1
|
Diniz IMA, de Oliveira RF, do Valle IB, Picoli CC, Jácome-Santos H, de Almeida Queiroz Ferreira L, Avelar GF, Diniz MG, Birbrair A. Photobiomodulation therapy induces NG2 activation through dermal adipocyte lipolysis during wound healing. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2025; 267:113151. [PMID: 40187095 DOI: 10.1016/j.jphotobiol.2025.113151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/05/2025] [Accepted: 03/15/2025] [Indexed: 04/07/2025]
Abstract
Photobiomodulation therapy (PBMT) is a rapidly advancing approach for restoring damaged tissues, particularly in skin and mucosal wounds. While its application is promising, the role of mature adipocytes in regenerating mesenchymal tissues after PBMT remains largely unexplored. This study demonstrates that PBMT applied to skin wounds significantly reduces the number and size of mature adipocytes. Additionally, PBMT modulates the upregulation of peroxisome proliferator-activated receptor γ (PPARγ), increasing the gene expression of fatty acid binding protein 4 (Fabp4) and perilipin 1, which are linked to enhanced lipolysis. The molecular activation of neural/glial antigen 2 (NG2) indicates the recruitment of progenitor cells following mature adipocytes lipolysis. In vitro, PBMT improved dermal skin cell proliferation, migration, inflammatory regulation, and differentiation capacities. These findings reveal a novel mechanistic pathway for skin regeneration, emphasizing the therapeutic potential of PBMT in modulating dermal fat tissue to facilitate wound healing. Collectively, this emerging knowledge provides valuable insights into managing dermal fat tissue to support wound healing.
Collapse
Affiliation(s)
- Ivana Márcia Alves Diniz
- Department of Restorative Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| | - Rafaela Férrer de Oliveira
- Department of Restorative Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Isabella Bittencourt do Valle
- Department of Pathology, School of Dentistry, Universidade Federal do Espírito Santo, Vitória, Espírito Santo, Brazil
| | | | - Humberto Jácome-Santos
- Department of Restorative Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Luiza de Almeida Queiroz Ferreira
- Department of Restorative Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Gleide Fernandes Avelar
- Department of Morphology, Biological Sciences Institute, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Marina Gonçalves Diniz
- Department of Pathology, Biological Sciences Institute, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Alexander Birbrair
- Department of Dermatology, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
2
|
Riley N, Kasza I, Hermsmeyer IDK, Trautman ME, Barrett-Wilt G, Jain R, Simcox JA, Yen CLE, MacDougald OA, Lamming DW, Alexander CM. Dietary lipids are largely deposited in skin and rapidly affect insulating properties. Nat Commun 2025; 16:4570. [PMID: 40379673 PMCID: PMC12084621 DOI: 10.1038/s41467-025-59869-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 05/07/2025] [Indexed: 05/19/2025] Open
Abstract
Skin is a regulatory hub for energy expenditure and metabolism, and alteration of lipid metabolism enzymes in skin impacts thermogenesis and obesogenesis in mice. Here we show that thermal properties of skin are highly reactive to diet: within three days, a high fat diet reduces heat transfer through skin. In contrast, a dietary manipulation that prevents obesity accelerates energy loss through skins. We find that skin is the largest target for dietary fat delivery, and that dietary triglyceride is assimilated by epidermis and dermal white adipose tissue, persisting for weeks after feeding. With caloric-restriction, mouse skins thin and assimilation of circulating lipids decreases. Using multi-modal lipid profiling, keratinocytes and sebocytes are implicated in lipid changes, which correlate with thermal function. We propose that skin should be routinely included in physiological studies of lipid metabolism, given the size of the skin lipid reservoir and its adaptable functionality.
Collapse
Affiliation(s)
- Nick Riley
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, USA
| | - Ildiko Kasza
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, USA
| | - Isabel D K Hermsmeyer
- Department of Molecular & Integrative Physiology, University of Michigan, Michigan, USA
| | - Michaela E Trautman
- Department of Medicine, University of Wisconsin-Madison, Madison, USA
- William S. Middleton Memorial Veterans Hospital, Madison, USA
| | | | - Raghav Jain
- Department of Biochemistry, University of Wisconsin-Madison, Madison, USA
| | - Judith A Simcox
- Department of Biochemistry, University of Wisconsin-Madison, Madison, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, USA
| | - Chi-Liang E Yen
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, USA
| | - Ormond A MacDougald
- Department of Molecular & Integrative Physiology, University of Michigan, Michigan, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, USA
- William S. Middleton Memorial Veterans Hospital, Madison, USA
| | - Caroline M Alexander
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, USA.
| |
Collapse
|
3
|
Hirpara MM, Woll J, Mesinkovska NA. Isotretinoin Effects on Nasal Contouring: The Role of Facial Landmark Detection Technology. Dermatol Surg 2025:00042728-990000000-01204. [PMID: 40304376 DOI: 10.1097/dss.0000000000004647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Affiliation(s)
- Milan M Hirpara
- Department of Dermatology, University of California, Irvine, Irvine, California
| | | | | |
Collapse
|
4
|
Ni X, Xu N, Shen J, Wan X, Shen S, Lei C, Wang S, Wang B, Shan X. SVF-gel application for the alleviation of full-thickness skin graft contraction: an experimental study in mice. Sci Rep 2025; 15:15082. [PMID: 40301501 PMCID: PMC12041231 DOI: 10.1038/s41598-025-99649-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 04/22/2025] [Indexed: 05/01/2025] Open
Abstract
Skin grafts often suffer from contracture, complicating recovery. SVF-gel shows promise in addressing scar contracture, but its therapeutic effects and mechanisms are not fully understood. This study evaluates the efficacy of SVF-gel in full-thickness skin grafting. Full-thickness skin grafts were harvested from mice dorsal skin, rotated and sutured. SVF-gel or saline was injected beneath the muscle fascia. Immunohistochemistry assessed SVF-gel's effects on angiogenesis, collagen deposition, fibrosis, and dermal adipocytes. Keloid-related genes from GSE92566 and GSE158395 were analyzed for functional enrichment and protein-protein interactions, with hub genes validated using GSE190626. SVF-gel significantly increased the grafted area, thickness of the epidermal and dermal adipose layers, and hair follicles compared to the control group. SVF-gel enhanced CD31 and perilipin expression, decreased α-SMA expression, and identified HLA+ cells around CD31+ cells in the dermal microvessels and adipose tissue of the graft. Sixty commonly downregulated keloid-related genes were identified, with KEGG pathway analysis indicating enrichment in the PPAR signaling pathway and lipolysis regulation. Five hub genes (ADIPOQ, FABP4, KRT7, LEP, and PIP) were validated. SVF-gel shows promise as a stem cell therapy for skin grafts, improving outcomes by enhancing revascularization, increasing collagen fiber density and regularity, accelerating myofibroblast turnover, promoting adipogenesis, and increasing hair follicles.
Collapse
Affiliation(s)
- Xuejun Ni
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Nating Xu
- Department of Burn and Plastic Surgery, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Jian Shen
- Department of Burn and Plastic Surgery, Ma'anshan People's Hospital, Ma'anshan, 243000, China
| | - Xiaofen Wan
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Shuyi Shen
- Department of Dermatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Chen Lei
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Songyu Wang
- Department of Dermatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Biao Wang
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China.
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
| | - Xiuying Shan
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China.
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
| |
Collapse
|
5
|
Braitenbach BPDF, Cerqueira CBSD, Lima SS, Hegouet IDC, Moreno BCL, Medrado ARAP. Effects of photobiomodulation on adipocytic infiltration in sites of skin healing: in vivo experimental study. Lasers Med Sci 2025; 40:153. [PMID: 40111643 DOI: 10.1007/s10103-025-04410-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 03/12/2025] [Indexed: 03/22/2025]
Abstract
Adipocyte infiltration consists of a temporary increase in the number of adipocytes in the microenvironment of tissue injury. There is already evidence in the literature of adipocytes' participation in promoting the inflammatory response, and parallelly laser photobiomodulation can benefit the tissue repair process. This study aimed to chronologically analyze adipocytic infiltration in the repair of photobiomodulated skin wounds experimentally induced in rats through histomorphometric analysis. The sample consisted of 20 rats divided into 2 groups: control group and group subjected to laser photobiomodulation. The skin portions of the back of rats were processed and stained with Hematoxylin-Eosin in 4 μm thick sections including the surgical wound 5 and 10 days after the proposed treatments. Qualitative and quantitative analyses were performed by capturing images of tissue sections, describing the organizational pattern of adipocytes around the surgical wound and counting individual adipocytes in the connective tissue in formation. Adipocytic infiltration was observed in both experimental groups on the 5th day, with a decrease on the 10th day. The group treated with photobiomodulation presented a greater number of adipocytes compared to the control group, in both periods analyzed. The findings of the present study seem to corroborate the literature, which indicates that adipose cells might stimulate inflammation and repair, and photobiomodulation can enhance these effects, since it aids the process of adipocytic infiltration in the injured area. Clinical trial number: Not applicable.
Collapse
|
6
|
Wu Z, Wang Z, Chen T, Wang D, Zhou F, Zhang G, Wei S, Wu Y. Dermal white adipose tissue: A new modulator in wound healing and regeneration. Regen Ther 2025; 28:115-125. [PMID: 39717110 PMCID: PMC11665542 DOI: 10.1016/j.reth.2024.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 11/06/2024] [Accepted: 11/20/2024] [Indexed: 12/25/2024] Open
Abstract
Dermal white adipose tissue (dWAT), distinguished by its origin from cells within the dermis and independence from subcutaneous fat tissue, has garnered significant attention for its non-metabolic functions. Characterized by strong communication with other components of the skin, dWAT mediates the proliferation and recruitment of various cell types by releasing adipogenic and inflammatory factors. Here, we focus on the modulatory role of dWAT at different stages during wound healing, highlighting its ability to mediate the adipocyte-to-myofibroblast transition which plays a pivotal role in the physiology and pathology processes of skin fibrosis, scarring, and aging. This review highlights the regulatory potential of dWAT in modulating wound healing processes and presents it as a target for developing therapeutic strategies aimed at reducing scarring and enhancing regenerative outcomes in skin-related disorders.
Collapse
Affiliation(s)
- Zhongyu Wu
- Department of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Zhanqi Wang
- Department of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun Avenue South, Haidian District, Beijing 100081, PR China
| | - Tao Chen
- Department of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Dongyang Wang
- Department of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Feng Zhou
- Department of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Guorui Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Shan Wei
- Huizhou Health Sciences Polytechnic, Huizhou 516025, Guangdong, PR China
| | - Yingying Wu
- Department of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| |
Collapse
|
7
|
Ertel A, Anderegg U, Franz S, Saalbach A. Dermal White Adipose Tissue-Derived Il-33 Regulates Il-4/13 Expression in Myeloid Cells during Inflammation. J Invest Dermatol 2025; 145:370-382. [PMID: 38909842 DOI: 10.1016/j.jid.2024.05.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/25/2024] [Accepted: 05/24/2024] [Indexed: 06/25/2024]
Abstract
Effective tissue response to infection and injury essentially relies on the fine-tuned induction and subsequent resolution of inflammation. Recent research highlighted multiple functions of dermal white adipose tissue (dWAT) beyond its traditional role as an energy reservoir. However, in contrast to other fat depots, there are only limited data about putative immune-regulatory functions of dWAT. Therefore, we investigated the impact of dWAT in the control of an acute skin inflammation. Skin inflammation triggers the activation of dWAT. In turn, soluble mediators of activated dWAT stimulate the expression of numerous genes controlling skin inflammation, including the T helper 2 cell cytokines Il4 and Il13, in myeloid cells in vitro. Consistently, myeloid cells isolated from inflamed skin showed a significant upregulation of Il-4/13 expression compared with those isolated from healthy skin. Mechanistically, we demonstrate that IL-33 released from activated dWAT is responsible for IL-4/13 stimulation in myeloid cells. Interestingly, obesity attenuates IL-33 secretion in dWAT during inflammation, resulting in decreased Il-4 and Il-13 expressions in myeloid cells. Our data reveal an IL-33-IL-4/13 signaling cascade initiated from dWAT in a T helper 2-independent context of inflammation that may contribute to limitation of inflammation. This cascade seems to be disturbed in individuals with obesity with prolonged inflammation.
Collapse
Affiliation(s)
- Anastasia Ertel
- Department of Dermatology, Venereology and Allergology, University of Leipzig Medical Center, Leipzig, Germany
| | - Ulf Anderegg
- Department of Dermatology, Venereology and Allergology, University of Leipzig Medical Center, Leipzig, Germany
| | - Sandra Franz
- Department of Dermatology, Venereology and Allergology, University of Leipzig Medical Center, Leipzig, Germany
| | - Anja Saalbach
- Department of Dermatology, Venereology and Allergology, University of Leipzig Medical Center, Leipzig, Germany.
| |
Collapse
|
8
|
Sun J, Ahmed I, Brown J, Khosrotehrani K, Shafiee A. The empowering influence of air-liquid interface culture on skin organoid hair follicle development. BURNS & TRAUMA 2025; 13:tkae070. [PMID: 39822647 PMCID: PMC11736897 DOI: 10.1093/burnst/tkae070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 10/25/2024] [Accepted: 10/30/2024] [Indexed: 01/19/2025]
Abstract
Background Rodent models have been widely used to investigate skin development, but do not account for significant differences in composition compared to human skin. On the other hand, two-dimensional and three-dimensional engineered skin models still lack the complex features of human skin such as appendages and pigmentation. Recently, hair follicle containing skin organoids (SKOs) with a stratified epidermis, and dermis layer have been generated as floating spheres from human-induced pluripotent stem cells (hiPSCs). Methods The current study aims to investigate the generation of hiPSCs-derived SKOs using an air-liquid interface (ALI) model on transwell membranes (T-SKOs) and compares their development with conventional floating culture in low-attachment plates (F-SKOs). Results Mature SKOs containing an epidermis, dermis, and appendages are created in both T-SKO and F-SKO conditions. It was found that the hair follicles are smaller and shorter in the F-SKO compared with T-SKOs. Additionally, the ALI conditions contribute to enhanced hair follicle numbers than conventional floating culture. Conclusions Together, this study demonstrates the significant influence of transwell culture on the morphogenesis of hair follicles within SKOs and highlights the potential for refinement of skin model engineering for advancing dermatology and skin research.
Collapse
Affiliation(s)
- Jane Sun
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4102Australia
| | - Imaan Ahmed
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4102Australia
| | - Jason Brown
- Herston Biofabrication Institute, Metro North Hospital and Health Service, Queensland Health, Brisbane, QLD, 4029Australia
- Royal Brisbane and Women's Hospital, Metro North Hospital and Health Service, Queensland Health, Brisbane, QLD, 4029Australia
| | - Kiarash Khosrotehrani
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4102Australia
| | - Abbas Shafiee
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4102Australia
- Herston Biofabrication Institute, Metro North Hospital and Health Service, Queensland Health, Brisbane, QLD, 4029Australia
- Royal Brisbane and Women's Hospital, Metro North Hospital and Health Service, Queensland Health, Brisbane, QLD, 4029Australia
| |
Collapse
|
9
|
Plum M, Beier JP, Ruhl T. Delayed cutaneous wound healing in young and old female mice is associated with differential growth factor release but not inflammatory cytokine secretion. Biogerontology 2025; 26:37. [PMID: 39775106 PMCID: PMC11711145 DOI: 10.1007/s10522-024-10179-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 12/26/2024] [Indexed: 01/11/2025]
Abstract
The capacity for tissue repair during wound healing declines with age. A chronic low but systemic inflammatory status, often called "inflammaging", is considered a key factor that contributes to impaired tissue regeneration. This phenomenon has been substantiated by an increased number of immune cells in wound-tissue of old mice. Although immune cells coordinate an inflammatory response by their secretome the composition of the wound milieu has not been examined. In young (2 months) and old (18 months) female mice, excision wounds were induced using a punch biopsy device, i.e., the healing progress occurred through secondary intention. The closure rate was analyzed for 7 days. At days 1, 3 and 7 post-surgery, wound specimen were investigated for immunohistochemical detection of granulocytes, M1-macrophages and mesenchymal stem cells of the skin. The concentrations of inflammatory cytokines and regenerative growth factors were determined in tissue homogenates by ELISA. The carbonyl assay was used to determine protein oxidation. In old mice, the wound closure was delayed between days 1 and 3 post-surgery, as was the peak of immune cell infiltration. There was no age effect on the concentration of inflammatory cytokines, but wounds of young animals contained higher number of mesenchymal stem cells and increased levels of growth factors. Protein oxidation was increased with age. The present study suggests that a reduced regenerative capacity rather than an enhanced inflammatory score affected the tissue regeneration process in old mice.
Collapse
Affiliation(s)
- Melissa Plum
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Justus P Beier
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Tim Ruhl
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany.
| |
Collapse
|
10
|
DeCostanza L, Grogan GM, Bruce AC, Peachey CM, Clark EA, Atkins K, Tylek T, Solga MD, Spiller KL, Peirce SM, Campbell CA, Cottler PS. Decellularized porcine dermal hydrogel enhances implant-based wound healing in the setting of irradiation. Acta Biomater 2025; 191:260-275. [PMID: 39522628 DOI: 10.1016/j.actbio.2024.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Acellular Dermal Matrix (ADM) provides mechanical and soft tissue support in implant-based breast reconstruction, and has shown to modulate the healing response. However, skin flap necrosis, edema, and previous radiation therapy can hinder ADM integration. Effective biomaterial integration requires regulating the immune response, fibrosis, and adipocyte-driven functionalization. Extracellular matrix (ECM) hydrogels have demonstrated utility in tissue regeneration, and decreasing inflammation and fibrosis in various tissues. Therefore, we hypothesized that a Decellularized Porcine Dermal (DPD) hydrogel to support ADM integration would prevent excessive fibrosis, regulate the macrophage response, and promote adipogenesis. Exploration of DPD hydrogel during ADM implantation in mice (healthy and radiated) revealed long-term effects of irradiation on implant wound healing. DPD hydrogel rescued radiation-induced fibrosis, restoring capsule thickness of healthy mice, and did not increase the fibroblast migration into the ADM. As a modulating soft tissue filler, DPD hydrogel also promoted adipocyte infiltration in healthy and irradiated mice. Detailed macrophage analysis showed that radiation led to the increase in pro-inflammatory, transition, and reparative markers. Despite relatively subtle effects on individual macrophage phenotype markers, multidimensional flow cytometry analysis revealed that DPD hydrogel temporally regulated two subpopulations. he presence of DPD resulted in significantly reduced CD9HiArg1HiCD301bLo and CD163HiCD38HiCD301bHi macrophages in healthy mice at one week, and a significant increase in CD9High macrophages with low expression of other markers at 6 weeks in irradiated mice. DPD hydrogel promotes a decreased fibrotic, and adipocyte-promoting coordination of wound healing in healthy and irradiated wound beds while not disrupting the immunomodulatory effects of ADM. STATEMENT OF SIGNIFICANCE: Acellular Dermal Matrix (ADM) provides mechanical and soft tissue support in post-mastectomy implant-based breast reconstruction, and positively affects wound healing. Following breast reconstruction, skin flap necrosis, edema, and previous radiation therapy can hinder ADM integration. Effective wound healing and biomaterial integration requires regulating the cellular immune response. Extracellular matrix hydrogels have demonstrated utility in tissue regeneration and decreasing inflammation and fibrosis in various tissues, but has yet to be utilized in the setting of breast reconstruction. Here, we demonstrated that a decellularized dermal hydrogel as an adjunct to ADM, decreases fibrosis and promotes adipogenesis during the coordination of wound healing in healthy and clinically relevant microenvironments that have received radiation therapy while not disrupting the immunomodulatory effects of implanted ADM.
Collapse
Affiliation(s)
- Lillian DeCostanza
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Graham M Grogan
- Department of Plastic Surgery, University of Virginia, Charlottesville, VA, USA
| | - Anthony C Bruce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Corrina M Peachey
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Evan A Clark
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Kristen Atkins
- Department of Pathology, University of Virginia, Charlottesville, VA, USA
| | - Tina Tylek
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Michael D Solga
- Flow Cytometry Core Facility, University of Virginia, Charlottesville, VA, USA
| | - Kara L Spiller
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Shayn M Peirce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | | | - Patrick S Cottler
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA; Department of Plastic Surgery, University of Virginia, Charlottesville, VA, USA; Department of Otolaryngology, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
11
|
Trimukhe AM, Melo JS, Chaturvedi D, Jain RD, Dandekar P, Deshmukh RR. RF pulsed plasma modified composite scaffold for enhanced anti-microbial activity and accelerated wound healing. Int J Pharm 2024; 667:124864. [PMID: 39461682 DOI: 10.1016/j.ijpharm.2024.124864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/15/2024] [Accepted: 10/20/2024] [Indexed: 10/29/2024]
Abstract
Infected wounds present significant challenges pertaining to healing and often demand administration of strong antibiotics to patients. Also, drug resistant microbes may alter the physiology of wounds to create biofilms, frequently leading to high morbidity and mortality. In this investigation, a biodegradable, microporous composite agarose-chitosan scaffold was fabricated. Furthermore, its surface was modified with diphenyldiselenide deposition, using low pressure pulsed plasma technology. The optimized plasma parameters, viz. 5ON/15OFF (ms) of plasma pulse rate and 80 min of treatment time resulted in scaffolds having enhanced anti-bacterial activity against gram positive microbes like Staphylococcus (S.) aureus and S. epidermidis. The scaffolds were non-toxic to skin cells, as confirmed by the MTT assay. Cell proliferation through plasma treated and untreated scaffolds was assessed by culturing primary human dermal fibroblasts (HdaF) and human keratinocytes (HaCaT) and visualizing via confocal microscopy. Moreover, in-vivo rat model confirmed accelerated wound healing with plasma treated scaffold (100 % on day 14), as compared to the untreated scaffold (100 % on day 16) when compared with over-the-counter (OTC) ointment Betadine (100 % on day 12).
Collapse
Affiliation(s)
- A M Trimukhe
- Department of Physics, Institute of Chemical Technology, Mumbai 400019, India
| | - J S Melo
- Enzyme Microbial Technology Section, Nuclear Agriculture and Biotechnology Division, Bhabha Atomic Research Centre, Mumbai 400085, India
| | - D Chaturvedi
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai 400019, India
| | - R D Jain
- Department of Biological Sciences and Biotechnology, Institute of Chemical Technology, Mumbai 400019, India
| | - P Dandekar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai 400019, India
| | - R R Deshmukh
- Department of Physics, Institute of Chemical Technology, Mumbai 400019, India.
| |
Collapse
|
12
|
Park HS, Song Y, Lee JH, Oh KR, Park H, Kang H. The role of exercise in promoting lymphangiogenesis and extracellular matrix synthesis in lymphedema-induced tissue injury. Mol Biol Rep 2024; 52:50. [PMID: 39676093 DOI: 10.1007/s11033-024-10149-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 12/03/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND Secondary lymphedema is a progressive condition caused by lipid- and protein-rich interstitial fluid accumulation resulting from compromised lymphatic function. It commonly occurs in cancer patients following surgical lymph node ablation and radiation treatment. This study aims to elucidate the effects of exercise on the myokine interleukin (IL)-6 and the molecular changes involved in lymphangiogenesis and extracellular matrix (ECM) synthesis using a lymphedema mouse model. METHODS AND RESULTS We induced lymphedema in male ICR mice by excising the inguinal, popliteal, and iliac lymph nodes and occluding the femoral lymphatic vessel in the right hind leg. We then conducted a time course analysis of swelling and targeted gene expression analysis of the affected leg tissue. We began the mice on involuntary wheel running exercise 2 days post-surgery. Over the subsequent 2 weeks, the swelling initially increased and then gradually declined. In contrast, the genes associated with lymphangiogenesis and ECM synthesis showed progressive upregulation, even after the swelling had subsided. Exercise reduced limb swelling and increased the genetic expression of IL-6 and key lymphangiogenesis markers, including Prox1, VEGF-C, VEGFR-3, and LYVE-1, in the affected limb. Exercise also increased the expression of FABP4 and αSMA, which are markers of active adipocytes and fibroblasts. CONCLUSIONS Our results suggest that exercise reduces lymphedema by promoting lymphangiogenesis and ECM synthesis, facilitating recovery. The insights obtained into the molecular changes underpinning exercise-induced improvements in lymphedema may contribute to the development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Hong Shik Park
- College of Physical Education, Kyung Hee University, Yongin, 17104, South Korea
| | - Youngju Song
- Department of Biomedical Science and Technology, Kyung Hee University, Seoul, 02447, South Korea
| | - Joon-Hee Lee
- College of Physical Education, Kyung Hee University, Yongin, 17104, South Korea
| | - Kyung-Rok Oh
- College of Physical Education, Kyung Hee University, Yongin, 17104, South Korea
| | - Hyon Park
- College of Physical Education, Kyung Hee University, Yongin, 17104, South Korea
| | - Hee Kang
- Humanitas College, Kyung Hee University, 1732 Deogyeongdae-Ro, Yongin, 17104, South Korea.
| |
Collapse
|
13
|
Sander EA, El-Hattab MY, Jacobson KR, Klingelhutz AJ, Ankrum JA, Calve S. Fibroblast-Adipocyte Lineage Cell Interactions Result in Differential Production of Extracellular Matrix Proteins. Cell Mol Bioeng 2024; 17:597-608. [PMID: 39926384 PMCID: PMC11799492 DOI: 10.1007/s12195-024-00829-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 10/08/2024] [Indexed: 02/11/2025] Open
Abstract
Introduction Scarring from traumatic injury, burns, and other complications remains a significant problem that diminishes quality of life for millions of people worldwide. A common target for the development of new therapies to promote healing and reduce scarring are myofibroblasts because of their central role in pathological scarring. Recent work indicates that adipocyte lineage cells also contribute to the wound healing process, including clinical reports that indicate that the placement of autologous adipose micrografts at the surgical site improves the appearance and pliability of existing scars. Methods To better understand how adipocyte lineage cells interact with fibroblasts to promote healing, we first utilized an in vitro model of wound healing to visualize fibroblast spheroid collagen deposition via time-lapse imaging. We then introduced pre-adipocyte and adipocyte spheroids to visualize pair-wise spheroid interactions and collagen deposition among all three cell types. Finally, we quantified differences in the extracellular matrix (ECM) proteins produced using liquid chromatography with tandem mass spectrometry (LC-MS/MS). Results We found that all three cell-types contribute to ECM deposition and that the composition of the ECM proteins, or matrisome, was significantly different depending on which cells were co-cultured together. Conclusions By better understanding the interactions among these cell types, novel adipose-tissue-based therapeutic approaches can be developed to improve wound healing and reduce scar tissue. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-024-00829-8.
Collapse
Affiliation(s)
- Edward A. Sander
- Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, 5629 Seamans Center, Iowa City, IA 52242 USA
| | - Mariam Y. El-Hattab
- Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, 5629 Seamans Center, Iowa City, IA 52242 USA
| | - Kathryn R. Jacobson
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, 1111 Engineering Dr, Boulder, CO USA
| | - Aloysius J. Klingelhutz
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA USA
| | - James A. Ankrum
- Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, 5629 Seamans Center, Iowa City, IA 52242 USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA USA
| | - Sarah Calve
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, 1111 Engineering Dr, Boulder, CO USA
| |
Collapse
|
14
|
Fujiwara H. Dynamic duo: Cell-extracellular matrix interactions in hair follicle development and regeneration. Dev Biol 2024; 516:20-34. [PMID: 39059679 DOI: 10.1016/j.ydbio.2024.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 06/20/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Ectodermal organs, such as hair follicles, originate from simple epithelial and mesenchymal sheets through a complex developmental process driven by interactions between these cell types. This process involves dermal condensation, placode formation, bud morphogenesis, and organogenesis, and all of these processes require intricate interactions among various tissues. Recent research has emphasized the crucial role of reciprocal and dynamic interactions between cells and the extracellular matrix (ECM), referred to as the "dynamic duo", in the development of ectodermal organs. These interactions provide spatially and temporally changing biophysical and biochemical cues within tissues. Using the hair follicle as an example, this review highlights two types of cell-ECM adhesion units-focal adhesion-type and hemidesmosome-type adhesion units-that facilitate communication between epithelial and mesenchymal cells. This review further explores how these adhesion units, along with other cell-ECM interactions, evolve during hair follicle development and regeneration, underscoring their importance in guiding both developmental and regenerative processes.
Collapse
|
15
|
Amuso VM, Haas MR, Cooper PO, Chatterjee R, Hafiz S, Salameh S, Gohel C, Mazumder MF, Josephson V, Kleb SS, Khorsandi K, Horvath A, Rahnavard A, Shook BA. Fibroblast-Mediated Macrophage Recruitment Supports Acute Wound Healing. J Invest Dermatol 2024:S0022-202X(24)02956-7. [PMID: 39581458 DOI: 10.1016/j.jid.2024.10.609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/26/2024]
Abstract
Epithelial and immune cells have long been appreciated for their contribution to the early immune response after injury; however, much less is known about the role of mesenchymal cells. Using single-nuclei RNA sequencing, we defined changes in gene expression associated with inflammation 1 day after wounding in mouse skin. Compared with those in keratinocytes and myeloid cells, we detected enriched expression of proinflammatory genes in fibroblasts associated with deeper layers of the skin. In particular, SCA1+ fibroblasts were enriched for numerous chemokines, including CCL2, CCL7, and IL-33, compared with SCA1- fibroblasts. Genetic deletion of Ccl2 in fibroblasts resulted in fewer wound-bed macrophages and monocytes during injury-induced inflammation, with reduced revascularization and re-epithelialization during the proliferation phase of healing. These findings highlight the important contribution of fibroblast-derived factors to injury-induced inflammation and the impact of immune cell dysregulation on subsequent tissue repair.
Collapse
Affiliation(s)
- Veronica M Amuso
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - MaryEllen R Haas
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Paula O Cooper
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Ranojoy Chatterjee
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, District of Columbia, USA
| | - Sana Hafiz
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Shatha Salameh
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Chiraag Gohel
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, District of Columbia, USA
| | - Miguel F Mazumder
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Violet Josephson
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Sarah S Kleb
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Khatereh Khorsandi
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Anelia Horvath
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Ali Rahnavard
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, District of Columbia, USA
| | - Brett A Shook
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA; The Department of Dermatology, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA.
| |
Collapse
|
16
|
Jin C, Zhao R, Hu W, Wu X, Zhou L, Shan L, Wu H. Topical hADSCs-HA Gel Promotes Skin Regeneration and Angiogenesis in Pressure Ulcers by Paracrine Activating PPARβ/δ Pathway. Drug Des Devel Ther 2024; 18:4799-4824. [PMID: 39478872 PMCID: PMC11523932 DOI: 10.2147/dddt.s474628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 10/09/2024] [Indexed: 11/02/2024] Open
Abstract
Background Pressure ulcer is common in the bedridden elderly with high mortality and lack of effective treatment. In this study, human-adipose-derived-stem-cells-hyaluronic acid gel (hADSCs-HA gel) was developed and applied topically to treat pressure ulcers, of which efficacy and paracrine mechanisms were investigated through in vivo and in vitro experiments. Methods Pressure ulcers were established on the backs of C57BL/6 mice and treated topically with hADSCs-HA gel, hADSCs, hyaluronic acid, and normal saline respectively. The rate of wound closure was observed continuously during the following 14 days and the wound samples were obtained for Western blot, histopathology, immunohistochemistry, and proteomic analysis. Human dermal fibroblasts (HDFs) and human venous endothelial cells (HUVECs) under normal or hypoxic conditions were treated with conditioned medium of human ADSCs (ADSC-CM), then CCK-8, scratch test, tube formation, and Western blot were conducted to evaluate the paracrine effects of hADSCs and to explore the underlying mechanism. Results The in vivo data demonstrated that hADSCs-HA gel significantly accelerated the healing of pressure ulcers by enhancing collagen expression, angiogenesis, and skin proliferation. The in vitro data revealed that hADSCs strengthened the proliferation and wound healing capabilities of HDFs and HUVECs, meanwhile promoted collagen secretion and tube formation through paracrine mode. ADSC-CM was also proved to exert protective effects on hypoxic HDFs and HUVECs. Besides, the results of proteomic analysis and Western blot elucidated that lipid metabolism and PPARβ/δ pathway mediated the healing effect of hADSCs-HA gel on pressure ulcers. Conclusion Our research showed that topical application of hADSCs-HA gel played an important role in dermal regeneration and angiogenesis. Therefore, hADSCs-HA gel exhibited the potential as a novel stem-cell-based therapeutic strategy of treating pressure ulcers in clinical practices.
Collapse
Affiliation(s)
- Chaoying Jin
- Department of Plastic and Aesthetic Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, People’s Republic of China
- School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310020, People’s Republic of China
| | - Ruolin Zhao
- Yichen Biotechnology Co., Ltd, Hangzhou, Zhejiang, 311200, People’s Republic of China
- Fuyang Academy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 311403, People’s Republic of China
| | - Weihang Hu
- Department of Critical Care Medicine, Zhejiang Hospital, Hangzhou, Zhejiang, 310013, People’s Republic of China
| | - Xiaolong Wu
- Fuyang Academy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 311403, People’s Republic of China
| | - Li Zhou
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310060, People’s Republic of China
| | - Letian Shan
- Fuyang Academy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 311403, People’s Republic of China
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310060, People’s Republic of China
| | - Huiling Wu
- Department of Plastic and Aesthetic Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, People’s Republic of China
- School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310020, People’s Republic of China
| |
Collapse
|
17
|
Karimi N, Ahmadi V. Aquaporin Channels in Skin Physiology and Aging Pathophysiology: Investigating Their Role in Skin Function and the Hallmarks of Aging. BIOLOGY 2024; 13:862. [PMID: 39596817 PMCID: PMC11592281 DOI: 10.3390/biology13110862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024]
Abstract
This study examines the critical role of aquaporins (AQPs) in skin physiology and aging pathophysiology. The skin plays a vital role in maintaining homeostasis by acting as a protective barrier against external pathogens and excessive water loss, while also contributing to the appearance and self-esteem of individuals. Key physiological features, such as elasticity and repair capability, are essential for its proper function. However, with aging, these characteristics deteriorate, reducing the skin's ability to tolerate environmental stressors which contribute to external aging as well as internal aging processes, which negatively affect barrier function, immune response, and overall well-being. AQPs, primarily known for facilitating water transport, are significant for normal skin functions, including hydration and the movement of molecules like glycerol and hydrogen peroxide, which influence various cellular processes and functions. In this context, we categorized aquaporin dysfunction into several hallmarks of aging, including mitochondrial dysfunction, cellular senescence, stem cell depletion, impaired macroautophagy, dysbiosis, and inflamm-aging. Eight aquaporins (AQP1, 3, 5, 7, 8, 9, 10, and 11) are expressed in various skin cells, regulating essential processes such as cell migration, proliferation, differentiation, and also immune response. Dysregulation or altered expression of these proteins can enhance skin aging and related pathologies by activating these hallmarks. This study provides valuable insights into the potential of targeting aquaporins to mitigate skin aging and improve skin physiologic functions.
Collapse
Affiliation(s)
- Nazli Karimi
- Physiology Department, Medical Faculty, Hacettepe University, Ankara 06800, Turkey
| | - Vahid Ahmadi
- Dermatology Department, Beytepe Murat Erdi Eker State Hospital, Ankara 06800, Turkey
| |
Collapse
|
18
|
Ridha Z, Fabi SG, Zubar R, Dayan SH. Decoding the Implications of Glucagon-like Peptide-1 Receptor Agonists on Accelerated Facial and Skin Aging. Aesthet Surg J 2024; 44:NP809-NP818. [PMID: 38874170 DOI: 10.1093/asj/sjae132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/04/2024] [Accepted: 06/12/2024] [Indexed: 06/15/2024] Open
Abstract
Following the advent of glucagon-like peptide-1 receptor agonists (GLP-1RAs), subsequent unintended effects such as accelerated facial aging and altered skin health have been noted. This review delves deeper into the causative underlying mechanisms and provides insights into the intricate relationship between GLP-1RAs, adipose tissue, and premature facial aging, thereby highlighting the need for a nuanced understanding of their effects on facial alterations and skin health. Studies exploring the potential effects of GLP-1RAs on facial alterations and offering insights into the possible underlying mechanisms, causes, and clinical implications were included. The accelerated facial aging and altered skin health observed in GLP-1RA patients appears to be multifactorial, involving loss of dermal and subcutaneous white adipose tissue, and altered proliferation and differentiation of adipose-derived stem cells (ADSCs), and impacts on the production and secretion of hormonal and metabolic factors. These changes compromise the structural integrity and barrier function of the skin and may lead to diminished facial muscle mass, further exacerbating the appearance of aging. The insights presented call for a paradigm shift in the clinical management of facial changes induced by GLP-1RAs, with a focus on treatment strategies aimed at targeting ADSC stimulation. These include autologous fat transfers to reintroduce cells rich in ADSCs for rejuvenation, composite fat grafting combining autologous fat with/without stromal vascular fraction, and the strategic use of soft tissue fillers for volume restoration and biostimulation. This review highlights the potential role of GLP-1RAs in modulating adipose tissue dynamics, thereby contributing to accelerated aging through metabolic, structural, and hormonal pathways. LEVEL OF EVIDENCE: 5
Collapse
|
19
|
Stojanovic M, Rai V, Agrawal DK. Effect of Electromagnetic Field on Proliferation and Migration of Fibroblasts and Keratinocytes: Implications in Wound Healing and Regeneration. JOURNAL OF BIOTECHNOLOGY AND BIOMEDICINE 2024; 7:387-399. [PMID: 39364330 PMCID: PMC11448841 DOI: 10.26502/jbb.2642-91280162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
Proliferation and migration of fibroblasts, keratinocytes, and endothelial cells are key events in the physiological process of wound healing. This process includes different but overlapping stages: hemostasis, inflammatory phase, the proliferative phase, and the remodeling phase. Traumatic brain injury (TBI) is defined as a mechanical insult to the brain from external mechanical force (primary injury), usually followed by the secondary injury including edema, inflammation, excitotoxicity, oxidative stress, or mitochondrial dysfunction. The process of tissue repair following TBI is based on the neuronal-glial interactions, where phagocytosis by microglia plays a crucial role. Low-frequency electromagnetic field (LF-EMF) has been shown to enhance tissue repair after TBI, however, there are limited studies investigating the effects of LF-EMF on the proliferation and migration of keratinocytes, fibroblasts, VSMCs, and endothelial cells in the context of wound healing and on neuronal cells and microglia in relation to healing after TBI. Better understanding of the effects of LF-EMF on the proliferation, migration, and differentiation of these cells is important to enhance tissue healing after injury. This review article comprehensively discussed the effect of EMF/LF-EMF on these cells. Results published by different authors are hardly comparable due to different methodological approach and experimental settings. EMF promotes migration and proliferation of fibroblasts, keratinocytes and endothelial cells (EC), and thus could improve wound healing. The pilot study preformed on a large animal model of TBI suggests anti-inflammatory effects of EMF stimulation following TBI. Therefore, EMF is recognized as a potential therapeutic option to accelerate the wound healing and improve cellular recovery and function after TBI. Nonetheless, future studies are needed to define the optimal parameters of EMF stimulation in terms of frequency or duration of exposure.
Collapse
Affiliation(s)
- Marija Stojanovic
- Department of Translational Research, Western University of Health Sciences, Pomona, California 91766, USA
- Institute of Medical Physiology "Richard Burian", Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Vikrant Rai
- Department of Translational Research, Western University of Health Sciences, Pomona, California 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, Pomona, California 91766, USA
| |
Collapse
|
20
|
Zhu B, Liang L, Hui L, Lu Y. Exploring the role of dermal sheath cells in wound healing and fibrosis. Wound Repair Regen 2024; 32:735-745. [PMID: 39129718 DOI: 10.1111/wrr.13206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 08/13/2024]
Abstract
Wound healing is a complex, dynamic process involving the coordinated interaction of diverse cell types, growth factors, cytokines, and extracellular matrix components. Despite emerging evidence highlighting their importance, dermal sheath cells remain a largely overlooked aspect of wound healing research. This review explores the multifunctional roles of dermal sheath cells in various phases of wound healing, including modulating inflammation, aiding in proliferation, and contributing to extracellular matrix remodelling. Special attention is devoted to the paracrine effects of dermal sheath cells and their role in fibrosis, highlighting their potential in improving healing outcomes, especially in differentiating between hairy and non-hairy skin sites. By drawing connections between dermal sheath cells activity and wound healing outcomes, this work proposes new insights into the mechanisms of tissue regeneration and repair, marking a step forward in our understanding of wound healing processes.
Collapse
Affiliation(s)
- Bing Zhu
- Translational Medicine Engineering Research Center of Inner Mongolia Autonomous Region, affiliated with Baotou Central Hospital, Baotou, China
| | - Lu Liang
- Translational Medicine Engineering Research Center of Inner Mongolia Autonomous Region, affiliated with Baotou Central Hospital, Baotou, China
| | - Lihua Hui
- Burn Research Institute of Inner Mongolia Autonomous Region, affiliated with Inner Mongolia Baogang Hospital, Baotou, China
| | - Yaojun Lu
- Translational Medicine Engineering Research Center of Inner Mongolia Autonomous Region, affiliated with Baotou Central Hospital, Baotou, China
| |
Collapse
|
21
|
Samarawickrama PN, Zhang G, Zhu E, Dong X, Nisar A, Zhu H, Ma Y, Zhou Z, Yang H, Gui L, Cao M, Li W, Chang Y, Zi M, Cui H, Duan Z, Zhang X, Li W, He Y. Clearance of senescent cells enhances skin wound healing in type 2 diabetic mice. Theranostics 2024; 14:5429-5442. [PMID: 39310100 PMCID: PMC11413796 DOI: 10.7150/thno.100991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 08/16/2024] [Indexed: 09/25/2024] Open
Abstract
Background: Diabetic foot ulcers (DFUs) pose a substantial healthcare challenge due to their high rates of morbidity, recurrence, disability, and mortality. Current DFU therapeutics continue to grapple with multiple limitations. Senescent cells (SnCs) have been found to have a beneficial effect on acute wound healing, however, their roles in chronic wounds, such as DFU, remain unclear. Methods and results: We collected skin, fat, and muscle samples from clinical patients with DFU and lower limb fractures. RNA-sequencing combined with qPCR analyses on these samples demonstrate a significant accumulation of SnCs at DFU, as indicated by higher senescence markers (e.g., p16 and p21) and a senescence-associated secretory phenotype (SASP). We constructed a type 2 diabetic model of db/db mice, fed with a high-fat diet (Db-HFD), which were wounded using a 6 mm punch to the dorsal skin. HFD slightly affected wound healing in wild-type (WT) mice, but high glucose significantly delayed wound healing in the Db-HFD mice. We injected the mice with a previously developed fluorescent probe (XZ1208), which allows the detection of SnCs in vivo, and observed a strong senescence signal at the wound site of the Db-HFD mice. Contrary to the beneficial effects of SnCs in acute wound healing, our results demonstrated that clearance of SnCs using the senolytic compound ABT263 significantly accelerated wound healing in Db-HFD mice. Conclusion: Collectively, these findings suggest that SnCs critically accumulate at wound sites, delaying the healing process in DFUs. Thus, targeting SnCs with senolytic therapy represents a promising approach for DFU treatment, potentially improving the quality of life for patients with DFUs.
Collapse
Affiliation(s)
- Priyadarshani Nadeeshika Samarawickrama
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guiqin Zhang
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China
| | - Enfang Zhu
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China
| | - Xin Dong
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Ayesha Nisar
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hong Zhu
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China
| | - Yuan Ma
- Department of Orthopedics, the Third People's Hospital of Yunnan Province, Kunming, Yunnan 650011, China
| | - Zheyan Zhou
- Department of Pathology, the Third People's Hospital of Yunnan Province, Kunming, Yunnan 650011, China
| | - Honglin Yang
- Department of Orthopedics, the Third People's Hospital of Yunnan Province, Kunming, Yunnan 650011, China
| | - Li Gui
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China
| | - Mei Cao
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China
| | - Wei Li
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China
| | - Yu Chang
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China
| | - Meiting Zi
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Haoling Cui
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China
| | - Zhongping Duan
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China
| | - Xuan Zhang
- Drug Discovery & Development Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Wen Li
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China
| | - Yonghan He
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
22
|
Riabinin A, Pankratova M, Rogovaya O, Vorotelyak E, Terskikh V, Vasiliev A. Ideal Living Skin Equivalents, From Old Technologies and Models to Advanced Ones: The Prospects for an Integrated Approach. BIOMED RESEARCH INTERNATIONAL 2024; 2024:9947692. [PMID: 39184355 PMCID: PMC11343635 DOI: 10.1155/2024/9947692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 04/18/2024] [Accepted: 07/20/2024] [Indexed: 08/27/2024]
Abstract
The development of technologies for the generation and transplantation of living skin equivalents (LSEs) is a significant area of translational medicine. Such functional equivalents can be used to model and study the morphogenesis of the skin and its derivatives, to test drugs, and to improve the healing of chronic wounds, burns, and other skin injuries. The evolution of LSEs over the past 50 years has demonstrated the leap in technology and quality and the shift from classical full-thickness LSEs to principled new models, including modification of classical models and skin organoids with skin derived from human-induced pluripotent stem cells (iPSCs) (hiPSCs). Modern methods and approaches make it possible to create LSEs that successfully mimic native skin, including derivatives such as hair follicles (HFs), sebaceous and sweat glands, blood vessels, melanocytes, and nerve cells. New technologies such as 3D and 4D bioprinting, microfluidic systems, and genetic modification enable achievement of new goals, cost reductions, and the scaled-up production of LSEs.
Collapse
Affiliation(s)
- Andrei Riabinin
- Department of Cell BiologyKoltzov Institute of Developmental Biology of the Russian Academy of Sciences, Moscow, Russia
| | - Maria Pankratova
- Department of Cell BiologyKoltzov Institute of Developmental Biology of the Russian Academy of Sciences, Moscow, Russia
| | - Olga Rogovaya
- Department of Cell BiologyKoltzov Institute of Developmental Biology of the Russian Academy of Sciences, Moscow, Russia
| | - Ekaterina Vorotelyak
- Department of Cell BiologyKoltzov Institute of Developmental Biology of the Russian Academy of Sciences, Moscow, Russia
| | - Vasiliy Terskikh
- Department of Cell BiologyKoltzov Institute of Developmental Biology of the Russian Academy of Sciences, Moscow, Russia
| | - Andrey Vasiliev
- Department of Cell BiologyKoltzov Institute of Developmental Biology of the Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
23
|
Amuso VM, Haas MR, Cooper PO, Chatterjee R, Hafiz S, Salameh S, Gohel C, Mazumder MF, Josephson V, Khorsandi K, Horvath A, Rahnavard A, Shook BA. Deep skin fibroblast-mediated macrophage recruitment supports acute wound healing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.09.607357. [PMID: 39149286 PMCID: PMC11326280 DOI: 10.1101/2024.08.09.607357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Epithelial and immune cells have long been appreciated for their contribution to the early immune response after injury; however, much less is known about the role of mesenchymal cells. Using single nuclei RNA-sequencing, we defined changes in gene expression associated with inflammation at 1-day post-wounding (dpw) in mouse skin. Compared to keratinocytes and myeloid cells, we detected enriched expression of pro-inflammatory genes in fibroblasts associated with deeper layers of the skin. In particular, SCA1+ fibroblasts were enriched for numerous chemokines, including CCL2, CCL7, and IL33 compared to SCA1- fibroblasts. Genetic deletion of Ccl2 in fibroblasts resulted in fewer wound bed macrophages and monocytes during injury-induced inflammation with reduced revascularization and re-epithelialization during the proliferation phase of healing. These findings highlight the important contribution of deep skin fibroblast-derived factors to injury-induced inflammation and the impact of immune cell dysregulation on subsequent tissue repair.
Collapse
Affiliation(s)
- Veronica M. Amuso
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - MaryEllen R. Haas
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Paula O. Cooper
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Ranojoy Chatterjee
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC 20052, USA
| | - Sana Hafiz
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Shatha Salameh
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Chiraag Gohel
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC 20052, USA
| | - Miguel F. Mazumder
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Violet Josephson
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Khatereh Khorsandi
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Anelia Horvath
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Ali Rahnavard
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC 20052, USA
| | - Brett A. Shook
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
- Department of Dermatology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| |
Collapse
|
24
|
Peña OA, Martin P. Cellular and molecular mechanisms of skin wound healing. Nat Rev Mol Cell Biol 2024; 25:599-616. [PMID: 38528155 DOI: 10.1038/s41580-024-00715-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2024] [Indexed: 03/27/2024]
Abstract
Wound healing is a complex process that involves the coordinated actions of many different tissues and cell lineages. It requires tight orchestration of cell migration, proliferation, matrix deposition and remodelling, alongside inflammation and angiogenesis. Whereas small skin wounds heal in days, larger injuries resulting from trauma, acute illness or major surgery can take several weeks to heal, generally leaving behind a fibrotic scar that can impact tissue function. Development of therapeutics to prevent scarring and successfully repair chronic wounds requires a fuller knowledge of the cellular and molecular mechanisms driving wound healing. In this Review, we discuss the current understanding of the different phases of wound healing, from clot formation through re-epithelialization, angiogenesis and subsequent scar deposition. We highlight the contribution of different cell types to skin repair, with emphasis on how both innate and adaptive immune cells in the wound inflammatory response influence classically studied wound cell lineages, including keratinocytes, fibroblasts and endothelial cells, but also some of the less-studied cell lineages such as adipocytes, melanocytes and cutaneous nerves. Finally, we discuss newer approaches and research directions that have the potential to further our understanding of the mechanisms underpinning tissue repair.
Collapse
Affiliation(s)
- Oscar A Peña
- School of Biochemistry, University of Bristol, Bristol, UK.
| | - Paul Martin
- School of Biochemistry, University of Bristol, Bristol, UK.
| |
Collapse
|
25
|
Boschi F, Negri A, Conti A, Bernardi P, Chirumbolo S, Sbarbati A. The human dermal white adipose tissue (dWAT) morphology: A multimodal imaging approach. Ann Anat 2024; 255:152289. [PMID: 38848928 DOI: 10.1016/j.aanat.2024.152289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 05/02/2024] [Accepted: 05/27/2024] [Indexed: 06/09/2024]
Abstract
BACKGROUND Dermal white adipose tissue (dWAT) in humans can be characterized as a relaxed dermal skin compartment consisting of functionally interlinked adipocytes. dWAT is typically discerned both in terms of morphology and function from subcutaneous white adipose tissue (sWAT). In particular in human thigh, the dWAT appears as thin extensions from the adipose panniculus to the dermis, and it is primarily associated with pilosebaceous units, hair follicles, sebaceous glands, and erector pili muscles. In this work, human fat tissue samples obtained post-mortem from the gluteo-femoral region were analyzed focusing on the thin extensions of dWAT named dermal cones. This anatomical region was chosen to deepen the dWAT morphological features of this site which is interesting both for clinical applications and genetical studies. The purpose of this exploratory methodological study was to gain deeper insights into the morphological features of human dWAT through a multimodal imaging approach. METHODS Optical microscopy, Magnetic Resonance Imaging (MRI) and Scanning Electron Microscopy (SEM), have been employed in this study. The cones' length and their distances were measured on the acquired images for optical microscopy and SEM. The cone's apparent regular distribution in MRI images was evaluated using a mathematical criterion, the conformity ratio, which is the ratio of the mean nearest-neighbor distance to its standard deviation. RESULTS The imaging techniques revealed white adipocytes forming a layer, referred to as sWAT, with cones measuring nearly 2 mm in size measured on SEM and Optical images (2.1 ± 0.4 mm), with the lower part embedded in the sWAT and the upper part extending into the dermis. The distance between the cones results about 1 mm measured on MRI images and they show an overall semiregular distribution. CONCLUSIONS MRI images demonstrated an orderly arrangement of cones, and their 3D reconstruction allowed to elucidate the dermal cones' disposition in the tissue sample and a more general comprehensive visualization of the entire fat structure within the dermis.
Collapse
Affiliation(s)
- Federico Boschi
- Department of Engineering for Innovation Medicine, Strada Le Grazie 8, Verona 37134, Italy.
| | - Alessandro Negri
- Department of Neurosciences, Biomedicine and Movement Sciences, Strada Le Grazie 8, Verona 37134, Italy
| | - Anita Conti
- Department of Neurosciences, Biomedicine and Movement Sciences, Strada Le Grazie 8, Verona 37134, Italy
| | - Paolo Bernardi
- Department of Neurosciences, Biomedicine and Movement Sciences, Strada Le Grazie 8, Verona 37134, Italy
| | - Salvatore Chirumbolo
- Department of Engineering for Innovation Medicine, Strada Le Grazie 8, Verona 37134, Italy
| | - Andrea Sbarbati
- Department of Neurosciences, Biomedicine and Movement Sciences, Strada Le Grazie 8, Verona 37134, Italy
| |
Collapse
|
26
|
Wareing N, Mills TW, Collum S, Wu M, Revercomb L, Girard R, Lyons M, Skaug B, Bi W, Ali MA, Koochak H, Flores AR, Yang Y, Zheng WJ, Swindell WR, Assassi S, Karmouty-Quintana H. Deletion of adipocyte Sine Oculis Homeobox Homolog 1 prevents lipolysis and attenuates skin fibrosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.22.595271. [PMID: 38826482 PMCID: PMC11142148 DOI: 10.1101/2024.05.22.595271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Dermal fibrosis is a cardinal feature of systemic sclerosis (SSc) for which there are limited treatment strategies. This is in part due to our fragmented understanding of how dermal white adipose tissue (DWAT) contributes to skin fibrosis. We identified elevated sine oculis homeobox homolog 1 (SIX1) expression in SSc skin samples from the GENISOS and PRESS cohorts, the expression of which correlated with adipose-associated genes and molecular pathways. SIX1 localization studies identified increased signals in the DWAT area in SSc and in experimental models of skin fibrosis. Global and adipocyte specific Six1 deletion abrogated end-stage fibrotic gene expression and dermal adipocyte shrinkage induced by SQ bleomycin treatment. Further studies revealed a link between elevated SIX1 and increased expression of SERPINE1 and its protein PAI-1 which are known pro-fibrotic mediators. However, SIX1 deletion did not appear to affect cellular trans differentiation. Taken together these results point at SIX1 as a potential target for dermal fibrosis in SSc.
Collapse
Affiliation(s)
- Nancy Wareing
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth Houston), TX, USA
- Division of Rheumatology, Department of Internal Medicine, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | - Tingting W Mills
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth Houston), TX, USA
| | - Scott Collum
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth Houston), TX, USA
| | - Minghua Wu
- Division of Rheumatology, Department of Internal Medicine, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | | | - Rene Girard
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth Houston), TX, USA
| | - Marka Lyons
- Division of Rheumatology, Department of Internal Medicine, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | - Brian Skaug
- Division of Rheumatology, Department of Internal Medicine, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | - Weizhen Bi
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth Houston), TX, USA
| | - Meer A. Ali
- D Bradley McWilliams School of Biomedical Informatics, Department of Internal Medicine, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | - Haniyeh Koochak
- Department of Pediatrics, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | - Anthony R Flores
- Department of Pediatrics, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | - Yuntao Yang
- D Bradley McWilliams School of Biomedical Informatics, Department of Internal Medicine, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | - W Jim Zheng
- D Bradley McWilliams School of Biomedical Informatics, Department of Internal Medicine, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | - William R Swindell
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| | - Shervin Assassi
- Division of Rheumatology, Department of Internal Medicine, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | - Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth Houston), TX, USA
- Divisions of Critical Care, Pulmonary and Sleep Medicine, Department of Internal Medicine, McGovern Medical School, UTHealth Houston, Houston TX, USA
| |
Collapse
|
27
|
Cooper PO, Kleb SS, Noonepalle SK, Amuso VM, Varshney R, Rudolph MC, Dhaliwal TK, Nguyen DV, Mazumder MF, Babirye NS, Gupta R, Nguyen BN, Shook BA. G-protein-coupled receptor 84 regulates acute inflammation in normal and diabetic skin wounds. Cell Rep 2024; 43:114288. [PMID: 38814782 PMCID: PMC11247419 DOI: 10.1016/j.celrep.2024.114288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 03/18/2024] [Accepted: 05/13/2024] [Indexed: 06/01/2024] Open
Abstract
Lipids have emerged as potent regulators of immune cell function. In the skin, adipocyte lipolysis increases the local pool of free fatty acids and is essential for coordinating early macrophage inflammation following injury. Here, we investigate G-protein-coupled receptor 84 (GPR84), a medium-chain fatty acid (MCFA) receptor, for its potential to propagate pro-inflammatory signaling after skin injury. GPR84 signaling was identified as a key component of regulating myeloid cell numbers and subsequent tissue repair through in vivo administration of a pharmacological antagonist and the MCFA decanoic acid. We found that impaired injury-induced dermal adipocyte lipolysis is a hallmark of diabetes, and lipidomic analysis demonstrated that MCFAs are significantly reduced in diabetic murine wounds. Furthermore, local administration of decanoic acid rescued myeloid cell numbers and tissue repair during diabetic wound healing. Thus, GPR84 is a readily targetable lipid signaling pathway for manipulating injury-induced tissue inflammation with beneficial effects on acute diabetic healing.
Collapse
Affiliation(s)
- Paula O Cooper
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Sarah S Kleb
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Satish K Noonepalle
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Veronica M Amuso
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Rohan Varshney
- Department of Biochemistry and Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Michael C Rudolph
- Department of Biochemistry and Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Tanvir K Dhaliwal
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Darlene V Nguyen
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Miguel F Mazumder
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Najuma S Babirye
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Ruchi Gupta
- Department of Surgery, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Bao-Ngoc Nguyen
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA; Department of Surgery, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Brett A Shook
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA; Department of Dermatology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA.
| |
Collapse
|
28
|
Liu M, Lu F, Feng J. Aging and homeostasis of the hypodermis in the age-related deterioration of skin function. Cell Death Dis 2024; 15:443. [PMID: 38914551 PMCID: PMC11196735 DOI: 10.1038/s41419-024-06818-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 02/01/2024] [Accepted: 06/10/2024] [Indexed: 06/26/2024]
Abstract
Adipose tissues in the hypodermis, the crucial stem cell reservoir in the skin and the endocrine organ for the maintenance of skin homeostasis undergo significant changes during skin aging. Dermal white adipose tissue (dWAT) has recently been recognized as an important organ for both non-metabolic and metabolic health in skin regeneration and rejuvenation. Defective differentiation, adipogenesis, improper adipocytokine production, and immunological dissonance dysfunction in dWAT lead to age-associated clinical changes. Here, we review age-related alterations in dWAT across levels, emphasizing the mechanisms underlying the regulation of aging. We also discuss the pathogenic changes involved in age-related fat dysfunction and the unfavorable consequences of accelerated skin aging, such as chronic inflammaging, immunosenescence, delayed wound healing, and fibrosis. Research has shown that adipose aging is an early initiation event and a potential target for extending longevity. We believe that adipose tissues play an essential role in aging and form a potential therapeutic target for the treatment of age-related skin diseases. Further research is needed to improve our understanding of this phenomenon.
Collapse
Affiliation(s)
- Meiqi Liu
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Feng Lu
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Jingwei Feng
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, People's Republic of China.
| |
Collapse
|
29
|
Li J, Liu Y, Zhang R, Yang Q, Xiong W, He Y, Ye Q. Insights into the role of mesenchymal stem cells in cutaneous medical aesthetics: from basics to clinics. Stem Cell Res Ther 2024; 15:169. [PMID: 38886773 PMCID: PMC11184751 DOI: 10.1186/s13287-024-03774-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 05/27/2024] [Indexed: 06/20/2024] Open
Abstract
With the development of the economy and the increasing prevalence of skin problems, cutaneous medical aesthetics are gaining more and more attention. Skin disorders like poor wound healing, aging, and pigmentation have an impact not only on appearance but also on patients with physical and psychological issues, and even impose a significant financial burden on families and society. However, due to the complexities of its occurrence, present treatment options cannot produce optimal outcomes, indicating a dire need for new and effective treatments. Mesenchymal stem cells (MSCs) and their secretomics treatment is a new regenerative medicine therapy that promotes and regulates endogenous stem cell populations and/or replenishes cell pools to achieve tissue homeostasis and regeneration. It has demonstrated remarkable advantages in several skin-related in vivo and in vitro investigations, aiding in the improvement of skin conditions and the promotion of skin aesthetics. As a result, this review gives a complete description of recent scientific breakthroughs in MSCs for skin aesthetics and the limitations of their clinical applications, aiming to provide new ideas for future research and clinical transformation.
Collapse
Affiliation(s)
- Junyi Li
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ye Liu
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Rui Zhang
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qianyu Yang
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wei Xiong
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Yan He
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, 430030, China.
| | - Qingsong Ye
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
30
|
Yin Y, Wang S, Xie D, Pan S, Fu H, Feng Z, Gao C, Ge X. Hyperbaric oxygen therapy promotes the browning of white fat and contributes to the healing of diabetic wounds. Int Wound J 2024; 21:e14867. [PMID: 38597295 PMCID: PMC11005105 DOI: 10.1111/iwj.14867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/11/2024] Open
Abstract
Non-healing wounds are one of the chronic complications of diabetes and have remained a worldwide challenge as one of the major health problems. Hyperbaric oxygen (HBO) therapy is proven to be very successful for diabetic wound treatment, for which the molecular basis is not understood. Adipocytes regulate multiple aspects of repair and may be therapeutic for inflammatory diseases and defective wound healing associated with aging and diabetes. Endothelial cell-derived extracellular vesicles could promote wound healing in diabetes. To study the mechanism by which HBO promotes wound healing in diabetes, we investigated the effect of HBO on fat cells in diabetic mice. A diabetic wound mouse model was established and treated with HBO. Haematoxylin and eosin (H&E) staining and immunofluorescence were used for the analysis of wound healing. To further explore the mechanism, we performed whole-genome sequencing on extracellular vesicles (EVs). Furthermore, we conducted in vitro experiments. Specifically, exosomes were collected from human umbilical vein endothelial cell (HUVEC) cells after HBO treatment, and then these exosomes were co-incubated with adipose tissue. The wound healing rate in diabetic mice treated with HBO was significantly higher. HBO therapy promotes the proliferation of adipose precursor cells. HUVEC-derived exosomes treated with HBO significantly promoted fat cell browning. These data clarify that HBO therapy may promote vascular endothelial cell proliferation and migration, and promote browning of fat cells through vascular endothelial cells derived exosomes, thereby promoting diabetic wound healing. This provides new ideas for the application of HBO therapy in the treatment of diabetic trauma.
Collapse
Affiliation(s)
- Yue Yin
- Department of Emergency MedicineXinhua Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shang‐Yuan Wang
- Department of Emergency MedicineXinhua Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Di Xie
- Department of Emergency MedicineXinhua Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shu‐Ming Pan
- Department of Emergency MedicineXinhua Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Hui‐min Fu
- Department of Emergency MedicineXinhua Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zhi‐hui Feng
- Department of Emergency MedicineXinhua Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Cheng‐Jin Gao
- Department of Emergency MedicineXinhua Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiao‐Li Ge
- Department of Emergency MedicineXinhua Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
31
|
Ninkovic N, Sparks HD, Ponjevic D, Muench G, Biernaskie JA, Krawetz RJ. Proteoglycan 4 (PRG4) treatment improves skin wound healing in a porcine model. FASEB J 2024; 38:e23547. [PMID: 38498368 DOI: 10.1096/fj.202301289rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 02/08/2024] [Accepted: 02/26/2024] [Indexed: 03/20/2024]
Abstract
Proteoglycan 4 (PRG4) is a boundary lubricant originally identified in articular cartilage and has been since shown to have immunomodulation and antifibrotic properties. Previously, we have demonstrated that recombinant human (rh)PRG4 treatment accelerates auricular cartilage injury closure through an inhibition of the fibrotic response, and promotion of tissue regeneration in mice. The purpose of the current study was to examine the effects of rhPRG4 treatment (vs. a DMSO carried control) on full-thickness skin wound healing in a preclinical porcine model. Our findings suggest that while rhPRG4 did not significantly accelerate nor impede full-thickness skin wound closure, it did improve repair quality by decreasing molecular markers of fibrosis and increasing re-vascularization. We also demonstrated that rhPRG4 treatment increased dermal adipose tissue during the healing process specifically by retaining adipocytes in the wound area but did not inhibit lipolysis. Overall, the results of the current study have demonstrated that rhPRG4 acts as antifibrotic agent and regulates dermal adipose tissue during the healing processes resulting in a tissue with a trajectory that more resembles the native skin vs. a fibrotic patch. This study provides strong rationale to examine if rhPRG4 can improve regeneration in human wounds.
Collapse
Affiliation(s)
- Nicoletta Ninkovic
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada
- McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, Alberta, Canada
| | - Holly D Sparks
- McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, Alberta, Canada
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Dragana Ponjevic
- McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, Alberta, Canada
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Greg Muench
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jeff A Biernaskie
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Roman J Krawetz
- McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, Alberta, Canada
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Biomedical Engineering, University of Calgary, Calgary, Alberta, Canada
- Department Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
32
|
Jussila A, Zhang B, Kirti S, Atit R. Tissue fibrosis associated depletion of lipid-filled cells. Exp Dermatol 2024; 33:e15054. [PMID: 38519432 PMCID: PMC10977660 DOI: 10.1111/exd.15054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 02/06/2024] [Accepted: 02/29/2024] [Indexed: 03/24/2024]
Abstract
Fibrosis is primarily described as the deposition of excessive extracellular matrix, but in many tissues it also involves a loss of lipid or lipid-filled cells. Lipid-filled cells are critical to tissue function and integrity in many tissues including the skin and lungs. Thus, loss or depletion of lipid-filled cells during fibrogenesis, has implications for tissue function. In some contexts, lipid-filled cells can impact ECM composition and stability, highlighting their importance in fibrotic transformation. Recent papers in fibrosis address this newly recognized fibrotic lipodystrophy phenomenon. Even in disparate tissues, common mechanisms are emerging to explain fibrotic lipodystrophy. These findings have implications for fibrosis in tissues composed of fibroblast and lipid-filled cell populations such as skin, lung, and liver. In this review, we will discuss the roles of lipid-containing cells, their reduction/loss during fibrotic transformation, and the mechanisms of that loss in the skin and lungs.
Collapse
Affiliation(s)
- Anna Jussila
- Department of Biology, College of Arts and Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Brian Zhang
- Department of Biology, College of Arts and Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Sakin Kirti
- Department of Biology, College of Arts and Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Radhika Atit
- Department of Biology, College of Arts and Sciences, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Dermatology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
33
|
Li Y, Long J, Zhang Z, Yin W. Insights into the unique roles of dermal white adipose tissue (dWAT) in wound healing. Front Physiol 2024; 15:1346612. [PMID: 38465261 PMCID: PMC10920283 DOI: 10.3389/fphys.2024.1346612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 01/31/2024] [Indexed: 03/12/2024] Open
Abstract
Dermal white adipose tissue (dWAT) is a newly recognized layer of adipocytes within the reticular dermis of the skin. In many mammals, this layer is clearly separated by panniculus carnosus from subcutaneous adipose tissue (sWAT). While, they concentrated around the hair shaft and follicle, sebaceous gland, and arrector pili muscle, and forms a very specific cone geometry in human. Both the anatomy and the histology indicate that dWAT has distinct development and functions. Different from sWAT, the developmental origin of dWAT shares a common precursor with dermal fibroblasts during embryogenesis. Therefore, when skin injury happens and mature adipocytes in dWAT are exposed, they may undergo lipolysis and dedifferentiate into fibroblasts to participate in wound healing as embryogenetic stage. Studies using genetic strategies to selectively ablate dermal adipocytes observed delayed revascularization and re-epithelialization in wound healing. This review specifically summarizes the hypotheses of the functions of dWAT in wound healing. First, lipolysis of dermal adipocytes could contribute to wound healing by regulating inflammatory macrophage infiltration. Second, loss of dermal adipocytes occurs at the wound edge, and adipocyte-derived cells then become ECM-producing wound bed myofibroblasts during the proliferative phase of repair. Third, mature dermal adipocytes are rich resources for adipokines and cytokines and could release them in response to injury. In addition, the dedifferentiated dermal adipocytes are more sensitive to redifferentiation protocol and could undergo expansion in infected wound. We then briefly introduce the roles of dWAT in protecting the skin from environmental challenges: production of an antimicrobial peptide against infection. In the future, we believe there may be great potential for research in these areas: (1) taking advantage of the plasticity of dermal adipocytes and manipulating them in wound healing; (2) investigating the precise mechanism of dWAT expansion in infected wound healing.
Collapse
Affiliation(s)
| | | | | | - Wen Yin
- *Correspondence: Ziang Zhang, ; Wen Yin,
| |
Collapse
|
34
|
Kita A, Yamamoto S, Saito Y, Chikenji TS. Cellular senescence and wound healing in aged and diabetic skin. Front Physiol 2024; 15:1344116. [PMID: 38440347 PMCID: PMC10909996 DOI: 10.3389/fphys.2024.1344116] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 02/05/2024] [Indexed: 03/06/2024] Open
Abstract
Cellular senescence is a biological mechanism that prevents abnormal cell proliferation during tissue repair, and it is often accompanied by the secretion of various factors, such as cytokines and chemokines, known as the senescence-associated secretory phenotype (SASP). SASP-mediated cell-to-cell communication promotes tissue repair, regeneration, and development. However, senescent cells can accumulate abnormally at injury sites, leading to excessive inflammation, tissue dysfunction, and intractable wounds. The effects of cellular senescence on skin wound healing can be both beneficial and detrimental, depending on the condition. Here, we reviewed the functional differences in cellular senescence that emerge during wound healing, chronic inflammation, and skin aging. We also review the latest mechanisms of wound healing in the epidermis, dermis, and subcutaneous fat, with a focus on cellular senescence, chronic inflammation, and tissue regeneration. Finally, we discuss the potential clinical applications of promoting and inhibiting cellular senescence to maximize benefits and minimize detrimental effects.
Collapse
Affiliation(s)
- Arisa Kita
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Plastic and Reconstructive Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Sena Yamamoto
- Graduate School of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Yuki Saito
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan
| | | |
Collapse
|
35
|
Cavallo I, Sivori F, Mastrofrancesco A, Abril E, Pontone M, Di Domenico EG, Pimpinelli F. Bacterial Biofilm in Chronic Wounds and Possible Therapeutic Approaches. BIOLOGY 2024; 13:109. [PMID: 38392327 PMCID: PMC10886835 DOI: 10.3390/biology13020109] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/24/2024]
Abstract
Wound repair and skin regeneration is a very complex orchestrated process that is generally composed of four phases: hemostasis, inflammation, proliferation, and remodeling. Each phase involves the activation of different cells and the production of various cytokines, chemokines, and other inflammatory mediators affecting the immune response. The microbial skin composition plays an important role in wound healing. Indeed, skin commensals are essential in the maintenance of the epidermal barrier function, regulation of the host immune response, and protection from invading pathogenic microorganisms. Chronic wounds are common and are considered a major public health problem due to their difficult-to-treat features and their frequent association with challenging chronic infections. These infections can be very tough to manage due to the ability of some bacteria to produce multicellular structures encapsulated into a matrix called biofilms. The bacterial species contained in the biofilm are often different, as is their capability to influence the healing of chronic wounds. Biofilms are, in fact, often tolerant and resistant to antibiotics and antiseptics, leading to the failure of treatment. For these reasons, biofilms impede appropriate treatment and, consequently, prolong the wound healing period. Hence, there is an urgent necessity to deepen the knowledge of the pathophysiology of delayed wound healing and to develop more effective therapeutic approaches able to restore tissue damage. This work covers the wound-healing process and the pathogenesis of chronic wounds infected by biofilm-forming pathogens. An overview of the strategies to counteract biofilm formation or to destroy existing biofilms is also provided.
Collapse
Affiliation(s)
- Ilaria Cavallo
- Microbiology and Virology Unit, San Gallicano Dermatological Institute IRCSS, 00144 Rome, Italy
| | - Francesca Sivori
- Microbiology and Virology Unit, San Gallicano Dermatological Institute IRCSS, 00144 Rome, Italy
| | - Arianna Mastrofrancesco
- Microbiology and Virology Unit, San Gallicano Dermatological Institute IRCSS, 00144 Rome, Italy
| | - Elva Abril
- Microbiology and Virology Unit, San Gallicano Dermatological Institute IRCSS, 00144 Rome, Italy
| | - Martina Pontone
- Microbiology and Virology Unit, San Gallicano Dermatological Institute IRCSS, 00144 Rome, Italy
| | - Enea Gino Di Domenico
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, 00185 Rome, Italy
| | - Fulvia Pimpinelli
- Microbiology and Virology Unit, San Gallicano Dermatological Institute IRCSS, 00144 Rome, Italy
| |
Collapse
|
36
|
Mohammed RN, Aziz Sadat SA, Hassan SMA, Mohammed HF, Ramzi DO. Combinatorial Influence of Bone Marrow Aspirate Concentrate (BMAC) and Platelet-Rich Plasma (PRP) Treatment on Cutaneous Wound Healing in BALB/c Mice. J Burn Care Res 2024; 45:59-69. [PMID: 37262317 PMCID: PMC11023107 DOI: 10.1093/jbcr/irad080] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Indexed: 06/03/2023]
Abstract
Bone marrow, a soft spongy tissue, is containing mesenchymal stem cells, that are well-recognized according to their self-renewability and stemness. Therefore, we hypothesized that bone marrow aspirate concentrate (BMAC) could have a pivotal influence on the process of wound healing in particular when it is combined with platelet-rich plasma (PRP). Thirty-six albino mice (BALB/c) were used in the study and they were grouped as negative-control, PRP treated, BMAC treated and BMAC plus PRP treated. An incisional wound (1 cm2) was made at the back of mouse and their wounds were treated according to their treatment plan and group allocations. Later, the skin at the treated wound sites was collected on days 7, 14, and 21 for histopathological investigation. The results showed that there was a statistically significant difference in BMAC+PRP-treated wounds over the rest of the treated groups in the acceleration of wound healing throughout the experiment by increasing the rate of wound contraction, re-epithelization process, and granulation tissue intensity with fluctuated infiltration in the number of the neutrophils, macrophages, and lymphocytes, also restoration of the epidermal and dermal thickness with less scarring and hair follicle regeneration vs to the negative-control, PRP and BMAC only treated groups. Our findings indicated that BMAC containing mesenchymal stem cells is an efficient approach, which can be used to enhance a smooth and physiopathological healing process, especially when it is used in combination with PRP.
Collapse
Affiliation(s)
- Rebar N Mohammed
- Medical Laboratory Analysis Department, College of Health Sciences, Cihan University of Sulaimaniya, Kurdistan Region, Iraq
- Department of Microbiology, College of Veterinary Medicine, University of Sulaimnai, Suleimanyah, Iraq
| | - Sadat Abdulla Aziz Sadat
- Department of Microbiology, College of Veterinary Medicine, University of Sulaimnai, Suleimanyah, Iraq
| | - Snur M A Hassan
- Department of Anatomy and Pathology, College of Veterinary Medicine, University of Sulaimnai, Suleimanyah, Iraq
| | - Hawraz Farhad Mohammed
- Department of Microbiology, College of Veterinary Medicine, University of Sulaimnai, Suleimanyah, Iraq
| | - Derin Omer Ramzi
- Department of Basic sciences, College of Veterinary Medicine, University of Sulaimnai, Suleimanyah, Iraq
| |
Collapse
|
37
|
Justynski O, Bridges K, Krause W, Forni MF, Phan QM, Sandoval-Schaefer T, Carter K, King DE, Hsia HC, Gazes MI, Vyce SD, Driskell RR, Miller-Jensen K, Horsley V. Apoptosis recognition receptors regulate skin tissue repair in mice. eLife 2023; 12:e86269. [PMID: 38127424 PMCID: PMC10735221 DOI: 10.7554/elife.86269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 11/17/2023] [Indexed: 12/23/2023] Open
Abstract
Apoptosis and clearance of apoptotic cells via efferocytosis are evolutionarily conserved processes that drive tissue repair. However, the mechanisms by which recognition and clearance of apoptotic cells regulate repair are not fully understood. Here, we use single-cell RNA sequencing to provide a map of the cellular dynamics during early inflammation in mouse skin wounds. We find that apoptotic pathways and efferocytosis receptors are elevated in fibroblasts and immune cells, including resident Lyve1+ macrophages, during inflammation. Interestingly, human diabetic foot wounds upregulate mRNAs for efferocytosis pathway genes and display altered efferocytosis signaling via the receptor Axl and its ligand Gas6. During early inflammation in mouse wounds, we detect upregulation of Axl in dendritic cells and fibroblasts via TLR3-independent mechanisms. Inhibition studies in vivo in mice reveal that Axl signaling is required for wound repair but is dispensable for efferocytosis. By contrast, inhibition of another efferocytosis receptor, Timd4, in mouse wounds decreases efferocytosis and abrogates wound repair. These data highlight the distinct mechanisms by which apoptotic cell detection coordinates tissue repair and provides potential therapeutic targets for chronic wounds in diabetic patients.
Collapse
Affiliation(s)
- Olivia Justynski
- Dept. of Molecular Cellular and Developmental Biology, Yale University, New Haven, United States
| | - Kate Bridges
- Dept. of Biomedical Engineering, Yale University, New Haven, United States
| | - Will Krause
- Dept. of Molecular Cellular and Developmental Biology, Yale University, New Haven, United States
| | - Maria Fernanda Forni
- Dept. of Molecular Cellular and Developmental Biology, Yale University, New Haven, United States
| | - Quan M Phan
- Washington State University, SMB, Pullman, United States
| | - Teresa Sandoval-Schaefer
- Dept. of Molecular Cellular and Developmental Biology, Yale University, New Haven, United States
| | - Kristyn Carter
- Dept. of Molecular Cellular and Developmental Biology, Yale University, New Haven, United States
| | - Diane E King
- Sunnycrest Bioinformatics, Flemington, United States
| | - Henry C Hsia
- Dept. of Surgery (Plastic), Yale School of Medicine, New Haven, United States
| | - Michael I Gazes
- Dept of Podiatric Surgery, Yale New Haven Hospital, New Haven, United States
| | - Steven D Vyce
- Dept of Podiatric Surgery, Yale New Haven Hospital, New Haven, United States
| | | | - Kathryn Miller-Jensen
- Dept. of Molecular Cellular and Developmental Biology, Yale University, New Haven, United States
- Dept. of Biomedical Engineering, Yale University, New Haven, United States
| | - Valerie Horsley
- Dept. of Molecular Cellular and Developmental Biology, Yale University, New Haven, United States
- Dept. of Dermatology, Yale School of Medicine, New Haven, United States
| |
Collapse
|
38
|
Yu N, Li T, Qiu Z, Xu J, Li Y, Huang J, Yang Y, Li Z, Long X, Zhang H. Wip1 regulates wound healing by affecting activities of keratinocytes and endothelial cells through ATM-p53 and mTOR signaling. Burns 2023; 49:1969-1982. [PMID: 37357059 DOI: 10.1016/j.burns.2023.05.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 06/27/2023]
Abstract
BACKGROUND As a p53-regulated gene, Wip1 regulates proliferation, migration, apoptosis, and senescence of several type cells, but its biological functions in keratinocytes and endothelial cells which are involved wound healing are not fully understood. This study aims to reveal the function and underlying mechanism of Wip1 in wound healing using models of transgenic animal, keratinocytes, and endothelial cells. METHODS Using Wip1 knockout C57 BL/6 mice, we investigated effect of Wip1 deficiency on wound healing and angiogenesis; And using HaCaT and HUVEC as keratinocytes and endothelial cells, combined using primary keratinocytes from Wip1 knockout mice, we studied the effects of Wip1 knockdown/knockout or overexpression on proliferation, migration, and protein expressions of signaling components in ATM-p53 and mTOR pathway. RESULTS Wip1 deficiency in mice impaired the wound repair and endothelial angiogenesis, reduced the thickness of granulation tissue, and decreased the number of Ki67-positive cells and CD31 positive vessels in granulation tissue. Knockdown of Wip1 by shRNAs suppressed the proliferation and migration of HaCaT and HUVEC cells and induced notably apoptosis in the two cells. In western blot, Wip1 knockdown enriched p53 and ATM proteins, while decreased activated AKT, mTOR and activated S6 ribosomal protein (pS6) levels in HaCaT and HUVEC cells. Ectopic expression of Wip1 decreased the p53 and ATM proteins, while increased activated AKT, mTOR and pS6 levels in HaCaT and HUVEC cells. And in primary keratinocytes from mice tail skin, Wip1 knockout increased p53 and ATM, while decreased activated AKT, mTOR and pS6 protein levels. CONCLUSION Our study directly supports that Wip1 regulated skin wound healing possibly by affecting bioactivities including proliferation, migration and apoptosis of keratinocytes and endothelial cells at least through by modulating ATM-p53 and mTOR signaling.
Collapse
Affiliation(s)
- Nanze Yu
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tianhao Li
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zikai Qiu
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Xu
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yunzhu Li
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiuzuo Huang
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yilan Yang
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhujun Li
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao Long
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Hongbing Zhang
- Department of Physiology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
39
|
Yao L, Jeong S, Kwon HR, Olson LE. Regulation of adipocyte dedifferentiation at the skin wound edge. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.22.568302. [PMID: 38045303 PMCID: PMC10690246 DOI: 10.1101/2023.11.22.568302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Adipocytes have diverse roles in energy storage and metabolism, inflammation, and tissue repair. Mature adipocytes have been assumed to be terminally differentiated cells. However, recent evidence suggests that adipocytes retain substantial phenotypic plasticity, with potential to dedifferentiate into fibroblast-like cells under physiological and pathological conditions. Here, we develop a two-step lineage tracing approach based on the observation that fibroblasts express platelet-derived growth factor receptor alpha ( Pdgfra ) while adipocytes express Adiponectin ( Adipoq ) but not Pdgfra . Our approach specifically traces Pdgfra + cells that originate from Adipoq + adipocytes. We find many traced adipocytes and fibroblast-like cells surrounding skin wounds, but only a few traced cells localize to the wound center. In agreement with adipocyte plasticity, traced adipocytes incorporate EdU, downregulate Plin1 and PPARγ, and upregulate αSMA. We also investigate the role of potential dedifferentiation signals using constitutively active PDGFRα mutation, Pdgfra knockout, or Tgfbr2 knockout models. We find that PDGF and TGFβ signaling both promote dedifferentiation, and PDGFRα does so independently of TGFβR2. These results demonstrate an intersectional genetic approach to trace the hybrid cell phenotype of Pdgfra + adipocytes, which may be important for wound repair, regeneration and fibrosis.
Collapse
|
40
|
Chen FZ, Tan PC, Yang Z, Li Q, Zhou SB. Identifying characteristics of dermal fibroblasts in skin homeostasis and disease. Clin Exp Dermatol 2023; 48:1317-1327. [PMID: 37566911 DOI: 10.1093/ced/llad257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/11/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023]
Abstract
Heterogeneous dermal fibroblasts are the main components that constitute the dermis. Distinct fibroblast subgroups show specific characteristics and functional plasticity that determine dermal structure during skin development and wound healing. Although researchers have described the roles of fibroblast subsets, this is not completely understood. We review recent evidence supporting understanding about the heterogeneity of fibroblasts. We summarize the origins and the identified profiles of fibroblast subpopulations. The characteristics of fibroblast subpopulations in both healthy and diseased states are highlighted, and the potential of subpopulations to be involved in wound healing in different ways was discussed. Additionally, we review the plasticity of subpopulations and the underlying signalling mechanisms. This review may provide greater insights into potential novel therapeutic targets and tissue regeneration strategies for the future.
Collapse
Affiliation(s)
- Fang-Zhou Chen
- Department of Plastic & Reconstructive Surgery, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People's Hospital, Shanghai, China
| | - Poh-Ching Tan
- Department of Plastic & Reconstructive Surgery, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People's Hospital, Shanghai, China
| | - Zihan Yang
- Department of Plastic & Reconstructive Surgery, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People's Hospital, Shanghai, China
- Department of Plastic and Burn Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Qingfeng Li
- Department of Plastic & Reconstructive Surgery, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People's Hospital, Shanghai, China
| | - Shuang-Bai Zhou
- Department of Plastic & Reconstructive Surgery, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People's Hospital, Shanghai, China
| |
Collapse
|
41
|
Rauchenwald T, Handle F, Connolly CE, Degen A, Seifarth C, Hermann M, Tripp CH, Wilflingseder D, Lobenwein S, Savic D, Pölzl L, Morandi EM, Wolfram D, Skvortsova II, Stoitzner P, Haybaeck J, Konschake M, Pierer G, Ploner C. Preadipocytes in human granulation tissue: role in wound healing and response to macrophage polarization. Inflamm Regen 2023; 43:53. [PMID: 37904253 PMCID: PMC10617061 DOI: 10.1186/s41232-023-00302-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 10/15/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND Chronic non-healing wounds pose a global health challenge. Under optimized conditions, skin wounds heal by the formation of scar tissue. However, deregulated cell activation leads to persistent inflammation and the formation of granulation tissue, a type of premature scar tissue without epithelialization. Regenerative cells from the wound periphery contribute to the healing process, but little is known about their cellular fate in an inflammatory, macrophage-dominated wound microenvironment. METHODS We examined CD45-/CD31-/CD34+ preadipocytes and CD68+ macrophages in human granulation tissue from pressure ulcers (n=6) using immunofluorescence, immunohistochemistry, and flow cytometry. In vitro, we studied macrophage-preadipocyte interactions using primary human adipose-derived stem cells (ASCs) exposed to conditioned medium harvested from IFNG/LPS (M1)- or IL4/IL13 (M2)-activated macrophages. Macrophages were derived from THP1 cells or CD14+ monocytes. In addition to confocal microscopy and flow cytometry, ASCs were analyzed for metabolic (OXPHOS, glycolysis), morphological (cytoskeleton), and mitochondrial (ATP production, membrane potential) changes. Angiogenic properties of ASCs were determined by HUVEC-based angiogenesis assay. Protein and mRNA levels were assessed by immunoblotting and quantitative RT-PCR. RESULTS CD45-/CD31-/CD34+ preadipocytes were observed with a prevalence of up to 1.5% of total viable cells in human granulation tissue. Immunofluorescence staining suggested a spatial proximity of these cells to CD68+ macrophages in vivo. In vitro, ASCs exposed to M1, but not to M2 macrophage secretome showed a pro-fibrotic response characterized by stress fiber formation, elevated alpha smooth muscle actin (SMA), and increased expression of integrins ITGA5 and ITGAV. Macrophage-secreted IL1B and TGFB1 mediated this response via the PI3K/AKT and p38-MAPK pathways. In addition, ASCs exposed to M1-inflammatory stress demonstrated reduced migration, switched to a glycolysis-dominated metabolism with reduced ATP production, and increased levels of inflammatory cytokines such as IL1B, IL8, and MCP1. Notably, M1 but not M2 macrophages enhanced the angiogenic potential of ASCs. CONCLUSION Preadipocyte fate in wound tissue is influenced by macrophage polarization. Pro-inflammatory M1 macrophages induce a pro-fibrotic response in ASCs through IL1B and TGFB1 signaling, while anti-inflammatory M2 macrophages have limited effects. These findings shed light on cellular interactions in chronic wounds and provide important information for the potential therapeutic use of ASCs in human wound healing.
Collapse
Affiliation(s)
- Tina Rauchenwald
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Florian Handle
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University Innsbruck, Innsbruck, Austria
| | - Catherine E Connolly
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Antonia Degen
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Christof Seifarth
- Institute of Clinical and Functional Anatomy, Medical University of Innsbruck, Innsbruck, Austria
| | - Martin Hermann
- Department of Anesthesiology and Critical Care Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Christoph H Tripp
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Doris Wilflingseder
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Susanne Lobenwein
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Dragana Savic
- Department of Therapeutic Radiology and Oncology, Medical University of Innsbruck, EXTRO-Lab, Tyrolean Cancer Research Institute, Innsbruck, Austria
| | - Leo Pölzl
- Department of Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Evi M Morandi
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Dolores Wolfram
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Ira-Ida Skvortsova
- Department of Therapeutic Radiology and Oncology, Medical University of Innsbruck, EXTRO-Lab, Tyrolean Cancer Research Institute, Innsbruck, Austria
| | - Patrizia Stoitzner
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Johannes Haybaeck
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University Innsbruck, Innsbruck, Austria
- Diagnostic and Research Center for Molecular Biomedicine, Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Marko Konschake
- Institute of Clinical and Functional Anatomy, Medical University of Innsbruck, Innsbruck, Austria
| | - Gerhard Pierer
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Christian Ploner
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
42
|
Cai Y, Tian J, Li J, Liu X, Li F, Zhang L, Xiao S, Jia C, Deng C. A novel combined technology for treating hypertrophic scars: adipose tissue extract combined with fractional CO 2 laser. Front Physiol 2023; 14:1284312. [PMID: 37965106 PMCID: PMC10642933 DOI: 10.3389/fphys.2023.1284312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/17/2023] [Indexed: 11/16/2023] Open
Abstract
Introduction: Owing to the need for liposuction and its unsuitability for allogeneic transplantation, the clinical application of stromal vascular fraction gel (SVF-gel) combined with fractional CO2 laser for scar treatment is limited. Adipose tissue extract (ATE), rich in cytokines and growth factors, offers a more convenient option for clinical practice as it can be easily prepared using purely physical methods and has low immunogenicity. We aimed to evaluate the effectiveness of ATE combined with fractional CO2 laser in the treatment of hypertrophic scars. Methods: ATE was prepared using discarded liposuction fluid from patients undergoing liposuction. A rabbit ear hypertrophic scar model was established and treated with ATE, fractional CO2 laser, or a combination. PBS was used as a control. The scar appearance and histological changes were observed. The immunohistochemistry method was used to evaluate the expression of α-SMA, while perilipin was detected using immunofluorescence. Additionally, the level of adipogenic signal C/EBPα and PPARγ mRNA was studied. Results: Following treatment, the volume of hypertrophic scar decreased, resulting in a softer texture and thinner dermis. Additionally, there was a decrease in the infiltration of inflammatory cells, and the collagen arrangement became looser and more regular, and the expression of α-SMA also decreased, with the combination of ATE and fractional laser showing the most significant improvement. Moreover, the combination group was found to promote subcutaneous fat regeneration and increase the expression of adipogenic signals C/EBPα and PPARγ. Conclusion: The combination of ATE and fractional CO2 laser treatment has been shown to inhibit the development of hypertrophic scars. This effect may be attributed to the enhancement of adipogenesis and decrease in collagen deposition.
Collapse
Affiliation(s)
- Yuan Cai
- Department of Dermatology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Jiao Tian
- Department of Surgery, Zunyi Medical College, Zunyi, Guizhou, China
- Department of Clinical Medicine, Zunyi Medical College, Zunyi, Guizhou, China
| | - Jianyi Li
- Department of Burns and Plastic Surgery, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, Guangxi, China
| | - Xing Liu
- Department of Burns and Plastic Surgery, Afliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Fang Li
- Department of Dermatology, Guiyang First People’s Hospital, Guiyang, Guizhou, China
| | - Lanfang Zhang
- Department of Dermatology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Shune Xiao
- Department of Burns and Plastic Surgery, Afliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Changsha Jia
- Department of Dermatology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Chengliang Deng
- Department of Burns and Plastic Surgery, Afliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
43
|
Tikhonova N, Milovanov AP, Aleksankina VV, Kulikov IA, Fokina TV, Aleksankin AP, Belousova TN, Mikhaleva LM, Niziaeva NV. Adipocytes in the Uterine Wall during Experimental Healing and in Cesarean Scars during Pregnancy. Int J Mol Sci 2023; 24:15255. [PMID: 37894936 PMCID: PMC10607476 DOI: 10.3390/ijms242015255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/09/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
We have suggested that adipocytes in uterine scars may affect the development of the placenta accrete spectrum (PAS). In the experimental part, we explored adipocytes in the uterine wall by the twelfth sexual cycle after surgery. In the clinical part, we investigated adipocyte clusters in the cesarean scar of pregnant women with and without PAS. The uterine wall was evaluated in gross and histological sections using morphometry, histochemistry (hematoxylin and eosin stain, Mallory stain), and immunohistochemistry for FABP4 (adipocyte markers), CD68, CD163, CD206 (macrophages), CD 34 (endothelium), cytokeratin 8 (epithelium), aSMA (smooth muscle cells). The design included an experimental study on Sprague-Dawley rats (n = 18) after a full-thickness surgical incision on the seventh (n = 6), 30th (n = 6), and 60th day (n = 6). The clinical groups include pregnant women without uterine scars (n = 10), pregnant women with a uterine scar after previous cesarean sections (n = 10), and women with PAS (n = 11). Statistical processing was carried out using nonparametric methods. Comparisons were conducted using the Mann-Whitney U-test and Kruskal-Wallis test. Statistical significance was considered at p < 0.05. On the seventh day, the rat uterine horn was enveloped by adipose tissue, which contained crown-like structures with FABP4+, CD68+, CD206+, and CD163+ cells. FABP4+ cells in the uterine wall were absent by the 30th day. The number of CD206+ and CD163+ cells in the adipose tissue decreased by the 30th day. On the 60th day, the attachment of fat tissue was revealed in the form of single strands. The serous layer around the damaged area totally recovered on the 60th day. FABP4+ cells were not detected in the uterine wall samples from pregnant women without a previous cesarean section. Adipocytes were found in the scar during non-complicated pregnancy and with PAS. Reducing the number of CD68+ cells in adipocyte clusters, there were in myometrium with PAS. Increased CD206+ and CD163+ cells were revealed in uterine adipocyte clusters of the group. According to the experimental finding, adipocytes should be absent in the uterine wall by the 12th sexual cycle after a full-thickness surgical incision. The presence of adipocyte clusters in cesarean scar indicated the disturbance of cell interaction. Differences in the numbers of CD206 and CD163 cells in adipocyte clusters between groups with and without PAS may be indirect evidence that uterine adipocytes affect the development of PAS.
Collapse
Affiliation(s)
- Natalia Tikhonova
- Avtsyn Research Institute of Human Morphology of FSBSI “Petrovsky National Research Centre of Surgery”, 119991 Moscow, Russia (T.V.F.); (A.P.A.); (N.V.N.)
| | - Andrey P. Milovanov
- Avtsyn Research Institute of Human Morphology of FSBSI “Petrovsky National Research Centre of Surgery”, 119991 Moscow, Russia (T.V.F.); (A.P.A.); (N.V.N.)
| | - Valentina V. Aleksankina
- Avtsyn Research Institute of Human Morphology of FSBSI “Petrovsky National Research Centre of Surgery”, 119991 Moscow, Russia (T.V.F.); (A.P.A.); (N.V.N.)
| | - Ilyas A. Kulikov
- SBHI of the Moscow Region “Vidnovsky Perinatal Center”, 142700 Moscow, Russia (T.N.B.)
| | - Tatiana V. Fokina
- Avtsyn Research Institute of Human Morphology of FSBSI “Petrovsky National Research Centre of Surgery”, 119991 Moscow, Russia (T.V.F.); (A.P.A.); (N.V.N.)
| | - Andrey P. Aleksankin
- Avtsyn Research Institute of Human Morphology of FSBSI “Petrovsky National Research Centre of Surgery”, 119991 Moscow, Russia (T.V.F.); (A.P.A.); (N.V.N.)
| | - Tamara N. Belousova
- SBHI of the Moscow Region “Vidnovsky Perinatal Center”, 142700 Moscow, Russia (T.N.B.)
| | - Ludmila M. Mikhaleva
- Avtsyn Research Institute of Human Morphology of FSBSI “Petrovsky National Research Centre of Surgery”, 119991 Moscow, Russia (T.V.F.); (A.P.A.); (N.V.N.)
| | - Natalya V. Niziaeva
- Avtsyn Research Institute of Human Morphology of FSBSI “Petrovsky National Research Centre of Surgery”, 119991 Moscow, Russia (T.V.F.); (A.P.A.); (N.V.N.)
| |
Collapse
|
44
|
Ru J, Zhang Q, Zhu S, Cai J, He Y, Lu F. Delivery of adipose-derived growth factors from heparinized adipose acellular matrix accelerates wound healing. Front Bioeng Biotechnol 2023; 11:1270618. [PMID: 37854882 PMCID: PMC10579818 DOI: 10.3389/fbioe.2023.1270618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/18/2023] [Indexed: 10/20/2023] Open
Abstract
Dermal white adipocytes are closely associated with skin homeostasis and wound healing. However, it has not been fully investigated whether adipose-derived products improve wound healing. Here, we obtained adipose acellular matrix (AAM) and adipose-derived growth factors (ADGFs) from human adipose tissue and fabricated an ADGF-loaded AAM via surface modification with heparin. The product, HEP-ADGF-AAM, contained an adipose-derived scaffold and released ADGFs in a controlled fashion. To test its efficacy in promoting wound healing, mice with full thickness wound received three different treatments: HEP-ADGF-AAM, AAM and ADM. Control mice received no further treatments. Among these treatments, HEP-ADGF-AAM best improved wound healing. It induced adipogenesis in situ after in vivo implantation and provided an adipogenic microenvironment for wounds by releasing ADGFs. HEP-ADGF-AAM not only induced adipocyte regeneration, but also enhanced fibroblast migration, promoted vessel formation, accelerated wound closure, and enhanced wound epithelialization. Moreover, there was a close interaction between HEP-ADGF-AAM and the wound bed, and collagen was turned over in HEP-ADGF-AAM. These results show that HEP-ADGF-AAM might substantially improve re-epithelialization, angiogenesis, and skin appendage regeneration, and is thus a promising therapeutic biomaterial for skin wound healing.
Collapse
Affiliation(s)
| | | | | | | | - Yunfan He
- *Correspondence: Yunfan He, ; Feng Lu,
| | - Feng Lu
- *Correspondence: Yunfan He, ; Feng Lu,
| |
Collapse
|
45
|
Walendzik K, Kopcewicz M, Wiśniewska J, Opyd P, Machcińska-Zielińska S, Gawrońska-Kozak B. Dermal white adipose tissue development and metabolism: The role of transcription factor Foxn1. FASEB J 2023; 37:e23171. [PMID: 37682531 DOI: 10.1096/fj.202300873rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/10/2023] [Accepted: 08/18/2023] [Indexed: 09/09/2023]
Abstract
Intradermal adipocytes form dermal white adipose tissue (dWAT), a unique fat depot localized in the lower layer of the dermis. However, recognition of molecular factors regulating dWAT development, homeostasis, and bioactivity is limited. Using Foxn1-/- and Foxn1+/+ mice, we demonstrated that epidermally expressed Foxn1 regulates dWAT development and defines the adipogenic capacity of dermal fibroblasts. In intact and post-wounded skin, Foxn1 contributes to the initial stimulation of dWAT adipogenesis and participates in the modulation of lipid metabolism processes. Furthermore, Foxn1 activity strengthens adipogenic processes through Bmp2 and Igf2 signaling and regulates lipid metabolism in differentiated dermal fibroblasts. The results reveal the contribution of Foxn1 to dWAT metabolism, thus identifying possible targets for modulation and regulation of dWAT in physiological and pathological processes in the skin.
Collapse
Affiliation(s)
- Katarzyna Walendzik
- Biological Function of Food, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Marta Kopcewicz
- Biological Function of Food, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Joanna Wiśniewska
- Biological Function of Food, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Paulina Opyd
- Department of Animal Nutrition and Feed Science, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Sylwia Machcińska-Zielińska
- Biological Function of Food, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Barbara Gawrońska-Kozak
- Biological Function of Food, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| |
Collapse
|
46
|
Singh P, Ali SA. Mature white adipocyte plasticity during mammary gland remodelling and cancer. CELL INSIGHT 2023; 2:100123. [PMID: 37771567 PMCID: PMC10522874 DOI: 10.1016/j.cellin.2023.100123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/15/2023] [Accepted: 08/15/2023] [Indexed: 09/30/2023]
Abstract
Mammary gland growth and differentiation predominantly rely on stromal-epithelial cellular communication. Specifically, mammary adipocytes play a crucial role in ductal morphogenesis, as well as in the proliferation and differentiation of mammary epithelial cells. The process of lactation entails a reduction in the levels of white adipose tissue associated with the MG, allowing for the expansion of milk-producing epithelial cells. Subsequently, during involution and the regression of the milk-producing unit, adipocyte layers resurface, occupying the vacated space. This dynamic phenomenon underscores the remarkable plasticity and expansion of adipose tissue. Traditionally considered terminally differentiated, adipocytes have recently been found to exhibit plasticity in certain contexts. Unraveling the significance of this cell type within the MG could pave the way for novel approaches to reduce the risk of breast cancer and enhance lactation performance. Moreover, a comprehensive understanding of adipocyte trans- and de-differentiation processes holds promise for the development of innovative therapeutic interventions targeting cancer, fibrosis, obesity, type 2 diabetes, and other related diseases. Additionally, adipocytes may find utility in the realm of regenerative medicine. This review article provides a comprehensive examination of recent advancements in our understanding of MG remodelling, with a specific focus on the tissue-specific functions of adipocytes and their role in the development of cancer. By synthesizing current knowledge in this field, it aims to consolidate our understanding of adipocyte biology within the context of mammary gland biology, thereby fostering further research and discovery in this vital area.
Collapse
Affiliation(s)
- Parul Singh
- Cell Biology and Proteomics Lab, Animal Biotechnology Center, ICAR-NDRI, 132001, India
- Division of Radiation Oncology, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Syed Azmal Ali
- Cell Biology and Proteomics Lab, Animal Biotechnology Center, ICAR-NDRI, 132001, India
- Division Proteomics of Stem Cells and Cancer, German Cancer Research Center, 69120, Heidelberg, Germany
| |
Collapse
|
47
|
Gawronska-Kozak B, Kopcewicz M, Machcinska-Zielinska S, Walendzik K, Wisniewska J, Drukała J, Wasniewski T, Rutkowska J, Malinowski P, Pulinski M. Gender Differences in Post-Operative Human Skin. Biomedicines 2023; 11:2653. [PMID: 37893027 PMCID: PMC10604277 DOI: 10.3390/biomedicines11102653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/25/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
Although the impact of age, gender, and obesity on the skin wound healing process has been extensively studied, the data related to gender differences in aspects of skin scarring are limited. The present study performed on abdominal human intact and scar skin focused on determining gender differences in extracellular matrix (ECM) composition, dermal white adipose tissue (dWAT) accumulation, and Foxn1 expression as a part of the skin response to injury. Scar skin of men showed highly increased levels of COLLAGEN 1A1, COLLAGEN 6A3, and ELASTIN mRNA expression, the accumulation of thick collagen I-positive fibers, and the accumulation of α-SMA-positive cells in comparison to the scar skin of women. However, post-injured skin of women displayed an increase (in comparison to post-injured men's skin) in collagen III accumulation in the scar area. On the contrary, women's skin samples showed a tendency towards higher levels of adipogenic-related genes (PPARγ, FABP4, LEPTIN) than men, regardless of intact or scar skin. Intact skin of women showed six times higher levels of LEPTIN mRNA expression in comparison to men intact (p < 0.05), men post-injured (p < 0.05), or women post-injured scar (p < 0.05) skin. Higher levels of FOXN1 mRNA and protein were also detected in women than in men's skin. In conclusion, the present data confirm and extend (dWAT layer) the data related to the presence of differences between men and women in the skin, particularly in scar tissues, which may contribute to the more effective and gender-tailored improvement of skin care interventions.
Collapse
Affiliation(s)
- Barbara Gawronska-Kozak
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748 Olsztyn, Poland; (M.K.); (S.M.-Z.); (K.W.); (J.W.)
| | - Marta Kopcewicz
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748 Olsztyn, Poland; (M.K.); (S.M.-Z.); (K.W.); (J.W.)
| | - Sylwia Machcinska-Zielinska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748 Olsztyn, Poland; (M.K.); (S.M.-Z.); (K.W.); (J.W.)
| | - Katarzyna Walendzik
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748 Olsztyn, Poland; (M.K.); (S.M.-Z.); (K.W.); (J.W.)
| | - Joanna Wisniewska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748 Olsztyn, Poland; (M.K.); (S.M.-Z.); (K.W.); (J.W.)
| | - Justyna Drukała
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 31-007 Krakow, Poland;
| | - Tomasz Wasniewski
- Department of Obstetrics, Perinatology and Gynecology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland;
| | - Joanna Rutkowska
- Department of Internal Medicine, Clinic of Endocrinology, Diabetology and Internal Medicine, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland;
| | - Piotr Malinowski
- Department of Surgery, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland
| | - Michał Pulinski
- Department of Surgery, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland
| |
Collapse
|
48
|
Turk CB, Baykara Ulusan M, Döş YM, Manav Baş V, Sarikaya Tellal E, Koku Aksu AE. The Effects of Oral Isotretinoin on Atrophic Acne Scars Measured by Shear-wave Elastography: An Observational, Single-center Study. THE JOURNAL OF CLINICAL AND AESTHETIC DERMATOLOGY 2023; 16:46-51. [PMID: 37720196 PMCID: PMC10503936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
Background Although the effects of oral isotretinoin (OI) on acne vulgaris and preventing further acne scars have been well-documented, the specific impact of OI alone on pre-existing atrophic acne scars (AAS) remains unclear. No clinical study has objectively evaluated the effect of OI on AAS yet. Objective We sought to investigate the OI effect on AAS quantitatively and reliably by shear-wave elastography (SWE). Methods This work is a single-center, prospective and observational study. Thirty patients with moderate and severe acne vulgaris accompanied by AAS were included. We started the OI with a standard dose regime. On Days 0 and 90 of treatment, patients' global acne grading system (GAGS) and the Goodman and Baron's Qualitative Global Scar Rating System (GSRS) were evaluated. The dermal thickness, subcutaneous tissue thickness, scar size, and scar and subcutaneous tissue's elastic modules were measured on both cheeks of each patient by SWE. Results The improvement in GSRS stages and GAGS scores in 90 days were statistically significant (respectively; p=0.029, <0.001). Scar size and dermal thickness decreased, while the subcutaneous tissue thickness and the elastic modulus of scar and subcutaneous tissue increased in bilateral cheeks. The thickness changes in the right side dermis, and subcutaneous tissue on both sides were noteworthy (p<0.05). Conclusion Besides its well-known effect on acne vulgaris, OI also could be an effective treatment option for reducing scar size and severity while improving skin elasticity. SWE may help follow skin and scar properties.
Collapse
Affiliation(s)
- Cemre Busra Turk
- Dr. Turk is with the Wellman Center for Photomedicine at Massachusetts General Hospital in Boston, Massachusetts
- Additionally, Dr. Turk is with the Department of Dermatology at Harvard Medical School in Boston, Massachusetts
| | - Melis Baykara Ulusan
- Dr. Baykara Ulusan is with the University of Health Sciences Istanbul Training and Research Hospital's Radiology Clinic in Istanbul, Turkey
| | - Yusuf Mert Döş
- Drs. Döş, Manav Baş, Sarıkaya Tellal, and Koku Aksu are with the University of Health Sciences Istanbul Training and Research Hospital's Dermatology Clinic in Istanbul, Turkey
| | - Vildan Manav Baş
- Drs. Döş, Manav Baş, Sarıkaya Tellal, and Koku Aksu are with the University of Health Sciences Istanbul Training and Research Hospital's Dermatology Clinic in Istanbul, Turkey
| | - Ebru Sarikaya Tellal
- Drs. Döş, Manav Baş, Sarıkaya Tellal, and Koku Aksu are with the University of Health Sciences Istanbul Training and Research Hospital's Dermatology Clinic in Istanbul, Turkey
| | - Ayse Esra Koku Aksu
- Drs. Döş, Manav Baş, Sarıkaya Tellal, and Koku Aksu are with the University of Health Sciences Istanbul Training and Research Hospital's Dermatology Clinic in Istanbul, Turkey
| |
Collapse
|
49
|
Knoedler S, Broichhausen S, Guo R, Dai R, Knoedler L, Kauke-Navarro M, Diatta F, Pomahac B, Machens HG, Jiang D, Rinkevich Y. Fibroblasts - the cellular choreographers of wound healing. Front Immunol 2023; 14:1233800. [PMID: 37646029 PMCID: PMC10461395 DOI: 10.3389/fimmu.2023.1233800] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 07/28/2023] [Indexed: 09/01/2023] Open
Abstract
Injuries to our skin trigger a cascade of spatially- and temporally-synchronized healing processes. During such endogenous wound repair, the role of fibroblasts is multifaceted, ranging from the activation and recruitment of innate immune cells through the synthesis and deposition of scar tissue to the conveyor belt-like transport of fascial connective tissue into wounds. A comprehensive understanding of fibroblast diversity and versatility in the healing machinery may help to decipher wound pathologies whilst laying the foundation for novel treatment modalities. In this review, we portray the diversity of fibroblasts and delineate their unique wound healing functions. In addition, we discuss future directions through a clinical-translational lens.
Collapse
Affiliation(s)
- Samuel Knoedler
- Department of Plastic Surgery and Hand Surgery, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
- Division of Plastic Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, United States
- Division of Plastic Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, Germany
| | - Sonja Broichhausen
- Department of Hand, Plastic and Reconstructive Surgery, Microsurgery, Burn Trauma Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Ruiji Guo
- Department of Plastic Surgery and Hand Surgery, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, Germany
| | - Ruoxuan Dai
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, Germany
| | - Leonard Knoedler
- Division of Plastic Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, United States
| | - Martin Kauke-Navarro
- Division of Plastic Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, United States
| | - Fortunay Diatta
- Division of Plastic Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, United States
| | - Bohdan Pomahac
- Division of Plastic Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, United States
| | - Hans-Guenther Machens
- Department of Plastic Surgery and Hand Surgery, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Dongsheng Jiang
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, Germany
| | - Yuval Rinkevich
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, Germany
| |
Collapse
|
50
|
Valenzuela PL, Carrera-Bastos P, Castillo-García A, Lieberman DE, Santos-Lozano A, Lucia A. Obesity and the risk of cardiometabolic diseases. Nat Rev Cardiol 2023; 20:475-494. [PMID: 36927772 DOI: 10.1038/s41569-023-00847-5] [Citation(s) in RCA: 170] [Impact Index Per Article: 85.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/08/2023] [Indexed: 03/18/2023]
Abstract
The prevalence of obesity has reached pandemic proportions, and now approximately 25% of adults in Westernized countries have obesity. Recognized as a major health concern, obesity is associated with multiple comorbidities, particularly cardiometabolic disorders. In this Review, we present obesity as an evolutionarily novel condition, summarize the epidemiological evidence on its detrimental cardiometabolic consequences and discuss the major mechanisms involved in the association between obesity and the risk of cardiometabolic diseases. We also examine the role of potential moderators of this association, with evidence for and against the so-called 'metabolically healthy obesity phenotype', the 'fatness but fitness' paradox or the 'obesity paradox'. Although maintenance of optimal cardiometabolic status should be a primary goal in individuals with obesity, losing body weight and, particularly, excess visceral adiposity seems to be necessary to minimize the risk of cardiometabolic diseases.
Collapse
Affiliation(s)
- Pedro L Valenzuela
- Physical Activity and Health Research Group (PaHerg), Research Institute of Hospital 12 de Octubre ("i + 12"), Madrid, Spain.
- Department of Systems Biology, University of Alcalá, Alcalá de Henares, Spain.
| | - Pedro Carrera-Bastos
- Center for Primary Health Care Research, Department of Clinical Sciences, Lund University, Malmö, Sweden
- Faculty of Sport Sciences, Universidad Europea de Madrid, Madrid, Spain
| | | | - Daniel E Lieberman
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Alejandro Santos-Lozano
- Physical Activity and Health Research Group (PaHerg), Research Institute of Hospital 12 de Octubre ("i + 12"), Madrid, Spain
- Department of Health Sciences, European University Miguel de Cervantes, Valladolid, Spain
| | - Alejandro Lucia
- Faculty of Sport Sciences, Universidad Europea de Madrid, Madrid, Spain.
- CIBER of Frailty and Healthy Aging (CIBERFES), Madrid, Spain.
| |
Collapse
|