell migration and coordination of their function by providing the EMC communication network in skin at this stage. For instance, the immunoglobulin superfamily glycoprotein, activated leukocyte cell adhesion molecule (ALCAM), has been implicated in the processes of cell adhesion and migration, and higher levels of ALCAM promote wound healing [149]. VE-Cad, an adhesion molecule of intracellular junction, is required for angiogenesis and has been shown to be associated with VEGF binding pathways [150,151]. This stage is marked not only by substantial and extended production of new undamaged collagen, but also by initiation of angiogenesis at the wound site resulting in new blood vessel formation. Studies in rodents and other models have reported that expression of the related tyrosine kinase with immunoglobulin and epidermal growth factor homology (Tie)-1 and -2 receptors, Ang-1 and -2, as well as FGFs, VEGFs, are important regulators of the angiogenesis [152,153,154,155]. That stage is followed by the epithelialization process by keratinocytes [156].
The final third stage of wound healing is the re-modeling phase. It is a prolonged phase that eventually results in gaining the skin’s integrity and properties. The transition from granulation to wound re-epithelialization marks the beginning of dermal tissue remodeling. It is associated with skin peeling. During this stage, the intermediate ECM structure composed of low strength, unorganized type III collagen and elastin become replaced by the ECM comprising of stronger type III collagen and structured elastin fibers. This event reconstitutes strength and resiliency of the dermis and provides for its elasticity. This phase of wound healing relies mainly on keratinocytes and how well they execute their functional potential. Surface receptors for many growth factors and cytokines expressed by keratinocytes make them capable of receiving signaling messages from multiple mediators such as KGF (FGF7), TGF-β, TNF-α, EGF, IFN-γ, as well as GM-CSF and IL-1 [156,157,158,159,160,161]. Scientific explorations of this stage of wound-healing strongly suggest the presence of a double paracrine loop where keratinocytes initially stimulate fibroblasts to synthesize growth factors and then this production of the growth factors burst triggers robust keratinocyte proliferation. Proliferation of keratinocytes further amplifies initial growth factors synthesis in fibroblasts. [162,163]. This stage of wound healing could be macroscopically (wound closure) and microscopically assessed by monitoring re-epithelialization, dermal-epidermal junction, skin appendage regeneration, granulation tissue, leukocyte infiltration, and density of dermal collagen fibers.
This last remodeling phase can last for several months and, in adults, usually leads to developing a non-functioning mass of fibrotic tissue known as a scar. This contrasts the early gestation in organism development, where the injured fetal tissues can be completely recreated without fibrosis due to skin regeneration. Deciphering differences in biological pathways guarding wound healing and regeneration has tremendous potential to improve therapeutic stem cells regenerative approaches to dermatological pathologies and boost aesthetic methods aimed to reverse the visible effects of skin aging [67].
Failure of one or several stages in the wound healing processes is generally linked to vascular disease, diabetes and many other clinical conditions that are all frequently associated with healing pathologies and ultimately aging.
Next, we will discuss one of the most promising and undoubtedly popular modern day sources used in clinical dermatology-adipose-derived stem cells to counteract this and several other pathologies.

6. From Lipoaspirate to Adipose-Derived Stem Cells (ADSCs)

Following the discovery that adipose tissue is a rich source of adult mesenchymal stem cells that exhibit regenerative potential [164] and wound healing properties [133,165,166], the lipoaspirated tissue began to be widely used in clinical applications in autologous or allogeneic fashion. Lipoaspirate is harvested by using tumescent abdominal liposuction techniques such as power assisted liposuction, surgical resection and laser-assisted liposuction [167]. In addition, the lipoaspirate harvested after surgery undergoes further processing using enzymatic [168,169] or mechanical cell separation [170,171,172]. Most SVF isolation protocols contain the following steps: washing the lipoaspirate, collagenase enzyme treatment, centrifugation, and removal of red blood cells [171]. After decantation or centrifugation, the processed lipoaspirate composed of the structural and stromal vascular cells, called stromal vascular fraction (SVF), can also be used for grafting. SVF contains a mixed composition of cells including pre-adipocytes, adipocytes, macrophages, endothelial progenitor cells (EPCs) and adipose-derived stem cells (ADSCs) [168,173,174] and has been shown to be a rich source of growth factors such as bFGF, IGF-1, VEGF, and PDGF-BB [175]. Despite the extensive and widespread use of adipose tissue in clinical applications, there is no single standard protocol available for isolation and preparation of adipose tissue. Recent reports have highlighted the differences in the protocols for harvesting, processing, and techniques for injections among clinicians and the challenges it introduces during comparison of the outcomes of clinical therapies [176]. A recent review article comparing some of the variables in isolation techniques reported no significant differences in delivery of the therapeutic benefits that depend on the donor fat site collection choice or pre-operational site preparation, fat harvesting methods, centrifugation speed or cannula size following the use of tumescent solution. However, a better comparison strategy should be deployed to substantiate such conclusions. For instance, it was demonstrated that better retention of the graft was reported in clinical studies that used centrifugation rather than sedimentation [177]. However, use of collagenase during SVF isolation steps introduces an additional regulatory burden. SVF and subsequent isolation of ADSCs from SVF is considered “more than minimal manipulation” according to U.S. Food and Drug Administration guidance documents and requires pre-market approvals of therapeutic applications of these biologic materials even for autologous cell therapies. These strict regulations are implemented due to observed variations in the composition of SVF isolated by use of different isolation protocol or devices [178,179,180]. Currently, significant effort is applied by researchers and clinicians to develop standardized methods for SVF isolation to implement quality control systems (QC) for SVF production in order to lower the regulatory barriers [168].
One of the methods to implement more stringent QC of clinical-grade material is an isolation of homogeneous population of the cells from SVF. Numerous new studies report that subsets of the cells isolated from the adipose niche (adipose-derived stem cells ADSCs) have a potent immunomodulatory activity and can robustly induce angiogenesis. It has been shown that developmental origin of ADSCs can be traced to mural cells (or pericytes) residing in the perivascular niche [181,182,183], thus explaining the remarkable capacity of ADSCs to communicate with a variety of the cell types and instruct the immune system through the paracrine signaling. It has been noted that the location of subcutaneous white adipose tissue deposits might influence stem cell recovery. For instance, in humans a much greater number of ADSCs can be recovered from the lipoaspirates of arm adipose tissue as compared with the tissue obtained from the thigh, breast, and the abdomen (reviewed in detail [184]).
The ADSCs from SVF can be expanded and passaged in culture, generating a homogeneous ADSCs population expressing the stromal surface markers similar to bone-marrow mesenchymal stem cells (BM-MSCs): CD29, CD44, CD73, CD90, and CD105 while staying negative for hematopoietic lineage markers CD31, CD34 and CD45 [185]. The secretory properties, cell surface expression markers, multipotent potential, and immunomodulatory properties of isolated ADSC and BM-MSCs have been rigorously compared and reported previously [11,23,186,187,188] concluding that the ADSCs, by and large, display secretory properties similar to those reported for BM-MSCs. However, it is important to note that prolonged ex vivo culturing of ADSCs (often required to create enough material for clinical application) results in significant and measurable changes in ADSCs due to the process of replicative senescence [23,189,190]. Senescence involves a coordination of diverse cellular processes ranging from inflammatory signaling, metabolic and cytoskeletal changes, ability of cells to cope with DNA damage, and dramatic changes in the regulatory properties of chromatin [23,189]. We and others have previously reported that isolated and ex vivo cultured hADSCs exhibit consistent self-renewing and, upon approaching replicative senescence, cultures accumulate large non-dividing cells expressing the lysozymal enzyme, senescence-associated β-galactosidase (SA-β-Gal), manifest a loss of control for chromatin organization, activate a persistent DNA damage response (DDR) and cause robust changes in transcriptional activity [189,191,192], while preserving ADSCs phenotypical stability. These changes not only result in the reduction of differentiation potential of ADSCs, but are also associated with severe changes in ADSCs secretory repertoire impacting ADSCs paracrine function and immunomodulation. Due to the profound secretory phenotype of senescent cells, their impact on organs and tissues after transplantation is far from neutral. The role of senescence in the aging of adult stem cells is tightly linked to tissue maintenance and homeostasis and often viewed as an irreversible barrier to immortalization and tumorigenesis under the assumption that cellular senescence evolved to suppress tumorigenesis [193,194]. This view has been intensely debated in recent years [195,196]. Contrary to the assumption that senescence and tumorigenicity are always permanently connected and mutually exclusive, recent data monitoring one of the well-established markers of senescence, p16INK4a, in mice indicates that the activation of this hallmark of cellular senescence is, in fact, a characteristic of all emerging cancers [197], thus suggesting that cellular senescence is a quasi-stable and/or plastic cellular state prone to oncogenesis rather than a cancer preventive mechanism.
Concurrently, there are considerable ongoing clinical activities in the development of ADSCs therapeutic applications. It is important to identify not only reliable markers of senescence that will translate into human settings, but also to better understand the intimate connections between oncogenic events and senescence in order to define a set of genetic targets that are able to elicit, modulate and, most importantly, balance both of these responses. These markers would allow one to implement an additional measurable QC system to the clinical applications of ADSCs, SVF and fat grafts in general, in order to increase efficacy and safety of the clinical applications.
Given the ease in isolation in large amounts for autologous fat, SVF, or ADSCs, these materials have been widely used for a broad spectrum of clinical applications. In this review, we will limit our discussion to the applications currently in trials for indications of clinical dermatology. We will discuss some of these applications in the next section.

7. Adipose-Derived Stem Cells and Their Applications in Clinical Dermatology

Autologous fat graft procedures, alternatively termed as autologous fat transfer or lipo injections, deliver processed lipoaspirate containing the ECM and SVF fractions as described in the previous section. Autologous fat performs like ideal soft tissue filler and has been applied in clinical applications ranging from structural [198], facial filling and rejuvenation [199,200,201], treatment of scars [202], aging-related dermatological indications [203,204], and wound healing [165,205]. The mechanistic explanations of the significant improvements achieved in these studies are deeply imbedded in the biology of the spatial and temporal relationship between adipose tissue and different stages of wound healing. Emerging data highlight the role of cells from the adipose deposit in skin homeostasis process. It was elegantly demonstrated that adipocyte precursor cells (also called preadipocytes, adipose-derived stem cells, adipose-derived stromal cells, see for details [184]) proliferate and differentiate, and mature adipocytes repopulate skin wounds following inflammatory stage of wound healing. This has been shown to occur in parallel with fibroblast migration [32]. Genetic and pharmacological ablation of adipose depots results in diminished fibroblast presence and ECM protein deposition in the regenerating dermis. All of the aforementioned defects result in long-term loss of skin homeostasis, compromising skin integrity and wound recurrence. In addition, further support was gained in studies demonstrating that stem-like cells from intradermal adipose depots, in parallel with their renewal, are necessary and sufficient to drive follicular stem cell activation [137]. Adipokines produced by adipose tissue such as adiponectin, visceral adipose tissue-derived serine protease inhibitor (vaspin) and leptin, exert hormone-like activities and can act on a systemic level [206]. These data highlight the importance of the intradermal adipogenic cells and their precursors as skin niche cells (shown in Figure 1). It also implies that adipogenic niche may alter epithelial stem cell function and have a clinical impact as a positive regulator of skin stem cell activity. Therefore, it is highly likely that the beneficial effects of lipoaspirate post fat grafting procedures are a result of this adipogenic niche and cells. Numerous technical advancements in the field have allowed for modifications in the protocol to further process lipoaspirate into microfat and nanofat thereby making the use of smaller gauge needles possible for fat injections to correct volume losses created by aging or diseases for softer more delicate areas. Microfat is generated using similar approaches as generating fat but using smaller multiport cannula [207]. Nanofat is generated by further processing microfat by mechanical emulsification using syringes leaving fine emulsified fat. Nanofat is generally used for injections using a 27 g needle for correcting delicate areas such as superficial rhytides, scars, eyelids and dark under-eye areas. Recent advances in techniques leading to combining routine fat grafting with nanofat may provide better outcomes for patients with problematic wound management where traditional fat grafts are unable to deliver desired clinical outcomes [208].
While autologous fat grafting is shown in numerous trials to be a safe and easy to perform technique (see Table 2), there are challenges that still remain. These challenges are associated with fat graft survival, necrosis, and loss in volume. Recently, it has been reported that clinical applications using mixed purified/processed fat with 50% or more of autologous platelet rich plasma (PRP) greatly improved fat grafting outcomes and it was no surprise that the improvement was contributed to better survival of fat cells and ADSCs differentiation triggered by the potent growth factors present in PRP such as PDGF, TGFβ, VEGF and EGF [209].
Many clinical applications have also tapped into the therapeutic properties of SVF considering SVF heterogeneous cellular composition. Mechanistically, multiple cell types of SVF might work together to provide a reconstituted niche where numerous signaling pathways listed in Table 1 come together to trigger the efficient healing process and improved therapeutical outcomes [210,211]. SVF and its application as a therapeutic have been extensively reviewed [212,213]. Homogeneous ADSCs isolated from SVF also reported to contribute to efficient regeneration, immunomodulation and wound healing either alone or when mixed in clinical prep with SVF to obtain ADSCs-enriched SVF [179,214]. Many preclinical and clinical studies have demonstrated that the administration of ADSCs improves wound healing by means of the increased accelerated wound closure, reduces scarring, promotes collagen synthesis, angiogenesis, and improves tensile strength. In these lines of clinical trials ADSCs have shown a great potential for the treatment of chronic wounds due to their enhanced growth factors secretion triggering angiogenesis. These factors include VEGF and many others listed in Table 1 and also reviewed by Toyserkani [215].
In addition, ADSCs have been reported to have therapeutic benefit in skin. ADSCs cultured in PRP have been used and are shown to be safe for many clinical applications listed in Table 2, [216]. Ex vivo data demonstrate that ADSCs maintained in PRP-containing media have been shown to have stimulatory effects on the proliferation and migration of dermal fibroblasts and keratinocytes, suggesting that mechanistic explanation of the success in this group of clinical trials might be embedded in ADSCs ability to support re-epithelialization potential by providing guidance and paracrine instructions for maintenance of epidermal organization and homeostasis [133,217,218]. The role of ADSCs in a double paracrine loop provide evidence of an intricate relationships between ADSCs and various cellular components of the dermis and epidermis (Figure 1) and, importantly, signaling pathways involved in skin homeostasis. Taking into consideration ADSCs’ secretory properties, one might suggest that such regulations are probably deployed through the same key growth factors and matrikines that we have listed in Table 1 [11,27]. However, further experiments are required to tease apart the mechanistic underpinning of these highly complex interactions and, importantly, to ensure reliable and reproducible results of these treatments.

8. Conclusions

There remain many concerns regarding the use of ADSCs in dermatology, including a lack of mechanistic details on how these cells act as precursors or influence keratinocytes, fibroblasts, and endothelial cells, or as a secretion vehicle of soluble factors. Additionally, while many preclinical and clinical studies have shown that a therapeutic effect results from the use of ADSC transplant or conditioned media, there is no systematic information currently available regarding whether donor age modifies ADSC regenerative or wound healing potential. Even though it has long been known that advanced age is negatively correlated with an organism’s reparative and regenerative potential; scarce, conflicting data that is available concerning the effects of donor age on ADSC function is not sufficient to warrant absence of long term adverse reaction to these cell use. Several ex vivo reports involving various species (rodents, monkeys, and humans) have indicated that aging is accompanied by many changes in biological processes affecting ADSCs (see in greater detail in [23,190,248]). One striking example highlights the importance of such investigations. In the recent study, Kato et al. evaluated whether donor age influences the potential of ADSCs deployed to assist wound healing. The reported data demonstrate that while the treatment with adult’s BM-MSCs or their ADSCs in cutaneous wound promotes better tissue repair/regeneration, efficacy of both of these therapeutic approaches was negatively affected by donor age [33]. It has been suggested that this might be due to changes in the SC paracrine factors secretion. Further studies are necessary to establish the optimal, long-lasting and, importantly, safe strategy for ADSCs applications in the treatment of skin pathological conditions and aging symptoms in patients.

Acknowledgments

We thank our many colleagues in the field who contributed to our understanding of skin homeostasis. We apologize to those whose papers are not cited owing to space constraints. We thank you Alexandra Amaro-Ortiz and Veronika Lunyak for the help in the manuscript preparation and James Tollervey for help with figures.

Author Contributions

Victoria V. Lunyak and Meenakshi Gaur conceived and wrote the review. Victoria V. Lunyak and Meenakshi Gaur generated figures. Victoria V. Lunyak, Meenakshi Gaur and Marek Dobke reviewed the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Makrantonaki, E.; Zouboulis, C.C. Molecular mechanisms of skin aging: State of the art. Ann. N. Y. Acad. Sci. 2007, 1119, 40–50. [Google Scholar] [CrossRef] [PubMed]
  2. Zouboulis, C.C.; Makrantonaki, E. Clinical aspects and molecular diagnostics of skin aging. Clin. Dermatol. 2011, 29, 3–14. [Google Scholar] [CrossRef] [PubMed]
  3. Zouboulis, C.C.; Boschnakow, A. Chronological ageing and photoageing of the human sebaceous gland. Clin. Exp. Dermatol. 2001, 26, 600–607. [Google Scholar] [CrossRef] [PubMed]
  4. Blanpain, C.; Fuchs, E. Epidermal homeostasis: A balancing act of stem cells in the skin. Nat. Rev. Mol. Cell Biol. 2009, 10, 207–217. [Google Scholar] [CrossRef] [PubMed]
  5. Blanpain, C.; Horsley, V.; Fuchs, E. Epithelial stem cells: Turning over new leaves. Cell 2007, 128, 445–458. [Google Scholar] [CrossRef] [PubMed]
  6. Blanpain, C.; Fuchs, E. Epidermal stem cells of the skin. Annu. Rev. Cell Dev. Biol. 2006, 22, 339–373. [Google Scholar] [CrossRef] [PubMed]
  7. Yi, R.; O’Carroll, D.; Pasolli, H.A.; Zhang, Z.; Dietrich, F.S.; Tarakhovsky, A.; Fuchs, E. Morphogenesis in skin is governed by discrete sets of differentially expressed microRNAs. Nat. Genet. 2006, 38, 356–362. [Google Scholar] [CrossRef] [PubMed]
  8. Tollervey, J.R.; Lunyak, V.V. Epigenetics: Judge, jury and executioner of stem cell fate. Epigenetics 2012, 7, 823–840. [Google Scholar] [CrossRef] [PubMed]
  9. Morgner, J.; Ghatak, S.; Jakobi, T.; Dieterich, C.; Aumailley, M.; Wickstrom, S.A. Integrin-linked kinase regulates the niche of quiescent epidermal stem cells. Nat. Commun. 2015, 6, 8198. [Google Scholar] [CrossRef] [PubMed]
  10. Odorisio, T. Epigenetic Control of Skin Re-Epithelialization: The NF-κB/JMJD3 Connection. J. Investig. Dermatol. 2016, 136, 738–740. [Google Scholar] [CrossRef] [PubMed]
  11. Kilroy, G.E.; Foster, S.J.; Wu, X.; Ruiz, J.; Sherwood, S.; Heifetz, A.; Ludlow, J.W.; Stricker, D.M.; Potiny, S.; Green, P.; et al. Cytokine profile of human adipose-derived stem cells: Expression of angiogenic, hematopoietic, and pro-inflammatory factors. J. Cell. Physiol. 2007, 212, 702–709. [Google Scholar] [CrossRef] [PubMed]
  12. Cappuzzello, C.; Doni, A.; Dander, E.; Pasqualini, F.; Nebuloni, M.; Bottazzi, B.; Mantovani, A.; Biondi, A.; Garlanda, C.; D’Amico, G. Mesenchymal Stromal Cell-Derived PTX3 Promotes Wound Healing via Fibrin Remodeling. J. Investig. Dermatol. 2016, 136, 293–300. [Google Scholar] [CrossRef] [PubMed]
  13. Ozpur, M.A.; Guneren, E.; Canter, H.I.; Karaaltin, M.V.; Ovali, E.; Yogun, F.N.; Baygol, E.G.; Kaplan, S. Generation of Skin Tissue Using Adipose Tissue-Derived Stem Cells. Plast. Reconstr. Surg. 2016, 137, 134–143. [Google Scholar] [CrossRef] [PubMed]
  14. Camilleri, E.T.; Gustafson, M.P.; Dudakovic, A.; Riester, S.M.; Garces, C.G.; Paradise, C.R.; Takai, H.; Karperien, M.; Cool, S.; Sampen, H.J.; et al. Identification and validation of multiple cell surface markers of clinical-grade adipose-derived mesenchymal stromal cells as novel release criteria for good manufacturing practice-compliant production. Stem Cell Res. Ther. 2016, 7, 107. [Google Scholar] [CrossRef] [PubMed]
  15. Menon, G.K. New insights into skin structure: Scratching the surface. Adv. Drug Deliv. Rev. 2002, 54 (Suppl. S1), S3–S17. [Google Scholar] [CrossRef]
  16. Elias, P.M.; Friend, D.S. The permeability barrier in mammalian epidermis. J. Cell Biol. 1975, 65, 180–191. [Google Scholar] [CrossRef] [PubMed]
  17. Nemes, Z.; Steinert, P.M. Bricks and mortar of the epidermal barrier. Exp. Mol. Med. 1999, 31, 5–19. [Google Scholar] [CrossRef] [PubMed]
  18. Barry, B.W. Novel mechanisms and devices to enable successful transdermal drug delivery. Eur. J. Pharm. Sci. Off. J. Eur. Fed. Pharm. Sci. 2001, 14, 101–114. [Google Scholar] [CrossRef]
  19. Foldvari, M. Non-invasive administration of drugs through the skin: Challenges in delivery system design. Pharm. Sci. Technol. Today 2000, 3, 417–425. [Google Scholar] [CrossRef]
  20. Bouwstra, J.A.; Ponec, M. The skin barrier in healthy and diseased state. Biochim. Biophys. Acta 2006, 1758, 2080–2095. [Google Scholar] [CrossRef] [PubMed]
  21. Mesa, K.R.; Rompolas, P.; Zito, G.; Myung, P.; Sun, T.Y.; Brown, S.; Gonzalez, D.G.; Blagoev, K.B.; Haberman, A.M.; Greco, V. Niche-induced cell death and epithelial phagocytosis regulate hair follicle stem cell pool. Nature 2015, 522, 94–97. [Google Scholar] [CrossRef] [PubMed]
  22. Fuchs, E.; Green, H. Changes in keratin gene expression during terminal differentiation of the keratinocyte. Cell 1980, 19, 1033–1042. [Google Scholar] [CrossRef]
  23. Niu, P.; Smagul, A.; Wang, L.; Sadvakas, A.; Sha, Y.; Perez, L.M.; Nussupbekova, A.; Amirbekov, A.; Akanov, A.A.; Galvez, B.G.; et al. Transcriptional profiling of interleukin-2-primed human adipose derived mesenchymal stem cells revealed dramatic changes in stem cells response imposed by replicative senescence. Oncotarget 2015, 6, 17938–17957. [Google Scholar] [CrossRef] [PubMed]
  24. Astori, G.; Vignati, F.; Bardelli, S.; Tubio, M.; Gola, M.; Albertini, V.; Bambi, F.; Scali, G.; Castelli, D.; Rasini, V.; et al. “In vitro” and multicolor phenotypic characterization of cell subpopulations identified in fresh human adipose tissue stromal vascular fraction and in the derived mesenchymal stem cells. J. Transl. Med. 2007, 5, 55. [Google Scholar] [CrossRef] [PubMed]
  25. Varma, M.J.; Breuls, R.G.; Schouten, T.E.; Jurgens, W.J.; Bontkes, H.J.; Schuurhuis, G.J.; van Ham, S.M.; van Milligen, F.J. Phenotypical and functional characterization of freshly isolated adipose tissue-derived stem cells. Stem Cells Dev. 2007, 16, 91–104. [Google Scholar] [CrossRef] [PubMed]
  26. Roh, N.K.; Kim, M.J.; Lee, Y.W.; Choe, Y.B.; Ahn, K.J. A Split-Face Study of the Effects of a Stabilized Hyaluronic Acid-Based Gel of Nonanimal Origin for Facial Skin Rejuvenation Using a Stamp-Type Multineedle Injector: A Randomized Clinical Trial. Plast. Reconstr. Surg. 2016, 137, 809–816. [Google Scholar] [CrossRef] [PubMed]
  27. Hocking, A.M.; Gibran, N.S. Mesenchymal stem cells: Paracrine signaling and differentiation during cutaneous wound repair. Exp. Cell Res. 2010, 316, 2213–2219. [Google Scholar] [CrossRef] [PubMed]
  28. Klein, J.; Permana, P.A.; Owecki, M.; Chaldakov, G.N.; Bohm, M.; Hausman, G.; Lapiere, C.M.; Atanassova, P.; Sowinski, J.; Fasshauer, M.; et al. What are subcutaneous adipocytes really good for? Exp. Dermatol. 2007, 16, 45–70. [Google Scholar] [PubMed]
  29. Jackson, W.M.; Nesti, L.J.; Tuan, R.S. Concise review: Clinical translation of wound healing therapies based on mesenchymal stem cells. Stem Cells Transl. Med. 2012, 1, 44–50. [Google Scholar] [CrossRef] [PubMed]
  30. Jackson, W.M.; Nesti, L.J.; Tuan, R.S. Mesenchymal stem cell therapy for attenuation of scar formation during wound healing. Stem Cell Res. Ther. 2012, 3, 20. [Google Scholar] [CrossRef] [PubMed]
  31. Naderi, N.; Combellack, E.J.; Griffin, M.; Sedaghati, T.; Javed, M.; Findlay, M.W.; Wallace, C.G.; Mosahebi, A.; Butler, P.E.; Seifalian, A.M.; et al. The regenerative role of adipose-derived stem cells (adsc) in plastic and reconstructive surgery. Int. Wound J. 2017, 14, 112–124. [Google Scholar] [CrossRef] [PubMed]
  32. Schmidt, B.A.; Horsley, V. Intradermal adipocytes mediate fibroblast recruitment during skin wound healing. Development 2013, 140, 1517–1527. [Google Scholar] [CrossRef] [PubMed]
  33. Kato, J.; Kamiya, H.; Himeno, T.; Shibata, T.; Kondo, M.; Okawa, T.; Fujiya, A.; Fukami, A.; Uenishi, E.; Seino, Y.; et al. Mesenchymal stem cells ameliorate impaired wound healing through enhancing keratinocyte functions in diabetic foot ulcerations on the plantar skin of rats. J. Diabetes Complicat. 2014, 28, 588–595. [Google Scholar] [CrossRef] [PubMed]
  34. Caplan, A.I.; Correa, D. PDGF in bone formation and regeneration: New insights into a novel mechanism involving MSCs. J. Orthop. Res. 2011, 29, 1795–1803. [Google Scholar] [CrossRef] [PubMed]
  35. Kim, W.S.; Park, B.S.; Sung, J.H.; Yang, J.M.; Park, S.B.; Kwak, S.J.; Park, J.S. Wound healing effect of adipose-derived stem cells: A critical role of secretory factors on human dermal fibroblasts. J. Dermatol. Sci. 2007, 48, 15–24. [Google Scholar] [CrossRef] [PubMed]
  36. Bernstein, E.F.; Underhill, C.B.; Hahn, P.J.; Brown, D.B.; Uitto, J. Chronic sun exposure alters both the content and distribution of dermal glycosaminoglycans. Br. J. Dermatol. 1996, 135, 255–262. [Google Scholar] [CrossRef] [PubMed]
  37. Waller, J.M.; Maibach, H.I. Age and skin structure and function, a quantitative approach (II): Protein, glycosaminoglycan, water, and lipid content and structure. Skin Res. Technol. 2006, 12, 145–154. [Google Scholar] [CrossRef] [PubMed]
  38. Farage, M.A.; Miller, K.W.; Elsner, P.; Maibach, H.I. Functional and physiological characteristics of the aging skin. Aging Clin. Exp. Res. 2008, 20, 195–200. [Google Scholar] [CrossRef] [PubMed]
  39. Suter-Widmer, J.; Elsner, P. The Irritant Contact Dermatitis Syndrome; CRC Press: Boca Raton, FL, USA, 1996; pp. 257–265. [Google Scholar]
  40. Waller, J.M.; Maibach, H.I. Age and skin structure and function, a quantitative approach (I): Blood flow, pH, thickness, and ultrasound echogenicity. Skin Res. Technol. 2005, 11, 221–235. [Google Scholar] [CrossRef] [PubMed]
  41. Oriba, H.A.; Bucks, D.A.; Maibach, H.I. Percutaneous absorption of hydrocortisone and testosterone on the vulva and forearm: Effect of the menopause and site. Br. J. Dermatol. 1996, 134, 229–233. [Google Scholar] [CrossRef] [PubMed]
  42. Duncan, K.O.; Leffell, D.J. Preoperative assessment of the elderly patient. Dermatol. Clin. 1997, 15, 583–593. [Google Scholar] [CrossRef]
  43. Kaya, G.; Saurat, J.H. Dermatoporosis: A chronic cutaneous insufficiency/fragility syndrome. Clinicopathological features, mechanisms, prevention and potential treatments. Dermatology 2007, 215, 284–294. [Google Scholar] [CrossRef] [PubMed]
  44. Kaya, G.; Tran, C.; Sorg, O.; Hotz, R.; Grand, D.; Carraux, P.; Didierjean, L.; Stamenkovic, I.; Saurat, J.H. Hyaluronate fragments reverse skin atrophy by a CD44-dependent mechanism. PLoS Med. 2006, 3, e493. [Google Scholar] [CrossRef] [PubMed]
  45. Nagai, R.; Mori, T.; Yamamoto, Y.; Kaji, Y.; Yonei, Y. Significance of advanced glycation end products in ageing-related disease. Anti Ageing Med. 2010, 7, 112–119. [Google Scholar] [CrossRef]
  46. Komosinska-Vassev, K.; Olczyk, P.; Winsz-Szczotka, K.; Klimek, K.; Olczyk, K. Plasma biomarkers of oxidative and AGE-mediated damage of proteins and glycosaminoglycans during healthy ageing: A possible association with ECM metabolism. Mech. Ageing Dev. 2012, 133, 538–548. [Google Scholar] [CrossRef] [PubMed]
  47. Nedic, O.; Rattan, S.I.; Grune, T.; Trougakos, I.P. Molecular effects of advanced glycation end products on cell signalling pathways, ageing and pathophysiology. Free Radic. Res. 2013, 47 (Suppl. S1), 28–38. [Google Scholar] [CrossRef] [PubMed]
  48. Farage, M.A.; Miller, K.W.; Elsner, P.; Maibach, H.I. Characteristics of the Aging Skin. Adv. Wound Care 2013, 2, 5–10. [Google Scholar] [CrossRef] [PubMed]
  49. Alkilani, A.Z.; McCrudden, M.T.; Donnelly, R.F. Transdermal Drug Delivery: Innovative Pharmaceutical Developments Based on Disruption of the Barrier Properties of the stratum corneum. Pharmaceutics 2015, 7, 438–470. [Google Scholar] [CrossRef] [PubMed]
  50. Hahn, H.J.; Jung, H.J.; Schrammek-Drusios, M.C.; Lee, S.N.; Kim, J.H.; Kwon, S.B.; An, I.S.; An, S.; Ahn, K.J. Instrumental evaluation of anti-aging effects of cosmetic formulations containing palmitoyl peptides, Silybum marianum seed oil, vitamin E and other functional ingredients on aged human skin. Exp. Ther. Med. 2016, 12, 1171–1176. [Google Scholar] [CrossRef] [PubMed]
  51. Sotoodian, B.; Maibach, H.I. Noninvasive test methods for epidermal barrier function. Clin. Dermatol. 2012, 30, 301–310. [Google Scholar] [CrossRef] [PubMed]
  52. Seok, J.; Hong, J.Y.; Choi, S.Y.; Park, K.Y.; Kim, B.J. A potential relationship between skin hydration and stamp-type microneedle intradermal hyaluronic acid injection in middle-aged male face. J. Cosmet. Dermatol. 2016. [Google Scholar] [CrossRef] [PubMed]
  53. Eklouh-Molinier, C.; Gaydou, V.; Froigneux, E.; Barlier, P.; Couturaud, V.; Manfait, M.; Piot, O. In vivo confocal Raman microspectroscopy of the human skin: Highlighting of spectral markers associated to aging via a research of correlation between Raman and biometric mechanical measurements. Anal. Bioanal. Chem. 2015, 407, 8363–8372. [Google Scholar] [CrossRef] [PubMed]
  54. Franzen, L.; Windbergs, M. Applications of Raman spectroscopy in skin research—From skin physiology and diagnosis up to risk assessment and dermal drug delivery. Adv. Drug Deliv. Rev. 2015, 89, 91–104. [Google Scholar] [CrossRef] [PubMed]
  55. Callaghan, T.M.; Wilhelm, K.P. A review of ageing and an examination of clinical methods in the assessment of ageing skin. Part I: Cellular and molecular perspectives of skin ageing. Int. J. Cosmet. Sci. 2008, 30, 313–322. [Google Scholar] [CrossRef] [PubMed]
  56. Kottner, J.; Schario, M.; Garcia Bartels, N.; Pantchechnikova, E.; Hillmann, K.; Blume-Peytavi, U. Comparison of two in vivo measurements for skin surface topography. Skin Res. Technol. 2013, 19, 84–90. [Google Scholar] [CrossRef] [PubMed]
  57. Trojahn, C.; Schario, M.; Dobos, G.; Blume-Peytavi, U.; Kottner, J. Reliability and validity of two in vivo measurements for skin surface topography in aged adults. Skin Res. Technol. 2015, 21, 54–60. [Google Scholar] [CrossRef] [PubMed]
  58. Myer, K.; Maibach, H. Stratum corneum evaluation methods: Overview. Skin Res. Technol. 2013, 19, 213–219. [Google Scholar] [CrossRef] [PubMed]
  59. Savory, S.A.; Agim, N.G.; Mao, R.; Peter, S.; Wang, C.; Maldonado, G.; Bearden Dietert, J.; Lieu, T.J.; Pretzlaff, K.; Das, S.; et al. Reliability assessment and validation of the postacne hyperpigmentation index (PAHPI), a new instrument to measure postinflammatory hyperpigmentation from acne vulgaris. J. Am. Acad. Dermatol. 2014, 70, 108–114. [Google Scholar] [CrossRef] [PubMed]
  60. Gniadecka, M.; Jemec, G.B. Quantitative evaluation of chronological ageing and photoageing in vivo: Studies on skin echogenicity and thickness. Br. J. Dermatol. 1998, 139, 815–821. [Google Scholar] [CrossRef] [PubMed]
  61. Irfan-Maqsood, M.; Matin, M.M.; Heirani-Tabasi, A.; Bahrami, M.; Naderi-Meshkin, H.; Mirahmadi, M.; Hassanzadeh, H.; Sanjar Moussavi, N.; Raza-Shah, H.; Raeesolmohaddeseen, M.; et al. Adipose derived mesenchymal stem cells express keratinocyte lineage markers in a co-culture model. Cell Mol. Biol. 2016, 62, 44–54. [Google Scholar] [PubMed]
  62. Hanel, K.H.; Cornelissen, C.; Luscher, B.; Baron, J.M. Cytokines and the skin barrier. Int. J. Mol. Sci. 2013, 14, 6720–6745. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Kulukian, A.; Holland, A.J.; Vitre, B.; Naik, S.; Cleveland, D.W.; Fuchs, E. Epidermal development, growth control, and homeostasis in the face of centrosome amplification. Proc. Natl. Acad. Sci. USA 2015, 112, E6311–E6320. [Google Scholar] [CrossRef] [PubMed]
  64. Pesenacker, A.M.; Broady, R.; Levings, M.K. Control of tissue-localized immune responses by human regulatory T cells. Eur. J. Immunol. 2015, 45, 333–343. [Google Scholar] [CrossRef] [PubMed]
  65. Rompolas, P.; Mesa, K.R.; Kawaguchi, K.; Park, S.; Gonzalez, D.; Brown, S.; Boucher, J.; Klein, A.M.; Greco, V. Spatiotemporal coordination of stem cell commitment during epidermal homeostasis. Science 2016, 352, 1471–1474. [Google Scholar] [CrossRef] [PubMed]
  66. Sayedyahossein, S.; Rudkouskaya, A.; Leclerc, V.; Dagnino, L. Integrin-linked kinase is indispensable for keratinocyte differentiation and epidermal barrier function. J. Investig. Dermatol. 2016, 136, 425–435. [Google Scholar] [CrossRef] [PubMed]
  67. Fitzpatrick, R.E.; Rostan, E.F. Reversal of photodamage with topical growth factors: A pilot study. J. Cosmet. Laser Ther. 2003, 5, 25–34. [Google Scholar] [CrossRef] [PubMed]
  68. Gold, M.H.; Goldman, M.P.; Biron, J. Efficacy of novel skin cream containing mixture of human growth factors and cytokines for skin rejuvenation. J. Drugs Dermatol. 2007, 6, 197–201. [Google Scholar] [PubMed]
  69. Ehrlich, M.; Rao, J.; Pabby, A.; Goldman, M.P. Improvement in the appearance of wrinkles with topical transforming growth factor β(1) and l-ascorbic acid. Dermatol. Surg. 2006, 32, 618–625. [Google Scholar] [PubMed]
  70. Lee, H.J.; Lee, E.G.; Kang, S.; Sung, J.H.; Chung, H.M.; Kim, D.H. Efficacy of microneedling plus human stem cell conditioned medium for skin rejuvenation: A randomized, controlled, blinded split-face study. Ann. Dermatol. 2014, 26, 584–591. [Google Scholar] [CrossRef] [PubMed]
  71. Kim, W.S.; Park, S.H.; Ahn, S.J.; Kim, H.K.; Park, J.S.; Lee, G.Y.; Kim, K.J.; Whang, K.K.; Kang, S.H.; Park, B.S.; et al. Whitening effect of adipose-derived stem cells: A critical role of TGF-β 1. Biol. Pharm. Bull. 2008, 31, 606–610. [Google Scholar] [CrossRef] [PubMed]
  72. Kim, W.S.; Park, B.S.; Park, S.H.; Kim, H.K.; Sung, J.H. Antiwrinkle effect of adipose-derived stem cell: Activation of dermal fibroblast by secretory factors. J. Dermatol. Sci. 2009, 53, 96–102. [Google Scholar] [CrossRef] [PubMed]
  73. Seo, K.Y.; Kim, D.H.; Lee, S.E.; Yoon, M.S.; Lee, H.J. Skin rejuvenation by microneedle fractional radiofrequency and a human stem cell conditioned medium in Asian skin: A randomized controlled investigator blinded split-face study. J. Cosmet. Laser Ther. 2013, 15, 25–33. [Google Scholar] [CrossRef] [PubMed]
  74. Lee, M.J.; Kim, J.; Lee, K.I.; Shin, J.M.; Chae, J.I.; Chung, H.M. Enhancement of wound healing by secretory factors of endothelial precursor cells derived from human embryonic stem cells. Cytotherapy 2011, 13, 165–178. [Google Scholar] [CrossRef] [PubMed]
  75. Marikovsky, M.; Breuing, K.; Liu, P.Y.; Eriksson, E.; Higashiyama, S.; Farber, P.; Abraham, J.; Klagsbrun, M. Appearance of heparin-binding EGF-like growth factor in wound fluid as a response to injury. Proc. Natl. Acad. Sci. USA 1993, 90, 3889–3893. [Google Scholar] [CrossRef] [PubMed]
  76. Werner, S.; Duan, D.S.; de Vries, C.; Peters, K.G.; Johnson, D.E.; Williams, L.T. Differential splicing in the extracellular region of fibroblast growth factor receptor 1 generates receptor variants with different ligand-binding specificities. Mol. Cell. Biol. 1992, 12, 82–88. [Google Scholar] [CrossRef] [PubMed]
  77. Abraham, J.A.; Whang, J.L.; Tumolo, A.; Mergia, A.; Friedman, J.; Gospodarowicz, D.; Fiddes, J.C. Human basic fibroblast growth factor: Nucleotide sequence and genomic organization. EMBO J. 1986, 5, 2523–2528. [Google Scholar] [PubMed]
  78. Eriksson, A.; Siegbahn, A.; Westermark, B.; Heldin, C.H.; Claesson-Welsh, L. PDGF α- and β-receptors activate unique and common signal transduction pathways. EMBO J. 1992, 11, 543–550. [Google Scholar] [PubMed]
  79. Rappolee, D.A.; Mark, D.; Banda, M.J.; Werb, Z. Wound macrophages express TGF-α and other growth factors in vivo: Analysis by mRNA phenotyping. Science 1988, 241, 708–712. [Google Scholar] [CrossRef] [PubMed]
  80. Mueller, A.R.; Platz, K.P.; Nalesnik, M.A.; Langrehr, J.M.; Neuhaus, P.; Schraut, W.H. Small bowel preservation causes basement membrane and endothelial cell injury. Transp. Proc. 1994, 26, 1498–1500. [Google Scholar]
  81. Madlener, M.; Mauch, C.; Conca, W.; Brauchle, M.; Parks, W.C.; Werner, S. Regulation of the expression of stromelysin-2 by growth factors in keratinocytes: Implications for normal and impaired wound healing. Biochem. J. 1996, 320, 659–664. [Google Scholar] [CrossRef] [PubMed]
  82. Frank, S.; Madlener, M.; Werner, S. Transforming growth factors β1, β2, and β3 and their receptors are differentially regulated during normal and impaired wound healing. J. Biol. Chem. 1996, 271, 10188–10193. [Google Scholar] [PubMed]
  83. Shah, M.; Foreman, D.M.; Ferguson, M.W. Neutralisation of TGF-β1 and TGF-β2 or exogenous addition of TGF-β3 to cutaneous rat wounds reduces scarring. J. Cell Sci. 1995, 108, 985–1002. [Google Scholar] [PubMed]
  84. Hubner, G.; Brauchle, M.; Smola, H.; Madlener, M.; Fassler, R.; Werner, S. Differential regulation of pro-inflammatory cytokines during wound healing in normal and glucocorticoid-treated mice. Cytokine 1996, 8, 548–556. [Google Scholar] [CrossRef] [PubMed]
  85. Hubner, G.; Werner, S. Serum growth factors and proinflammatory cytokines are potent inducers of activin expression in cultured fibroblasts and keratinocytes. Exp. Cell Res. 1996, 228, 106–113. [Google Scholar] [CrossRef] [PubMed]
  86. Leung, G.; Wang, A.; Fernando, M.; Phan, V.C.; McKay, D.M. Bone marrow-derived alternatively activated macrophages reduce colitis without promoting fibrosis: Participation of IL-10. American journal of physiology. Gastrointest. Liver Physiol. 2013, 304, G781–G792. [Google Scholar] [CrossRef] [PubMed]
  87. Peranteau, W.H.; Zhang, L.; Muvarak, N.; Badillo, A.T.; Radu, A.; Zoltick, P.W.; Liechty, K.W. IL-10 overexpression decreases inflammatory mediators and promotes regenerative healing in an adult model of scar formation. J. Investig. Dermatol. 2008, 128, 1852–1860. [Google Scholar] [CrossRef] [PubMed]
  88. Krishnamurthy, P.; Thal, M.; Verma, S.; Hoxha, E.; Lambers, E.; Ramirez, V.; Qin, G.; Losordo, D.; Kishore, R. Interleukin-10 deficiency impairs bone marrow-derived endothelial progenitor cell survival and function in ischemic myocardium. Circ. Res. 2011, 109, 1280–1289. [Google Scholar] [CrossRef] [PubMed]
  89. King, A.; Balaji, S.; Le, L.D.; Crombleholme, T.M.; Keswani, S.G. Regenerative Wound Healing: The Role of Interleukin-10. Adv. Wound Care 2014, 3, 315–323. [Google Scholar] [CrossRef] [PubMed]
  90. Mattey, D.L. Interleukin-4 induces myofibroblast differentiation is synovial fibroblasts. Biochem. Soc. Trans. 1997, 25, 290S. [Google Scholar] [CrossRef] [PubMed]
  91. Salmon-Ehr, V.; Serpier, H.; Nawrocki, B.; Gillery, P.; Clavel, C.; Kalis, B.; Birembaut, P.; Maquart, F.X. Expression of interleukin-4 in scleroderma skin specimens and scleroderma fibroblast cultures. Potential role in fibrosis. Arch. Dermatol. 1996, 132, 802–806. [Google Scholar] [CrossRef] [PubMed]
  92. Lacraz, S.; Nicod, L.; Galve-de Rochemonteix, B.; Baumberger, C.; Dayer, J.M.; Welgus, H.G. Suppression of metalloproteinase biosynthesis in human alveolar macrophages by interleukin-4. J. Clin. Investig. 1992, 90, 382–388. [Google Scholar] [CrossRef] [PubMed]
  93. Serpier, H.; Gillery, P.; Salmon-Ehr, V.; Garnotel, R.; Georges, N.; Kalis, B.; Maquart, F.X. Antagonistic effects of interferon-gamma and interleukin-4 on fibroblast cultures. J. Investig. Dermatol. 1997, 109, 158–162. [Google Scholar] [CrossRef] [PubMed]
  94. Salmon-Ehr, V.; Ramont, L.; Godeau, G.; Birembaut, P.; Guenounou, M.; Bernard, P.; Maquart, F.X. Implication of interleukin-4 in wound healing. Lab. Investig. J. Tech. Methods Pathol. 2000, 80, 1337–1343. [Google Scholar] [CrossRef]
  95. Matias, M.A.; Saunus, J.M.; Ivanovski, S.; Walsh, L.J.; Farah, C.S. Accelerated wound healing phenotype in Interleukin 12/23 deficient mice. J. Inflamm. 2011, 8, 39. [Google Scholar] [CrossRef] [PubMed]
  96. Fujimoto, M.; Nakano, M.; Terabe, F.; Kawahata, H.; Ohkawara, T.; Han, Y.; Ripley, B.; Serada, S.; Nishikawa, T.; Kimura, A.; et al. The influence of excessive IL-6 production in vivo on the development and function of FOXP3+ regulatory T cells. J. Immunol. 2011, 186, 32–40. [Google Scholar] [CrossRef] [PubMed]
  97. Lee, J.Y.; Jeong, H.J.; Kim, M.K.; Wee, W.R. Bone marrow-derived mesenchymal stem cells affect immunologic profiling of interleukin-17-secreting cells in a chemical burn mouse model. Korean J. Ophthalmol. 2014, 28, 246–256. [Google Scholar] [CrossRef] [PubMed]
  98. Frank, S.; Stallmeyer, B.; Kampfer, H.; Kolb, N.; Pfeilschifter, J. Leptin enhances wound re-epithelialization and constitutes a direct function of leptin in skin repair. J. Clin. Investig. 2000, 106, 501–509. [Google Scholar] [CrossRef] [PubMed]
  99. Torsney, E.; Charlton, R.; Parums, D.; Collis, M.; Arthur, H.M. Inducible expression of human endoglin during inflammation and wound healing in vivo. Inflamm. Res. 2002, 51, 464–470. [Google Scholar] [CrossRef] [PubMed]
  100. Shibata, S.; Tada, Y.; Hau, C.S.; Mitsui, A.; Kamata, M.; Asano, Y.; Sugaya, M.; Kadono, T.; Masamoto, Y.; Kurokawa, M.; et al. Adiponectin regulates psoriasiform skin inflammation by suppressing IL-17 production from gammadelta-T cells. Nat. Commun. 2015, 6, 7687. [Google Scholar] [CrossRef] [PubMed]
  101. Qa’aty, N.; Vincent, M.; Wang, Y.; Wang, A.; Mitts, T.F.; Hinek, A. Synthetic ligands of the elastin receptor induce elastogenesis in human dermal fibroblasts via activation of their IGF-1 receptors. J. Dermatol. Sci. 2015, 80, 175–185. [Google Scholar] [CrossRef] [PubMed]
  102. Prokop, I.; Kononczuk, J.; Surazynski, A.; Palka, J. Cross-talk between integrin receptor and insulin-like growth factor receptor in regulation of collagen biosynthesis in cultured fibroblasts. Adv. Med. Sci. 2013, 58, 292–297. [Google Scholar] [CrossRef] [PubMed]
  103. Noordam, R.; Gunn, D.A.; Tomlin, C.C.; Maier, A.B.; Griffiths, T.; Catt, S.D.; Ogden, S.; Slagboom, P.E.; Westendorp, R.G.; Griffiths, C.E.; et al. Serum insulin-like growth factor 1 and facial ageing: High levels associate with reduced skin wrinkling in a cross-sectional study. Br. J. Dermatol. 2013, 168, 533–538. [Google Scholar] [CrossRef] [PubMed]
  104. Robertson, S.A.; Seamark, R.F. Granulocyte macrophage colony stimulating factor (GM-CSF) in the murine reproductive tract: Stimulation by seminal factors. Reprod. Fertil. Dev. 1990, 2, 359–368. [Google Scholar] [CrossRef] [PubMed]
  105. Iida, N.; Haisa, M.; Igarashi, A.; Pencev, D.; Grotendorst, G.R. Leukocyte-derived growth factor links the PDGF and CXC chemokine families of peptides. FASEB J. 1996, 10, 1336–1345. [Google Scholar] [PubMed]
  106. Schmitt, S.; Safferling, K.; Westphal, K.; Hrabowski, M.; Muller, U.; Angel, P.; Wiechert, L.; Ehemann, V.; Muller, B.; Holland-Cunz, S.; et al. Stathmin regulates keratinocyte proliferation and migration during cutaneous regeneration. PLoS ONE 2013, 8, e75075. [Google Scholar] [CrossRef]
  107. Bevan, D.; Gherardi, E.; Fan, T.P.; Edwards, D.; Warn, R. Diverse and potent activities of HGF/SF in skin wound repair. J. Pathol. 2004, 203, 831–838. [Google Scholar] [CrossRef] [PubMed]
  108. Peters, E.M.; Liotiri, S.; Bodo, E.; Hagen, E.; Biro, T.; Arck, P.C.; Paus, R. Probing the effects of stress mediators on the human hair follicle: Substance P holds central position. Am. J. Pathol. 2007, 171, 1872–1886. [Google Scholar] [CrossRef] [PubMed]
  109. Wu, Z.; Tang, Y.; Fang, H.; Su, Z.; Xu, B.; Lin, Y.; Zhang, P.; Wei, X. Decellularized scaffolds containing hyaluronic acid and EGF for promoting the recovery of skin wounds. J. Mater. Sci. Mater. Med. 2015, 26, 5322. [Google Scholar] [CrossRef] [PubMed]
  110. Gainza, G.; Bonafonte, D.C.; Moreno, B.; Aguirre, J.J.; Gutierrez, F.B.; Villullas, S.; Pedraz, J.L.; Igartua, M.; Hernandez, R.M. The topical administration of rhEGF-loaded nanostructured lipid carriers (rhEGF-NLC) improves healing in a porcine full-thickness excisional wound model. J. Control. Release 2015, 197, 41–47. [Google Scholar] [CrossRef] [PubMed]
  111. Norouzi, M.; Shabani, I.; Ahvaz, H.H.; Soleimani, M. PLGA/gelatin hybrid nanofibrous scaffolds encapsulating EGF for skin regeneration. J. Biomed. Mater. Res. Part A 2015, 103, 2225–2235. [Google Scholar] [CrossRef] [PubMed]
  112. Zhang, M.; Sun, L.; Wang, X.; Chen, S.; Jia, Q.; Liu, N.; Chen, Y.; Kong, Y.; Zhang, L.; Zhang, A.L. Activin B promotes BM-MSC-mediated cutaneous wound healing by regulating cell migration via the JNK-ERK signaling pathway. Cell Transp. 2013. [Google Scholar] [CrossRef]
  113. Zaragosi, L.E.; Wdziekonski, B.; Villageois, P.; Keophiphath, M.; Maumus, M.; Tchkonia, T.; Bourlier, V.; Mohsen-Kanson, T.; Ladoux, A.; Elabd, C.; et al. Activin A plays a critical role in proliferation and differentiation of human adipose progenitors. Diabetes 2010, 59, 2513–2521. [Google Scholar] [CrossRef] [PubMed]
  114. Moura, J.; da Silva, L.; Cruz, M.T.; Carvalho, E. Molecular and cellular mechanisms of bone morphogenetic proteins and activins in the skin: Potential benefits for wound healing. Arch. Dermatol. Res. 2013, 305, 557–569. [Google Scholar] [CrossRef] [PubMed]
  115. Dani, C. Activins in adipogenesis and obesity. Int. J. Obes. 2013, 37, 163–166. [Google Scholar] [CrossRef] [PubMed]
  116. Gragnani, A.; Cezillo, M.V.; da Silva, I.D.; de Noronha, S.M.; Correa-Noronha, S.A.; Ferreira, L.M. Gene expression profile of cytokines and receptors of inflammation from cultured keratinocytes of burned patients. Burns J. Int. Soc. Burn Inj. 2014, 40, 947–956. [Google Scholar] [CrossRef] [PubMed]
  117. Adachi, T.; Kobayashi, T.; Sugihara, E.; Yamada, T.; Ikuta, K.; Pittaluga, S.; Saya, H.; Amagai, M.; Nagao, K. Hair follicle-derived IL-7 and IL-15 mediate skin-resident memory T cell homeostasis and lymphoma. Nat. Med. 2015, 21, 1272–1279. [Google Scholar] [CrossRef] [PubMed]
  118. Knipper, J.A.; Willenborg, S.; Brinckmann, J.; Bloch, W.; Maass, T.; Wagener, R.; Krieg, T.; Sutherland, T.; Munitz, A.; Rothenberg, M.E.; et al. Interleukin-4 Receptor α Signaling in Myeloid Cells Controls Collagen Fibril Assembly in Skin Repair. Immunity 2015, 43, 803–816. [Google Scholar] [CrossRef] [PubMed]
  119. Crane, J.D.; MacNeil, L.G.; Lally, J.S.; Ford, R.J.; Bujak, A.L.; Brar, I.K.; Kemp, B.E.; Raha, S.; Steinberg, G.R.; Tarnopolsky, M.A. Exercise-stimulated interleukin-15 is controlled by AMPK and regulates skin metabolism and aging. Aging Cell 2015, 14, 625–634. [Google Scholar] [CrossRef] [PubMed]
  120. Kim, D.W.; Jeon, B.J.; Hwang, N.H.; Kim, M.S.; Park, S.H.; Dhong, E.S.; Yoon, E.S.; Lee, B.I. Adipose-derived stem cells inhibit epidermal melanocytes through an interleukin-6-mediated mechanism. Plast. Reconstr. Surg. 2014, 134, 470–480. [Google Scholar] [CrossRef] [PubMed]
  121. Brandt, K.; Grunler, J.; Brismar, K.; Wang, J. Effects of IGFBP-1 and IGFBP-2 and their fragments on migration and IGF-induced proliferation of human dermal fibroblasts. Growth Horm. IGF Res. 2015, 25, 34–40. [Google Scholar] [CrossRef] [PubMed]
  122. Kim, M.S.; Song, H.J.; Lee, S.H.; Lee, C.K. Comparative study of various growth factors and cytokines on type I collagen and hyaluronan production in human dermal fibroblasts. J. Cosmet. Dermatol. 2014, 13, 44–51. [Google Scholar] [CrossRef] [PubMed]
  123. Tzellos, T.G.; Klagas, I.; Vahtsevanos, K.; Triaridis, S.; Printza, A.; Kyrgidis, A.; Karakiulakis, G.; Zouboulis, C.C.; Papakonstantinou, E. Extrinsic ageing in the human skin is associated with alterations in the expression of hyaluronic acid and its metabolizing enzymes. Exp. Dermatol. 2009, 18, 1028–1035. [Google Scholar] [CrossRef] [PubMed]
  124. Nagaoka, A.; Yoshida, H.; Nakamura, S.; Morikawa, T.; Kawabata, K.; Kobayashi, M.; Sakai, S.; Takahashi, Y.; Okada, Y.; Inoue, S. Regulation of Hyaluronan (HA) Metabolism Mediated by HYBID (Hyaluronan-binding Protein Involved in HA Depolymerization, KIAA1199) and HA Synthases in Growth Factor-stimulated Fibroblasts. J. Biol. Chem. 2015, 290, 30910–30923. [Google Scholar] [CrossRef] [PubMed]
  125. Anitua, E.; Sanchez, M.; Zalduendo, M.M.; de la Fuente, M.; Prado, R.; Orive, G.; Andia, I. Fibroblastic response to treatment with different preparations rich in growth factors. Cell Prolif. 2009, 42, 162–170. [Google Scholar] [CrossRef] [PubMed]
  126. Poimenidi, E.; Theodoropoulou, C.; Koutsioumpa, M.; Skondra, L.; Droggiti, E.; van den Broek, M.; Koolwijk, P.; Papadimitriou, E. Vascular endothelial growth factor A (VEGF-A) decreases expression and secretion of pleiotrophin in a VEGF receptor-independent manner. Vasc. Pharmacol. 2016, 80, 11–19. [Google Scholar] [CrossRef] [PubMed]
  127. Rezza, A.; Sennett, R.; Tanguy, M.; Clavel, C.; Rendl, M. PDGF signalling in the dermis and in dermal condensates is dispensable for hair follicle induction and formation. Exp. Dermatol. 2015, 24, 468–470. [Google Scholar] [CrossRef] [PubMed]
  128. Ezure, T.; Amano, S. Adiponectin and leptin up-regulate extracellular matrix production by dermal fibroblasts. BioFactors 2007, 31, 229–236. [Google Scholar] [CrossRef] [PubMed]
  129. Tadokoro, S.; Ide, S.; Tokuyama, R.; Umeki, H.; Tatehara, S.; Kataoka, S.; Satomura, K. Leptin promotes wound healing in the skin. PLoS ONE 2015, 10, e0121242. [Google Scholar] [CrossRef] [PubMed]
  130. Kypriotou, M.; Rivero, D.; Haller, S.; Mariotto, A.; Huber, M.; Acha-Orbea, H.; Werner, S.; Hohl, D. Activin A inhibits antigen-induced allergy in murine epicutaneous sensitization. Front. Immunol. 2013, 4, 246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Dugrillon, A.; Eichler, H.; Kern, S.; Kluter, H. Autologous concentrated platelet-rich plasma (cPRP) for local application in bone regeneration. Int. J. Oral Maxillofac. Surg. 2002, 31, 615–619. [Google Scholar] [CrossRef] [PubMed]
  132. Rendu, F.; Brohard-Bohn, B. The platelet release reaction: Granules’ constituents, secretion and functions. Platelets 2001, 12, 261–273. [Google Scholar] [CrossRef] [PubMed]
  133. Hassan, W.U.; Greiser, U.; Wang, W. Role of adipose-derived stem cells in wound healing. Wound Repair Regen. 2014, 22, 313–325. [Google Scholar] [CrossRef] [PubMed]
  134. Ojeh, N.; Pastar, I.; Tomic-Canic, M.; Stojadinovic, O. Stem Cells in Skin Regeneration, Wound Healing, and Their Clinical Applications. Int. J. Mol. Sci. 2015, 16, 25476–25501. [Google Scholar] [CrossRef] [PubMed]
  135. Nathan, C.F. Neutrophil activation on biological surfaces. Massive secretion of hydrogen peroxide in response to products of macrophages and lymphocytes. J. Clin. Investig. 1987, 80, 1550–1560. [Google Scholar] [CrossRef] [PubMed]
  136. Singer, A.J.; Clark, R.A. Cutaneous wound healing. N. Engl. J. Med. 1999, 341, 738–746. [Google Scholar] [PubMed]
  137. Festa, E.; Fretz, J.; Berry, R.; Schmidt, B.; Rodeheffer, M.; Horowitz, M.; Horsley, V. Adipocyte lineage cells contribute to the skin stem cell niche to drive hair cycling. Cell 2011, 146, 761–771. [Google Scholar] [CrossRef] [PubMed]
  138. Gomez, D.E.; Alonso, D.F.; Yoshiji, H.; Thorgeirsson, U.P. Tissue inhibitors of metalloproteinases: Structure, regulation and biological functions. Eur. J. Cell Biol. 1997, 74, 111–122. [Google Scholar] [PubMed]
  139. Fisher, G.J.; Varani, J.; Voorhees, J.J. Looking older: Fibroblast collapse and therapeutic implications. Arch. Dermatol. 2008, 144, 666–672. [Google Scholar] [CrossRef] [PubMed]
  140. Rittie, L.; Berton, A.; Monboisse, J.C.; Hornebeck, W.; Gillery, P. Decreased contraction of glycated collagen lattices coincides with impaired matrix metalloproteinase production. Biochem. Biophys. Res. Commun. 1999, 264, 488–492. [Google Scholar] [CrossRef] [PubMed]
  141. Eming, S.A.; Werner, S.; Bugnon, P.; Wickenhauser, C.; Siewe, L.; Utermohlen, O.; Davidson, J.M.; Krieg, T.; Roers, A. Accelerated wound closure in mice deficient for interleukin-10. Am. J. Pathol. 2007, 170, 188–202. [Google Scholar] [CrossRef] [PubMed]
  142. McCartney-Francis, N.; Mizel, D.; Wong, H.; Wahl, L.; Wahl, S. TGF-β regulates production of growth factors and TGF-β by human peripheral blood monocytes. Growth Factors 1990, 4, 27–35. [Google Scholar] [CrossRef] [PubMed]
  143. Kiritsy, C.P.; Lynch, A.B.; Lynch, S.E. Role of growth factors in cutaneous wound healing: A review. Crit. Rev. Oral Biol. Med. 1993, 4, 729–760. [Google Scholar] [CrossRef] [PubMed]
  144. Mateo, R.B.; Reichner, J.S.; Albina, J.E. Interleukin-6 activity in wounds. Am. J. Physiol. 1994, 266, R1840–R1844. [Google Scholar] [PubMed]
  145. Moulin, V. Growth factors in skin wound healing. Eur. J. Cell Biol. 1995, 68, 1–7. [Google Scholar] [PubMed]
  146. Nishikawa, K.; Asai, T.; Shigematsu, H.; Shimizu, K.; Kato, H.; Asano, Y.; Takashima, S.; Mekada, E.; Oku, N.; Minamino, T. Development of anti-HB-EGF immunoliposomes for the treatment of breast cancer. J. Control. Release 2012, 160, 274–280. [Google Scholar] [CrossRef] [PubMed]
  147. Lee, J.Y.; Yun, M.; Paik, J.S.; Lee, S.B.; Yang, S.W. PDGF-BB Enhances the Proliferation of Cells in Human Orbital Fibroblasts by Suppressing PDCD4 Expression via Up-Regulation of microRNA-21. Investig. Ophthalmol. Vis. Sci. 2016, 57, 908–913. [Google Scholar] [CrossRef] [PubMed]
  148. Scholz, F.; Schulte, A.; Adamski, F.; Hundhausen, C.; Mittag, J.; Schwarz, A.; Kruse, M.L.; Proksch, E.; Ludwig, A. Constitutive expression and regulated release of the transmembrane chemokine CXCL16 in human and murine skin. J. Investig. Dermatol. 2007, 127, 1444–1455. [Google Scholar] [CrossRef] [PubMed]
  149. Sanders, A.J.; Jiang, D.G.; Jiang, W.G.; Harding, K.G.; Patel, G.K. Activated leukocyte cell adhesion molecule impacts on clinical wound healing and inhibits HaCaT migration. Int. Wound J. 2011, 8, 500–507. [Google Scholar] [CrossRef] [PubMed]
  150. Chen, L.; Ren, G.S.; Li, F.; Sun, S.Q. Expression of livin and vascular endothelial growth factor in different clinical stages of human esophageal carcinoma. World J. Gastroenterol. 2008, 14, 5749–5754. [Google Scholar] [CrossRef] [PubMed]
  151. Zanetti, A.; Lampugnani, M.G.; Balconi, G.; Breviario, F.; Corada, M.; Lanfrancone, L.; Dejana, E. Vascular endothelial growth factor induces SHC association with vascular endothelial cadherin: A potential feedback mechanism to control vascular endothelial growth factor receptor-2 signaling. Arterioscler. Thromb. Vasc. Biol. 2002, 22, 617–622. [Google Scholar] [CrossRef] [PubMed]
  152. Park, I.S.; Chung, P.S.; Ahn, J.C. Adipose-derived stromal cell cluster with light therapy enhance angiogenesis and skin wound healing in mice. Biochem. Biophys. Res. Commun. 2015, 462, 171–177. [Google Scholar] [CrossRef] [PubMed]
  153. Kampfer, H.; Pfeilschifter, J.; Frank, S. Expressional regulation of angiopoietin-1 and -2 and the tie-1 and -2 receptor tyrosine kinases during cutaneous wound healing: A comparative study of normal and impaired repair. Lab. Investig. J. Tech. Methods Pathol. 2001, 81, 361–373. [Google Scholar] [CrossRef]
  154. Staton, C.A.; Valluru, M.; Hoh, L.; Reed, M.W.; Brown, N.J. Angiopoietin-1, angiopoietin-2 and Tie-2 receptor expression in human dermal wound repair and scarring. Br. J. Dermatol. 2010, 163, 920–927. [Google Scholar] [CrossRef] [PubMed]
  155. Joao de Masi, E.C.; Campos, A.C.; Joao de Masi, F.D.; Ratti, M.A.; Ike, I.S.; Joao de Mais, R.D. The influence of growth factors on skin wound healing in rats. Braz. J. Otorhinolaryngol. 2016, 82, 512–521. [Google Scholar] [CrossRef] [PubMed]
  156. Peplow, P.V.; Chatterjee, M.P. A review of the influence of growth factors and cytokines in in vitro human keratinocyte migration. Cytokine 2013, 62, 1–21. [Google Scholar] [CrossRef] [PubMed]
  157. Werner, S.; Krieg, T.; Smola, H. Keratinocyte-fibroblast interactions in wound healing. J. Investig. Dermatol. 2007, 127, 998–1008. [Google Scholar] [CrossRef] [PubMed]
  158. Ansel, J.; Perry, P.; Brown, J.; Damm, D.; Phan, T.; Hart, C.; Luger, T.; Hefeneider, S. Cytokine modulation of keratinocyte cytokines. J. Investig. Dermatol. 1990, 94 (Suppl. S6), 101S–107S. [Google Scholar] [CrossRef] [PubMed]
  159. Finch, P.W.; Rubin, J.S.; Miki, T.; Ron, D.; Aaronson, S.A. Human KGF is FGF-related with properties of a paracrine effector of epithelial cell growth. Science 1989, 245, 752–755. [Google Scholar] [CrossRef] [PubMed]
  160. Grossman, R.M.; Krueger, J.; Yourish, D.; Granelli-Piperno, A.; Murphy, D.P.; May, L.T.; Kupper, T.S.; Sehgal, P.B.; Gottlieb, A.B. Interleukin 6 is expressed in high levels in psoriatic skin and stimulates proliferation of cultured human keratinocytes. Proc. Natl. Acad. Sci. USA 1989, 86, 6367–6371. [Google Scholar] [CrossRef] [PubMed]
  161. Freedberg, I.M.; Tomic-Canic, M.; Komine, M.; Blumenberg, M. Keratins and the keratinocyte activation cycle. J. Investig. Dermatol. 2001, 116, 633–640. [Google Scholar] [CrossRef] [PubMed]
  162. Babu, P.S.; Danilovich, N.; Sairam, M.R. Hormone-induced receptor gene splicing: Enhanced expression of the growth factor type I follicle-stimulating hormone receptor motif in the developing mouse ovary as a new paradigm in growth regulation. Endocrinology 2001, 142, 381–389. [Google Scholar] [CrossRef] [PubMed]
  163. Florin, L.; Maas-Szabowski, N.; Werner, S.; Szabowski, A.; Angel, P. Increased keratinocyte proliferation by JUN-dependent expression of PTN and SDF-1 in fibroblasts. J. Cell Sci. 2005, 118, 1981–1989. [Google Scholar] [CrossRef] [PubMed]
  164. Kapur, S.K.; Dos-Anjos Vilaboa, S.; Llull, R.; Katz, A.J. Adipose tissue and stem/progenitor cells: Discovery and development. Clin. Plast. Surg. 2015, 42, 155–167. [Google Scholar] [CrossRef] [PubMed]
  165. Conde-Green, A.; Marano, A.A.; Lee, E.S.; Reisler, T.; Price, L.A.; Milner, S.M.; Granick, M.S. Fat Grafting and Adipose-Derived Regenerative Cells in Burn Wound Healing and Scarring: A Systematic Review of the Literature. Plast. Reconstr. Surg. 2016, 137, 302–312. [Google Scholar] [CrossRef] [PubMed]
  166. Negenborn, V.L.; Groen, J.W.; Smit, J.M.; Niessen, F.B.; Mullender, M.G. The Use of Autologous Fat Grafting for Treatment of Scar Tissue and Scar-Related Conditions: A Systematic Review. Plast. Surg. Nurs. 2016, 36, 131–143. [Google Scholar] [CrossRef] [PubMed]
  167. Shridharani, S.M.; Broyles, J.M.; Matarasso, A. Liposuction devices: Technology update. Med. Devices 2014, 7, 241–251. [Google Scholar] [CrossRef] [PubMed]
  168. 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] [PubMed]
  169. Banyard, D.A.; Salibian, A.A.; Widgerow, A.D.; Evans, G.R. Implications for human adipose-derived stem cells in plastic surgery. J. Cell. Mol. Med. 2015, 19, 21–30. [Google Scholar] [CrossRef] [PubMed]
  170. Aronowitz, J.A.; Lockhart, R.A.; Hakakian, C.S. Mechanical versus enzymatic isolation of stromal vascular fraction cells from adipose tissue. SpringerPlus 2015, 4, 713. [Google Scholar] [CrossRef] [PubMed]
  171. Cleveland, E.C.; Albano, N.J.; Hazen, A. Roll, Spin, Wash, or Filter? Processing of Lipoaspirate for Autologous Fat Grafting: An Updated, Evidence-Based Review of the Literature. Plast. Reconstr. Surg. 2015, 136, 706–713. [Google Scholar] [CrossRef] [PubMed]
  172. Conde-Green, A.; Kotamarti, V.S.; Sherman, L.S.; Keith, J.D.; Lee, E.S.; Granick, M.S.; Rameshwar, P. Shift toward Mechanical Isolation of Adipose-derived Stromal Vascular Fraction: Review of Upcoming Techniques. Plastic and reconstructive surgery. Glob. Open 2016, 4, e1017. [Google Scholar] [CrossRef] [PubMed]
  173. Hanke, A.; Prantl, L.; Wenzel, C.; Nerlich, M.; Brockhoff, G.; Loibl, M.; Gehmert, S. Semi-automated extraction and characterization of Stromal Vascular Fraction using a new medical device. Clin. Hemorheol. Microcirc. 2016. [Google Scholar] [CrossRef] [PubMed]
  174. van Dongen, J.A.; Stevens, H.P.; Parvizi, M.; van der Lei, B.; Harmsen, M.C. The fractionation of adipose tissue procedure to obtain stromal vascular fractions for regenerative purposes. Wound Repair Regen. 2016, 24, 994–1003. [Google Scholar] [CrossRef] [PubMed]
  175. Grasys, J.; Kim, B.S.; Pallua, N. Content of Soluble Factors and Characteristics of Stromal Vascular Fraction Cells in Lipoaspirates from Different Subcutaneous Adipose Tissue Depots. Aesthet. Surg. J. 2016, 36, 831–841. [Google Scholar] [CrossRef] [PubMed]
  176. Kakagia, D.; Pallua, N. Autologous fat grafting: In search of the optimal technique. Surg. Innov. 2014, 21, 327–336. [Google Scholar] [CrossRef] [PubMed]
  177. Strong, A.L.; Cederna, P.S.; Rubin, J.P.; Coleman, S.R.; Levi, B. The Current State of Fat Grafting: A Review of Harvesting, Processing, and Injection Techniques. Plast. Reconstr. Surg. 2015, 136, 897–912. [Google Scholar] [CrossRef] [PubMed]
  178. Chen, Y.W.; Wang, J.R.; Liao, X.; Li, S.H.; Xiao, L.L.; Cheng, B.; Xie, G.H.; Song, J.X.; Liu, H.W. Effect of suction pressures on cell yield and functionality of the adipose-derived stromal vascular fraction. J. Plast. Reconstr. Aesthet. Surg. 2016. [Google Scholar] [CrossRef] [PubMed]
  179. Guo, J.; Nguyen, A.; Banyard, D.A.; Fadavi, D.; Toranto, J.D.; Wirth, G.A.; Paydar, K.Z.; Evans, G.R.; Widgerow, A.D. Stromal vascular fraction: A regenerative reality? Part 2: Mechanisms of regenerative action. J. Plast. Reconstr. Aesthet. Surg. 2016, 69, 180–188. [Google Scholar] [CrossRef] [PubMed]
  180. Streit, L.; Jaros, J.; Sedlakova, V.; Sedlackova, M.; Drazan, L.; Svoboda, M.; Pospisil, J.; Vyska, T.; Vesely, J.; Hampl, A. A Comprehensive In Vitro Comparison of Preparation Techniques for Fat Grafting. Plast. Reconstr. Surg. 2016. [Google Scholar] [CrossRef] [PubMed]
  181. Jiang, Y.; Berry, D.C.; Tang, W.; Graff, J.M. Independent stem cell lineages regulate adipose organogenesis and adipose homeostasis. Cell Rep. 2014, 9, 1007–1022. [Google Scholar] [CrossRef] [PubMed]
  182. Gimble, J.M.; Guilak, F. Differentiation potential of adipose derived adult stem (ADAS) cells. Curr. Top. Dev. Biol. 2003, 58, 137–160. [Google Scholar] [PubMed]
  183. Katz, A.J.; Tholpady, A.; Tholpady, S.S.; Shang, H.; Ogle, R.C. Cell surface and transcriptional characterization of human adipose-derived adherent stromal (hADAS) cells. Stem Cells 2005, 23, 412–423. [Google Scholar] [CrossRef] [PubMed]
  184. 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] [PubMed]
  185. 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] [PubMed]
  186. Strioga, M.; Viswanathan, S.; Darinskas, A.; Slaby, O.; Michalek, J. Same or not the same? Comparison of adipose tissue-derived versus bone marrow-derived mesenchymal stem and stromal cells. Stem Cells Dev. 2012, 21, 2724–2752. [Google Scholar] [CrossRef] [PubMed]
  187. Melief, S.M.; Schrama, E.; Brugman, M.H.; Tiemessen, M.M.; Hoogduijn, M.J.; Fibbe, W.E.; Roelofs, H. Multipotent stromal cells induce human regulatory T cells through a novel pathway involving skewing of monocytes toward anti-inflammatory macrophages. Stem Cells 2013, 31, 1980–1991. [Google Scholar] [CrossRef] [PubMed]
  188. Li, C.Y.; Wu, X.Y.; Tong, J.B.; Yang, X.X.; Zhao, J.L.; Zheng, Q.F.; Zhao, G.B.; Ma, Z.J. Comparative analysis of human mesenchymal stem cells from bone marrow and adipose tissue under xeno-free conditions for cell therapy. Stem Cell Res. Ther. 2015, 6, 55. [Google Scholar] [CrossRef] [PubMed]
  189. Wang, J.; Geesman, G.J.; Hostikka, S.L.; Atallah, M.; Blackwell, B.; Lee, E.; Cook, P.J.; Pasaniuc, B.; Shariat, G.; Halperin, E.; et al. Inhibition of activated pericentromeric SINE/Alu repeat transcription in senescent human adult stem cells reinstates self-renewal. Cell Cycle 2011, 10, 3016–3030. [Google Scholar] [CrossRef] [PubMed]
  190. Lopez, M.; Niu, P.; Wang, L.; Vogelsang, M.; Gaur, M.; Krastins, B.; Zhao, Y.; Smagul, A.; Nussupbekova, A.; Akanov, A.; et al. Opposing activities of oncogenic MIR17HG and tumor suppressive MIR100HG clusters and their gene targets regulate replicative senescence in human adult stem cells. Aging Mech. Dis. 2017, in press. [Google Scholar]
  191. Gruber, H.E.; Somayaji, S.; Riley, F.; Hoelscher, G.L.; Norton, H.J.; Ingram, J.; Hanley, E.N., Jr. Human adipose-derived mesenchymal stem cells: Serial passaging, doubling time and cell senescence. Biotech. Histochem. 2012, 87, 303–311. [Google Scholar] [CrossRef] [PubMed]
  192. Jun, H.S.; Dao, L.T.; Pyun, J.C.; Cho, S. Effect of cell senescence on the impedance measurement of adipose tissue-derived stem cells. Enzym. Microb. Technol. 2013, 53, 302–306. [Google Scholar] [CrossRef] [PubMed]
  193. von Mering, C.; Jensen, L.J.; Snel, B.; Hooper, S.D.; Krupp, M.; Foglierini, M.; Jouffre, N.; Huynen, M.A.; Bork, P. STRING: Known and predicted protein-protein associations, integrated and transferred across organisms. Nucleic Acids Res. 2005, 33, D433–D437. [Google Scholar] [CrossRef]
  194. Jeyapalan, J.C.; Sedivy, J.M. Cellular senescence and organismal aging. Mech. Ageing Dev. 2008, 129, 467–474. [Google Scholar] [CrossRef] [PubMed]
  195. Campisi, J. Cellular senescence as a tumor-suppressor mechanism. Trends Cell Biol. 2001, 11, S27–S31. [Google Scholar] [CrossRef]
  196. Lin, A.W.; Barradas, M.; Stone, J.C.; van Aelst, L.; Serrano, M.; Lowe, S.W. Premature senescence involving p53 and p16 is activated in response to constitutive MEK/MAPK mitogenic signaling. Genes Dev. 1998, 12, 3008–3019. [Google Scholar] [CrossRef] [PubMed]
  197. Kishi, S.; Bayliss, P.E.; Hanai, J. A prospective epigenetic paradigm between cellular senescence and epithelial-mesenchymal transition in organismal development and aging. Transl. Res. J. Lab. Clin. Med. 2015, 165, 241–249. [Google Scholar] [CrossRef] [PubMed]
  198. Agostini, T.; Lazzeri, D.; Pini, A.; Marino, G.; Li Quattrini, A.; Bani, D.; Dini, M. Wet and dry techniques for structural fat graft harvesting: Histomorphometric and cell viability assessments of lipoaspirated samples. Plast. Reconstr. Surg. 2012, 130, 331e–339e. [Google Scholar] [CrossRef] [PubMed]
  199. Coleman, S.R.; Katzel, E.B. Fat Grafting for Facial Filling and Regeneration. Clin. Plast. Surg. 2015, 42, 289–300. [Google Scholar] [CrossRef] [PubMed]
  200. Marten, T.J.; Elyassnia, D. Fat grafting in facial rejuvenation. Clin. Plast. Surg. 2015, 42, 219–252. [Google Scholar] [CrossRef] [PubMed]
  201. Sinno, S.; Mehta, K.; Simmons, C.; Stuzin, J.M. Current Trends in Facial Rejuvenation: An Assessment of ASPS Members Utilization of Fat Grafting During Facelifting. Plast. Reconstr. Surg. 2015, 136 (Suppl. S4), 141. [Google Scholar] [CrossRef] [PubMed]
  202. Maione, L.; Memeo, A.; Pedretti, L.; Verdoni, F.; Lisa, A.; Bandi, V.; Giannasi, S.; Vinci, V.; Mambretti, A.; Klinger, M. Autologous fat graft as treatment of post short stature surgical correction scars. Injury 2014, 45 (Suppl. S6), S126–S132. [Google Scholar] [CrossRef] [PubMed]
  203. Bernardini, F.P.; Gennai, A.; Izzo, L.; Zambelli, A.; Repaci, E.; Baldelli, I.; Fraternali-Orcioni, G.; Hartstein, M.E.; Santi, P.L.; Quarto, R. Superficial Enhanced Fluid Fat Injection (SEFFI) to Correct Volume Defects and Skin Aging of the Face and Periocular Region. Aesthet. Surg. J. 2015, 35, 504–515. [Google Scholar] [CrossRef] [PubMed]
  204. Giunta, R.E.; Eder, M.; Machens, H.G.; Muller, D.F.; Kovacs, L. Structural fat grafting for rejuvenation of the dorsum of the hand. Handchir. Mikrochir. Plast. Chir. 2010, 42, 143–147. [Google Scholar] [CrossRef] [PubMed]
  205. Luu, C.A.; Larson, E.; Rankin, T.M.; Pappalardo, J.L.; Slepian, M.J.; Armstrong, D.G. Plantar Fat Grafting and Tendon Balancing for the Diabetic Foot Ulcer in Remission. Plast. Reconstr. Surg. Glob. Open 2016, 4, e810. [Google Scholar] [CrossRef] [PubMed]
  206. Zvonic, S.; Lefevre, M.; Kilroy, G.; Floyd, Z.E.; DeLany, J.P.; Kheterpal, I.; Gravois, A.; Dow, R.; White, A.; Wu, X.; et al. Secretome of primary cultures of human adipose-derived stem cells: Modulation of serpins by adipogenesis. Mol. Cell. Proteom. 2007, 6, 18–28. [Google Scholar] [CrossRef] [PubMed]
  207. Jansma, J.; Schepers, R.H.; Vissink, A. Microfat transfer in cosmetic facial procedures. Ned. Tijdschr. Tandheelk. 2014, 121, 330–335. [Google Scholar] [CrossRef]
  208. Kemaloglu, C.A. Nanofat grafting under a split-thickness skin graft for problematic wound management. SpringerPlus 2016, 5, 138. [Google Scholar] [CrossRef] [PubMed]
  209. Modarressi, A. Platlet rich plasma (prp) improves fat grafting outcomes. World J Plast Surg 2013, 2, 6–13. [Google Scholar] [PubMed]
  210. 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]
  211. Karagianni, M.; Kraneburg, U.; Kluter, H.; Machens, H.G.; Bieback, K.; Schantz, J.T.; Papadopulos, N. Autologous fat grafts and supportive enrichment with adipose tissue stromal cells. Handchir. Mikrochir. Plast. Chir. 2013, 45, 93–98. [Google Scholar] [PubMed]
  212. Han, S.; Sun, H.M.; Hwang, K.C.; Kim, S.W. Adipose-derived stromal vascular fraction cells: Update on clinical utility and efficacy. Crit. Rev. Eukaryot. Gene Expr. 2015, 25, 145–152. [Google Scholar] [CrossRef] [PubMed]
  213. Nguyen, A.; Guo, J.; Banyard, D.A.; Fadavi, D.; Toranto, J.D.; Wirth, G.A.; Paydar, K.Z.; Evans, G.R.; Widgerow, A.D. Stromal vascular fraction: A regenerative reality? Part 1: Current concepts and review of the literature. J. Plast. Reconstr. Aesthet. Surg. 2016, 69, 170–179. [Google Scholar] [CrossRef] [PubMed]
  214. Rigotti, G.; Marchi, A.; Galie, 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]
  215. Toyserkani, N.M.; Christensen, M.L.; Sheikh, S.P.; Sorensen, J.A. Adipose-Derived Stem Cells: New Treatment for Wound Healing? Ann. Plast. Surg. 2015, 75, 117–123. [Google Scholar] [CrossRef] [PubMed]
  216. Marques, L.F.; Stessuk, T.; Camargo, I.C.; Sabeh Junior, N.; dos Santos, L.; Ribeiro-Paes, J.T. Platelet-rich plasma (PRP): Methodological aspects and clinical applications. Platelets 2015, 26, 101–113. [Google Scholar] [CrossRef] [PubMed]
  217. Stessuk, T.; Puzzi, M.B.; Chaim, E.A.; Alves, P.C.; de Paula, E.V.; Forte, A.; Izumizawa, J.M.; Oliveira, C.C.; Frei, F.; Ribeiro-Paes, J.T. Platelet-rich plasma (PRP) and adipose-derived mesenchymal stem cells: Stimulatory effects on proliferation and migration of fibroblasts and keratinocytes in vitro. Arch. Dermatol. Res. 2016, 308, 511–520. [Google Scholar] [CrossRef] [PubMed]
  218. Son, W.C.; Yun, J.W.; Kim, B.H. Adipose-derived mesenchymal stem cells reduce MMP-1 expression in UV-irradiated human dermal fibroblasts: Therapeutic potential in skin wrinkling. Biosci. Biotechnol. Biochem. 2015, 79, 919–925. [Google Scholar] [CrossRef] [PubMed]
  219. Dos Anjos, S.; Matas-Palau, A.; Mercader, J.; Katz, A.J.; Llull, R. Reproducible Volume Restoration and Efficient Long-term Volume Retention after Point-of-care Standardized Cell-enhanced Fat Grafting in Breast Surgery. Plast. Reconstr. Surg. Glob. Open 2015, 3, e547. [Google Scholar] [CrossRef] [PubMed]
  220. Wankhade, U.D.; Shen, M.; Kolhe, R.; Fulzele, S. Advances in Adipose-Derived Stem Cells Isolation, Characterization, and Application in Regenerative Tissue Engineering. Stem Cells Int. 2016, 2016, 3206807. [Google Scholar] [CrossRef] [PubMed]
  221. Gentile, P.; de Angelis, B.; Pasin, M.; Cervelli, G.; Curcio, C.B.; Floris, M.; Di Pasquali, C.; Bocchini, I.; Balzani, A.; Nicoli, F.; et al. Adipose-derived stromal vascular fraction cells and platelet-rich plasma: Basic and clinical evaluation for cell-based therapies in patients with scars on the face. J. Craniofac. Surg. 2014, 25, 267–272. [Google Scholar] [CrossRef] [PubMed]
  222. Trojahn Kolle, 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] [PubMed]
  223. Tonnard, P.; Verpaele, A.; Peeters, G.; Hamdi, M.; Cornelissen, M.; Declercq, H. Nanofat grafting: Basic research and clinical applications. Plast. Reconstr. Surg. 2013, 132, 1017–1026. [Google Scholar] [CrossRef] [PubMed]
  224. Ahn, S.H.; Lee, H.J.; Lee, J.S.; Yoon, H.; Chun, W.; Kim, G.H. A novel cell-printing method and its application to hepatogenic differentiation of human adipose stem cell-embedded mesh structures. Sci. Rep. 2015, 5, 13427. [Google Scholar] [CrossRef] [PubMed]
  225. Atalay, S.; Coruh, A.; Deniz, K. Stromal vascular fraction improves deep partial thickness burn wound healing. Burns J. Int. Soc. Burn Inj. 2014, 40, 1375–1383. [Google Scholar] [CrossRef] [PubMed]
  226. 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 J. Int. Soc. Burn Inj. 2015, 41, 1504–1516. [Google Scholar] [CrossRef] [PubMed]
  227. Cheng, C.; Sheng, L.; Li, H.; Mao, X.; Zhu, M.; Gao, B.; Li, Q. Cell-Assisted Skin Grafting: Improving Texture and Elasticity of Skin Grafts through Autologous Cell Transplantation. Plast. Reconstr. Surg. 2016, 137, 58e–66e. [Google Scholar] [CrossRef] [PubMed]
  228. Klar, A.S.; Guven, S.; Zimoch, J.; Zapiorkowska, N.A.; Biedermann, T.; Bottcher-Haberzeth, S.; Meuli-Simmen, C.; Martin, I.; Scherberich, A.; Reichmann, E.; et al. Characterization of vasculogenic potential of human adipose-derived endothelial cells in a three-dimensional vascularized skin substitute. Pediatr. Surg. Int. 2016, 32, 17–27. [Google Scholar] [CrossRef] [PubMed]
  229. Brohem, C.A.; de Carvalho, C.M.; Radoski, C.L.; Santi, F.C.; Baptista, M.C.; Swinka, B.B.; de, A.U.C.; de Araujo, L.R.; Graf, R.M.; Feferman, I.H.; et al. Comparison between fibroblasts and mesenchymal stem cells derived from dermal and adipose tissue. Int. J. Cosmet. Sci. 2013, 35, 448–457. [Google Scholar] [CrossRef] [PubMed]
  230. Tissiani, L.A.; Alonso, N. A Prospective and Controlled Clinical Trial on Stromal Vascular Fraction Enriched Fat Grafts in Secondary Breast Reconstruction. Stem Cells Int. 2016, 2016, 2636454. [Google Scholar] [CrossRef] [PubMed]
  231. Chatterjee, S.; Laliberte, M.; Blelloch, S.; Ratanshi, I.; Safneck, J.; Buchel, E.; Raouf, A. Adipose-Derived Stromal Vascular Fraction Differentially Expands Breast Progenitors in Tissue Adjacent to Tumors Compared to Healthy Breast Tissue. Plast. Reconstr. Surg. 2015, 136, 414e–425e. [Google Scholar] [CrossRef] [PubMed]
  232. Toyserkani, N.M.; Quaade, M.L.; Sorensen, J.A. Cell-Assisted Lipotransfer: A Systematic Review of Its Efficacy. Aesthet. Plast. Surg. 2016, 40, 309–318. [Google Scholar] [CrossRef] [PubMed]
  233. Wang, L.; Luo, X.; Lu, Y.; Fan, Z.H.; Hu, X. Is the Resorption of Grafted Fat Reduced in Cell-Assisted Lipotransfer for Breast Augmentation? Ann. Plast. Surg. 2015, 75, 128–134. [Google Scholar] [CrossRef] [PubMed]
  234. Kozlik, M.; Wojcicki, P. The use of stem cells in plastic and reconstructive surgery. Adv. Clin. Exp. Med. Off. Organ Wroc. Med. Univ. 2014, 23, 1011–1017. [Google Scholar] [CrossRef] [PubMed]
  235. Minteer, D.; Marra, K.G.; Rubin, J.P. Adipose-derived mesenchymal stem cells: Biology and potential applications. Adv. Biochem. Eng. Biotechnol. 2013, 129, 59–71. [Google Scholar] [PubMed]
  236. Mestak, O.; Sukop, A.; Hsueh, Y.S.; Molitor, M.; Mestak, J.; Matejovska, J.; Zarubova, L. Centrifugation versus PureGraft for fatgrafting to the breast after breast-conserving therapy. World J. Surg. Oncol. 2014, 12, 178. [Google Scholar] [CrossRef] [PubMed]
  237. Tran, T.T.; Kahn, C.R. Transplantation of adipose tissue and stem cells: Role in metabolism and disease. Nat. Rev. Endocrinol. 2010, 6, 195–213. [Google Scholar] [CrossRef] [PubMed]
  238. Rigotti, G.; Charles-de-Sa, 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. Aesthet. Surg. J. Am. Soc. Aesthet. Plast. Surg. 2016, 36, 261–270. [Google Scholar] [CrossRef] [PubMed]
  239. Charles-de-Sa, 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] [PubMed]
  240. Fukuoka, H.; Suga, H. Hair Regeneration Treatment Using Adipose-Derived Stem Cell Conditioned Medium: Follow-up With Trichograms. Eplasty 2015, 15, e10. [Google Scholar] [PubMed]
  241. Asero, R.; Tedeschi, A.; Lorini, M.; Gerosa, M.; Meroni, P.; Riboldi, P. Circulating stem cell factor in patients with chronic idiopathic urticaria. Ann. Allergy Asthma Immunol. 2003, 91, 79–81. [Google Scholar] [CrossRef]
  242. Kesten, S.; Fraser, J.K. Autologous Adipose Derived Regenerative Cells: A Platform for Therapeutic Applications. Surg. Technol. Int. 2016, XXIX, 38–44. [Google Scholar]
  243. Granel, B.; Daumas, A.; Jouve, E.; Harle, J.R.; Nguyen, P.S.; Chabannon, C.; Colavolpe, N.; Reynier, J.C.; Truillet, R.; Mallet, S.; et al. Safety, tolerability and potential efficacy of injection of autologous adipose-derived stromal vascular fraction in the fingers of patients with systemic sclerosis: An open-label phase I trial. Ann. Rheum. Dis. 2015, 74, 2175–2182. [Google Scholar] [CrossRef] [PubMed]
  244. Liew, A.; O’Brien, T. Therapeutic potential for mesenchymal stem cell transplantation in critical limb ischemia. Stem Cell Res. Ther. 2012, 3, 28. [Google Scholar] [CrossRef] [PubMed]
  245. Kim, I.; Lee, S.K.; Yoon, J.I.; Kim, D.E.; Kim, M.; Ha, H. Fibrin glue improves the therapeutic effect of MSCs by sustaining survival and paracrine function. Tissue Eng. Part A 2013, 19, 2373–2381. [Google Scholar] [CrossRef] [PubMed]
  246. Marston, W.A.; Hanft, J.; Norwood, P.; Pollak, R. The efficacy and safety of Dermagraft in improving the healing of chronic diabetic foot ulcers: Results of a prospective randomized trial. Diabetes Care 2003, 26, 1701–1705. [Google Scholar] [CrossRef] [PubMed]
  247. Claudio-da-Silva, C.; Baptista, L.S.; Carias, R.B.; Menezes Neto Hda, 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] [PubMed]
  248. Tollervey, J.R.; Lunyak, V.V. Adult stem cells: Simply a tool for regenerative medicine or an additional piece in the puzzle of human aging? Cell Cycle 2011, 10, 4173–4176. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Skin Structure. The skin is composed of 3 main layers-Epidermis, Dermis and Subcutaneous layers containing (1) hair shaft; (2) stratum corneum; (3) sweat-pore; (4) hair follicle; (5) arrector pili muscle; (6) sebaceous gland; (7) nerve; (8) eccrine sweat gland; (9) cutaneous vascular plexes; (10) adipose depot. Detailed structure of (A) epidermis is shown with the stratum layers and (B) cellular composition and dermis is shown.
Figure 1. Skin Structure. The skin is composed of 3 main layers-Epidermis, Dermis and Subcutaneous layers containing (1) hair shaft; (2) stratum corneum; (3) sweat-pore; (4) hair follicle; (5) arrector pili muscle; (6) sebaceous gland; (7) nerve; (8) eccrine sweat gland; (9) cutaneous vascular plexes; (10) adipose depot. Detailed structure of (A) epidermis is shown with the stratum layers and (B) cellular composition and dermis is shown.
Ijms 18 00208 g001
Figure 2. A Schematic representation of dermal fibroblast in regulation of skin homeostasis. Fibroblasts have been shown to influence the signaling pathways for the synthesis of extracellular matrix (ECM), collagen and growth factors.
Figure 2. A Schematic representation of dermal fibroblast in regulation of skin homeostasis. Fibroblasts have been shown to influence the signaling pathways for the synthesis of extracellular matrix (ECM), collagen and growth factors.
Ijms 18 00208 g002
Figure 3. Stages of Wound Healing: Wound healing is a 3-step process. (1) Inflammation stage initiated by migration of inflammatory and stem cells to the wound-site; (2) Proliferation and Granularization stage involving secretion of growth factors and migration of fibroblasts, keratinocytes and endothelial cells to initiate ECM production and repair and; (3) Re-epithelialization stage is the final stage of wound healing process marked by the synthesis and organization of new collagen and elastin fibers resulting in healed epidermis.
Figure 3. Stages of Wound Healing: Wound healing is a 3-step process. (1) Inflammation stage initiated by migration of inflammatory and stem cells to the wound-site; (2) Proliferation and Granularization stage involving secretion of growth factors and migration of fibroblasts, keratinocytes and endothelial cells to initiate ECM production and repair and; (3) Re-epithelialization stage is the final stage of wound healing process marked by the synthesis and organization of new collagen and elastin fibers resulting in healed epidermis.
Ijms 18 00208 g003
Table 1. Growth factors and their function in skin homeostasis, regeneration and repair.
Table 1. Growth factors and their function in skin homeostasis, regeneration and repair.
Growth FactorPrimary Target Cells and EffectReferences
HB-EGFKeratinocyte and fibroblast mitogen[75]
FGFs 1, 2, and 4Angiogenic and fibroblast mitogen[76,77]
PDGFChemotactic for macrophages, fibroblasts; macrophage activation; fibroblast mitogen, and matrix production[78]
IGF-1Endothelial cell fibroblast mitogen[79,80]
TGF-β1 and β2Keratinocyte migration; chemotactic for macrophages and fibroblasts[81]
TGF-β3Wounded mouse skin, cutaneous rat wounds; Antiscarring[82,83]
IL-1α and –βEarly activators of growth factor expression in macrophages, keratinocytes and fibroblasts; function in wound healing[84,85]
IL-10Regenerative wound repair, Optimization of fibroblast and endothelial progenitor cells (EPC) function in differentiation, Attenuation of inflammatory response[86,87,88,89]
IL-4Induction of fibroblast migration and differentiation, ECM production, control of MMP, Lower dermis maintenance, Wound healing[90,91,92,93,94]
IL-12Epithelial cells under mouse tongue; Early inflammatory response and vascularization in wound healing, Regulation of Growth factors synthesis[95]
IL-6, IL-8, IL-17aPromote connection between immune cells and keratinocytes, fibroblasts Pro-inflammatory response, All the stages of wound healing,[96,97]
LEP and LEPRKeratinocytes proliferation, fibroblasts migration, Hyaluronic acid and collagen synthesis, Re-epithelialization processes[32,98]
EndoglinHuman skin, lung and liver; Co-receptor for TGF-β, Promotes wound healing[99]
AdipoqRecruitment of fibroblasts and endothelial cells; Dermis regeneration, Wound healing, Suppression of inflammation[32,100]
IGFBP1, IGFBP3Human dermal fibroblasts, human skin Collagen synthesis, Elastogenesis in dermal fibroblasts, Angiogenesis, Proliferation and differentiation[101,102,103]
CSF1, CSF3 and receptor CSFR1Rat dermis Stimulation of TGFβ, Anti-inflammatory, Wound healingPatent US8273712 B2 [104]
PPBP/NAP-2Fibroblast; mitogenic activity, Modulation of immune cells[103,105]
HGFKeratinocytes migration and differentiation; Scarless wound healing[30,106,107]
NGFRHuman hair follicle cells, ECM remodeling, Wound healing, Modulation of immune cells[108]
EGFRabbit skin cells, White pig skin, human fibroblasts, rat skin, Activation of dormant stem cells, Wound healing, Skin rejuvenation, Production of collagen, elastin, ECM remodeling[33,109,110,111]
TNF-αMice epithelial cells, Similar to the IL-1[84]
Activin A and Activin BRat; Cutaneous wound healing, Human adipose progenitors proliferation and differentiation[112,113,114,115]
Table 2. ADSCs in Clinical Trials for Dermatological Applications.
Table 2. ADSCs in Clinical Trials for Dermatological Applications.
ConditionTitleTrial NumberStatusReferences
ScarsA Phase I/II Study of Autologous Fat Transfer for Scar Prevention and Remodeling.NCT01119326REGISTERED[219,220]
A Phase II/III Clinical Study of Autologous Cultured Adipocytes (ANTG-adip) for the Treatment of Depressed Scar to Evaluate Safety and Efficacy.NCT00992147COMPLETED
A Phase I Open-labeled, Single-arm, Single-centred Study to Test the Safety of ADSC-SVF-002 in Subjects With Soft Tissue Defects or Abnormal Wound Healing.NCT02590042RECRUITING
Facial AtrophyA Phase II Double-blind, Randomized, Study to Assess the Efficacy of Facial Fat Grafts Supplemented With Autologous, Adipose Derived Stromal Vascular Fraction (SVF).NCT02526576RECRUITING[210,221,222,223]
A Phase One, Open Label, Single Arm Study to Demonstrate the Safety of Antria Cell Preparation Process During Facial Fat Grafting Assisted With Autologous, Adipose Derived Stromal Vascular Fraction (SVF).NCT01828723COMPLETED
Preliminary Investigation of the Effect of Human Adipose Tissue-derived Mesenchymal Stem Cell (MSC) in Progressive Hemifacial Atrophy (Romberg’s Disease).NCT01309061COMPLETED
Potential of Mesenchymal Stem Cell Enriched Adipose Tissue Grafting for Contour Deformities of Face.NCT02494752REGISTERED
Burn and wound healingA Phase 1 Clinical Study to Evaluate the Safety of Allogeneic Adipose-derived Stem Cells in the Subjects With Deep Second-degree Burn Wound.NCT02394873COMPLETED[208,224,225,226]
Evaluation of Tissue Regeneration Potential (in the Skin) of Child’s Adipose Cells During the Development.NCT02779205COMPLETED
Feasibility of Obtaining Adipose Derived Regenerative Cells (ADRCs) From Discarded Thermal Burn Eschar Tissue Using Investigational Celution® System for Autologous Treatment of Thermal Burn Injury (The FAST Trial).NCT02362386COMPLETED
Skin graftPhase 1/2 Study of Autologous Stromal Vascular Fraction in Adipose Tissue Transplantation in Improving Skin Grafting.NCT02546882RECRUITING[227,228,229]
A Phase I, Two Armed, Open, Prospective and Multicentre Study to Evaluate the Safety of Autologous Tissue-engineered Dermal Substitutes and Dermo-epidermal Skin Substitutes for the Treatment of Large Deep Partial and Full Thickness Skin Defects in Children and Adults.NCT02145130RECRUITING
Reconstructive surgeryImmunophenotyping of Fresh Stromal Vascular Fraction From Adipose Derived Stem Cells (ADSC) Enriched Fat Grafts for Refinements of Reconstructed Breasts.NCT01771913COMPLETED[230,231,232,233,234,235,236,237,238,239]
Skin improvement in Breast.NCT01801878REGISTERED
Adipose Stromal Cell Enriched Autologous Fat Grafting for Treating Pain at Amputation Sites: A Single Center Site, Prospective, Randomized, Pilot Outcomes Trial.NCT02076022RECRUITING
Enriched Autologous Fat Grafting for Treating Pain at Amputation Sites.NCT01645722REGISTERED
Structural Fat Grafting for Craniofacial Trauma: Effect of Concentrating Endogenous Stromal Cells in the Fat Graft.NCT01564524REGISTERED
Outcomes After Centrifugation Versus PureGraft for Fatgrafting to the Breast After Breast-conserving Therapy.NCT01979757COMPLETED
A Clinical Evaluation Of Adipose Derived Regenerative Cells In The Treatment Of Patients With Breast Deformities Post Segmental Breast Resection (Lumpectomy) With Or Without Radiation Therapy. A Phase IV Post Market Study.NCT00616135COMPLETED
AlopeciaSubcutaneous Transplantation of Autologous Cell Enriched Adipose Tissue For Follicular Niche Stimulation in Early Stage Alopecia Androgenetica (STYLE): a Randomized, Blinded, Controlled Trial.NCT02503852RECRUITING[137,240]
Adipose-derived Stromal Vascular Fraction (SVF) Injections to Stimulate Hair Regrowth for Androgenetic Alopecia.NCT02626780REGISTERED
Biocellular Regenerative Therapy in Hair Loss: Use of High Density Platelet-Rich Plasma Concentrates and Cell-Enriched Emulsified Adipose-Derived Tissue Stromal Vascular Fraction.NCT02849470RECRUITING
The Effect of Allogeneic Human Adipose Derived Stem Cell Component Extract on Androgenic Alopecia.NCT02594046REGISTERED
Point-of-Care Adipose-derived Cells for Hair Growth.NCT02729415REGISTERED
Adipose Tissue Derived Stem Cell Based Hair Restoration Therapy for Alopecia.NCT02865421REGISTERED
Urticaria or SclerodermaExperimental Autologous Mesenchymal Stem Cell Therapy in Treatment of Chronic Autoimmune Urticaria.NCT02824393REGISTERED[241,242,243]
Scleroderma Treatment With Celution Processed Adipose Derived Regenerative Cells (ADRCs) Registry.NCT02328625WITHDRAWN
Scleroderma Treatment With Celution Processed Adipose Derived Regenerative Cells [134]: A Randomized, Double-Blind, Placebo-Controlled Trial With Incomplete Crossover.NCT02396238REGISTERED
Subcutaneous Injection of Autologous Adipose Tissue-derived SVF Into the Fingers of Patients With Systemic Sclerosis: Controlled Clinical Trial With Efficacy Assessment.NCT02558543RECRUITING
Pressure UlcerTreatment of Hypertensive Leg Ulcer by Adipose Tissue Grafting.NCT01932021REGISTERED[133,244,245,246]
A Pilot Study: Evaluating the Safety/Feasibility of ADSC on Adults With Stage III or IV Pressure Ulcers.NCT02375802RECRUITING
Adipose Derived Regenerative Cellular Therapy of Chronic Wounds.NCT02092870RECRUITING
LipodystrophiesPhase I Study of a Filler Agent Composed of Mesenchymal Stem Cells Obtained From Autologous Adipose Tissue Associated With Hyaluronic Acid.NCT02034786REGISTERED[247]

Share and Cite

MDPI and ACS Style

Gaur, M.; Dobke, M.; Lunyak, V.V. Mesenchymal Stem Cells from Adipose Tissue in Clinical Applications for Dermatological Indications and Skin Aging. Int. J. Mol. Sci. 2017, 18, 208. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18010208

AMA Style

Gaur M, Dobke M, Lunyak VV. Mesenchymal Stem Cells from Adipose Tissue in Clinical Applications for Dermatological Indications and Skin Aging. International Journal of Molecular Sciences. 2017; 18(1):208. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18010208

Chicago/Turabian Style

Gaur, Meenakshi, Marek Dobke, and Victoria V. Lunyak. 2017. "Mesenchymal Stem Cells from Adipose Tissue in Clinical Applications for Dermatological Indications and Skin Aging" International Journal of Molecular Sciences 18, no. 1: 208. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18010208

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