Anti-fibrotic effect of adipose-derived stem cells on fibrotic scars

BACKGROUND Sustained injury, through radiotherapy, burns or surgical trauma, can result in fibrosis, displaying an excessive deposition of extracellular matrix (ECM), persisting inflammatory reaction, and reduced vascularization. The increasing recognition of fibrosis as a cause for disease and mortality, and increasing use of radiotherapy causing fibrosis, stresses the importance of a decent anti-fibrotic treatment. AIM To obtain an in-depth understanding of the complex mechanisms underlying fibrosis, and more specifically, the potential mechanisms-of-action of adipose-derived stomal cells (ADSCs) in realizing their anti-fibrotic effect. METHODS A systematic review of the literature using PubMed, Embase and Web of Science was performed by two independent reviewers. RESULTS The injection of fat grafts into fibrotic tissue, releases ADSC into the environment. ADSCs’ capacity to directly differentiate into key cell types (e.g., ECs, fibroblasts), as well as to secrete multiple paracrine factors (e.g., hepatocyte growth factor, basis fibroblast growth factor, IL-10), allows them to alter different mechanisms underlying fibrosis in a combined approach. ADSCs favor ECM degradation by impacting the fibroblast-to-myofibroblast differentiation, favoring matrix metalloproteinases over tissue inhibitors of metalloproteinases, positively influencing collagen organization, and inhibiting the pro-fibrotic effects of transforming growth factor-β1. Furthermore, they impact elements of both the innate and adaptive immune response system, and stimulate angiogenesis on the site of injury (through secretion of pro-angiogenic cytokines like stromal cell-derived factor-1 and vascular endothelial growth factor). CONCLUSION This review shows that understanding the complex interactions of ECM accumulation, immune response and vascularization, is vital to fibrosis treatments’ effectiveness like fat grafting. It details how ADSCs intelligently steer this complex system in an anti-fibrotic or pro-angiogenic direction, without falling into extreme dilation or stimulation of a single aspect. Detailing this combined approach, has brought fat grafting one step closer to unlocking its full potential as a non-anecdotal treatment for fibrosis.

[1]  M. Karsdal,et al.  Dermal fibroblasts have different extracellular matrix profiles induced by TGF-β, PDGF and IL-6 in a model for skin fibrosis , 2020, Scientific Reports.

[2]  S. Coleman Long-Term Survival of Fat Transplants: Controlled Demonstrations , 2020, Aesthetic Plastic Surgery.

[3]  B. Weber,et al.  Global trends in clinical trials involving pluripotent stem cells: a systematic multi-database analysis , 2020, npj Regenerative Medicine.

[4]  M. Saint-Cyr,et al.  How Fat Grafting Works , 2020, Plastic and reconstructive surgery. Global open.

[5]  Kevin C. Wang,et al.  The antifibrotic adipose‐derived stromal cell: Grafted fat enriched with CD74+ adipose‐derived stromal cells reduces chronic radiation‐induced skin fibrosis , 2020, Stem cells translational medicine.

[6]  M. Nowicki,et al.  The Proliferation and Differentiation of Adipose-Derived Stem Cells in Neovascularization and Angiogenesis , 2020, International journal of molecular sciences.

[7]  D. Greening,et al.  Fat Therapeutics: The Clinical Capacity of Adipose-Derived Stem Cells and Exosomes for Human Disease and Tissue Regeneration , 2020, Frontiers in Pharmacology.

[8]  D. Mangieri,et al.  Neovascularization is a key feature of liver fibrosis progression: anti-angiogenesis as an innovative way of liver fibrosis treatment , 2020, Molecular Biology Reports.

[9]  Amina El Ayadi,et al.  Current Approaches Targeting the Wound Healing Phases to Attenuate Fibrosis and Scarring , 2020, International journal of molecular sciences.

[10]  B. Hinz,et al.  Evasion of apoptosis by myofibroblasts: a hallmark of fibrotic diseases , 2019, Nature Reviews Rheumatology.

[11]  S. Macparland,et al.  Lifting the veil on macrophage diversity in tissue regeneration and fibrosis , 2019, Science Immunology.

[12]  Fei-Fei Liu,et al.  Targeting metabolic dysregulation for fibrosis therapy , 2019, Nature Reviews Drug Discovery.

[13]  J. Rubin,et al.  Adipose‐Derived Stem Cell Therapy Ameliorates Ionizing Irradiation Fibrosis via Hepatocyte Growth Factor‐Mediated Transforming Growth Factor‐β Downregulation and Recruitment of Bone Marrow Cells , 2019, Stem cells.

[14]  M. Albersen,et al.  The mechanisms and potential of stem cell therapy for penile fibrosis , 2018, Nature Reviews Urology.

[15]  P. Blondeel,et al.  Autologous fat grafting: Latest insights , 2018, Annals of medicine and surgery.

[16]  G. Opdenakker,et al.  Immunomodulation as Rescue for Chronic Atonic Skin Wounds. , 2018, Trends in immunology.

[17]  Mary E. Ziegler,et al.  Adipose-Derived Tissue in the Treatment of Dermal Fibrosis: Antifibrotic Effects of Adipose-Derived Stem Cells , 2018, Annals of plastic surgery.

[18]  Masaaki Ii,et al.  Anti-inflammatory and anti-fibrotic effects of intravenous adipose-derived stem cell transplantation in a mouse model of bleomycin-induced interstitial pneumonia , 2017, Scientific Reports.

[19]  F. Lu,et al.  Early Macrophage Infiltration Improves Fat Graft Survival by Inducing Angiogenesis and Hematopoietic Stem Cell Recruitment , 2017, Plastic and reconstructive surgery.

[20]  F. Lu,et al.  Macrophage infiltration regulates the adipose ECM reconstruction and the fibrosis process after fat grafting. , 2017, Biochemical and biophysical research communications.

[21]  W. Liu,et al.  Inhibition of Pathological Phenotype of Hypertrophic Scar Fibroblasts Via Coculture with Adipose-Derived Stem Cells. , 2017, Tissue engineering. Part A.

[22]  Maurizio Memo,et al.  Clinical potentials of human pluripotent stem cells , 2017, Cell Biology and Toxicology.

[23]  M. Harmsen,et al.  The power of fat and its adipose‐derived stromal cells: emerging concepts for fibrotic scar treatment , 2017, Journal of tissue engineering and regenerative medicine.

[24]  A. Seifalian,et al.  The regenerative role of adipose‐derived stem cells (ADSC) in plastic and reconstructive surgery , 2017, International wound journal.

[25]  Clifford M. Babbey,et al.  Human Adipose-Derived Stem Cells Suppress Elastase-Induced Murine Abdominal Aortic Inflammation and Aneurysm Expansion through Paracrine Factors , 2017, Cell transplantation.

[26]  H. Yu,et al.  Indoleamine 2,3-Dioxygenase Is Not a Pivotal Regulator Responsible for Suppressing Allergic Airway Inflammation through Adipose-Derived Stem Cells , 2016, PloS one.

[27]  Deok‐Ho Kim,et al.  Molecular networks underlying myofibroblast fate and fibrosis. , 2016, Journal of molecular and cellular cardiology.

[28]  A. Theocharis,et al.  Extracellular matrix structure. , 2016, Advanced drug delivery reviews.

[29]  S. Peirce,et al.  Macrophage Recruitment and Polarization During Collateral Vessel Remodeling in Murine Adipose Tissue , 2016, Microcirculation.

[30]  M. Alcaraz,et al.  Paracrine in vivo inhibitory effects of adipose tissue-derived mesenchymal stromal cells in the early stages of the acute inflammatory response. , 2015, Cytotherapy.

[31]  Li-Na Liu,et al.  Intralesional injection of adipose-derived stem cells reduces hypertrophic scarring in a rabbit ear model , 2015, Stem Cell Research & Therapy.

[32]  S. Corvera,et al.  Alternatively Activated M2 Macrophages Improve Autologous Fat Graft Survival in a Mouse Model through Induction of Angiogenesis. , 2015, Plastic and reconstructive surgery.

[33]  R. Reis,et al.  Polyhydroxybutyrate-co-hydroxyvalerate structures loaded with adipose stem cells promote skin healing with reduced scarring. , 2015, Acta biomaterialia.

[34]  Hong-Mian Li,et al.  Human Breast Adipose-Derived Stem Cells Transfected with the Stromal Cell-Derived Factor-1 Receptor CXCR4 Exhibit Enhanced Viability in Human Autologous Free Fat Grafts , 2014, Cellular Physiology and Biochemistry.

[35]  M. Harmsen,et al.  Adipose Tissue–Derived Stromal Cells Inhibit TGF-&bgr;1–Induced Differentiation of Human Dermal Fibroblasts and Keloid Scar–Derived Fibroblasts in a Paracrine Fashion , 2014, Plastic and reconstructive surgery.

[36]  H. Mizuno,et al.  The Effect of Bone-Marrow-Derived Stem Cells and Adipose-Derived Stem Cells on Wound Contraction and Epithelization. , 2014, Advances in wound care.

[37]  C. Feghali-Bostwick,et al.  Fibroblasts in fibrosis: novel roles and mediators , 2014, Front. Pharmacol..

[38]  T. Shimada,et al.  siRNA knockdown of tissue inhibitor of metalloproteinase-1 in keloid fibroblasts leads to degradation of collagen type I. , 2014, The Journal of investigative dermatology.

[39]  F. Lu,et al.  The Survival Condition and Immunoregulatory Function of Adipose Stromal Vascular Fraction (SVF) in the Early Stage of Nonvascularized Adipose Transplantation , 2013, PloS one.

[40]  G. M. Di Guglielmo,et al.  Dynamics of Transforming Growth Factor Beta Signaling in Wound Healing and Scarring. , 2013, Advances in wound care.

[41]  S. Bellusci,et al.  The Anti-Scar Effects of Basic Fibroblast Growth Factor on the Wound Repair In Vitro and In Vivo , 2013, PloS one.

[42]  X. Bai,et al.  Protection against TGF-β1-induced fibrosis effects of IL-10 on dermal fibroblasts and its potential therapeutics for the reduction of skin scarring , 2013, Archives of Dermatological Research.

[43]  D. K. Rah,et al.  Effect of Human Adipose Derived Stem Cells on Scar Formation and Remodeling in a Pig Model: A Pilot Study , 2012, Dermatologic surgery : official publication for American Society for Dermatologic Surgery [et al.].

[44]  T. Wynn,et al.  Mechanisms of fibrosis: therapeutic translation for fibrotic disease , 2012, Nature Medicine.

[45]  Y. Tabata,et al.  Therapeutic potential of fibroblast growth factor-2 for hypertrophic scars: upregulation of MMP-1 and HGF expression , 2012, Laboratory Investigation.

[46]  A. Ghosh,et al.  PAI‐1 in tissue fibrosis , 2012, Journal of cellular physiology.

[47]  T. Wynn,et al.  Protective and pathogenic functions of macrophage subsets , 2011, Nature Reviews Immunology.

[48]  Y. Taniyama,et al.  Hepatocyte growth factor attenuates renal fibrosis through TGF-β1 suppression by apoptosis of myofibroblasts , 2010, Journal of hypertension.

[49]  Benoit Hendrickx,et al.  Integration of Blood Outgrowth Endothelial Cells in Dermal Fibroblast Sheets Promotes Full Thickness Wound Healing , 2010, Stem cells.

[50]  C. Denton,et al.  Animal models of scleroderma: lessons from transgenic and knockout mice , 2009, Current opinion in rheumatology.

[51]  T. Murohara,et al.  Autologous adipose-derived regenerative cells for therapeutic angiogenesis. , 2009, Current pharmaceutical design.

[52]  S. Dickens,et al.  A plasma-based biomatrix mixed with endothelial progenitor cells and keratinocytes promotes matrix formation, angiogenesis, and reepithelialization in full-thickness wounds. , 2009, Tissue engineering. Part A.

[53]  A. Smith,et al.  Arginase-1–Expressing Macrophages Suppress Th2 Cytokine–Driven Inflammation and Fibrosis , 2009, PLoS pathogens.

[54]  S. Dickens,et al.  Regulable vascular endothelial growth factor165 overexpression by ex vivo expanded keratinocyte cultures promotes matrix formation, angiogenesis, and healing in porcine full-thickness wounds. , 2008, Tissue engineering. Part A.

[55]  Y. Tada,et al.  Antifibrotic effects of hepatocyte growth factor on scleroderma fibroblasts and analysis of its mechanism , 2006, Modern rheumatology.

[56]  D. Broide,et al.  Reduced peribronchial fibrosis in allergen-challenged MMP-9-deficient mice. , 2006, American journal of physiology. Lung cellular and molecular physiology.

[57]  S. Forbes,et al.  Selective depletion of macrophages reveals distinct, opposing roles during liver injury and repair. , 2005, The Journal of clinical investigation.

[58]  L. Wakefield,et al.  IL-13 Activates a Mechanism of Tissue Fibrosis That Is Completely TGF-β Independent , 2004, The Journal of Immunology.

[59]  T. Wynn Fibrotic disease and the TH1/TH2 paradigm , 2004, Nature Reviews Immunology.

[60]  F. Brombacher,et al.  Alternative macrophage activation is essential for survival during schistosomiasis and downmodulates T helper 1 responses and immunopathology. , 2004, Immunity.

[61]  S. Fujimoto,et al.  [Vasculogenesis and angiogenesis]. , 2003, Journal of UOEH.

[62]  D. Sheppard,et al.  Transforming Growth Factor-β-Dependent and -Independent Pathways of Induction of Tubulointerstitial Fibrosis in β6−/− Mice , 2003 .

[63]  W. Laug,et al.  Increased plasminogen activator inhibitor-1 in keloid fibroblasts may account for their elevated collagen accumulation in fibrin gel cultures. , 2003, The American journal of pathology.

[64]  Min Zhu,et al.  Human adipose tissue is a source of multipotent stem cells. , 2002, Molecular biology of the cell.

[65]  B. Vaillant,et al.  Regulation of Hepatic Fibrosis and Extracellular Matrix Genes by the Th Response: New Insight into the Role of Tissue Inhibitors of Matrix Metalloproteinases , 2001, Journal of Immunology.

[66]  R. Homer,et al.  Interleukin-13 Induces Tissue Fibrosis by Selectively Stimulating and Activating Transforming Growth Factor β1 , 2001, The Journal of experimental medicine.

[67]  D. Anthony,et al.  Transient expression of IL-1beta induces acute lung injury and chronic repair leading to pulmonary fibrosis. , 2001, The Journal of clinical investigation.

[68]  H. Lorenz,et al.  Multilineage cells from human adipose tissue: implications for cell-based therapies. , 2001, Tissue engineering.

[69]  T. Wynn,et al.  IL-10 and the Dangers of Immune Polarization: Excessive Type 1 and Type 2 Cytokine Responses Induce Distinct Forms of Lethal Immunopathology in Murine Schistosomiasis , 2000, The Journal of Immunology.

[70]  J. Ward,et al.  MT1-MMP-Deficient Mice Develop Dwarfism, Osteopenia, Arthritis, and Connective Tissue Disease due to Inadequate Collagen Turnover , 1999, Cell.

[71]  Anita B. Roberts,et al.  Mice lacking Smad3 show accelerated wound healing and an impaired local inflammatory response , 1999, Nature Cell Biology.

[72]  E. Rieber,et al.  Local production of interleukin-4 during radiation-induced pneumonitis and pulmonary fibrosis in rats: macrophages as a prominent source of interleukin-4. , 1997, American journal of respiratory cell and molecular biology.

[73]  R. Seder,et al.  Regulation of transforming growth factor‐β production by interleukin‐12 , 1997 .

[74]  A. Wangoo,et al.  Interleukin‐10‐ and corticosteroid‐induced reduction in type I procollagen in a human ex vivo scar culture , 1997, International journal of experimental pathology.

[75]  A. Sher,et al.  An IL-12-based vaccination method for preventing fibrosis induced by schistosome infection , 1995, Nature.

[76]  J. Whitsett,et al.  Expression of a tumor necrosis factor-alpha transgene in murine lung causes lymphocytic and fibrosing alveolitis. A mouse model of progressive pulmonary fibrosis. , 1995, The Journal of clinical investigation.

[77]  T. Wynn Fibrotic disease and the T(H)1/T(H)2 paradigm. , 2004, Nature reviews. Immunology.

[78]  D. Sheppard,et al.  Transforming growth factor-beta-dependent and -independent pathways of induction of tubulointerstitial fibrosis in beta6(-/-) mice. , 2003, The American journal of pathology.

[79]  R. Seder,et al.  Regulation of transforming growth factor-beta production by interleukin-12. , 1997, European journal of immunology.