Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Exploring the mechanisms behind autologous lipotransfer for radiation-induced fibrosis: A systematic review

  • Nikhil Pattani ,

    Contributed equally to this work with: Nikhil Pattani, Jaspinder Sanghera

    Roles Data curation, Formal analysis, Investigation, Methodology, Writing – original draft, Writing – review & editing

    Affiliation Epsom and St Helier NHS Trust, London, United Kingdom

  • Jaspinder Sanghera ,

    Contributed equally to this work with: Nikhil Pattani, Jaspinder Sanghera

    Roles Data curation, Formal analysis, Investigation, Methodology, Writing – original draft, Writing – review & editing

    Affiliation Walsall Healthcare NHS Trust, Birmingham, United Kingdom

  • Benjamin J. Langridge ,

    Roles Conceptualization, Project administration, Supervision, Writing – review & editing

    b.langridge1@nhs.net

    Affiliations Department of Plastic Surgery, Royal Free Hospital, London, United Kingdom, Division of Surgery & Interventional Sciences, University College London, London, United Kingdom, Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London, United Kingdom

  • Marvin L. Frommer,

    Roles Supervision, Writing – review & editing

    Affiliations Division of Surgery & Interventional Sciences, University College London, London, United Kingdom, Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London, United Kingdom

  • Jeries Abu-Hanna,

    Roles Supervision

    Affiliations Division of Surgery & Interventional Sciences, University College London, London, United Kingdom, Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London, United Kingdom, Division of Medical Sciences, University of Oxford, Oxford, United Kingdom

  • Peter Butler

    Roles Conceptualization, Supervision

    Affiliations Department of Plastic Surgery, Royal Free Hospital, London, United Kingdom, Division of Surgery & Interventional Sciences, University College London, London, United Kingdom, Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London, United Kingdom

Abstract

Aim

Radiation-induced fibrosis is a recognised consequence of radiotherapy, especially after multiple and prolonged dosing regimens. There is no definitive treatment for late-stage radiation-induced fibrosis, although the use of autologous fat transfer has shown promise. However, the exact mechanisms by which this improves radiation-induced fibrosis remain poorly understood. We aim to explore existing literature on the effects of autologous fat transfer on both in-vitro and in-vivo radiation-induced fibrosis models, and to collate potential mechanisms of action.

Method

PubMed, Cochrane reviews and Scopus electronic databases from inception to May 2023 were searched. Our search strategy combined both free-text terms with Boolean operators, derived from synonyms of adipose tissue and radiation-induced fibrosis.

Results

The search strategy produced 2909 articles. Of these, 90 underwent full-text review for eligibility, yielding 31 for final analysis. Nine conducted in-vitro experiments utilising a co-culture model, whilst 25 conducted in-vivo experiments. Interventions under autologous fat transfer included adipose-derived stem cells, stromal vascular function, whole fat and microfat. Notable findings include downregulation of fibroblast proliferation, collagen deposition, epithelial cell apoptosis, and proinflammatory processes. Autologous fat transfer suppressed hypoxia and pro-inflammatory interferon-γ signalling pathways, and tissue treated with adipose-derived stem cells stained strongly for anti-inflammatory M2 macrophages. Although largely proangiogenic initially, studies show varying effects on vascularisation. There is early evidence that adipose-derived stem cell subgroups may have different functional properties.

Conclusion

Autologous fat transfer functions through pro-angiogenic, anti-fibrotic, immunomodulatory, and extracellular matrix remodelling properties. By characterising these mechanisms, relevant drug targets can be identified and used to further improve clinical outcomes in radiation-induced fibrosis. Further research should focus on adipose-derived stem cell sub-populations and augmentation techniques such as cell-assisted lipotransfer.

Introduction

Radiotherapy serves as a cornerstone of cancer treatment. The procedure involves projecting high energy photons, or ionising radiation, towards a tumour to irreversibly damage the DNA of its malignant cells and prevent further neoplastic progression [1]. However, radiotherapy can indiscriminately damage surrounding healthy tissue leading to unwanted side effects [2]. With the growing number of cancer survivors due to general advances in cancer treatment, clinicians increasingly deal with these dreadful side effects.

Radiation-induced fibrosis (RIF) is a recognised consequence of external beam radiotherapy, especially after multiple, prolonged dosing regimens. This can be defined as the overgrowth, hardening, and/or scarring of the skin, underlying soft tissue and/or organs to which radiation has been applied. Its pathophysiology is characterised by acute injury followed by misguided healing, involving excessive deposition of extracellular matrix (ECM) components such as collagen [2]. This reaction can occur in several areas, including but not limited to the skin, subcutaneous tissue, lungs, breasts and gastrointestinal tract.

Radiation-induced skin fibrosis is commonly seen, since the skin and subcutaneous tissue overlying the target tumours are particularly radiosensitive. This is due to the high proliferative capacity and oxygenation requirements of the basal epidermal cells [3]. Over 90% of patients receiving radiotherapy develop moderate to severe skin reactions, on a spectrum from acute radiodermatitis to chronic RIF [4]. The impact of this condition on tissue form and function can be immense and significantly worsen quality of life. For example, head and neck cancer patients may experience both cosmetic and functional impairment with skin tightening and thickening, trismus, neck pain and cervical dystonia in combination with volumetric defects [5].

Immediately post-radiotherapy, patients may experience acute dermatological signs such as mucositis, rashes, and erythema [1]. Instead, RIF of the skin is a chronic complication post-radiotherapy with its onset often being delayed by six months to several years. Initial warning signs may include pain, ulceration, and induration of the affected site, which may resolve without intervention [6]. As it progresses, features such as hypo-/hyper-pigmentation, skin atrophy, and loss of hair follicles, nails and sebaceous glands may present [7]. In severe late-stage RIF of the skin, the underlying subcutaneous tissue is affected resulting in contractures, necrosis and lymphoedema [8].

The pathogenesis of RIF of the skin is complex involving several different inflammatory and immunological mechanisms across a long timespan. Radiation injury results primarily from the production of reactive oxygen species, which damage surrounding cellular materials [9]. Cellular injury results in the release of chemoattractant molecules causing an acute inflammatory response, with fibroblast migration and ECM deposition [911]. Immune cells such as neutrophils are attracted to the site of injury and release further pro-inflammatory cytokines [12]. Later on, lymphocytes arrive and macrophages differentiate into their sub-types (M1 and M2) [13]. This eventually leads to the generation of pro- fibrotic molecules such as transforming growth factor beta (TGF-β), which results in fibroblast production and their subsequent differentiation into myofibroblasts. Myofibroblasts secrete excessive amounts of collagen and fibronectin, resulting in the late effects of RIF such as thickening, contractures etc. [14].

To date, there is no definitive treatment for late-stage RIF of the skin. For decades, it was considered an inevitable, progressive, and irreversible condition which must be tolerated in order to achieve complete cancer eradication. However, the use of autologous fat transfer (AFT), or autologous lipotransfer, has shown promise as treatment for RIF. Adipose tissue can easily be harvested in most patients [15]. From the first AFT procedure performed by Rigotti et al. in 2007, the field has expanded with multiple variations in graft content and surgical techniques [16]. Later, it was observed that fat grafting not only served as a filler, but also enhanced the quality of the surrounding tissue, seemingly making overlying scars fade. A study by Griffin et al. observed aesthetic and functional improvements in RIF of the skin in 97% of their patients treated with AFT [17]. It was further found to significantly improve psychological health. Histological research demonstrated a reduction, remodelling, and realignment of collagen fibres, increased vascularization, and reduction of contractile proteins, dermal thickness, and scar size [18,19].

To date, the exact mechanisms by which AFTs improve fibrosis remain poorly understood. Fat tissue mainly consists of adipocytes, but a variety of other cells are also present collectively named stromal vascular cells. The stromal vascular fraction (SVF) consists of a mix of cells including immune cells, endothelial cells, smooth muscle cells, pericytes, preadipocytes and adipose derived stem/stromal cells (ADSCs). Although only accounting for a small percentage of the cells present in lipotransfers, ADSCs are believed to be of central importance in the grafts anti-fibrotic effects. This is believed to be due to their inherent regenerative capacity, but especially their paracrine signalling. ADSCs are common to all lipotransfer techniques (including whole adipose grafts, stromal vascular fraction, cell-assisted lipotransfer (CAL) etc.).

In this review, we aim to explore existing literature on the use of AFT in both in-vitro and in-vivo RIF skin models, and to collate potential mechanisms by which this procedure takes effect against the condition.

Methods

A protocol was created according to the Preferred Reporting Items for Systematic Review and Meta-Analyses Protocols (PRISMA-P) statement [20]. The search strategy as follows was designed by BL, JAH, JS, and NP.

Information sources

PubMed, Cochrane reviews and Scopus electronic databases from inception to May 2023 were searched. Our search strategy for PubMed combined both free-text terms and Medical Subject Headings (MeSH) with Boolean operators, as shown in Table 1. The search strategies for the latter two databases consists of free-text terms and Boolean operators only. These are shown in Table 2.

Study selection

Two authors (NP and JS) screened the compiled list of search results. These were managed using Endnote 20 (Thomas Reuters, Toronto, Canada) and Excel (Microsoft, Redmond, Washington, USA). After removing duplicates, the articles were independently screened by two authors (NP and JS) in parallel, first by title and abstract. Any disagreements were reviewed by the third and fourth author (BL and JAH). Those remaining were studied in full, prior to a final shortlisting based on pre-specified inclusion/exclusion criteria listed below. To ensure all relevant studies were identified, the reference lists of all included articles and previous systematic reviews were routinely checked. This process is displayed diagrammatically as a PRISMA flowchart in Fig 1.

Eligibility criteria

Inclusion criteria:

  1. Studies utilising photon/electron radiation
  2. Studies utilising adipose tissue fractions or single cell groups isolated from adipose tissue as their intervention
  3. In vitro/in vivo studies with primary data
  4. Studies published in English language

Exclusion criteria:

  1. Studies solely focused on fibrosis of visceral organs
  2. Clinical trials
  3. Review papers and other non-primary data sources
  4. Studies focusing on differentiated cells or stem cells derived from non-adipose sources e.g., bone marrow- derived mesenchymal stem cells

Selected articles

The search was performed on 13th May 2023. The search strategy (Tables 1 and 2) yielded 2909 studies initially, and then 2273 after duplicates removed. After title and abstract screening, 90 articles underwent full-text review for eligibility. Of these articles, 59 were excluded in-line with our inclusion/exclusion criteria. In total, 31 papers underwent analysis in the final systematic review.

Data collection process

All data from the final studies was extracted onto an electronic proforma using Excel (Microsoft, Redmond, Washington, USA) by two authors (NP and JS). This proforma was pre-designed by BL and trialled on three articles. It was deemed to provide a consistent and structured method of data extraction and appraisal. Any disagreements with extracted data were rectified by BL and JAH.

For studies which provided information on multiple interventions, skin conditions and/or research outcomes, only data relating to the current research objective was extracted. Where appropriate, authors were contacted to provide missing information.

Data items

Data items extracted included study design (in vivo/in vitro), intervention preparation, cell types used, disease model, radiation protocol e.g., dosage regimens (Grays, fractionation), date of last follow-up and recorded outcomes. Where mechanisms behind AFT in RIF of the skin were suggested in studies, this and corresponding evidence were also collated.

Risk of bias assessment

All included studies involved a non-randomised design comparing both effector/intervention and comparator/control groups. Without randomisation these observational results may be inherently biased. The risk of bias for in-vivo studies was assessed using the Systematic Review Centre for Laboratory Animal Experimentation (SYRCLE) assessment tool [21]. The Office of Health Assessment and Translation (OHAT) tool designed by the United States national toxicology program was used for in-vitro studies [22].

Two authors (NP and JS) independently appraised each study, assigned individual scores for each tool domain, and then presented this information in risk of bias tables. These are shown in S1 File. Any discrepancies between authors were resolved by consensus with authors BL and JAH.

Data synthesis

As there is no standardised protocol or unit of analysis for assessing the effects of AFT in RIF, methodologies vary between studies. An overarching meta-analysis and quantitative synthesis was not conducted on account of this between-study heterogeneity. Instead, outcome measures were evaluated using simple descriptive statistics and results collated in a narrative synthesis. The results were split into in vivo and in vitro experiments and separated further by effector cell (fibroblast, immune cells, vascular endothelial cells (VEC) etc.). The effect of AFTs on vascular function, immunomodulation and fibrosis were summarised in the review.

Results

Literature search summary

In total, 31 studies were included for final analysis. Of these, nine conducted in vitro experiments utilising a co-culture model whilst 25 studies conducted in vivo experiments. The results are summarised below in Tables 3 and 4.

ADSC preparation and characterisation

Across all studies, ADSCs were prepared in a similar fashion. Adipose tissue was isolated from the target tissue and then digested with collagenases. After undergoing cycles of filtration and centrifugation, ADSCs were obtained and cultured through different passage numbers. In 18 studies, ADSCs were further characterised, and this was either achieved through flow cytometry, fluorescence-activated cell sorting (FACS) or differentiation assays to show ADSC cell surface markers or prove their differentiation potential.

In-vitro study design

All nine in vitro experiments used a co-culture set-up where two different cell types were cultured together either directly in the same well or indirectly by using well inserts [2331]. Four in vitro studies adopted ADSCs as their treatment. Only Xiao et al. utilised mature adipocytes. Shukla et al., Yao et al., Saijo et al., and Sörgel et al. used ADSC-cultured media (ADSC-CM). Yu et al. utilised SVF. A variety of different cell types were used in the co-culture experiments: human microvascular dermal endothelial cell (HDMEC) (n = 1), normal human dermal fibroblasts (NHDF)(n = 2), human foreskin fibroblasts (HFF)(n = 1), bone marrow derived stem cells (BMSC)(n = 1), human immortalized keratinocyte (HaCaT)(n = 3), normal human oral squamous epithelial cells (NOK)(n = 1), human dermal lymphatic endothelial cells (HDLEC)(n = 1), and human skin fibroblast cells (WS1)(n = 2). The radiation protocol generally involved a single exposure of 2–20 Grays (Gy) (median 6Gy), with some studies using varying radiation doses. The methodology of the different studies included proliferation assays (n = 4), enzyme-linked immunosorbent assays (ELISA) (n = 3), Western Blotting (n = 5), reverse transcriptase reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR) (n = 2), and migration assays (n = 3). Other experiments included: tube formation assay, Luminex assay, Terminal deoxynucleotidyl transferase dUTP Nick-End Labelling (TUNEL) apoptotic cell assay and Tandem Mass Tag (TMT)- based proteomic quantification.

In-vivo study design

In the twenty-five in vivo studies, different species were utilised: mice (n = 14), rats (n = 4), mini pigs (n = 6), rabbit (n = 1), and human tissue (n = 1) [26,27,30,3254]. Fifteen studies utilised a single exposure of 10 to 90Gy (median: 50.6Gy) and seven studies a cumulative exposure of 30Gy fractionated as six 5Gy doses over 12 days. Experimental design usually involved irradiation of animals and a transitionary period for RIF to develop (median 25 days), followed by one/multiple AFT procedures involving ADSC, SVF, whole fat or microfat. Control groups were either given phosphate buffered saline (PBS) or ringer’s lactate solution. Two studies looked at the augmentation of lipotransfer using ADSC sub-types. Furthermore, two studies investigated supplementing platelet-rich plasma (PRP) to improve AFT outcomes.The average observational period across studies was 76 days. Outcomes recorded included clinical evaluation, histological assessment (n = 20), mechanical strength testing (n = 5), fat graft volume retention using micro-CT (n = 2), immunohistochemistry (IHC) (n = 15), immunofluorescence (IF) (n = 4), Picrosirius red collagen staining (n = 3) and Massons trichrome collagen staining (n = 8).

Risk of bias assessment

For in-vitro studies (OHAT assessment tool), a low risk of bias was noted in the domains of experimental conditions, exposure characterisation, incomplete data, and reporting on all measured outcomes. Authors provided sufficient information on plate conditions, media and solvents, and they were similar across control and intervention groups. However, they score lower on domains two and five, only due to limited information on whether assessor blinding had taken place. Automated systems where the cells are transferred to assays without assessor handling would mitigate the need for blinding.

Across the in-vivo studies (SYRCLE assessment tool), there was a low risk of selection, attrition, and reporting biases highlighted. Authors ensured similar baseline animal characteristics, measured primary/secondary outcomes in all intervention and control groups, and included all animals in the final analyses. However, we noted that most studies had insufficient details to assess domains three to six e.g., allocation concealment and assessor blinding. As this issue was common, it was not grounds for exclusion, however, this is an area for future studies to improve on.

In-vitro studies

Fibroblasts.

As previously mentioned, three cell lines were used human skin fibroblasts (WS1), primary proliferating normal human dermal fibroblasts (NHDF), and human foreskin fibroblasts (HFF).

Neither co-culture of mature adipocytes with WS1 cells nor of ADSCs with NHDFs increased proliferation of the fibroblasts [25,28]. However, Shukla et al. observed that ADSC-CM treatment reduced NHDF hypermigration post-radiation by 17.5% in a scratch migration assay when compared to NHDFs without treatment [24]. On the contrary Xiao et al. found that co-culture with mature skin adipocytes increased the migration of WS1 cells [28].

Ejaz et al. demonstrated that co-culture of HFF and ADSCs led to a downregulation in TGF-β1 and Col-1 expression compared to irradiated monoculture (RT-qPCR) [27]. In a Luminex assay, HGF production was increased in irradiated co-culture compared to non-irradiated co-culture. Furthermore, addition of HGF protein in increasing concentrations to irradiated HFFs independently decreased TGF-B1 expression in a dose-dependent manner. Haubner et al. noted an increase in the secretion of matrix metalloproteinase (MMP) 1 and MMP2 but also tissue inhibitor of metalloproteinases (TIMP) 1 and TIMP2 when co-culturing ADSCs and NHDFs, relative to the non-irradiated control. Irradiated NHDFs in monoculture showed a significant reduction of MMP1 and an increase in MMP2 and TIMP2. Through TMT- based proteomic quantification, Yu et al. demonstrated significantly higher secretion of 239 proteins in WS1/ADSC co-culture relative to WS1 monoculture [30]. Of the upregulated proteins, 64% were extracellular proteins including MMP1 and collagen subtypes. Kyoto Encyclopaedia of Genes and Genomes (KEGG)-based pathway enrichment analysis revealed that the ECM receptor interaction and focal adhesion pathways were also significantly upregulated.

Endothelial cells.

Saijo et al. discovered that co-culturing irradiated human dermal lymphatic endothelial cells (HDLECs) with ADSCs led to increased proliferation, compared to HDLEC monoculture [29]. They further noted that adding ADSC-CM resulted in smaller scratches (scratch migration assay) and approximately two-fold longer tubes (tube formation assay) [29]. Haubner et al. demonstrated that when co-culturing HDMECs with ADSCs, intercellular adhesion molecule 1 (ICAM-1), vascular cell adhesion protein 1 (VCAM-1), IL -6, and fibroblast growth factor 2 (FGF2) secretion were significantly reduced compared to HDMEC mono-culture [23].

Epithelial cells.

Using a TUNEL apoptosis assay, Yao et al. demonstrated that the proportion of HaCaT and NOK undergoing apoptosis was lower in the ADSC-CM treated group than in the control group treated with normal medium [26]. Furthermore, the secretion of pro-apoptotic CTSF, BAX, Bid, and caspase 9 was downregulated in cells treated with ADSC culture supernatant (Western blotting). Xiao et al. showed that mature skin adipocytes in co-culture had no effect on keratinocyte proliferation but increased their migration [28]. On the other hand, Sörgel et al. reported that ADSC-CM had no impact on the viability and migration of keratinocytes [31].

BMSCs.

Ejaz et al. noted that there was a significant downregulation in the expression of TGF-β1, collagen (COL)1, connective tissue growth factor (CTGF), tumour necrosis factor (TNF), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), and COL4 genes in ADSC/BMSC co-culture relative to BMSC monoculture [27].

In-vivo studies

ECM remodelling.

Sultan et al. noted that the fat-grafted animal group showed less Smad3 activity accompanied by a 42% lower scar index compared with the saline-grafted group eight weeks post-graft [33]. Similarly, Garza et al. discovered that irradiated mice scalp tissue displayed a significant decrease in collagen amount at eight weeks post-fat graft compared to irradiated tissue pre-fat graft [36]. However, Yu et al. showed that TGF-β1 levels (IHC) were increased and IL-6 levels decreased in SVF-grafted rats relative to PBS-grafted controls at 2 weeks post radiation [30].

Sun et al. showed that there was a decrease in collagen fibres (Masson’s trichrome) as well as lower amounts of TGF-β1 (IHC) in ADSC- treated rabbit muscle at 8 and 26 weeks compared to the PBS-treated control [46]. Yao et al. demonstrated that collagen fibre density (Masson’s trichrome) and hydroxyproline levels was significantly decreased in the ADSC-grafted rat group at 4- and 12-weeks post irradiation compared to the PBS control group [26]. Both Adem et al. and Sowa et al. showed reduced dermal thickness and collagen density in irradiated mice grafted with lipoaspirate, relative to controls [52,53].

On histological analysis, Khademi et al. found no difference in fibrosis and collagen level between ADSC- and PBS-injected rats [43]. Further, Ejaz et al. showed that collagen staining in ADSC-treated mice 30 days post-radiation was similar to non-irradiated control mouse skin [27]. By using GFP+ Luc+ ASDCs they additionally demonstrated that injected ADSCs were still present at the irradiated sites 77 days post-injection. However, Riccobono et al. noted increased staining of alpha smooth muscle actin (α-SMA) (IF) in ADSC-grafted muscle tissue when compared to the PBS-grafted group at day 76 [35].

Epithelial cells.

Forcheron et al. showed that in ADSC-grafted animals, there was an increase in cytokeratin expression (IF) after irradiation [34]. This corresponded with increased proliferation of keratinocytes recorded at day 119, two weeks after the fourth ADSC graft (Ki-67 staining). Q-dot labelling revealed that ADSCs were not present in the epidermis.

Endothelial cells.

Lindegren et al. demonstrated that AFT had the second greatest effect on the hypoxia gene pathway in irradiated breast tissue [39]. The authors noted decreases in CTFG, NDST1, FOSL2 and PPFIA4 mRNA gene expression alongside with increases in FAM162A, GPC3, INHA, GPC4 and FOX03 mRNA expression.

Immunomodulation.

Kim et al. demonstrated lower expression of CD68 (monocyte lineage marker) and CD11b (leukocyte marker) in ADSC-seeded spheroid-grafted mice relative to irradiated controls [51]. Lindegren et al. noted that AFT decreased the macrophage density (CD86 IHC marker) in irradiated human breast tissue [39]. However, in contrast, Khademi et al. discovered on histological examination after 8 weeks that there was no difference in median macrophage number between ADSC- injected and PBS control-injected rats [43].

Riccobono et al. showed that IL-10 (Western Blotting) was present in irradiated muscle tissue in an ADSC-injected group, but not detected in the PBS-injected group at day 76 post irradiation [45]. Through IF, they also demonstrated that ADSC-treated muscle tissue stained strongly for M2 macrophage markers such as CD206, whilst weakly staining for M1 macrophage markers such as CD68 at day 76 post-irradiation [44].

Yao et al. found that upon histological examination, the number of lymphocytes in ADSC-treated rats at 4 and 12 weeks was lower than PBS-grafted controls [26]. Controversially, Forcheron et al. demonstrated earlier lymphocyte infiltration in ADSC-injected minipigs relative to the PBS-injected controls [34]. Using Fluorescent Quantum dot (Q-dot) labelled ADSCs, authors demonstrated amassing ADSCs at the subcutis/dermis junction post-grafting.

Lindegren et al. demonstrated that AFT greatly suppressed the pro-inflammatory interferon-γ gene pathway in irradiated breasts [39].

BMSCs.

Ejaz et al. generated GFP+ Luc+ bone marrow chimeric mice through total body irradiation and subsequent bone marrow transplantation [27]. Four weeks later, one leg was irradiated and injected with ADSCs, whilst the contralateral leg served as a control. GFP+ Luc+ bone marrow-derived stem cells were detected at the injection site 21 days post-irradiation but not detected in the non-irradiated contralateral leg. FACS analysis of irradiated tissue demonstrated an increase in cells expressing CD34 and Sca1 (haematopoietic), CD45 (lymphocytes), and CD31 (VEC).

Vascularisation.

Garza et al. demonstrated increased CD31 staining in irradiated mice scalp tissue at two- and eight-weeks post-fat grafting compared with irradiated tissue prior to fat grafting [36]. Adem et al. showed higher CD31 staining (IHC) in fat-grafted mice compared to irradiated controls [52]. Kim et al. noted higher CD31 staining (IHC) and pro-angiogenic gene expression in ADSC-seeded spheroid-grafted mice compared to controls [51]. Huang et al. also noted increased CD31 staining (IHC) and vascular density in the ASDC-treated rat group compared with the control group three weeks after fat grafting [47]. ADSCs utilised in the experiment were labelled with CM-Dil to track movement two weeks after injections. Authors noted that a proportion of labelled ADSCs co-localized with the vascular markers von Willebrand factor (vWF) and CD31, which were found in dendritic vascular structures.

On the contrary, Forcheron et al. demonstrated that after wound healing was complete in ADSC-grafted mini pigs, there was no evidence of increased vascular proliferation [34]. Khademi et al. showed that there was no significant difference in angiogenesis between rats injected with PBS and ADSCs at four weeks post-grafting [43]. Sultan et al. discovered that there was a significant decrease in CD31 staining (IHC) in fat-grafted mice compared to the saline-grafted mice at eight weeks post-grafting, while no significant difference was observed at four weeks post-grafting [33]. However, both sham and fat-grafted animals demonstrated greater vascular density than non-irradiated controls.

Augmentation of AFT.

Luan et al. enriched their fat grafts with ADSCs, known as cell-assisted lipotransfer (CAL). Results included reduced collagen staining (Masson’s trichrome), increased CD31 staining (IHC), and decreased skin stiffness at eight weeks post-grafting in mice compared to fat alone [38].

As shown by Abbas et al., Vitamin E-treated fat grafts resulted in greater CD31 staining, lower 8-isoprostane (oxidative injury marker), and lower pro-inflammatory cytokine expression compared to fat grafts without Vitamin E treatment [50]. Pentoxifylline treatment also reduced dermal thickness and collagen density.

Bertrand et al. demonstrated that, in mice injected with microfat and SVF, there was greater increase in diameter and density of vessel at 12 weeks post-graft than microfat alone [40]. Furthermore, on average a greater wound area underwent successful healing in the microfat/SVF-injected subgroup compared to the microfat-injected subgroup. In the study by Sowa et al., the SVF-injected subgroup showed reduced periphilin staining relative to the fat-only subgroup [53].

Borrelli et al. demonstrated that grafting with fat enriched with stromal vascular cells (SVC) resulted in less dense collagen staining than mice receiving saline, sham injection, or fat-alone treatment at 12 weeks post-graft (Masson’s trichrome) [41]. There was also increased CD31 staining (IHC) in fat-grafted mice relative to sham or saline injection, However, there was no significant difference between fat-only and fat/SVC subgroups. Furthermore, immunostaining showed decreased CD26+ and protein delta homolog 1 (Dlk+) fibroblast subpopulations in fat grafted mice relative to sham injection. However, there was no difference noted in CD26+ and Dlk staining between fat/SVC and fat-only subgroups.

Evin et al. and Bertrand et al. studied the effects of PRP augmentation [40,49]. The former demonstrated the highest activation of proliferation (Ki-67), anti-apoptosis (Bcl-2), differentiation and signalling (β-catenin) within hair follicles by the ADSCs+ PRP subgroup, followed by ADSCs only. The latter reported the greatest area of wound healing in their microfat/PRP subgroup compared to both microfat/SVF and microfat-injected subgroups.

Chen et al. investigated the effects of platelet-rich fibrin (PRF) augmentation on irradiated minipig parotid glands, compared to ADSC-only, PRF-only and sham control groups [48]. Six months post-injection, defects in the combined group were the smallest and shallowest. This group also showed the highest rate of neoangiogenesis (Factor 8) and anti-apoptosis (TUNEL assay).

Kim et al., Sowa et al., and Adem et al. studied the impact of 3-D biological structures on graft outcomes, with regards to ADSC-seeded spheroids, micronized cellular adipose matrices (MCAM), and decellularized adipose matrices (DAM) respectively [5153]. In irradiated murine salivary glands, spheroid treatment appeared to restore both cellular architecture and functionality (mucin secretion). MCAM-grafted tissue displayed reduced dermal hypertrophy, fat atrophy and dermal fibrosis relative to lipoaspirate and controls. DAM-grafted tissue had increasedCD31 staining and lower collagen staining compared to irradiated control, however, fat grafting was superior.

ADSC subpopulations.

With use of FACS, some studies sought to further characterise ADSC by their sub-populations and test their efficacy in AFTs individually. For example, in irradiated mice Borelli et al. compared fat graft injections (200 μL) enriched with either CD74+, CD74−, unsorted ADSCs (CD34+ only) and a lipoaspirate-only control [32]. The authors discovered that CD74+ enrichment led to significantly decreased collagen (Masson’s trichrome) and Versican staining (IF) as well as skin stiffness at eight weeks compared to other sub-groups. Through FACS, CD74+ enrichment also significantly decreased the proportion of fibrotic papillary and reticular fibroblasts, and significantly increased the proportions of anti-fibrotic zigzag and lipofibroblasts [55]. Finally, fat grafts enriched with CD74+ led to reduced inflammation, increased integrity and less fibrosis compared to all other sub-groups on histological analysis.

Deleon et al. also characterised and tested ADSC sub-populations with enrichment for CD146 in fat graft (200 μl) injections [42]. Grafts enriched with CD146+ were noted to have significantly increased staining for fibrillin, perilipin and CD31. In addition, this sub-group showed the greatest decrease in collagen (Masson’s trichrome), TGF-β1 and elastin staining compared to other sub-groups at eight weeks post-fat grafting.

Riccobono et al. compared autologous ADSC grafts to allogenic ADSC grafts in a mini-pig cutaneous radiation model. Using a clinical score, autologous ADSCs were noted to have improved radiation wound healing at the final study endpoint relative to the PBS control group, however, allogenic ADSCs had not [35].

Discussion

This systematic review has summarised in-vitro and in-vivo findings on the effect of AFTs, but also graft-inherent cell types such as ADSCs on RIF of the skin (Fig 2). In-vitro research has mainly focused on the impact of ADSCs with very limited research on other cell types within the fat graft, such as endothelial cells, smooth muscle cells, preadipocytes and fibroblasts [56]. For in-vivo research, whole fat, micro fat, SVF as well as ADSCs alone were investigated for efficacy. Modifications of the fat grafts such as CAL with isolated ADSCs led to improved outcomes [32,38,40]. An interest in ADSC sub-populations led to findings of CD146+ and CD74+ ADSCs possibly being more effective [41,42].

thumbnail
Fig 2. Diagrammatic summary of AFT mechanisms of action.

AFT treats RIF through various mechanisms of action, including improved vascularisation, lymphangiogenesis, cell regeneration, immunomodulation, fibroblast modulation and ECM remodelling. Certain ADSC subpopulations have been shown to possess unique beneficial characteristics. Augmentation techniques such as cell-assisted lipotransfer and platelet-rich plasma have also shown promise.

https://doi.org/10.1371/journal.pone.0292013.g002

In the following, we will discuss different possible effects of AFTs and its inherent cell types on RIF of the skin. These can be roughly categorized into effects on vascularisation, immunomodulation, and the extracellular matrix.

Vascularisation

Radiotherapy is known to induce a local inflammatory microenvinroment with increased vascular endothelial dysfunction and permeability. A microscopic hallmark of RIF is microvascular obliteration–a consequence of induced local inflammation and excessive collagen deposition. The disruption and loss of micro-vessels in combination with an increased demand for oxygen, due to an influx of inflammatory and mesenchymal cells to the site of injury, leads to the accumulation of reactive oxygen species (ROS). While ROS are necessary due to their antimicrobial activity in normal wound healing, they cause damage to the tissue in the dysregulated fibrotic environment. Furthermore, ROS strongly activate the TGF-β/SMAD axis and lead to additional downstream collagen accumulation [57]. Hypoxia, caused by post-radiation vasculopathy, is considered a major stimulus of the fibrotic response seen in RIF [58]. Hence, it would seem beneficial for AFTs to enhance tissue vascularisation to counteract this phenomenon.

However, it may not be a question of vascular density but rather the quality and functionality of the present vasculature, indicated for example by their patency [59]. Sultan et al. found increased vascular density four weeks after irradiation injury compared to non-irradiated controls, indicated by increased staining for CD31 and vWF both in sham and fat-grafted mice [60]. In addition, levels of pro-angiogenic molecules such as vascular-endothelial growth factor (VEGF) and stromal cell-derived factor-1 (SDF-1) were higher in the fat-grafted group [60]. After eight weeks, however, less CD31 and vWF were measured in the fat-grafted group, while presenting with a 42% lower scar index compared to the sham-grafted group. This possibly indicates that AFT promotes more proficient vascularisation, leading to closer resemblance to normal skin in the pre-irradiation state. Similarly in other scarring and fibrotic skin conditions, AFT normalized abnormal microvasculature architecture [61,62]. In addition, Lindegren et al. studied genetic pathways in irradiated human breast samples, finding that the hypoxia gene pathway was severely aberrant after radiation and AFT restored this back to normal [39].

ADSCs alone showed ambiguous effects on vascularisation in vivo. One study noted them to increase vascular density three weeks post-grafting, while another study claimed there was no difference between PBS- and ADSC-injected rats after four weeks [43,47]. Huang et al. showed that ADSCs co-localized with vWF and CD31 markers at day 14 post-fat graft [47]. Unrelated to RIF of the skin, Nie et al. demonstrated similar findings using an excisional wound healing model [63]. In this study, ADSCs activated wound healing processes and angiogenesis, as well as being present in newly formed capillaries. It remained unclear whether the ADSCs directly differentiated into VEC, increased VEC proliferation, or supported VEC function by fusing with them. Although the potential of ADSCs to differentiate into VEC in vivo has been demonstrated repeatedly, it remains unclear if this mechanism attenuates RIF of the skin by promoting or regulating angiogenesis [63,64].

Another potential way ADSCs may promote angiogenesis is through the secretion of growth factors at the injection site. Previous research has proven the ability of ADSCs to secrete proangiogenic factors including different VEGFs as well as basic fibroblast growth factor (FGF2) and hepatocyte growth factor (HGF), which have also been found to promote angiogenesis [23,63,65,66]. HGF may play an important role in the angiogenic potential of ADSCs. Cai et al. achieved HGF-knockdown in ADSCs and observed a significantly reduced capacity to promote endothelial cell maturation, migration, and angiogenesis in a mouse hindlimb ischemia model [67]. Yet, no in vivo study has examined the expression of HGF and VEGF in skin harvested from fat-grafted animals for RIF. This warrants further investigation. Haubner et al. also found that FGF2 levels were increased in ADSC and HDMEC co-culture experiments, however, the relative increase was lower in the co-culture as compared to ADSCs alone [23]. Whilst this does suggest a role for FGF2, overexpression might be counterproductive with ADSCs potentially regulating ideal levels. Similarly, FGF2 is believed to have positive effects on wound healing by accelerating proliferation and granulation, while overexpression may lead to uncontrolled formation of granulation tissue [68].

Radiotherapy is known to induce a local inflammatory microenvinroment with increased vascular endothelial dysfunction and permeability. Coupled with the secretion of pro-inflammatory chemotactic cyotkines such as IL-6, leukocytes adhere to and transmigrate across the vascular endothelium into surrounding cells with more ease [69]. In particular, monocyte chemoattractant protein-1 (MCP-1) and IL-8 can trigger firm adhesion of monocytes to the vascular endothelium. Haubner et al. also noted that radiation-induced HDMEC dysfunction involved the upregulation of pro-atherogenic adhesion molecules ICAM-1 and VCAM-1 [70]. This upregulation was attenuated by ADSC co-culture, as well as IL-6 expression. ADSCs could help ameliorate VEC dysfunction in this way.

Lymphangiogenesis

Radiotherapy is also an independent risk factor for developing lymphoedema, and ADSCs may also play a significant role in lymphangiogenesis post-irradiation [71]. Saijo et al. demonstrated that, when co-cultured with ADSCs and ADSC-CM, irradiated HDLECs efficiently proliferated, migrated, and formed tubes [29]. Outside of our study sample, Ahmadzadeh et al. showed a similar effect of ADSCs on non-irradiated HDLECs. They demonstrated more effective proliferation, migration and formation of tube-like structures than after stimulation with either VEGF-C, HGF or FGF alone–suggesting the combination of factors in the ADSC-CM to be most effective [72]. Saijo et al. theorised a paracrine mechanism of action through upregulation of FGF2, which is both a pro-lymphangiogenic and pro-angiogenic factor. FGF2-knockdown in these HDLECs significantly decreased its lymphangiogenic capabilities.

Reactive oxygen species

Outside of our study sample, Kim et al. measured the anti-oxidative properties of ADSC-CM and found it be equally potent as 100 μM ascorbic acid [73]. They treated human fibroblasts with tert­-butyl hydroperoxide (mimicking the oxidising effects of radiotherapy) and incubated them with ADSC-CM, resulting in improved survival and increased SOD and glutathione peroxidase (GPx) activity in fibroblasts. Proteomic analysis of the cultured media further showed different anti-oxidative compounds including superoxide dismutase (SOD), insulin-like growth factor (IGF), hepatocyte growth factor (HGF), platelet-derived growth factor (PDGF), keratinocyte growth factor (KGF), and FGF among others.

Immunomodulation

Cytokines, chemokines, and damage-associated molecular patterns (DAMPs) are produced by epithelial and endothelial cells damaged by radiation. This causes an influx of inflammatory cells, predominately macrophages and neutrophils, which are supposed to eliminate dead cells, debris, and invading pathogens [2,74]. Repetitive trauma or radiation and a disproportionate immune response with prolonged, unresolved inflammation ultimately lead to excessive amounts of ECM deposition [2]. Macrophages are usually found near fibroblasts and repeatedly described as master regulators of fibrosis [75,76]. Depending on their phenotype, they have diverse functions ranging from coordinating wound healing by promoting angiogenesis, tissue remodelling, and resolving inflammation (M2 type), to recruiting immune cells, causing inflammation, and tissue destruction (M1 type) [77]. However, while early fibrosis is characterised by high numbers of inflammatory cells and activated, large fibroblasts with less compact collagen fibres, later stages of fibrosis tend to show compact and organized collagen fibres and often less inflammation with less inflammatory immune cells [78]. Hence, possible immunomodulatory effects of AFT may also depend on what state of RIF is present–early vs late.

The SVF as part of a lipotransfer consists of a variety of cell types including macrophages, endothelial cells, ADSCs, and so on. However, lymphocytic immune cells account for the largest group of cells in the SVF [79,80]. This results in a variety of possible mechanisms by which AFTs can restore tissue homeostasis in RIF. The immunomodulatory effects of AFTs and its inherent cell types have been investigated in several studies. In the context of radiation injury, Lindegren et al. explored the expression of a multitude of gene pathways in irradiated human breast samples, finding that the interferon-gamma (IFN-γ) gene pathway was severely dysregulated post-radiation and subsequently downregulated the most by AFT [39]. IFN-γ is a pro-inflammatory cytokine secreted by T helper 1 (Th1) cells and heavily involved in the development of RIF [8183].

ADSCs have demonstrated great immunomodulatory capacity. They have been found to suppress lymphocyte proliferation, suppress natural killer (NK) cell cytotoxicity, inhibit differentiation of monocyte-derived immature dendritic cells, and promote macrophage polarization towards an anti-inflammatory M2 phenotype [26,84]. In RIF, some of these effects have been reproduced. In irradiated minipig muscle, Riccobono et al. displayed that ADSC injection led to a greater proportion of M2 to M1 macrophages relative to irradiated PBS-injected control, as well as higher amounts of IL-10 and TGF-β [44]. Both IL-10 and TFG-β have been linked to M2 polarization, however, TGF-β is also one of the main drivers of tissue fibrosis [85]. Other study settings have proposed IL-6 and colony-stimulating factor 1 (CSF1) as possible mediators of ADSC-induced M2 polarization [86,87]. These M2 macrophages then help drive repair of radiated tissues. If large numbers of M1 macrophages were present, they would likely to shift the skin from a state of repair to continued inflammation, myofibroblast survival, and fibrosis [44,88].

M2 macrophages can be split further into sub-types (M2a, M2b, M2c and M2d) with varying roles [85]. Further research is required into these specific subtypes and how ADSCs can modulate their function, since literature to date has only focused on M2 macrophages as a collective whole. Furthermore, expression of tumour necrosis factor-inducible gene 6 protein (TSG-6) and other anti-inflammatory chemokines should be studied in greater detail. In microglia, Hu et al. demonstrated that ADSC-secreted micro-RNAs upregulated TSG-6 expression, which then inhibited immune-mediated inflammation and encouraged tissue repair [89]. Whilst this immunomodulation shows promise in future treatment of inflammatory central nervous disorders such as multiple sclerosis, it is not known whether this applies to RIF.

Extracellular matrix remodelling

Excessive extracellular matrix (ECM) deposition–especially collagen–is the hallmark feature of fibrosis. Remodelling of the ECM after injury is essential for tissue homeostasis and in the case of RIF is strongly dysregulated with fibroblasts producing vast amounts of ECM. AFTs could possibly remodel the ECM directly through ECM synthesis, degradation, and modification or indirectly by changing the behaviour of tissue resident cells which produce ECM e.g., fibroblasts.

AFTs have been shown to reduce, remodel, and realign collagen fibres, as well as reduce α-SMA, dermal thickness and scar size [18]. For RIF of the skin specifically, this review provides further evidence that collagen density and dermal thickness significantly decrease after fat grafting (20, 23, 27) as well as ADSC injections alone (32). ADSCs have shown to prevent fibroblast to myofibroblast transition, cause myofibroblast apoptosis as well as generally decrease the production of collagen from fibroblasts in research of fibrotic skin conditions and scarring [18,19,90]. Further, ADSCs in co-culture with irradiated fibroblasts led to a decrease in α-SMA, collagen, and TGF-β production [23,27].

Secreted HGF, as well as FGF2, have been suggested as possible ways by which ADSCs can modulate fibroblast activity and remodel the ECM. They are believed to be part of an antifibrotic FGF2-JNK-HGF pathway [91]. FGF2 supposedly causes myofibroblast apoptosis and inhibits the TGF-β1/Smad–pathway with subsequent reduction of α-SMA expression and collagen production. It is also believed to increase MMP-1 and decrease TIMP-1 [9294]. FGF2 administration to hypertrophic scars in animal models reduced epidermal thickness and collagen levels [95]. HGF was present in ADSC monoculture alone and co-culture of ADSCs with irradiated fibroblasts increased HGF levels. When recombinant HGF was added to an irradiated fibroblast monoculture, a dose-dependent reduction in TGF-β levels was also recorded [27]. According to Ejaz et al., ADSC-secreted HGF may also stimulate the influx of endothelial, bone marrow stem cells and other cell types that collectively remodel the extracellular matrix [27]. The authors showed an influx of GFP+ Luc+ labelled BMSC in AFT-treated mice, but none were identified in saline-grafted mice. HGF has been shown to recruit BMSCs via interactions with their c-met receptor [96]. After they are recruited, ADSCs may affect the expression profiles of these BMSCs, as Ejaz et al. noted significant downregulation in the expression of pro-fibrotic proteins (TGF-β, COL1, CTGF) in ADSC/BMSC co-culture.

Irradiation leads to a dysregulation of MMPs and TIMPs–a further driver of RIF [97,98]. In chronic RIF of the skin, it would seem beneficial if AFTs or ADSCs would increase the amount of active MMPs while decreasing TIMPs. This would enable tissue remodelling with a decrease in ECM. Indeed, a liquid chromatography (LC-MS) study of the ADSC secretome revealed the highest enrichment score and protein counts for ECM proteins, cell adhesion proteins as well as MMPs and TIMPs [95]. However, the functions of TIMPs and MMPs go beyond tissue remodelling and their regulation is complex [99]. They can promote inflammation as well as decrease inflammation. Further, MMPs are tissue specific, regulated by immune and stromal cells, and their functions depend on disease and tissue context [100]. For example, some MMPs–such as MMP8 –are profibrotic while others are believed to be antifibrotic [99]. Further, ECM degradation by MMPs also leads to the release of dormant TGF-β. Excessive MMP activity mat even facilitate the development of chronic wounds, by breaking down newly formed ECM of granulation tissue too quickly, before healing can begin [101]. Hence, it is not as simple as upregulating MMPs and downregulating TIMPs to treat fibrosis. TGF-β decreases the activity of MMPs, specifically MMP-2, MMP-9, and MMP-1, within fibroblasts [102,103]. ADSCs counteracted this effect with increased MMP1 and MMP2 levels, but also increased TIMP1 and TIMP2 levels detected in co-cultures with irradiated fibroblasts [25].

The dynamics of ECM remodelling in fibrotic skin conditions are complex and no simple answer exists on how AFTs or ADSCs may reinstate tissue homeostasis. However, we assume RIF is counteracted by reinstating the physiological balance between ECM synthesis and degradation. This is achieved directly by downregulating profibrotic MMPs and secreting anti-fibrotic MMPs, as well as indirectly by inhibiting excessive ECM production from fibroblasts. This is by preventing their activation and transition into myofibroblasts.

Regenerative ability

The variety of cell types such as endothelial cells or immune in AFTs could possibly replace damaged cells in RIF of the skin. Here, emphasis has been placed mainly on ADSCs. As a form of mesenchymal stem cell (MSCs) they are capable of self-renewal, show long-term viability, and multilineage potential. They can differentiate into various cell types of the mesenchymal lineage including adipocytes, fibroblasts, myocytes, keratinocytes, chondrocytes, osteoblasts but also of other lineages such as neural cells, endothelial cells, and hepatocytes [104,105]. An in vivo study by Haubner et al. showed that when ADSCs were cultured in HDMEC medium, ADSC count decreased progressively during the follow-up period. One suggested theory was the ADSCs were actively differentiating into endothelial cells [23]. Hence, ADSCs could possibly directly replace cell types damaged by radiation. It remains unclear if this mechanism is part of the clinical effect observed after AFTs.

Anti-apoptosis

Radiation-induced cell death of epithelial and endothelial cells is a key driver of RIF of the skin. This leads to the production of cytokines, chemokines, and DAMPs, and subsequent inflammation and influx of immune cells; all of which impair tissue function [106]. AFT may decrease cell death and henceforth prevent further disease progression. When added to irradiated fibroblasts, SVF upregulated DMTF1, MFAP5, and P116, all of which regulate key cellular apoptotic pathways [30]. Similarly, ADSCs led to downregulation of pro-apoptotic proteins (CTSF, Bid, BAX) and fewer cells undergoing apoptosis as a result in co-culture with human immortalized keratinocytes (HaCat) and human oral squamous epithelial cells (Nok) (14). Evin et al. showed increased anti-apoptotic Bcl-2 expression by hair follicles was in their ADSC/PRP and ADSC groups, relative to the control [49]. Rehman et al. also reported direct secretion of anti-apoptotic factors by ADSCs [66]. Huang et al. showed that injected ADSCs co-localized with the cell proliferation marker Ki67 in vivo, providing further evidence that ADSCs may promote cell survival and preserve tissue function and integrity [47].

Augmentation of fat grafts

A prominent limitation of fat grafting in RIF is its retention in hostile, irradiated tissue. In studies of autologous fat grafts in non-irradiated sites, retention rates already vary between 40–60% [107]. On the other hand, irradiated tissue is a fibrotic, hypovascular and inflammatory environment. Grafted fat is therefore susceptible to central necrosis and subsequent inflammation [15]. This can lead to cyst formation, further fibrosis and local infection [108]. Augmentation techniques may help overcome these limitations.

CAL is the enrichment of fat grafts with cells from SVF or with culture-expanded ADSCs. This technique has already shown efficacy in human trials for autologous fat grafts and cosmetic breast augmentation [109,110]. CAL resulted in greater fat volume retention, reduced post-operative atrophy and better cosmetic outcomes. Recently, Luan et al. has also demonstrated its efficacy in RIF treatment, with regards to reduced collagen deposition and increased angiogenesis (CD31) [38].

SVF consists of several cell types, including pericytes, endothelial, haematopoietic, stromal and stem cells. ADSCs only make up less than 5% of SVF. As Yu et al. demonstrated, all these cell types can affect the extracellular matrix through various pathways. Upregulated proteins included microfibrillar-associated proteins (MFAPs) and MMP-1 which complement the regenerative effects of ADSCs [30]. Practical advantages of using SVF over ADSCs alone stems from its simpler isolation process, meaning higher yields can be produced. In addition, no ex-vivo amplification is required which thereby reduces the risk of cell contamination and potential neoplasia [111].

PRP is a concentrate of platelets, obtained after centrifugation of whole blood. It is abundant in pro-regenerative growth factors including PDGF, TGFB, VEGF and epidermal growth factor [49]. Augmentation with PRP is an exciting area of research because its fibrin provides a three-dimensional structure which helps to retain both ADSCs and their secreted growth factors. This allows for appropriate cell migration and enhanced paracrine effects of these growth factors [112]. In particular, the work by Evin et al. on the effects of ADSC/PRP on irradiated hair follicles is promising for the treatment of radiation-induced alopecia in head and neck cancer. The potential benefits of PRF goes even further. This is a second-generation platelet concentrate which is easier to prepare, less expensive and carries a lower risk of immunological rejection compared to PRP [113].

Following from above, several bioactive structural supports in injectable form are currently under investigation, in attempts to better simulate ECM properties. In particular, in vitro research with conventional monolayer cell cultures is significantly limited, as it cannot recreate the mechanical and biochemical communication networks which aid ADSC proliferation and differentiation. Decellularized adipose matrices (DAMs) are acellular, albeit natural 3-D structures that have shown promise even if they do not resemble the donor site’s anatomy [114]. In our included study by Adem et al., demonstrated retention of anti-fibrotic cytokines FGF-2 and MIF, and pro-angiogenic cytokines such as PDGF-AA [52,115]. However, DAMs were inferior to fat grafting alone. This highlights the importance of ADSCs in facilitating de-novo adipogenesis and tissue regeneration. DAMs enriched with further growth factors and ADSCs to levels higher than native grafts may be an avenue to explore. For example, micronized cellular adipose matrices (MCAMs) contain a high proportion of ADSCs, minimal adipocytes and some capillaries [116]. Sowa et al. demonstrated MCAMs had similar anti-fibrotic effects to SVF and superior adipogenic capability to fat grafting alone [53]. In addition, the preparation of MCAMs is less intensive, quicker, and not subject to the same regulations as SVF. Finally, Kim et al. proved the efficacy of cultured, ADSC-seeded spheroids in restoring functionality of irradiated murine salivary glands [51]. Prior to transplant, these cellular microenvironments and its biochemical pathways can be sequentially engineered. For example, the authors treated the spheroids initially with a Wnt/β-catenin pathway activator followed by FGF7 at intervals, to stimulate ADSC differentiation into salivary gland stem cells and paracrine factor secretion respectively.

Additional supplementation of these structures with biocompatible materials and compounds may prove beneficial. Research has shown how ADSCs embedded fibrin glue can enhance wound healing through stimulating angiogenesis, fibroblast migration and ECM organisation [117]. Abbas et al. have highlighted the potent anti-inflammatory and anti-oxidative effects of Vitamin E [50]. Although it has no pro-angiogenic properties itself, Vitamin E likely enhanced ADSC-induced angiogenesis through the above effects on the local microenvironment. In addition, its fat solubility is inherently advantageous in lipotransfer.

ADSC subpopulations

Although often considered as a single entity, ADSCs are a heterogeneous mix of subpopulations with different cellular functions and secretomes. Previous research has identified angiogenic (CD248+), adipogenic (BMPR-1A+), osteogenic (CD105-/‘endoglin’) ADSCs [107]. The CD90+/CD73+/CD105+/CD45-/CD31-/ CD146+(−) sub-types are also known to secrete a large variety of ECM proteins [95].

In our study sample, Borrelli et al. identified promising antifibrotic and anti-inflammatory potential of CD74+ ADSCs in RIF [32]. CD74 activates the anti-inflammatory AMPK signalling pathway through the macrophage inhibitory factor (MIF) receptor [118]. The antifibrotic effects were theorised to be a result of increased fibrillin expression. Fibrillin-1 proteins form the structure of connective tissue microfibrils and are known to regulate TGF-β levels in the ECM [119]. Mutations in these proteins, pathognomonic to Marfan’s syndrome, results in elevated TGF-β and increased susceptibility to lung and kidney fibrosis in particular [120,121]. However, its exact role in RIF of the skin is unclear.

Deleon et al. noted increased expression of CD31 (angiogenesis), perilipin (adipogenesis) and fibrillin by CD146+ ADSCs [42]. The authors theorised that increased fibrillin expression may have enhanced elasticity of irradiated tissue overlying their CD146+ ASC-enriched fat grafts. Perilipin 1-deficient adipocytes have been previously shown to promote inflammation by stimulating monocyte migration and IL-6, iNOS, and IL-1β expression by macrophages [122]. Increased perilipin expression may counteract these inflammatory changes. Continued research into CD146+ ADSCs has shown further proangiogenic potential, through upregulation of VEGF and ANGPTI [123]. The latter stabilises VEGF-induced vascular endothelial cell proliferation and prevents blood vessel leakage [124].

Further proteomic studies are warranted to investigate individual ADSC subtypes (categorised by cell surface markers) and how they can be applied to treatment of RIF of the skin. These purified cells may improve lipotransfer outcomes whilst reducing potential side effects.

Future advancements

Autologous fat grafting shows great potential for treatment of RIF, where it has been proven as a safe and effective treatment [17]. However, although many mechanisms of action of AFTs and especially ADSCs in attenuating RIF have been proposed, a comprehensive explanation of the underlying antifibrotic mechanism is still missing. The pathophysiology of RIF of the skin is complex and whether findings from other fibrotic skin conditions such as scleroderma can be adopted, remains to be proven.

Strikingly, most research has focused on the effects of ADSCs on fibrosis, while they only account for less than 5% of cells in the SVF [125]. They show undoubtable potential for regenerative medicine and this review has once again demonstrated their ability to seemingly attenuate hallmark features of fibrosis both in vitro as well as in vivo. However, future research should adopt a more wholesome approach to investigating how other cell types in the transplanted fat grafts could attenuate fibrosis. This includes macrophages, T cells, but also endothelial cells or preadipocytes. It also remains to be investigated whether ADSCs are a more or less effective treatment option than whole fat grafts. Furthermore, rising importance is being placed on identifying subsets of ADSCs, because they have been shown to be a heterogenous mixture of cells with different subpopulations. Identifying more effective subsets could further improve treatment efficacy and give insight into fibrotic and inflammatory pathways.

Large scale ex-vivo expansion of ADSCs and/or other stem cell types suitable for human use is a significant barrier for transition into mainstream clinical practice. In current literature, conventional growth media utilises foetal bovine serum (FBS) [126]. Subsequent FBS-components retained in the stem cells may then induce immune rejection reactions in human recipients [127]. However, human serum (autologous and pooled allogeneic) cannot provide the large outputs required for therapeutic use, as has already been attempted for human mesenchymal stem cells (hMSCs) [128,129]. Instead, research efforts should focus on developing serum-free (‘chemically-defined’) media for ADSCs. For example, Mark et al. demonstrated comparable efficacy of hMSC-specific serum-free growth media [126]. However, they did concede that favourable CD105+ expression for cardioregenerative potential was reduced and its chemical composition requires further alterations.

There are strict restrictions on such advanced medicine and therapeutic products (AMTPs) set by regulatory bodies such as the Medicines and Healthcare products Regulatory Agency (MHRA), Federal Drugs Agency (FDA) and Human Tissue Authority (HTA) [130]. For example, in the USA, collagenase use to digest adipose tissue and derive ADSCs results in ‘HCT/P-351’ classification [111]. This is the drug/biologics pathway and requires FDA approval with a high barrier to entry in the clinical market. In addition, there are no standardised protocols for assessing these products and patient safety may be impacted as a result.

The work by Lindegren et al., using irradiated human breast tissue and conducting a gene expression analysis, is an important step forward in the field and similar studies should be conducted for corroboration [39]. Fibrotic mechanisms may vary across species and so these results are the most relevant for future clinical practice. Nonetheless, human recruitment in these surgical trials is difficult and only a small sample size was possible. The Royal College of Surgeons of England recently published a study citing service pressures, complicated bureaucracy, and lack of infrastructure (e.g. no trial nurses) as the most common barriers [131]. Gene expression analysis of tissue biopsies can be taken further with whole transcriptome analysis with total RNA sequencing, which will help identify single nucleotide polymorphisms (SNPs) in patients [132]. These slight variations in genetic coding can alter function of proteins involved in pro-/anti-fibrotic pathways, which may enhance or inhibit the efficacy of AFT against RIF of the skin. This form of ‘personalised medicine’ will help to improve individual patient outcomes.

Ultimately, research will continue on discovering the underlying mechanisms by which fibrosis is attenuated or even reversed. The identified factors could be used to enrich fat grafts or even be used as therapeutic options by themselves to entirely bypass the strict regulations currently in place for cellular therapies.

Limitations

A significant limitation in this field of research is that most studies use varying methodologies, which restricts our ability to compare study results and conduct a statistical meta-analysis. These differences included in vitro cell types, in vivo animal models, radiation protocols, measured outcomes and their quantification techniques. Although their methods for ADSC-derivation were generally similar, some studies had no mention of how they characterised these stem cells and confirmed their regenerative capacity. A consensus should be reached on key ADSC markers, which will help to standardise future research and increase validity. The same should be done for all factors of study heterogeneity as aforementioned.

Conclusion

In conclusion, AFT shows promising efficacy in RIF of the skin and likely functions through a combination of immunomodulatory, vasculogenic and anti-fibrotic pathways to reach their clinical effects. Further characterization of these mechanisms will enable identification of potential drug targets, and thereby improve outcomes of future AFT therapies. Future research should explore other cell types within the fat graft such as adipocytes, endothelial cells, stromal cells and their associated progenitor cells.

References

  1. 1. Ryan JL. Ionizing radiation: the good, the bad, and the ugly. J Invest Dermatol. 2012;132(3 Pt 2):985–93. pmid:22217743
  2. 2. Wynn TA. Cellular and molecular mechanisms of fibrosis. J Pathol. 2008;214(2):199–210. pmid:18161745
  3. 3. Rachidi W, Harfourche G, Lemaitre G, Amiot F, Vaigot P, Martin MT. Sensing radiosensitivity of human epidermal stem cells. Radiother Oncol. 2007;83(3):267–76. pmid:17540468
  4. 4. Salvo N, Barnes E, van Draanen J, Stacey E, Mitera G, Breen D, et al. Prophylaxis and management of acute radiation-induced skin reactions: a systematic review of the literature. Curr Oncol. 2010;17(4):94–112. pmid:20697521
  5. 5. Ramia P, Bodgi L, Mahmoud D, Mohammad MA, Youssef B, Kopek N, et al. Radiation-Induced Fibrosis in Patients with Head and Neck Cancer: A Review of Pathogenesis and Clinical Outcomes. Clin Med Insights Oncol. 2022;16:11795549211036898. pmid:35125900
  6. 6. Delanian S, Balla-Mekias S, Lefaix JL. Striking regression of chronic radiotherapy damage in a clinical trial of combined pentoxifylline and tocopherol. J Clin Oncol. 1999;17(10):3283–90. pmid:10506631
  7. 7. Bray FN, Simmons BJ, Wolfson AH, Nouri K. Acute and Chronic Cutaneous Reactions to Ionizing Radiation Therapy. Dermatol Ther (Heidelb). 2016;6(2):185–206. pmid:27250839
  8. 8. Harper JL, Franklin LE, Jenrette JM, Aguero EG. Skin toxicity during breast irradiation: pathophysiology and management. South Med J. 2004;97(10):989–93. pmid:15558927
  9. 9. Travis EL. Organizational response of normal tissues to irradiation. Semin Radiat Oncol. 2001;11(3):184–96. pmid:11447575
  10. 10. Denham JW, Hauer-Jensen M. The radiotherapeutic injury—a complex ’wound’. Radiother Oncol. 2002;63(2):129–45. pmid:12063002
  11. 11. Williams JP, Johnston CJ, Finkelstein JN. Treatment for radiation-induced pulmonary late effects: spoiled for choice or looking in the wrong direction? Curr Drug Targets. 2010;11(11):1386–94. pmid:20583979
  12. 12. Lefaix JL, Daburon F. Diagnosis of acute localized irradiation lesions: review of the French experimental experience. Health Phys. 1998;75(4):375–84. pmid:9753360
  13. 13. Gordon S, Martinez FO. Alternative activation of macrophages: mechanism and functions. Immunity. 2010;32(5):593–604. pmid:20510870
  14. 14. Martin M, Lefaix J, Delanian S. TGF-beta1 and radiation fibrosis: a master switch and a specific therapeutic target? Int J Radiat Oncol Biol Phys. 2000;47(2):277–90. pmid:10802350
  15. 15. Shukla L, Yuan Y, Shayan R, Greening DW, Karnezis T. Fat Therapeutics: The Clinical Capacity of Adipose-Derived Stem Cells and Exosomes for Human Disease and Tissue Regeneration. Front Pharmacol. 2020;11:158. pmid:32194404
  16. 16. Rigotti G, Marchi A, Galie M, Baroni G, Benati D, Krampera M, et al. Clinical treatment of radiotherapy tissue damage by lipoaspirate transplant: a healing process mediated by adipose-derived adult stem cells. Plast Reconstr Surg. 2007;119(5):1409–22. pmid:17415234
  17. 17. Griffin MF, Drago J, Almadori A, Kalavrezos N, Butler PE. Evaluation of the efficacy of lipotransfer to manage radiation-induced fibrosis and volume defects in head and neck oncology. Head Neck. 2019;41(10):3647–55. pmid:31389085
  18. 18. Borovikova AA, Ziegler ME, Banyard DA, Wirth GA, Paydar KZ, Evans GRD, et al. Adipose-Derived Tissue in the Treatment of Dermal Fibrosis: Antifibrotic Effects of Adipose-Derived Stem Cells. Ann Plast Surg. 2018;80(3):297–307. pmid:29309331
  19. 19. Spiekman M, van Dongen JA, Willemsen JC, Hoppe DL, van der Lei B, Harmsen MC. The power of fat and its adipose-derived stromal cells: emerging concepts for fibrotic scar treatment. J Tissue Eng Regen Med. 2017;11(11):3220–35. pmid:28156060
  20. 20. Shamseer L, Moher D, Clarke M, Ghersi D, Liberati A, Petticrew M, et al. Preferred reporting items for systematic review and meta-analysis protocols (PRISMA-P) 2015: elaboration and explanation. BMJ. 2015;350:g7647. pmid:25555855
  21. 21. Hooijmans CR, Rovers MM, de Vries RB, Leenaars M, Ritskes-Hoitinga M, Langendam MW. SYRCLE’s risk of bias tool for animal studies. BMC Med Res Methodol. 2014;14:43. pmid:24667063
  22. 22. Rooney A. Extending a Risk-of-Bias Approach to Address In Vitro Studies2015.
  23. 23. Haubner F, Leyh M, Ohmann E, Pohl F, Prantl L, Gassner HG. Effects of external radiation in a co-culture model of endothelial cells and adipose-derived stem cells. Radiat Oncol. 2013;8:66. pmid:23514369
  24. 24. Shukla L, Luwor R, Ritchie ME, Akbarzadeh S, Zhu HJ, Morrison W, et al. Therapeutic Reversal of Radiotherapy Injury to Pro-fibrotic Dysfunctional Fibroblasts In Vitro Using Adipose-derived Stem Cells. Plast Reconstr Surg Glob Open. 2020;8(3):e2706. pmid:32537359
  25. 25. Haubner F, Muschter D, Pohl F, Schreml S, Prantl L, Gassner HG. A Co-Culture Model of Fibroblasts and Adipose Tissue-Derived Stem Cells Reveals New Insights into Impaired Wound Healing After Radiotherapy. Int J Mol Sci. 2015;16(11):25947–58. pmid:26528967
  26. 26. Yao C, Zhou Y, Wang H, Deng F, Chen Y, Zhu X, et al. Adipose-derived stem cells alleviate radiation-induced dermatitis by suppressing apoptosis and downregulating cathepsin F expression. Stem Cell Res Ther. 2021;12(1):447. pmid:34372921
  27. 27. Ejaz A, Epperly MW, Hou W, Greenberger JS, Rubin JP. Adipose-Derived Stem Cell Therapy Ameliorates Ionizing Irradiation Fibrosis via Hepatocyte Growth Factor-Mediated Transforming Growth Factor-beta Downregulation and Recruitment of Bone Marrow Cells. Stem Cells. 2019;37(6):791–802.
  28. 28. Xiao Y, Mo W, Jia H, Yu D, Qiu Y, Jiao Y, et al. Ionizing radiation induces cutaneous lipid remolding and skin adipocytes confer protection against radiation-induced skin injury. J Dermatol Sci. 2020;97(2):152–60. pmid:32001116
  29. 29. Saijo H, Suzuki K, Yoshimoto H, Imamura Y, Yamashita S, Tanaka K. Paracrine Effects of Adipose-Derived Stem Cells Promote Lymphangiogenesis in Irradiated Lymphatic Endothelial Cells. Plast Reconstr Surg. 2019;143(6):1189e–200e. pmid:30907807
  30. 30. Yu D, Zhang S, Mo W, Jiang Z, Wang M, An L, et al. Transplantation of the Stromal Vascular Fraction (SVF) Mitigates Severe Radiation-Induced Skin Injury. Radiat Res. 2021;196(3):250–60. pmid:34107043
  31. 31. Sorgel CA, Schmid R, Stadelmann N, Weisbach V, Distel L, Horch RE, et al. IGF-I and Hyaluronic Acid Mitigate the Negative Effect of Irradiation on Human Skin Keratinocytes. Cancers (Basel). 2022;14(3). pmid:35158856
  32. 32. Borrelli MR, Patel RA, Adem S, Diaz Deleon NM, Shen AH, Sokol J, et al. The antifibrotic adipose-derived stromal cell: Grafted fat enriched with CD74+ adipose-derived stromal cells reduces chronic radiation-induced skin fibrosis. Stem Cells Transl Med. 2020;9(11):1401–13. pmid:32563212
  33. 33. Sultan SM, Stern CS, Allen RJ Jr., Thanik VD, Chang CC, Nguyen PD, et al. Human fat grafting alleviates radiation skin damage in a murine model. Plast Reconstr Surg. 2011;128(2):363–72. pmid:21502909
  34. 34. Forcheron F, Agay D, Scherthan H, Riccobono D, Herodin F, Meineke V, et al. Autologous adipocyte derived stem cells favour healing in a minipig model of cutaneous radiation syndrome. PLoS One. 2012;7(2):e31694. pmid:22348120
  35. 35. Riccobono D, Agay D, Scherthan H, Forcheron F, Vivier M, Ballester B, et al. Application of adipocyte-derived stem cells in treatment of cutaneous radiation syndrome. Health Phys. 2012;103(2):120–6. pmid:22951469
  36. 36. Garza RM, Paik KJ, Chung MT, Duscher D, Gurtner GC, Longaker MT, et al. Studies in fat grafting: Part III. Fat grafting irradiated tissue—improved skin quality and decreased fat graft retention. Plast Reconstr Surg. 2014;134(2):249–57. pmid:25068325
  37. 37. Riccobono D, Forcheron F, Agay D, Scherthan H, Meineke V, Drouet M. Transient gene therapy to treat cutaneous radiation syndrome: development in a minipig model. Health Phys. 2014;106(6):713–9. pmid:24776904
  38. 38. Luan A, Duscher D, Whittam AJ, Paik KJ, Zielins ER, Brett EA, et al. Cell-Assisted Lipotransfer Improves Volume Retention in Irradiated Recipient Sites and Rescues Radiation-Induced Skin Changes. Stem Cells. 2016;34(3):668–73. pmid:26661694
  39. 39. Lindegren A, Schultz I, Sinha I, Cheung L, Khan AA, Tekle M, et al. Autologous fat transplantation alters gene expression patterns related to inflammation and hypoxia in the irradiated human breast. Br J Surg. 2019;106(5):563–73. pmid:30802303
  40. 40. Bertrand B, Eraud J, Velier M, Cauvin C, Macagno N, Boucekine M, et al. Supportive use of platelet-rich plasma and stromal vascular fraction for cell-assisted fat transfer of skin radiation-induced lesions in nude mice. Burns. 2020;46(7):1641–52. pmid:32475796
  41. 41. Borrelli MR, Diaz Deleon NM, Adem S, Patel RA, Mascharak S, Shen AH, et al. Fat grafting rescues radiation-induced joint contracture. Stem Cells. 2020;38(3):382–9. pmid:31793745
  42. 42. Deleon NMD, Adem S, Lavin CV, Abbas DB, Griffin M, King ME, et al. Angiogenic CD34+CD146+ adipose-derived stromal cells augment recovery of soft tissue after radiotherapy. J Tissue Eng Regen Med. 2021;15(12):1105–17. pmid:34582109
  43. 43. Khademi B, Safari S, Mosleh-Shirazi MA, Mokhtari M, Chenari N, Razmkhah M. Therapeutic effect of adipose-derived mesenchymal stem cells (ASCs) on radiation-induced skin damage in rats. Stem Cell Investig. 2020;7:12. pmid:32832535
  44. 44. Riccobono D, Nikovics K, Francois S, Favier AL, Jullien N, Schrock G, et al. First Insights Into the M2 Inflammatory Response After Adipose-Tissue-Derived Stem Cell Injections in Radiation-Injured Muscles. Health Phys. 2018;115(1):37–48. pmid:29787429
  45. 45. Riccobono D, Agay D, Francois S, Scherthan H, Drouet M, Forcheron F. Contribution of INTRAMUSCULAR Autologous Adipose Tissue-Derived Stem Cell Injections to Treat Cutaneous Radiation Syndrome: Preliminary Results. Health Phys. 2016;111(2):117–26. pmid:27356055
  46. 46. Sun W, Ni X, Sun S, Cai L, Yu J, Wang J, et al. Adipose-Derived Stem Cells Alleviate Radiation-Induced Muscular Fibrosis by Suppressing the Expression of TGF-beta1. Stem Cells Int. 2016;2016:5638204.
  47. 47. Huang SP, Huang CH, Shyu JF, Lee HS, Chen SG, Chan JY, et al. Promotion of wound healing using adipose-derived stem cells in radiation ulcer of a rat model. J Biomed Sci. 2013;20:51. pmid:23876213
  48. 48. Chen Y, Niu Z, Xue Y, Yuan F, Fu Y, Bai N. Improvement in the repair of defects in maxillofacial soft tissue in irradiated minipigs by a mixture of adipose-derived stem cells and platelet-rich fibrin. Br J Oral Maxillofac Surg. 2014;52(8):740–5. pmid:24993354
  49. 49. Evin N, Tosun Z, Aktan TM, Duman S, Harmankaya I, Yavas G. Effects of Adipose-Derived Stem Cells and Platelet-Rich Plasma for Prevention of Alopecia and Other Skin Complications of Radiotherapy. Ann Plast Surg. 2021;86(5):588–97. pmid:33141771
  50. 50. Abbas DB, Lavin CV, Fahy EJ, Griffin M, Guardino NJ, Nazerali RS, et al. Fat Grafts Augmented With Vitamin E Improve Volume Retention and Radiation-Induced Fibrosis. Aesthet Surg J. 2022;42(8):946–55. pmid:35350074
  51. 51. Kim J, Eom MR, Ji Jeong E, Choi JS, Kwon SK. Multiple stimulation with spheroids comprising salivary gland and adipose-derived stem cells enhances regeneration of radiation-damaged salivary glands. Journal of Industrial and Engineering Chemistry. 2022;107:493–504.
  52. 52. Adem S, Abbas DB, Lavin CV, Fahy EJ, Griffin M, Diaz Deleon NM, et al. Decellularized Adipose Matrices Can Alleviate Radiation-Induced Skin Fibrosis. Adv Wound Care (New Rochelle). 2022;11(10):524–36. pmid:34346243
  53. 53. Sowa Y, Inafuku N, Kishida T, Mori M, Mazda O, Yoshimura K. Prophylactic application of human adipose tissue-derived products to prevent radiation disorders. Plast Reconstr Surg. 2023. pmid:36728661
  54. 54. Chen X, Chen Y, Wang Z, Dong Z, Yao Y, Li Y, et al. Adipose-derived stem cells regulate CD4+ T-cell-mediated macrophage polarization and fibrosis in fat grafting in a mouse model. Heliyon. 2022;8(11):e11538. pmid:36406697
  55. 55. Griffin MF, desJardins-Park HE, Mascharak S, Borrelli MR, Longaker MT. Understanding the impact of fibroblast heterogeneity on skin fibrosis. Dis Model Mech. 2020;13(6). pmid:32541065
  56. 56. Xue EY, Narvaez L, Chu CK, Hanson SE. Fat Processing Techniques. Semin Plast Surg. 2020;34(1):11–6. pmid:32071574
  57. 57. Kim KK, Sheppard D, Chapman HA. TGF-beta1 Signaling and Tissue Fibrosis. Cold Spring Harb Perspect Biol. 2018;10(4).
  58. 58. Halle M, Gabrielsen A, Paulsson-Berne G, Gahm C, Agardh HE, Farnebo F, et al. Sustained inflammation due to nuclear factor-kappa B activation in irradiated human arteries. J Am Coll Cardiol. 2010;55(12):1227–36. pmid:20298930
  59. 59. Kischer CW, Thies AC, Chvapil M. Perivascular myofibroblasts and microvascular occlusion in hypertrophic scars and keloids. Hum Pathol. 1982;13(9):819–24. pmid:7106747
  60. 60. Sultan SM, Barr JS, Butala P, Davidson EH, Weinstein AL, Knobel D, et al. Fat grafting accelerates revascularisation and decreases fibrosis following thermal injury. J Plast Reconstr Aesthet Surg. 2012;65(2):219–27. pmid:21962530
  61. 61. Del Papa N, Caviggioli F, Sambataro D, Zaccara E, Vinci V, Di Luca G, et al. Autologous fat grafting in the treatment of fibrotic perioral changes in patients with systemic sclerosis. Cell Transplant. 2015;24(1):63–72. pmid:25606975
  62. 62. Bruno A, Delli Santi G, Fasciani L, Cempanari M, Palombo M, Palombo P. Burn scar lipofilling: immunohistochemical and clinical outcomes. J Craniofac Surg. 2013;24(5):1806–14. pmid:24036785
  63. 63. Nie C, Yang D, Xu J, Si Z, Jin X, Zhang J. Locally administered adipose-derived stem cells accelerate wound healing through differentiation and vasculogenesis. Cell Transplant. 2011;20(2):205–16. pmid:20719083
  64. 64. Planat-Benard V, Silvestre JS, Cousin B, Andre M, Nibbelink M, Tamarat R, et al. Plasticity of human adipose lineage cells toward endothelial cells: physiological and therapeutic perspectives. Circulation. 2004;109(5):656–63. pmid:14734516
  65. 65. Cao R, Eriksson A, Kubo H, Alitalo K, Cao Y, Thyberg J. Comparative Evaluation of FGF-2, VEGF-A, and VEGF-C: Induced Angiogenesis, Lymphangiogenesis, Vascular Fenestrations, and Permeability. Circulation Research. 2004;94(5):664–70. pmid:14739162
  66. 66. Rehman J, Traktuev D, Li J, Merfeld-Clauss S, Temm-Grove CJ, Bovenkerk JE, et al. Secretion of angiogenic and antiapoptotic factors by human adipose stromal cells. Circulation. 2004;109(10):1292–8. pmid:14993122
  67. 67. Cai L, Johnstone BH, Cook TG, Liang Z, Traktuev D, Cornetta K, et al. Suppression of hepatocyte growth factor production impairs the ability of adipose-derived stem cells to promote ischemic tissue revascularization. Stem Cells. 2007;25(12):3234–43. pmid:17901400
  68. 68. Hom DB, Unger GM, Pernell KJ, Manivel JC. Improving surgical wound healing with basic fibroblast growth factor after radiation. Laryngoscope. 2005;115(3):412–22. pmid:15744149
  69. 69. Gaugler MH, Squiban C, van der Meeren A, Bertho JM, Vandamme M, Mouthon MA. Late and persistent up-regulation of intercellular adhesion molecule-1 (ICAM-1) expression by ionizing radiation in human endothelial cells in vitro. Int J Radiat Biol. 1997;72(2):201–9. pmid:9269313
  70. 70. Haubner F, Ohmann E, Pohl F, Prantl L, Strutz J, Gassner HG. Effects of radiation on the expression of adhesion molecules and cytokines in a static model of human dermal microvascular endothelial cells. Clin Hemorheol Microcirc. 2013;54(4):371–9. pmid:23089880
  71. 71. Hinrichs CS, Watroba NL, Rezaishiraz H, Giese W, Hurd T, Fassl KA, et al. Lymphedema secondary to postmastectomy radiation: incidence and risk factors. Ann Surg Oncol. 2004;11(6):573–80. pmid:15172932
  72. 72. Ahmadzadeh N, Robering JW, Kengelbach-Weigand A, Al-Abboodi M, Beier JP, Horch RE, et al. Human adipose-derived stem cells support lymphangiogenesis in vitro by secretion of lymphangiogenic factors. Experimental Cell Research. 2020;388(2):111816. pmid:31923426
  73. 73. Kim W-S, Park B-S, Kim H-K, Park J-S, Kim K-J, Choi J-S, et al. Evidence supporting antioxidant action of adipose-derived stem cells: Protection of human dermal fibroblasts from oxidative stress. Journal of Dermatological Science. 2008;49(2):133–42. pmid:17870415
  74. 74. Ahamed J, Laurence J. Role of Platelet-Derived Transforming Growth Factor-beta1 and Reactive Oxygen Species in Radiation-Induced Organ Fibrosis. Antioxid Redox Signal. 2017;27(13):977–88.
  75. 75. Wynn TA, Barron L. Macrophages: master regulators of inflammation and fibrosis. Semin Liver Dis. 2010;30(3):245–57. pmid:20665377
  76. 76. Lucas T, Waisman A, Ranjan R, Roes J, Krieg T, Muller W, et al. Differential roles of macrophages in diverse phases of skin repair. J Immunol. 2010;184(7):3964–77. pmid:20176743
  77. 77. Liu YC, Zou XB, Chai YF, Yao YM. Macrophage polarization in inflammatory diseases. Int J Biol Sci. 2014;10(5):520–9. pmid:24910531
  78. 78. Straub JM, New J, Hamilton CD, Lominska C, Shnayder Y, Thomas SM. Radiation-induced fibrosis: mechanisms and implications for therapy. J Cancer Res Clin Oncol. 2015;141(11):1985–94. pmid:25910988
  79. 79. Nguyen A, Guo J, Banyard DA, Fadavi D, Toranto JD, Wirth GA, et al. Stromal vascular fraction: A regenerative reality? Part 1: Current concepts and review of the literature. Journal of Plastic, Reconstructive & Aesthetic Surgery. 2016;69(2):170–9. pmid:26565755
  80. 80. Hildreth AD, Ma F, Wong YY, Sun R, Pellegrini M, O’Sullivan TE. Single-cell sequencing of human white adipose tissue identifies new cell states in health and obesity. Nature Immunology. 2021;22(5):639–53. pmid:33907320
  81. 81. Di Maggio FM, Minafra L, Forte GI, Cammarata FP, Lio D, Messa C, et al. Portrait of inflammatory response to ionizing radiation treatment. J Inflamm (Lond). 2015;12:14. pmid:25705130
  82. 82. Schaue D, Kachikwu EL, McBride WH. Cytokines in radiobiological responses: a review. Radiat Res. 2012;178(6):505–23. pmid:23106210
  83. 83. Lee SH, Kwon JY, Kim SY, Jung K, Cho ML. Interferon-gamma regulates inflammatory cell death by targeting necroptosis in experimental autoimmune arthritis. Sci Rep. 2017;7(1):10133. pmid:28860618
  84. 84. Ceccarelli S, Pontecorvi P, Anastasiadou E, Napoli C, Marchese C. Immunomodulatory Effect of Adipose-Derived Stem Cells: The Cutting Edge of Clinical Application. Frontiers in Cell and Developmental Biology. 2020;8. pmid:32363193
  85. 85. Yao Y, Xu XH, Jin L. Macrophage Polarization in Physiological and Pathological Pregnancy. Front Immunol. 2019;10:792. pmid:31037072
  86. 86. Pilny E, Smolarczyk R, Jarosz-Biej M, Hadyk A, Skorupa A, Ciszek M, et al. Human ADSC xenograft through IL-6 secretion activates M2 macrophages responsible for the repair of damaged muscle tissue. Stem Cell Res Ther. 2019;10(1):93. pmid:30867059
  87. 87. Lo Sicco C, Reverberi D, Balbi C, Ulivi V, Principi E, Pascucci L, et al. Mesenchymal Stem Cell-Derived Extracellular Vesicles as Mediators of Anti-Inflammatory Effects: Endorsement of Macrophage Polarization. Stem Cells Transl Med. 2017;6(3):1018–28. pmid:28186708
  88. 88. Sica A, Mantovani A. Macrophage plasticity and polarization: in vivo veritas. J Clin Invest. 2012;122(3):787–95. pmid:22378047
  89. 89. Hu Y, Li G, Zhang Y, Liu N, Zhang P, Pan C, et al. Upregulated TSG-6 Expression in ADSCs Inhibits the BV2 Microglia-Mediated Inflammatory Response. Biomed Res Int. 2018;2018:7239181. pmid:30584538
  90. 90. Wang L, Hu L, Zhou X, Xiong Z, Zhang C, Shehada HMA, et al. Exosomes secreted by human adipose mesenchymal stem cells promote scarless cutaneous repair by regulating extracellular matrix remodelling. Sci Rep. 2017;7(1):13321. pmid:29042658
  91. 91. Suga H, Eto H, Shigeura T, Inoue K, Aoi N, Kato H, et al. IFATS collection: Fibroblast growth factor-2-induced hepatocyte growth factor secretion by adipose-derived stromal cells inhibits postinjury fibrogenesis through a c-Jun N-terminal kinase-dependent mechanism. Stem Cells. 2009;27(1):238–49. pmid:18772314
  92. 92. Abe M, Yokoyama Y, Ishikawa O. A possible mechanism of basic fibroblast growth factor-promoted scarless wound healing: the induction of myofibroblast apoptosis. Eur J Dermatol. 2012;22(1):46–53. pmid:22370167
  93. 93. Xie J, Bian H, Qi S, Xu Y, Tang J, Li T, et al. Effects of basic fibroblast growth factor on the expression of extracellular matrix and matrix metalloproteinase-1 in wound healing. Clin Exp Dermatol. 2008;33(2):176–82. pmid:18257838
  94. 94. Shi HX, Lin C, Lin BB, Wang ZG, Zhang HY, Wu FZ, et al. The anti-scar effects of basic fibroblast growth factor on the wound repair in vitro and in vivo. PLoS One. 2013;8(4):e59966. pmid:23565178
  95. 95. Kalinina N, Kharlampieva D, Loguinova M, Butenko I, Pobeguts O, Efimenko A, et al. Characterization of secretomes provides evidence for adipose-derived mesenchymal stromal cells subtypes. Stem Cell Res Ther. 2015;6:221. pmid:26560317
  96. 96. Rosu-Myles M, Stewart E, Trowbridge J, Ito CY, Zandstra P, Bhatia M. A unique population of bone marrow cells migrates to skeletal muscle via hepatocyte growth factor/c-met axis. J Cell Sci. 2005;118(Pt 19):4343–52. pmid:16144866
  97. 97. Mueller CK, Thorwarth M, Schultze-Mosgau S. Late changes in cutaneous gene expression patterns after adjuvant treatment of oral squamous cell carcinoma (OSCC) by radiation therapy. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 2010;109(5):694–9. pmid:20185344
  98. 98. Walton KL, Johnson KE, Harrison CA. Targeting TGF-beta Mediated SMAD Signaling for the Prevention of Fibrosis. Front Pharmacol. 2017;8:461.
  99. 99. Giannandrea M, Parks WC. Diverse functions of matrix metalloproteinases during fibrosis. Dis Model Mech. 2014;7(2):193–203. pmid:24713275
  100. 100. Loffek S, Schilling O, Franzke CW. Series "matrix metalloproteinases in lung health and disease": Biological role of matrix metalloproteinases: a critical balance. Eur Respir J. 2011;38(1):191–208. pmid:21177845
  101. 101. Eming S, Smola H, Hartmann B, Malchau G, Wegner R, Krieg T, et al. The inhibition of matrix metalloproteinase activity in chronic wounds by a polyacrylate superabsorber. Biomaterials. 2008;29(19):2932–40. pmid:18400293
  102. 102. Goffin L, Seguin-Estevez Q, Alvarez M, Reith W, Chizzolini C. Transcriptional regulation of matrix metalloproteinase-1 and collagen 1A2 explains the anti-fibrotic effect exerted by proteasome inhibition in human dermal fibroblasts. Arthritis Res Ther. 2010;12(2):R73. pmid:20429888
  103. 103. Krstic J, Santibanez JF. Transforming growth factor-beta and matrix metalloproteinases: functional interactions in tumor stroma-infiltrating myeloid cells. ScientificWorldJournal. 2014;2014:521754. pmid:24578639
  104. 104. Cawthorn WP, Scheller EL, MacDougald OA. Adipose tissue stem cells meet preadipocyte commitment: going back to the future. J Lipid Res. 2012;53(2):227–46. pmid:22140268
  105. 105. Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ, et al. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng. 2001;7(2):211–28. pmid:11304456
  106. 106. Mendelsohn FA, Divino CM, Reis ED, Kerstein MD. Wound care after radiation therapy. Adv Skin Wound Care. 2002;15(5):216–24. pmid:12368711
  107. 107. Borrelli MR, Shen AH, Lee GK, Momeni A, Longaker MT, Wan DC. Radiation-Induced Skin Fibrosis: Pathogenesis, Current Treatment Options, and Emerging Therapeutics. Ann Plast Surg. 2019;83(4S Suppl 1):S59-S64. pmid:31513068
  108. 108. Coleman SR. Long-term survival of fat transplants: controlled demonstrations. Aesthetic Plast Surg. 1995;19(5):421–5. pmid:8526158
  109. 109. Kolle SF, Fischer-Nielsen A, Mathiasen AB, Elberg JJ, Oliveri RS, Glovinski PV, et al. Enrichment of autologous fat grafts with ex-vivo expanded adipose tissue-derived stem cells for graft survival: a randomised placebo-controlled trial. Lancet. 2013;382(9898):1113–20. pmid:24075051
  110. 110. Yoshimura K, Sato K, Aoi N, Kurita M, Hirohi T, Harii K. Cell-assisted lipotransfer for cosmetic breast augmentation: supportive use of adipose-derived stem/stromal cells. Aesthetic Plast Surg. 2008;32(1):48–55; discussion 6–7. pmid:17763894
  111. 111. Mazini L, Ezzoubi M, Malka G. Overview of current adipose-derived stem cell (ADSCs) processing involved in therapeutic advancements: flow chart and regulation updates before and after COVID-19. Stem Cell Res Ther. 2021;12(1):1. pmid:33397467
  112. 112. Tobita M, Tajima S, Mizuno H. Adipose tissue-derived mesenchymal stem cells and platelet-rich plasma: stem cell transplantation methods that enhance stemness. Stem Cell Res Ther. 2015;6:215. pmid:26541973
  113. 113. Sunitha Raja V, Munirathnam Naidu E. Platelet-rich fibrin: evolution of a second-generation platelet concentrate. Indian J Dent Res. 2008;19(1):42–6. pmid:18245923
  114. 114. Porzionato A, Stocco E, Barbon S, Grandi F, Macchi V, De Caro R. Tissue-Engineered Grafts from Human Decellularized Extracellular Matrices: A Systematic Review and Future Perspectives. Int J Mol Sci. 2018;19(12). pmid:30567407
  115. 115. Dolivo DM, Larson SA, Dominko T. Fibroblast Growth Factor 2 as an Antifibrotic: Antagonism of Myofibroblast Differentiation and Suppression of Pro-Fibrotic Gene Expression. Cytokine Growth Factor Rev. 2017;38:49–58. pmid:28967471
  116. 116. Feng J, Doi K, Kuno S, Mineda K, Kato H, Kinoshita K, et al. Micronized cellular adipose matrix as a therapeutic injectable for diabetic ulcer. Regen Med. 2015;10(6):699–708. pmid:26440061
  117. 117. Araujo S, Sganzella MF, Sagiorato RN, Leite MN, Caetano GF, Aparecida de Aro A, et al. Human adipose-derived stem cells in fibrin glue carrier modulate wound healing phases in rats. Current Research in Biotechnology. 2022;4:503–13.
  118. 118. Thuy le TT, Kawada N. Antifibrotic role of macrophage migration inhibitory factor: discovery of an unexpected function. Hepatology. 2012;55(4):1295–7. pmid:22461077
  119. 119. Isogai Z, Ono RN, Ushiro S, Keene DR, Chen Y, Mazzieri R, et al. Latent transforming growth factor beta-binding protein 1 interacts with fibrillin and is a microfibril-associated protein. J Biol Chem. 2003;278(4):2750–7. pmid:12429738
  120. 120. Neptune ER, Frischmeyer PA, Arking DE, Myers L, Bunton TE, Gayraud B, et al. Dysregulation of TGF-beta activation contributes to pathogenesis in Marfan syndrome. Nat Genet. 2003;33(3):407–11. pmid:12598898
  121. 121. Suarez EM, Knackstedt RJ, Jenrette JM. Significant fibrosis after radiation therapy in a patient with Marfan syndrome. Radiat Oncol J. 2014;32(3):208–12. pmid:25324993
  122. 122. Sohn JH, Lee YK, Han JS, Jeon YG, Kim JI, Choe SS, et al. Perilipin 1 (Plin1) deficiency promotes inflammatory responses in lean adipose tissue through lipid dysregulation. J Biol Chem. 2018;293(36):13974–88. pmid:30042231
  123. 123. Borrelli MR, Patel RA, Blackshear C, Vistnes S, Diaz Deleon NM, Adem S, et al. CD34+CD146+ adipose-derived stromal cells enhance engraftment of transplanted fat. Stem Cells Transl Med. 2020;9(11):1389–400. pmid:32543083
  124. 124. Thurston G, Suri C, Smith K, McClain J, Sato TN, Yancopoulos GD, et al. Leakage-resistant blood vessels in mice transgenically overexpressing angiopoietin-1. Sci. 1999;286(5449):2511–4. pmid:10617467
  125. 125. Hildreth AD, Ma F, Wong YY, Sun R, Pellegrini M, O’Sullivan TE. Single-cell sequencing of human white adipose tissue identifies new cell states in health and obesity. Nat Immunol. 2021;22(5):639–53. pmid:33907320
  126. 126. Mark P, Kleinsorge M, Gaebel R, Lux CA, Toelk A, Pittermann E, et al. Human Mesenchymal Stem Cells Display Reduced Expression of CD105 after Culture in Serum-Free Medium. Stem Cells Int. 2013;2013:698076. pmid:24194767
  127. 127. Spees JL, Gregory CA, Singh H, Tucker HA, Peister A, Lynch PJ, et al. Internalized antigens must be removed to prepare hypoimmunogenic mesenchymal stem cells for cell and gene therapy. Mol Ther. 2004;9(5):747–56. pmid:15120336
  128. 128. Kuznetsov SA, Mankani MH, Robey PG. Effect of serum on human bone marrow stromal cells: ex vivo expansion and in vivo bone formation. Transplantation. 2000;70(12):1780–7. pmid:11152111
  129. 129. Shahdadfar A, Fronsdal K, Haug T, Reinholt FP, Brinchmann JE. In vitro expansion of human mesenchymal stem cells: choice of serum is a determinant of cell proliferation, differentiation, gene expression, and transcriptome stability. Stem Cells. 2005;23(9):1357–66. pmid:16081661
  130. 130. Labedz-Maslowska A, Szkaradek A, Mierzwinski T, Madeja Z, Zuba-Surma E. Processing and Ex Vivo Expansion of Adipose Tissue-Derived Mesenchymal Stem/Stromal Cells for the Development of an Advanced Therapy Medicinal Product for use in Humans. Cells. 2021;10(8). pmid:34440677
  131. 131. RL OC, T R, RI C, SA M, S P, DR L, et al. Barriers to clinical trial recruitment and recommendations for supporting breast surgeons in the workplace. The Bulletin of the Royal College of Surgeons of England. 2020;102(7):320–3.
  132. 132. Jiang Z, Zhou X, Li R, Michal JJ, Zhang S, Dodson MV, et al. Whole transcriptome analysis with sequencing: methods, challenges and potential solutions. Cell Mol Life Sci. 2015;72(18):3425–39. pmid:26018601