We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Therapeutic potential of adipose tissue derivatives in skin photoaging

    Meiqi Liu

    Department of Plastic & Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong 510515, PR China

    ,
    Feng Lu

    Department of Plastic & Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong 510515, PR China

    &
    Jingwei Feng

    *Author for correspondence:

    E-mail Address: feng.jingwei@qq.com

    Department of Plastic & Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong 510515, PR China

    Published Online:https://doi.org/10.2217/rme-2023-0098

    Abstract

    Photoaging, the primary cause of exogenous skin aging and predominantly caused by ultraviolet radiation, is an essential type of skin aging characterized by chronic skin inflammation. Recent studies have shown that oxidative stress, inflammation, skin barrier homeostasis, collagen denaturation and pigmentation are the main contributors to it. As a composite tissue rich in matrix and vascular components, adipose tissue derivatives have been recently gaining attention as potential therapeutic agents for various human diseases with fat-processing technology upgrades. This review analyzes both ‘minimally treated’ and ‘nonminimally treated’ fat derivatives to give an overview of the preclinical and clinical relevance of adipose tissue derivatives for antiphotoaging application, highlighting their good clinical prospects as well as discussing their safety and potential risks.

    Tweetable abstract

    This review analyzes both ‘minimally treated’ and ‘nonminimally treated’ fat derivatives to provide an overview of the preclinical and clinical relevance of adipose tissue derivatives for photoaging applications.

    Graphical abstract

    Background: classic & novel adipose tissue derivatives

    Since the maturation of fat-grafting technology in the 1990s, fat grafting has been widely used in the field of plastic surgery and has been gaining increasing attention in the field of regenerative medicine, including dermatology. In the recent decade, many new theories and technologies have emerged on methods of fat harvesting and processing, such as stromal vascular fraction (SVF), purified adipose-derived stem cells (ASCs), nanofat, SVF-gel, and so on (Figure 1). Different fat preparations are physically processed to meet corresponding clinical needs depending on patient characteristics and the filling effect desired in the treatment of photoaging (Table 1 & Figure 1).

    Figure 1. Classic and novel fat-harvesting techniques and their effects in antiphotoaging.

    The Coleman procedure is the current standard for harvesting adipose tissue, and it involves liposuction followed by a process of centrifugation separation to retrieve adipocytes and stromal vascular fraction for re-engraftment into the patient. Microfragmented fat techniques, namely the commercialized Lipogems system, are produced by gradient filtration and stainless steel bead homogenization methods. Nanofat is extracted by a small aperture liposuction needle, rinsed and filtered, then placed between two syringes for emulsification to be injected through a fine needle. Adipose matrix complex is obtained by intercepting fat aspirates through a macroporous filter with a stiffness up to about 6 kPa. Stromal vascular fraction-gel is generated by a removal step of water and oil on the basis of enzymation. Adipose collagen fragment is a sustained-release system for adipokines to enhance angiogenesis, antiapoptosis, antioxidant activity and collagen synthesis.

    ACF: Adipose collagen fragment; AMC: Adipose matrix comple; ASC: Adipose stem cell; ECM: Extracellular matrix; SVF: Stromal vascular fraction; SVF-gel: Stromal vascular fraction gel.

    Table 1. Classic and novel adipose tissue derivatives and their effects in preclinical and clinical applications.
    Product/derivative typeDerivative nameProcessing technologyApplication advantagesApplication pitfallsRef.
    Minimally processed adipose tissue derivativesColeman fatPrimary fat-extraction technologyRapid and easy restoration of subcutaneous soft-tissue volume lossLarge adipose tissue particles difficult for precise filling in the face; abundant free lipid droplets causing post-transplantation lipolysis[1–4]
    Microfragmented fatMechanically processed – lipogems systemGood maintenance of AT histology; contains more perivascular cells and better secretory capacityFat particles are still large, with a significant amount of lipid droplets interpreted[5–9]
    Nano fatFat emulsification techniqueCan be injected by a fine syringe needleAbundant free lipid droplets shed from necrotic adipocytes[10–12]
    SVFEffective components extracted from adipose tissue after effective removal of oil droplets from adipose tissue by physical methodsHigh-density autologous adipose-derived stem cells; contains a variety of cells with regeneration functionsGene contamination[13–16]
    SVF-gelOn the basis of emulsification, the step of removing water and oil are added to concentrate the fat matrix components into a fine colloidal injectionPrecise injection; high-density autologous adipose-derived stem cellsSoft texture, unable to fill deep wrinkles and large areas of soft tissue[12,17]
    Adipose tissue matrix productsDATPreparation processes and chemical agents introduced for tissue engineeringEngineered adipose tissue constructionLack of clinical application[18–21]
    ACFCell-free extracellular matrix-enriched glueSustained release systems for adipokines; trophic effect[22,23]
    AMCObtained by intercepting through a large pore strainer after fat aspirate filtrationHigh stiffness for long-term in vivo retention[24,25]
    Adipose tissue secretory productsATESheared fresh adipose tissue immersed in serum-free DMEM/F12 for 15 minRich in proangiogenesis and proadipogenesis factors; good effects in promoting angiogenesis and adipogenesis[26]
    ALEAcquisition of secretory products during fat emulsification[27]
    CeffeNanofat centrifugal layered products[28–30]
    LF-EVsExtracellular vesicle-enriched after LAF ultracentrifugation[31]
    ATEVsEnrichment of secretory vesicles after fat homogenization[32,33]
    Adipose stem cells and stem cell-derived secretory productsASCStems cells derived from adipose tissuePromotes denaturated collagen degradation and collagen neoformation; stimulates proinflammatory and antioxidant-related signal pathways; mitophagy promotion effectsGene contamination, unsafe and uncontrollable application in clinics[34–39]
    ASC-Exo
    ASC-EVs
    ASC-CM
    Rich in a variety of miRNAs and proteins; high security and allogeneic applicationNo volume-restoration effects[40–45]

    ACF: Adipose collagen fragment; ALE: Adipose liquid extract; AMC: Adipose matrix complex; ASC: Adipose-derived stem cell; ASC-CM: Adipose-derived stem cell medium; ASC-EV: Adipose-derived stem cell extracellular vesicle; ASC-Exo: Adipose-derived stem cell exosome; AT: Adipose tissue; ATEV: Adipose tissue extracellular vesicle; ATE: Adipose tissue extract; DAT: Decellularized adipose matrix; Ceffe: Cell free fat extract; LAF: Lipoaspirate fluid; LF-EV: Lipoaspirate liquid extracellular vesicle; SVF: Stromal vascular fraction; SVF-gel: Stromal vascular fraction gel.

    Particle fat grafting

    In 1992, American fat transplant expert Coleman proposed a revolutionary fat-extraction technique. In his first literature on structural fat grafting, he mentioned that patients showed improvements in skin texture and elasticity after treatment [1], which was broadly recognized by plastic surgeons in their clinical work. Particle fat transplantation, as a key technology in the case of volumetric defects, has the advantage of quick and easy restoration of soft-tissue volume loss (Table 1). However, Coleman's fat particles were reported to be large and difficult to precisely fill the face, leading to irregular fat accumulation [2]. Moreover, they tend to release abundant free lipid droplets after post-transplantation lipolysis, developing side effects such as facial swelling, fat cysts and even lipogranulomas (Table 1) [3].

    Minimally manipulated adipose tissue derivatives

    With the gradual standardization and management of stem cell applications, the concept of minimal manipulation was proposed by the US FDA in 2005, which involved treating adipose tissue through pure physical methods such as injection, centrifugation and homogenization [4]. To avoid severe ischaemia and fat necrosis occurring with excessive volume injection on the treatment plane, requirements for the fineness of adipose tissue particles are rising. Millifat applying 2.4 mm or fewer holes and microfat applying 1.2 mm or fewer holes have been devised with more delicate particles compared with traditional fat grafts, which use 2–3-mm holes [10,46,47].

    Mechanically fragmented fat

    To obtain more exquisite fat particles with better regeneration ability, simple physical fat homogenization technology without enzyme digestion has rapidly been developed (Figure 1). ‘One-stop’ ready-to-use systems such as condensation, Lipogems and squeezed fat have been put into use in the operating room (Figure 1) [48]. Among them, supported by more clinical and experimental literature are the microfragmented fat technique, reported by the Italian scholar Bianchi in 2013 [49]. Microfragmented fat with a positive effect on the maintenance of AT histology has not only been used in the field of orthopedic and aesthetic surgery, but has also been reported in the field of fecal incontinence, degenerative osteoarthritis [5] and chemotherapeutic sustained release (Table 1) [6]. Unlike SVF cells obtained by enzymatic digestion, microfragmented fat contains more perivascular cells and better secretory capacity in tissue repair and regeneration (Table 1) [7], as it contains more natural niches that can be recognized by host immune-mediated cells in the first stage of fat grafting [8].

    Fat enzymation technique

    The representative technique for fat enzymation is the emulsification nanofat technique, published by Tonnard et al. in 2013. The adipose tissue is rinsed, filtered and then placed between two syringes for emulsification [10]. ASCs have been observed in nanofat [10]. However, nanofat also contains a large number of lipids shed from broken necrotic adipocytes, implying a prolonged local inflammatory response (Table 1) [9]. Based on the technique, in 2016 our team reported on the adipose extracellular matrix (ECM), namely SVF-gel, showing good skin-regeneration ability [11]. On the basis of enzymation, a removal step of water and oil was added to concentrate the fat matrix components into a delicate colloidal injection (Figure 1) [11]. Currently, adipose tissue derivatives that have been reported to contain abundant ASCs or SVF are: squeezed fat (1.1  ×  106ASC/ml) [12], SVF-gel (1.8 × 105ASC/ml) [11] and nanofat (1.2  ×  106 SVF/ml) (Figure 1).

    Adipose-derived secretary products

    In addition to the adipose tissue-derived products described above, lipoaspirates can also be prepared as cell-free adipose secretory products that are highly effective in stimulating angiogenesis and adipogenesis (Table 1). The aims of extracellular elements of adipose tissue are to promote an autogenous regenerative microenvironment at the recipient site. Adipose tissue extract (ATE), an immersion product of minced fresh adipose tissue, has shown dose-dependent proangiogenic and proadipogenic potential [50]. Adipose liquid extract, obtained during fat emulsification, can promote the healing of chronic skin ulcers [26]. Cell-free fat extract is a liquid component purified by centrifugation from nanofat containing proinflammatory and proapoptotic growth factors [27,28]. Its subcutaneous injection has been shown to increase capillary density, which subsequently promotes skin regeneration [29]. Lipoaspirate liquid extracellular vesicles are an enrichment of extracellular vesicles obtained after ultracentrifugation of lipoaspirate fluid, and they also play a proangiogenic and proadipogenic role in studies of adjuvant fat grafting [30]. Adipose tissue extracellular vesicles (ATEVs) are then enriched in secretory vesicles after fat homogenization, and through the combination of hydrogel material, ATEVs can promote de novo synthesis of adipose tissue [31,32].

    In addition to stimulating the expression of antioxidant enzymes, adipose tissue secretomes also exhibit good inherent superoxide dismutase and catalase activity [33]. The main difference between this type of product and purified stem cells in terms of anti-inflammatory function is that they not only contain growth factor-like cytokines, but also contain an immense number of inflammatory factor signals that initiate repair mechanisms (Table 1). High-throughput analysis of adipose tissue extract revealed that they contained classical proinflammatory factors such as IL-2, IL-6 and IL-8, neutrophil-activating protein, TIMP-2 and giant cell-related inflammatory protein [50].

    ASC secretome

    Abundant in adipose tissue, mesenchymal stem cells play a key role in maintaining skin homeostasis through paracrine effects. Under solar radiation, premature ASC aging can gradually lead to the loss of their ability to maintain homeostasis and the biological function of skin tissue. Therefore, the ‘regeneration’ of ASCs and other skin-resident cells are highly attractive strategies to combat photoaging and premature senility. ASC-secretome encourages fibroblast, epithelial and endothelial cell proliferation, migration and secretory activity [51,52]. This may considerably benefit skin damage, requiring minimal or no volumetric correction remodeling.

    SVF and ASCs have great application value in indications such as scars, refractory wounds, burns, auxiliary fat grafting, anal fistula, and so on. SVF, as an effective component extracted from adipose tissue after efficient removal of oil droplets, is rich in a variety of cells with repair functions as well as a mixture of bioactive factors, and has shown promising applications in the treatment of diabetic foot, cartilage regeneration in arthritis and gastrointestinal syndromes [13,14,53]. ASC medium, ASC exosomes and ASC extracellular vesicles (ASC-EVs) have been reported to protect against ultraviolet B (UVB)-induced skin photoaging [15]. Cell secretary products have gained attention due to their better safety profile and commercialization potential of allogeneic applications (Table 1).

    Decellularized adipose matrix products

    Decellularized adipose matrix (DAT) has been applied to the construction of engineered fat for combatting clinical dilemmas such as traumatic injury or tumor resection [54,55]. The application of stem cell-mediated therapies in regenerative medicine, along with its biomaterials and growth factors, is expanding beyond the dermatological sector at an alarming pace (Table 1). Since adipose tissue-derived substances are relatively soft, they often perform poor supportive shaping effects for the loss of soft tissue in the middle of the face, wrinkles, dry lines, and so on. Based on this, DAT-hydrogel has been introduced into clinical application, which can be injected subcutaneously and has plasticity for irregular soft tissue defects, and is considered to be one of the most suitable materials for soft tissue filling [22,56].

    Concerning insufficient early angiogenesis after DAT transplantation, DAT hydrogel loaded with bFGF and extracellular vesicles were developed [57]. bFGF-DAT can significantly induce capillary formation within neoadipose tissue and has a mature morphology quite similar to that of endogenous adipose tissue [18,19,57]. DAT enriched with EVs also shows significant proangiogenic and proadipogenesis effects in the early post-transplantation period (Table 1 & Figure 2) [20]. In the administration form of hydrogel dressing, γ-PGA hydrogels loading cell-free fat extracts have promoted diabetic ulcers in a mouse model [21]. This may become a potential avenue for adipose-derived secretary products to participate in tissue engineering to construct anti-photoaging strategies.

    Figure 2. Mechanism of adipose tissue derivatives delaying skin photoaging.

    ECM: Extracellular matrix; IL-1RA: Interleukin 1 receptor antagonist; MMP: Matrix metalloproteinase; ROS: Reactive oxygen species; UV: Ultraviolet.

    Concurrent with the continued approval of commercialized collagen products, autologous adipose matrix products that require only minimal processing have joined the ranks of dermal fillers (Table 1). Adipose collagen fragment (ACF), as a sustained-release system for adipokines, exerts strong therapeutic effects to enhance angiogenesis, antiapoptosis, antioxidant activity and collagen synthesis in photoaging models [56]. ACF has also shown long-term filling effects for deep wrinkles in clinical studies [58]. Adipose matrix complex is a fiber-rich adipose tissue derivative obtained by intercepting fat aspirates through a macroporous filter [23]. With a stiffness up to about 6 kPa (adipose tissue 2–4 kPa [24]) and rich in fibrous tissue of the fascia layer, it has the clinical practice potential for better supportive filling effects for sites such as the mentum and the dorsum of nose [23].

    Therapeutic potential & strategies of adipose tissue derivatives in antiphotoaging

    UV radiation can deeply penetrate the skin and induce profound alterations of the dermal connective tissue. With further purification of bioactive components from the adipose tissue fraction, the effects of adipose tissue derivatives on resistance to photoaging can have remarkable biomedical and clinical relevance. Adipose tissue derivatives have shown different degrees of therapeutic efficacy in multiple aspects such as clearance of denatured collagen fibers, regeneration of collagen fibers, inflammation regulation and inhibition of reactive oxygen species (ROS) (Figure 2).

    Antiphotoaging effects of classical minimally processed adipose tissue derivatives

    For minimally manipulated adipose tissue derivatives, with the integrity of the tissue structure, stem cells embedded in their natural niche may perform better antiphotoaging effects in the short and long term. Crucial to the success of fat transplantation is the percentage of fat cells that survive. A comparative study showed that compared with plain nanofat, SVF and ASC-enriched nanofat demonstrated superior improvements in reducing wrinkles and increasing dermal thickness, collagen content and neocollagen morphology (Table 1 & Figure 1) [25]. Another comparative study revealed that compared with nanofat and microfat in vitro culture, the SVF-gel group had higher long-term cell density and activity, while in the clinical trial of antiwrinkle injections in the neck and face, the microfat group restored better instant subcutaneous volume and better skin texture owing to a more integrated extracellular structure (Table 1 & Figure 1) [16]. While SVF-gel has a higher density and survival rate of ASCs, it is more conducive to the recovery of subcutaneous adipose tissue content, which may affect the survival of adipocytes and play a lasting secretory role at a later stage [17,59]. The therapeutic effect of SVF-gel on photoaging and wrinkles was considered mainly through promoting TGF-β1 expression and activation of fibroblasts to produce type I collagen prepreptide, thereby increasing the density of collagen in the dermis [60], which corresponds to the collagen-egeneration effects of adipose-derived stem cells described below (Figure 2).

    Antiphotoaging mechanism of ASCs & their derivatives

    Collagen protection & antioxidation

    Long-term exposure to sunlight will produce harmful intracellular stress and accumulate ROS and proinflammatory cytokines, accelerating skin aging and resulting in premature skin senescence. UV radiation causes direct damage to the structure of epidermal lipids, fibroblast DNA, mitochondria, telomeres and other structures, further forming peroxidized lipids, pyrimidine dimers and photoproducts, and leading to a decrease in the viability or even apoptosis of skin-resident cells [61]. Simultaneously, a large amount of ROS is generated in the skin irradiated by UVB and UVA. UV-induced ROS accumulation stimulates proinflammatory, antioxidant-related signal pathways in cells, which subsequently induces cell apoptosis and reduces the synthesis and increases the degradation of collagen and elastin, and finally forms wrinkles, pigmentation, coarser skin and other premature manifestations (Figure 2) [62–64].

    The antiphotoaging effect of ASCs has been gradually recognized in studies involving humans and animal photoaging models. In a small-scale, self-controlled clinical study it was found that after autologous ASC therapy were applied, the number of anti-inflammatory M2 phenotype macrophages in the skin was significantly increased compared with the control group. Meanwhile, ASCs were observed to stimulate cathepsin K and matrix metalloproteinase (MMP)-2 pathways to better clear denaturated fibers, promote the deposition of newly produced collagen and deepen the papillary structure of the basement membrane [65]. ASCs overexpressing TGF-β1-related genes ameliorated UVB-induced photoaging by increasing the collagen content and decreasing protein levels of MMP-1, MMP-3 and p-P38 [34]. The TGF-β pathway could be relevant to upstream signaling (Table 1 & Figure 2).

    ASCs improve collagen degradation and neoformation [35] thanks to their abundant exocrine factors (Figure 2). ASC medium, on the one hand, upregulates antioxidant response elements and inhibits ROS; on the other hand, it can increase the expression of inhibitors that promote TGF-β1, MMP-1 and inhibit the expression of type I procollagen synthesis inhibitors such as IL-6 [36]. ASC-EVs enriched with a variety of miRNAs and proteins contribute to fibrocyte proliferation, collagen synthesis and regulation of cellular senescence [40]. Unlike ASCs, which are proinflammatory and highly efficient at participating in denatured tissue removal, ASC-EVs have antioxidant and anti-inflammatory functions (i.e., preventing macrophages from differentiating into the proinflammatory M1 phenotype) and are involved in ECM synthesis through upregulating TIMP-1 and increasing fibroblast activity and TGF-β secretion to maintain collagen fiber content (Table 1) [40–42]. The ASC-secreted derivatives mentioned above, compared with ASCs, have better allogeneic therapeutic value and higher potential for commercialization and clinical translation [43,44].

    Mitophagy promotion effects of ASCs

    When the skin is under harsh conditions, it is autophagy that effectively maintains intracellular homeostasis and is one of the important survival mechanisms for cells to resist internal and external stresses. Mitochondrial membrane damage and DNA deletions are common in photoaged skin [45]. This damage can lead to abnormal mitochondrial metabolism and aggravate skin oxidative stress [45,66]. Cells under photosensitive oxidative attack face intense redox stress and rely on autophagy and mitophagy to maintain the survival or replenishment of cells within the skin [67,68]. Mitophagy removes oxidized and depolarized mitochondria and prevents the release of proapoptotic proteins, the production of ROS, and the futile hydrolysis of ATP [69,70]. The self-saving process of senescence and autophagy of skin-resident cells may be linked through the MAPK pathway [71]. Studies have shown that autophagic dysfunction in cells after parallel UV damage to the mitochondrial and lysosomal membrane can greatly increase the efficiency of cell death by photosensitizers [72].

    In addition to the classical antiphotoaging mechanisms described above, recent studies have revealed that ASCs could maintain an orderly renewal of skin-resident cells by promoting solar-induced autophagic apoptosis. ASCs can delay premature skin senescence by promoting mitophagy. In a mouse model of premature aging with mitochondrial DNA polymerase PolG gene knockout, ASCs have been shown to facilitate mitochondrial phagocytosis and then delay aging [73]. ASCs may break the cascade of oxidative stress, improve the quality of mitochondria in skin-resident cells, achieve REDOX balance, and increase macromolecular synthesis and cell proliferation in the long term.

    Immunomodulation of ASCs against inflammaging & immunosenescence

    Inflammaging is the key term in the exponentially growing field of aging in the last decade. The presence of chronic inflammation causes an imbalance of epidermal and dermal homeostasis, leading to premature aging caused by cellular dysfunction. Gene-expression profiling studies involving multiple human species have linked immunity and inflammation to photoaging [74,75]. Senescence-associated secretory phenotype marker proteins IL-8 and IL-1RA/IL-1 are consistently elevated at UV exposed areas (Figure 2) [76]. UV-induced aged fibroblasts produce senescence-associated secretory phenotype factors that not only stimulate chemokines to release from surrounding nonsenescent fibroblasts, but also interfere with TLR2, TLR6 and CD36 to reduce the phagocytic capacity of macrophages, thereby promoting immune evasion of photoaged damaged cells and resulting in prolonged low-grade chronic inflammation within the skin [77]. There is an infiltration of more monocytes in chronically UV exposed skin [78], and they can directly differentiate into macrophages, inducing ROS accumulation [79], induce MMP transcription in dermal fibroblasts and degrade the ECM [80,81]. Skin inflammaging also results in functional decline of innate and acquired immunity in the dermal layer during aging [82]. It has been suggested that both photoaging and endogenous aging trigger inflammatory aging resulting from remodeling of the immune system [83]. Photoaging as an inflammatory stimulus greatly increases the infiltration of immunosuppressive cells, such as regulatory T cells, myeloid derived suppressor cells and regulatory dendritic cells, which coincides with the manifestation of immunosenescence [84–86].

    ASCs as well-sourced mesenchymal stem cells are often considered an important tool for treating autoimmune diseases or as antiaging therapy. The widely recognized powerful immunomodulatory and exocrine effects of ASCs also have definite inhibitory effects on key marker proteins of inflammaging [87]. Studies have found that ASCs can improve the phagocytotic function and polarization ability of aging macrophages (Table 1) [37,38]. In terms of acquired immunity, ASC transplantation can improve regulatory T-cell proliferation and Th1 to Th2 cell transformation of T-cell senescence in acquired immunity [39,88], thereby improving necessary immune senescence.

    Reconstruction potential of dermal white adipose tissue against photoaging

    Adipose stem cells and preadipocytes resident in white adipose tissue are considered to be the main stem cell population contributing to skin tissue regeneration [89], and over the past decades dermal white adipose tissue (dWAT) has been viewed as an antiaging target that was distinguished from the deeper subcutaneous fat. In an adipocyte progenitor cell derivation observation study, only one group of seven progenitors contained a mixed population of both subcutaneous and visceral adipose tissue-derived cells [90]. dWAT is a unique adipose layer within the reticular dermis of the skin, where fat and precursor cells have been found to play unique roles other than antimicrobial, antifibrotic, healing and energy metabolism [91,92]. The thickness of the adipose layer beneath UV-exposed skin decreases with age [93]. Decreased adipogenesis and increased fibrosis were also found in the thinned dermis of photoaged skin [94]. The mechanism may be related to the high concentration of IL-11, IL-1α, IL-6 and TNF-α secreted at the dermal junction after sun exposure, which can greatly inhibit the lipogenic differentiation of precursor cells [95]. In addition, repeated UV radiation, drug and other stimuli can lead to the conversion of precursor cells into myofibroblasts [96]. It has also been suggested that the regulatory mechanism in the differentiation of subcutaneous ASCs originates from epigenetic modification without changing their basic DNA sequence [97], which are less easily affected by ROS, UV radiation and other aging factors than other skin stem cells are, making them an excellent cell target of antiphotoaging. Replacement of dermal fat by fibrous tissue will lead to mechanical abnormalities caused by microscopic skin structure and loss of true skin volume, namely true wrinkles and skin relaxation [98]. Whereas these various types of ingredients enriched, intradermally injectable fat products offer great possibilities for remodeling of dWAT.

    Attempts at antiphotoaging of adipose tissue derivatives

    Cell-free adipose tissue secretory product effects in photoaging

    Stem cells extracted from lipoaspirate fluid have shown osteogenic and adipogenic potential in studies [99,100], and the antiphotoaging function of lipoaspirate liquid extracellular vesicles is perhaps related to these stem cells. ATEVs are then enriched in secretory vesicles after fat homogenization, and through the combination of this and hydrogel material, ATEVs can promote de novo synthesis in adipose tissue [20]. Platelet-rich plasma (PRP), with a similar healing capacity as fat derivatives, is also commonly used clinically as an adjunct to skin therapy as a growth factor cocktail. In a model of PRP combined with ASC transplantation to treat photoaging, PRP caused marked inflammatory infiltration and vascular reactivity [101]. Meanwhile, the dermal thickening caused by PRP is an unnatural deposition of elastic fibers in the reticular dermis layer, and the tight adhesion between subcutaneous fat and appendages is no longer maintained after treatment, thus some researchers have suggested that the dermal thickening effect of PRP is actually a kind of dermal fibrosis manifestation caused by it [102,103].

    DAT product effects in photoaging

    DAT, as an important tissue engineering scaffold tool, is used to piggyback on different types of stem cells to achieve tissue generation. Studies have shown that DAT has low cytotoxicity and immunogenicity, and contains functional proteins involved in ASC metabolism and collagen neoformation, providing a good microenvironment for inducing adipogenesis (Figure 2) [104]. ACF, rich in type I collagen, type IV collagen and Lamin, showed favorable anti-inflammatory ability in a UVB-induced aging mouse model, characterized by inhibiting the expression of SA-β-gal and ROS, and promoting high expression of antioxidant enzymes [105]. In a clinical study, the results of precise neck wrinkles filling with ACF on 21 women showed that 85.7% of patients were satisfied with the treatment outcome, and no infection, injection site nodules or other adverse side effects were observed [58]. Meanwhile, ACF, as a sustained-release system for adipokines, exerts a rejuvenating effect beyond the filling effect by the sustained release of cytokines through local survival [105].

    Adipose tissue derivatives combined with microneedles for photoaging

    Adipokine treatment of skin photoaging has shortcomings such as poor permeability and poor biological stability. Meanwhile, skin photoaging is a gradual process, so multiple subcutaneous or intracutaneous injections become inevitable for photoaging treatment [106]. Microneedles combined with cell-free adipose tissue derivatives have the advantage of small wound surface and subdermal administration through the skin barrier, which is a more acceptable treatment method for patients in the future. Percutaneous microneedles promote neocollagen formation in a manner that produces minimal damage and initiates skin self-repair. Treatment in combination with ASC-EVs and microneedles can accelerate the recovery mechanism with minimal invasive incisions and is more effective in treating coarse wrinkles and erythema caused by photoaging [107]. ACF-loaded long-acting microneedle patch-treated skin could release ACF components over a long period, having a long-term antiphotoaging effect [108]. Therefore, the long-term adipokine-microneedle system mediated by microtrauma could be a potential strategy to prevent skin photoaging in the future.

    Safety & risks for the application of adipose tissue derivatives

    For autologous fat transfer, there isn't yet a definitive gold standard for the procedure. Because the patient's own fat is used, the technique has been considered to be low risk of hypersensitivity reactions, foreign body reactions or autoinflammatory reactions [109,110]. However, like any other facial filler or medical intervention, adipose tissue derivatives containing intracellular and extracellular structures can be subjected to adverse events and complications including embolism, skin necrosis, hematoma, bruising, swelling, dysesthesia and contour imperfection at both the donor and the recipient site [111,112]. In addition, adipose tissue derivatives intended for filling face the risk of poor survival after grafting, resulting in nodules, oil cysts, calcifications, and so on [113,114].

    A meta-analysis involving microfat and nanofat demonstrated that minimally processed adipose tissue derivatives had lower complication rates than unprocessed fat [115]. Nanofat and SVF-gel, as minimally processed products with a delicate texture, have fewer complications of lumpiness after grafting than normal donor site-harvested fat [116,117]. Nanofat has been reported to have a longer erythematous phase in only the recovery phase of grafting [10], which may be the result that the effects of nanofat therapy are based on upregulated inflammatory signals [118].

    Multiple processing technologies of adipose tissue can be regarded as ways of screening fat components. This makes adipose tissue derivatives endowed with different biological and physical properties from simply harvested fat, and the outcome after transplantation is no longer limited to the form of fat lumps. For instance, although nanofat has no outstanding volume-filling effect, even difficult-to-fill deep wrinkles [119], it can be used as a dermal filler to improve pockmarks and skin texture, and produce a clear thickening effect in the dermis [116,120].

    On the other hand, long-term volume filling effects with no serious adverse events during the follow-up period have also been validated in other types of adipose tissue-derived products. ACF can be stored subcutaneously in the form of collagen for a long time and can maintain stable volume after 14 weeks [58]. Adipose matrix complex has been shown to maintain its hardness after 3 months of subcutaneous transplantation in mice, and the survival graft was a type I collagen dominated fibrous tissue mass interspersed with mature live adipose tissue rather than fibrotic mass [23]. It can be said that the model of applying fat derivatives to restore soft tissue is a strategy of supplying a biological scaffold to maintain the original microenvironment of stem progenitor cells, thus making the final fate of the graft predictable.

    However, some products that lack scaffolds have potential risks worth noting. It is generally accepted that SVF-assisted fat grafting enhances tissue resistance to ischemia and hypoxia, which in turn improves graft survival after large-volume engrafting [121,122] without causing more complications [122]. However, these complex fat products rich in dissociative stem progenitor cells may also have stem cell-related risks after transplantation such as heterotopic fibrosis, lymphatic ganglia and other abnormal differentiation outside the transplant site. Patients who underwent SVF-assisted fat grafting were reported to develop postoperative lymphadenopathy of the abdominal wall and inguinal region [123]. Stem cells in nonphysiological environments are difficult to control, which is also the reason why transplanting fat derivatives is required by various laws and regulations to satisfy the ‘minimum treated’ standard [124].

    Secreted products such as EVs and the exosomes of stem cells have the corresponding advantages of simple composition, a well-defined pathway and low tumorigenicity. However, when it comes to secretory derivatives of infected or malignant cells, those safe secresomes can transfer substances that initiate infection or play a role in innating the autoimmune response [125,126]. In a randomized, controlled, split-face trial of 25 people receiving CO2 laser treatment, the side of the face treated with adipose-derived exosome gel showed better erythema reduction [127]. Most of the novel adipose tissue derivatives are in the preclinical exploration phase, and there are currently a lack of clinical cases and follow-up reports of a certain scale. Most importantly, there is still a lack of standardized procedures for harvesting, processing and injection techniques worldwide.

    Conclusion

    With the increasing longevity of global population aging, the focus of medical research has gradually shifted to the prevention and management of aging and the optimization of an overall healthy life span. Therefore, more and more attention has been given to the treatment of skin photoaging in the field of cosmetic minimally invasive surgery. As a widely used, readily available and inexpensive autogenous material in clinical practice, adipose tissue derivatives show great application potential by promoting tissue regeneration through various functions, including regulating the immune response, providing differentiation, migration and enzyme reaction signals, and promoting tissue balance. Cumulative evidence shows that adipose derivatives can inhibit oxidative stress, remove solar degenerative collagen, induce endogenous collagen regeneration, increase dermal thickness and improve skin firmness in antiphotoaging regeneration of skin. Adipose tissue engineering technology combined with adipose tissue derivatives also shows attractive antiaging and application potential. However, at present there is still a lack of high-level evidence-based medical evidence for adipose tissue derivatives in the treatment of photoaging. The optimal therapeutic introduction of adipose tissue derivatives, the safety of tissue engineering products and the mechanism of action of different adipose components remain to be clarified. We believe that with the introduction of the orderly opening and access system for cell therapy and biological drugs, medicinal adipose technology will become an important regenerative tool in the field of antiaging skin repair.

    Future perspective

    The evolution of fat processing has not only offered valuable autologous fillers to treat cell-depleted conditions, but has also received tremendous commercial success in antiaging cosmetic therapeutics. Fat tissue manipulation allows better injection accessibility such as superficial or accurate delivery in filling fine lines or fibrotic skin while introducing stem cells or trophic factors to the site. Future studies should focus on each adipose component's role in fat retention in the multiple aged tissue microenvironment with respect to providing reliable and reproducible outcomes for different patients with various accelerated aging conditions. However, for many cosmetic surgeons, preserving the intact adipose microstructure may still be a key principle in successful fat grafting procedures, especially for tissue defects that require large-volume delivery. Decision-making should balance out the volume-gain ratio in a donor site shortage situation. Simultaneously, with the development of adipose medicine, induced or manipulated adipose tissue may also be valuable as a metabolically and hormonally active drug in reconstituting soft tissue homeostasis.

    Executive summary
    • This article reviews the current state and prospects of adipose tissue derivatives for skin photoaging.

    • The advantages, challenges and applications of current antiphotoaging strategies used in fat-processing technology, as well as the recent advances and future directions in the field, are discussed.

    Classic & novel adipose tissue derivatives

    • The effects of different adipose tissue derivatives on skin regeneration and rejuvenation in vitro and in vivo are evaluated.

    • The advantages and disadvantages of different fat-harvesting and -processing technologies are compared, namely particle fat grafting, mechanically fragmented fat, fat enzymation technique, adipose-derived secretary products and decellularized adipose matrix products in terms of filling effects, secretory capacity, angiogenesis and adipogenesis properties, and antiphotoageing potential.

    Therapeutic potential & strategies for antiphotoaging

    • Adipose tissue derivatives have different degrees of therapeutic efficacy in multiple aspects such as clearance of denatured collagen fibers, regeneration of collagen fibers, inflammation regulation and inhibition of reactive oxygen species.

    • Even though fat harvesting techniques have made significant progress in the laboratory, it is crucial to further evaluate the long-term safety and antiphotoaging effects of these derivatives in randomized controlled clinical trials.

    Future directions & perspectives

    • Future directions and perspectives for the field of adipose tissue derivatives for photoaging are discussed.

    • One the one hand, future studies should focus on each adipose component's role in fat retention in the multiple-aged tissue microenvironment with respect to providing reliable and reproducible outcomes for different patients of various conditions in accelerated aging.

    • One the other hand, preserving an intact adipose microstructure will still be the key principle for a successful fat-grafting procedure, especially for tissue defects that requires large-volume delivery.

    Financial & competing interests disclosure

    Funding was received from the National Nature Science Foundation of China (grant no. 82002058). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    No writing assistance was utilized in the production of this manuscript.

    Open access

    This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Coleman WP 3rd. The history of dermatologic liposuction. Dermatol. Clin. 8(3), 381–383 (1990). •• An essential background source for this article, because it first mentioned that structural fat grafting showed improvements in skin texture and elasticity after treatment.
    • 2. Pallua N, Kim BS. Microfat and lipoconcentrate for the treatment of facial scars. Clin. Plast. Surg. 47(1), 139–145 (2020).
    • 3. Azoury SC, Shakir S, Bucky LP, Percec I. Modern fat grafting techniques to the face and neck. Plast. Reconstr. Surg. 148(4), 620e–633e (2021).
    • 4. Couture LA, Carpenter MK. 2005 donor eligibility requirements: unintended consequences for stem cell development. Stem Cells Transl. Med. 4(10), 1097–1100 (2015).
    • 5. Casari G, Resca E, Giorgini A et al. Microfragmented adipose tissue is associated with improved ex vivo performance linked to HOXB7 and b-FGF expression. Stem Cell Res. Ther. 12(1), 481 (2021).
    • 6. Alessandri G, Cocce V, Pastorino F et al. Microfragmented human fat tissue is a natural scaffold for drug delivery: potential application in cancer chemotherapy. J. Control. Rel. 302, 2–18 (2019).
    • 7. Vezzani B, Shaw I, Lesme H et al. Higher pericyte content and secretory activity of microfragmented human adipose tissue compared to enzymatically derived stromal vascular fraction. Stem Cells Transl. Med. 7(12), 876–886 (2018).
    • 8. Yokota N, Hattori M, Ohtsuru T et al. Comparative clinical outcomes after intra-articular injection with adipose-derived cultured stem cells or noncultured stromal vascular fraction for the treatment of knee osteoarthritis. Am. J. Sports Med. 47(11), 2577–2583 (2019).
    • 9. Molitor M, Travnickova M, Mestak O et al. The influence of low- and high-negative-pressure liposuction and different harvesting sites on the viability and yield of adipocytes and other nucleated cells. Aesthetic Plast. Surg. 45(6), 2952–2970 (2021).
    • 10. Tonnard P, Verpaele A, Peeters G, Hamdi M, Cornelissen M, Declercq H. Nanofat grafting: basic research and clinical applications. Plast. Reconstr. Surg. 132(4), 1017–1026 (2013).
    • 11. Yao Y, Dong Z, Liao Y et al. Adipose extracellular matrix/stromal vascular fraction gel: a novel adipose tissue-derived injectable for stem cell therapy. Plast. Reconstr. Surg. 139(4), 867–879 (2017).
    • 12. Mashiko T, Wu SH, Feng J et al. Mechanical micronization of lipoaspirates: squeeze and emulsification techniques. Plast. Reconstr. Surg. 139(1), 79–90 (2017).
    • 13. Pak J, Lee JH, Pak N et al. Cartilage regeneration in humans with adipose tissue-derived stem cells and adipose stromal vascular fraction cells: updated status. Int. J. Mol. Sci. 19(7), (2018).
    • 14. Bensemmane L, Squiban C, Demarquay C et al. The stromal vascular fraction mitigates radiation-induced gastrointestinal syndrome in mice. Stem Cell Res. Ther. 12(1), 309 (2021).
    • 15. Cai Y, Li J, Jia C, He Y, Deng C. Therapeutic applications of adipose cell-free derivatives: a review. Stem Cell Res. Ther. 11(1), 312 (2020).
    • 16. Yang Z, Jin S, He Y, Zhang X, Han X, Li F. Comparison of microfat, nanofat, and extracellular matrix/stromal vascular fraction gel for skin rejuvenation: basic research and clinical applications. Aesthet. Surg. J. 41(11), NP1557–NP1570 (2021).
    • 17. Zhu Y, Yang F, Yang Y, Cheng B, Zuo J. A retrospective study of SVF-gel compared with nanofat combined with high-density fat in the treatment of early periorbital aging. Ophthalmic Plast. Reconstr. Surg. 38(4), 340–347 (2022).
    • 18. Zhang S, Lu Q, Cao T, Toh WS. Adipose tissue and extracellular matrix development by injectable decellularized adipose matrix loaded with basic fibroblast growth factor. Plast. Reconstr. Surg. 137(4), 1171–1180 (2016).
    • 19. Zhang G, Ci H, Ma C et al. Additively manufactured macroporous chambers facilitate large volume soft tissue regeneration from adipose-derived extracellular matrix. Acta Biomater. 148, 90–105 (2022).
    • 20. Nie JY, Zhu YZ, Wang JW et al. Preparing adipogenic hydrogel with neo-mechanical isolated adipose-derived extracellular vesicles for adipose tissue engineering. Plast. Reconstr. Surg. 148(2), 212e–222e (2021).
    • 21. Yin M, Wang X, Yu Z et al. gamma-PGA hydrogel loaded with cell-free fat extract promotes the healing of diabetic wounds. J. Mater. Chem. B 8(36), 8395–8404 (2020).
    • 22. Jeon EY, Joo KI, Cha HJ. Body temperature-activated protein-based injectable adhesive hydrogel incorporated with decellularized adipose extracellular matrix for tissue-specific regenerative stem cell therapy. Acta Biomater. 114, 244–255 (2020).
    • 23. Li Y, Zhang P, Zhang X et al. Adipose matrix complex: a high-rigidity collagen-rich adipose-derived material for fat grafting. Aging (Albany NY) 13(11), 14910–14923 (2021).
    • 24. Li Y, Wu M, Zhang Z et al. Application of external force regulates the migration and differentiation of adipose-derived stem/progenitor cells by altering tissue stiffness. Tissue Eng. Part A. 25(23-24), 1614–1622 (2019).
    • 25. Zheng H, Qiu L, Su Y, Yi C. Conventional nanofat and SVF/ADSC-concentrated nanofat: a comparative study on improving photoaging of nude mice skin. Aesthet. Surg. J. 39(11), 1241–1250 (2019).
    • 26. He Y, Xia J, Chen H, Wang L, Deng C, Lu F. Human adipose liquid extract induces angiogenesis and adipogenesis: a novel cell-free therapeutic agent. Stem Cell Res. Ther. 10(1), 252 (2019).
    • 27. Cai Y, Yu Z, Yu Q et al. Fat extract improves random pattern skin flap survival in a rat model. Aesthet. Surg. J. 39(12), NP504–NP514 (2019).
    • 28. Yu Z, Cai Y, Deng M et al. Fat extract promotes angiogenesis in a murine model of limb ischemia: a novel cell-free therapeutic strategy. Stem Cell Res. Ther. 9(1), 294 (2018).
    • 29. Xu Y, Deng M, Cai Y et al. Cell-free fat extract increases dermal thickness by enhancing angiogenesis and extracellular matrix production in nude mice. Aesthet. Surg. J. 40(8), 904–913 (2020).
    • 30. Nie F, Ding P, Zhang C, Zhao Z, Bi H. Extracellular vesicles derived from lipoaspirate fluid promote fat graft survival. Adipocyte 10(1), 293–309 (2021).
    • 31. Pomatto M, Gai C, Negro F et al. Differential therapeutic effect of extracellular vesicles derived by bone marrow and adipose mesenchymal stem cells on wound healing of diabetic ulcers and correlation to their cargoes. Int. J. Mol. Sci. 22(8), (2021).
    • 32. Tsiapalis D, O'driscoll L. Mesenchymal stem cell derived extracellular vesicles for tissue engineering and regenerative medicine applications. Cells 9(4), (2020).
    • 33. Ali Khan A, Hansson J, Weber P et al. Comparative secretome analyses of primary murine white and brown adipocytes reveal novel adipokines. Mol. Cell. Proteomics 17(12), 2358–2370 (2018).
    • 34. Fan F, Li Y, Liu Y, Shao L, Yu J, Li Z. Overexpression of klotho in adipose-derived stem cells protects against UVB-induced photoaging in co-cultured human fibroblasts. Mol. Med. Rep. 18(6), 5473–5480 (2018).
    • 35. Gentile P. New strategies in plastic surgery: autologous adipose-derived mesenchymal stem cells contained in fat grafting improves symptomatic scars. Front. Biosci. (Landmark Ed.) 26(8), 255–257 (2021).
    • 36. Li L, Ngo HTT, Hwang E et al. Conditioned medium from human adipose-derived mesenchymal stem cell culture prevents UVB-induced skin aging in human keratinocytes and dermal fibroblasts. Int. J. Mol.Sci. 21(1), (2019). • Direct laboratory evidence of adipose tissue derivative effects in skin photoaging; shows improvements in UVB-induced skin aging when applying conditioned medium from human adipose-derived mesenchymal stem cell culture.
    • 37. Liu F, Qiu H, Xue M et al. MSC-secreted TGF-beta regulates lipopolysaccharide-stimulated macrophage M2-like polarization via the Akt/FoxO1 pathway. Stem Cell Res. Ther. 10(1), 345 (2019).
    • 38. Li Y, Sheng Q, Zhang C et al. STAT6 up-regulation amplifies M2 macrophage anti-inflammatory capacity through mesenchymal stem cells. Int. Immunopharmacol. 91, 107266 (2021).
    • 39. Li W, Chen W, Huang S, Yao G, Tang X, Sun L. Mesenchymal stem cells prevent overwhelming inflammation and reduce infection severity via recruiting CXCR3(+) regulatory T cells. Clin. Transl. Immunol. 9(10), e1181 (2020).
    • 40. Choi JS, Cho WL, Choi YJ et al. Functional recovery in photo-damaged human dermal fibroblasts by human adipose-derived stem cell extracellular vesicles. J. Extracell. Vesicles 8(1), 1565885 (2019).
    • 41. Xu P, Xin Y, Zhang Z et al. Extracellular vesicles from adipose-derived stem cells ameliorate ultraviolet B-induced skin photoaging by attenuating reactive oxygen species production and inflammation. Stem Cell Res. Ther. 11(1), 264 (2020). • Direct laboratory evidence of adipose tissue derivative effects in skin photoaging. Reports on improvements in UVB-Induced skin photoaging by attenuating reactive oxygen species production and inflammation when applying extracellular vesicles from adipose-derived stem cells.
    • 42. Yano F, Takeda T, Kurokawa T et al. Effects of conditioned medium obtained from human adipose-derived stem cells on skin inflammation. Regen. Ther. 20, 72–77 (2022).
    • 43. Wang M, Wang C, Chen M et al. Efficient angiogenesis-based diabetic wound healing/skin reconstruction through bioactive antibacterial adhesive ultraviolet shielding nanodressing with exosome release. ACS Nano. 13(9), 10279–10293 (2019).
    • 44. Shiekh PA, Singh A, Kumar A. Exosome laden oxygen releasing antioxidant and antibacterial cryogel wound dressing OxOBand alleviate diabetic and infectious wound healing. Biomaterials 249, 120020 (2020).
    • 45. Wang M, Charareh P, Lei X, Zhong JL. Autophagy: multiple mechanisms to protect skin from ultraviolet radiation-driven photoaging. Oxid. Med. Cell. Longev. 2019, 8135985 (2019).
    • 46. Dasiou-Plakida D. Fat injections for facial rejuvenation: 17 years experience in 1720 patients. J. Cosmet. Dermatol. 2(3-4), 119–125 (2003).
    • 47. Trivisonno A, Alexander RW, Baldari S et al. Intraoperative strategies for minimal manipulation of autologous adipose tissue for cell- and tissue-based therapies: concise review. Stem Cells Transl. Med. 8(12), 1265–1271 (2019).
    • 48. Yang H, Lee H. Successful use of squeezed-fat grafts to correct a breast affected by Poland syndrome. Aesthetic Plast. Surg. 35(3), 418–425 (2011).
    • 49. Bianchi F, Maioli M, Leonardi E et al. A new nonenzymatic method and device to obtain a fat tissue derivative highly enriched in pericyte-like elements by mild mechanical forces from human lipoaspirates. Cell. Transplant. 22(11), 2063–2077 (2013).
    • 50. Sarkanen JR, Kaila V, Mannerstrom B et al. Human adipose tissue extract induces angiogenesis and adipogenesis in vitro. Tissue Eng. Part A. 18(1-2), 17–25 (2012).
    • 51. Bellei B, Migliano E, Tedesco M et al. Adipose tissue-derived extracellular fraction characterization: biological and clinical considerations in regenerative medicine. Stem Cell Res. Ther. 9(1), 207 (2018).
    • 52. Dubey NK, Mishra VK, Dubey R, Deng YH, Tsai FC, Deng WP. Revisiting the advances in isolation, characterization and secretome of adipose-derived stromal/stem cells. Int. J. Mol. Sci. 19(8), (2018).
    • 53. Zhao X, Guo J, Zhang F et al. Therapeutic application of adipose-derived stromal vascular fraction in diabetic foot. Stem Cell Res. Ther. 11(1), 394 (2020).
    • 54. Zhang Q, Johnson JA, Dunne LW et al. Decellularized skin/adipose tissue flap matrix for engineering vascularized composite soft tissue flaps. Acta Biomater. 35, 166–184 (2016).
    • 55. Banyard DA, Borad V, Amezcua E, Wirth GA, Evans GR, Widgerow AD. Preparation, characterization, and clinical implications of human decellularized adipose tissue extracellular matrix (hDAM): a comprehensive review. Aesthet. Surg. J. 36(3), 349–357 (2016).
    • 56. An JY, Um W, You DG et al. Gold-installed hyaluronic acid hydrogel for ultrasound-triggered thermal elevation and on-demand cargo release. Int. J. Biol. Macromol. 193(Pt A), 553–561 (2021).
    • 57. Lu Q, Li M, Zou Y, Cao T. Delivery of basic fibroblast growth factors from heparinized decellularized adipose tissue stimulates potent de novo adipogenesis. J. Control. Rel. 174, 43–50 (2014).
    • 58. Zhang Y, Zhang X, Jin X et al. Adipose collagen fragment: a novel adipose-derived extracellular matrix concentrate for skin filling. Aesthet. Surg. J. 42(5), NP337–NP350 (2022).
    • 59. Yao Y, Cai J, Zhang P et al. Adipose stromal vascular fraction gel grafting: a new method for tissue volumization and rejuvenation. Dermatol. Surg. 44(10), 1278–1286 (2018).
    • 60. Zhu S, Cai J, Wang J, Feng J, Lu F. Protective effect of fat-tissue-derived products against ultraviolet irradiation-induced photoaging in mouse skin. Plast. Reconstr. Surg. 148(6), 1290–1299 (2021).
    • 61. Camillo L, Gironi LC, Zavattaro E, Esposto E, Savoia P. Nicotinamide attenuates UV-induced stress damage in human primary keratinocytes from cancerization fields. J. Invest. Dermatol. 142(5), 1466–1477; e1461 (2022).
    • 62. Kammeyer A, Luiten RM. Oxidation events and skin aging. Ageing Res. Rev. 21, 16–29 (2015).
    • 63. Lephart ED. Skin aging and oxidative stress: Equol's anti-aging effects via biochemical and molecular mechanisms. Ageing Res. Rev. 31, 36–54 (2016).
    • 64. Wenk J, Brenneisen P, Meewes C et al. UV-induced oxidative stress and photoaging. Curr. Probl. Dermatol. 29, 83–94 (2001).
    • 65. Charles-De-Sa L, Gontijo-De-Amorim NF, Rigotti G et al. Photoaged skin therapy with adipose-derived stem cells. Plast. Reconstr. Surg. 145(6), 1037e–1049e (2020).
    • 66. Klapan K, Simon D, Karaulov A et al. Autophagy and skin diseases. Front. Pharmacol. 13, 844756 (2022).
    • 67. Chen Q, Zhang H, Yang Y et al. Metformin attenuates UVA-induced skin photoaging by suppressing mitophagy and the PI3K/AKT/mTOR pathway. Int. J. Mol.Sci. 23(13), (2022).
    • 68. Zhang C, Gao X, Li M et al. The role of mitochondrial quality surveillance in skin aging: focus on mitochondrial dynamics, biogenesis and mitophagy. Ageing Res. Rev. 87, 101917 (2023).
    • 69. Sreedhar A, Aguilera-Aguirre L, Singh KK. Mitochondria in skin health, aging, and disease. Cell Death Dis. 11(6), 444 (2020).
    • 70. Kim I, Lemasters JJ. Mitophagy selectively degrades individual damaged mitochondria after photoirradiation. Antioxid. Redox Signal. 14(10), 1919–1928 (2011).
    • 71. Gu Y, Han J, Jiang C, Zhang Y. Biomarkers, oxidative stress and autophagy in skin aging. Ageing Res. Rev. 59, 101036 (2020).
    • 72. Martins WK, Santos NF, Rocha CS et al. Parallel damage in mitochondria and lysosomes is an efficient way to photoinduce cell death. Autophagy 15(2), 259–279 (2019).
    • 73. Lv M, Zhang S, Jiang B et al. Adipose-derived stem cells regulate metabolic homeostasis and delay aging by promoting mitophagy. FASEB J. 35(7), e21709 (2021).
    • 74. Robinson MK, Binder RL, Griffiths CE. Genomic-driven insights into changes in aging skin. J. Drugs Dermatol. 8(Suppl. 7), s8–11 (2009).
    • 75. Yan W, Zhang LL, Yan L et al. Transcriptome analysis of skin photoaging in chinese females reveals the involvement of skin homeostasis and metabolic changes. PLoS One 8(4), e61946 (2013).
    • 76. Bradley B, Jarrold CYRT, Ho CY et al. Inflammaging in human photoexposed skin: early onset of senescence and imbalanced epidermal homeostasis across the decades. Experim. Dermatol. doi: 10.1111/exd.14654 (2022).
    • 77. Narzt MS, Pils V, Kremslehner C et al. Epilipidomics of senescent dermal fibroblasts identify lysophosphatidylcholines as pleiotropic senescence-associated secretory phenotype (SASP) factors. J. Invest. Dermatol. 141(4S), 993–1006; e1015 (2021).
    • 78. Bosset S, Bonnet-Duquennoy M, Barre P et al. Photoageing shows histological features of chronic skin inflammation without clinical and molecular abnormalities. Br. J. Dermatol. 149(4), 826–835 (2003).
    • 79. Weiskirchen R, Weiskirchen S, Tacke F. Organ and tissue fibrosis: Molecular signals, cellular mechanisms and translational implications. Mol. Aspects Med. 65, 2–15 (2019).
    • 80. Taddese S, Weiss AS, Neubert RH, Schmelzer CE. Mapping of macrophage elastase cleavage sites in insoluble human skin elastin. Matrix Biol. 27(5), 420–428 (2008).
    • 81. Kaur A, Ecker BL, Douglass SM et al. Remodeling of the collagen matrix in aging skin promotes melanoma metastasis and affects immune cell motility. Cancer Discov. 9(1), 64–81 (2019).
    • 82. Pilkington SM, Bulfone-Paus S, Griffiths CEM, Watson REB. Inflammaging and the Skin. J. Invest. Dermatol. 141(4S), 1087–1095 (2021).
    • 83. Salminen A, Kaarniranta K, Kauppinen A. Photoaging: UV radiation-induced inflammation and immunosuppression accelerate the aging process in the skin. Inflamm. Res. 71(7-8), 817–831 (2022).
    • 84. Maeda A, Beissert S, Schwarz T, Schwarz A. Phenotypic and functional characterization of ultraviolet radiation-induced regulatory T cells. J. Immunol. 180(5), 3065–3071 (2008).
    • 85. Benayoun BA, Pollina EA, Singh PP et al. Remodeling of epigenome and transcriptome landscapes with aging in mice reveals widespread induction of inflammatory responses. Genome Res. 29(4), 697–709 (2019).
    • 86. Salminen A. Activation of immunosuppressive network in the aging process. Ageing Res. Rev. 57, 100998 (2020).
    • 87. Govindasamy V, Rajendran A, Lee ZX et al. The potential role of mesenchymal stem cells in modulating antiageing process. Cell Biol. Int. 45(10), 1999–2016 (2021).
    • 88. Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood 105(4), 1815–1822 (2005).
    • 89. Zhang D, He S, Wang Q, Pu S, Zhou Z, Wu Q. Impact of aging on the characterization of brown and white adipose tissue-derived stem cells in mice. Cells Tissues Organs 209(1), 26–36 (2020).
    • 90. Vijay J, Gauthier MF, Biswell RL et al. Single-cell analysis of human adipose tissue identifies depot and disease specific cell types. Nat. Metab. 2(1), 97–109 (2020).
    • 91. Chen SX, Zhang LJ, Gallo RL. Dermal white adipose tissue: a newly recognized layer of skin innate defense. J. Invest. Dermatol. 139(5), 1002–1009 (2019).
    • 92. Martins V, Gonzalez De Los Santos F, Wu Z, Capelozzi V, Phan SH, Liu T. FIZZ1-induced myofibroblast transdifferentiation from adipocytes and its potential role in dermal fibrosis and lipoatrophy. Am. J. Pathol. 185(10), 2768–2776 (2015).
    • 93. Kahn DM, Shaw RB. Overview of current thoughts on facial volume and aging. Facial Plast. Surg. 26(5), 350–355 (2010).
    • 94. Kruglikov IL, Scherer PE. Dermal adipocytes and hair cycling: is spatial heterogeneity a characteristic feature of the dermal adipose tissue depot? Exp. Dermatol. 25(4), 258–262 (2016).
    • 95. Li WH, Pappas A, Zhang L, Ruvolo E, Cavender D. IL-11, IL-1alpha, IL-6, and TNF-alpha are induced by solar radiation in vitro and may be involved in facial subcutaneous fat loss in vivo. J. Dermatol. Sci. 71(1), 58–66 (2013).
    • 96. Liu SY, Wu JJ, Chen ZH et al. Insight into the role of dermal white adipose tissue loss in dermal fibrosis. J. Cell. Physiol. 237(1), 169–177 (2022).
    • 97. Sasahara Y, Kubota Y, Kosaka K et al. Adipose-derived stem cells and ceiling culture-derived preadipocytes cultured from subcutaneous fat tissue differ in their epigenetic characteristics and osteogenic potential. Plast. Reconstr. Surg. 144(3), 644–655 (2019).
    • 98. Kruglikov IL, Scherer PE. Skin aging: are adipocytes the next target? Aging (Albany NY) 8(7), 1457–1469 (2016).
    • 99. Tarallo M, Fino P, Ribuffo D et al. Liposuction aspirate fluid adipose-derived stem cell injection and secondary healing in fingertip injury: a pilot study. Plast. Reconstr. Surg. 142(1), 136–147 (2018).
    • 100. Duscher D, Atashroo D, Maan ZN et al. Ultrasound-assisted liposuction does not compromise the regenerative potential of adipose-derived stem cells. Stem Cells Transl. Med. 5(2), 248–257 (2016).
    • 101. Rigotti G, Charles-De-Sa L, Gontijo-De-Amorim NF et al. 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. 36(3), 261–270 (2016).
    • 102. Charles-De-Sa L, Gontijo-De-Amorim N, Sbarbati A et al. Photoaging skin therapy with PRP and ADSC: a comparative study. Stem Cells Int. 2020, 2032359 (2020). •• The article is the clinical research of adipose tissue derivatives' effects in skin photoaging. It highlighted the improvements in UVB-induced skin photoaging by adipose-derived stem cells.
    • 103. Charles-De-Sa L, Gontijo-De-Amorim NF, Takiya CM et al. Effect of use of platelet-rich plasma (PRP) in skin with intrinsic aging process. Aesthet. Surg. J. 38(3), 321–328 (2018).
    • 104. Pati F, Jang J, Ha DH et al. Printing three-dimensional tissue analogues with decellularized extracellular matrix bioink. Nat. Commun. 5, 3935 (2014).
    • 105. Jin X, Zhang Y, Zhang X et al. An adipose-derived injectable sustained-release collagen scaffold of adipokines prepared through a fast mechanical processing technique for preventing skin photoaging in mice. Front. Cell. Dev. Biol. 9, 722427 (2021).
    • 106. Jiang C, Hu Y, Liang Y, Chang J. Photoaged skin therapy with adipose-derived stem cells. Plast. Reconstr. Surg. 148(1), 144e–145e (2021).
    • 107. Cao Z, Jin S, Wang P et al. Microneedle based adipose derived stem cells-derived extracellular vesicles therapy ameliorates UV-induced photoaging in SKH-1 mice. J. Biomed. Mater. Res. A. 109(10), 1849–1857 (2021).
    • 108. Jin X, Zhang X, Li Y et al. Long-acting microneedle patch loaded with adipose collagen fragment for preventing the skin photoaging in mice. Biomater. Adv. 135, 212744 (2022). • Good preclinical attempt of adipose tissue-derived portable material for antiphotoaging. Reported on improvements in UVB-induced skin photoaging by applying a long-acting microneedle patch loaded with adipose collagen fragment.
    • 109. Toledo LS, Mauad R. Fat injection: a 20-year revision. Clin. Plast. Surg. 33(1), 47–53; vi (2006).
    • 110. De Boulle K, Heydenrych I. Patient factors influencing dermal filler complications: prevention, assessment, and treatment. Clin. Cosmet. Investig. Dermatol. 8, 205–214 (2015).
    • 111. Groen JW, Krastev TK, Hommes J, Wilschut JA, Ritt M, Van Der Hulst R. Autologous fat transfer for facial rejuvenation: a systematic review on technique, efficacy, and satisfaction. Plast. Reconstr. Surg. Glob Open 5(12), e1606 (2017).
    • 112. Kadouch J, Schelke LW, Swift A. Ultrasound to improve the safety and efficacy of lipofilling of the temples. Aesthet. Surg. J. 41(5), 603–612 (2021).
    • 113. Orholt M, Larsen A, Hemmingsen MN et al. Complications after breast augmentation with fat grafting: a systematic review. Plast. Reconstr. Surg. 145(3), 530e–537e (2020).
    • 114. Yoshimura K, Coleman SR. Complications of fat grafting: how they occur and how to find, avoid, and treat them. Clin. Plast. Surg. 42(3), 383–388; ix (2015).
    • 115. Yang F, Ji Z, Peng L et al. Efficacy, safety and complications of autologous fat grafting to the eyelids and periorbital area: a systematic review and meta-analysis. PLOS ONE 16(4), e0248505 (2021).
    • 116. Tenna S, Cogliandro A, Barone M et al. Comparative study using autologous fat grafts plus platelet-rich plasma with or without fractional CO2 laser resurfacing in treatment of acne scars: analysis of outcomes and satisfaction with FACE-Q. Aesthetic Plast. Surg. 41(3), 661–666 (2017).
    • 117. Jiang S, Quan Y, Wang J, Cai J, Lu F. Fat grafting for facial rejuvenation using stromal vascular fraction gel injection. Clin. Plast. Surg. 47(1), 73–79 (2020).
    • 118. Sanchez-Macedo N, Mcluckie M, Grunherz L, Lindenblatt N. Protein profiling of mechanically processed lipoaspirates: discovering wound healing and antifibrotic biomarkers in nanofat. Plast. Reconstr. Surg. 150(2), 341e–354e (2022).
    • 119. Uyulmaz S, Sanchez Macedo N, Rezaeian F, Giovanoli P, Lindenblatt N. Nanofat grafting for scar treatment and skin quality improvement. Aesthet. Surg. J. 38(4), 421–428 (2018).
    • 120. Menkes S, Luca M, Soldati G, Polla L. Subcutaneous injections of nanofat adipose-derived stem cell grafting in facial rejuvenation. Plast. Reconstr. Surg. Glob Open 8(1), e2550 (2020).
    • 121. Yoshimura K. Cell-assisted lipotransfer and therapeutic use of adipose stem cells thereafter. Aesthetic Plast. Surg. 44(4), 1266–1267 (2020).
    • 122. Li M, Chen C. The efficacy of cell-assisted lipotransfer versus conventional lipotransfer in breast augmentation: a systematic review and meta-analysis. Aesthetic Plast. Surg. 45(4), 1478–1486 (2021).
    • 123. Yoshimura K, Aoi N, Suga H et al. Ectopic fibrogenesis induced by transplantation of adipose-derived progenitor cell suspension immediately after lipoinjection. Transplantation 85(12), 1868–1869 (2008).
    • 124. Mcallister TN, Audley D, L'heureux N. Autologous cell therapies: challenges in US FDA regulation. Regen. Med. 7(Suppl. 6), 94–97 (2012).
    • 125. Kim SM, Yang Y, Oh SJ, Hong Y, Seo M, Jang M. Cancer-derived exosomes as a delivery platform of CRISPR/Cas9 confer cancer cell tropism-dependent targeting. J. Control. Rel. 266, 8–16 (2017).
    • 126. Liu S, Wu X, Chandra S et al. Extracellular vesicles: Emerging tools as therapeutic agent carriers. Acta Pharm. Sin. B. 12(10), 3822–3842 (2022).
    • 127. Kwon HH, Yang SH, Lee J et al. Combination treatment with human adipose tissue stem cell-derived exosomes and fractional CO2 laser for acne scars: a 12-week prospective, double-blind, randomized, split-face study. Acta Derm. Venereol. 100(18), adv00310 (2020).