Immunomodulation of Skin Repair: Cell-Based Therapeutic Strategies for Skin Replacement (A Comprehensive Review)

Biomedicines - Tập 10 Số 1 - Trang 118
Shima Tavakoli1, Marta Kisiel2, Thomas Biedermann3,4, Agnes S. Klar3,4
1Division of Polymer Chemistry, Department of Chemistry-Ångstrom Laboratory, Uppsala University, 75121 Uppsala, Sweden;
2Environmental and Occupational Medicine, Medical Sciences, Uppsala University Hospital, 75237 Uppsala, Sweden;
3Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
4Tissue Biology Research Unit, University Children's Hospital Zurich, University of Zurich, 8952 Schlieren, Switzerland;

Tóm tắt

The immune system has a crucial role in skin wound healing and the application of specific cell-laden immunomodulating biomaterials emerged as a possible treatment option to drive skin tissue regeneration. Cell-laden tissue-engineered skin substitutes have the ability to activate immune pathways, even in the absence of other immune-stimulating signals. In particular, mesenchymal stem cells with their immunomodulatory properties can create a specific immune microenvironment to reduce inflammation, scarring, and support skin regeneration. This review presents an overview of current wound care techniques including skin tissue engineering and biomaterials as a novel and promising approach. We highlight the plasticity and different roles of immune cells, in particular macrophages during various stages of skin wound healing. These aspects are pivotal to promote the regeneration of nonhealing wounds such as ulcers in diabetic patients. We believe that a better understanding of the intrinsic immunomodulatory features of stem cells in implantable skin substitutes will lead to new translational opportunities. This, in turn, will improve skin tissue engineering and regenerative medicine applications.

Từ khóa


Tài liệu tham khảo

Vig, K., Chaudhari, A., Tripathi, S., Dixit, S., Sahu, R., Pillai, S., Dennis, V.A., and Singh, S.R. (2017). Advances in Skin Regeneration Using Tissue Engineering. Int. J. Mol. Sci., 18.

Gaur, M., Dobke, M., Lunyak, V.V., Piatelli, A., and Zavan, B. (2017). Molecular Sciences Mesenchymal Stem Cells from Adipose Tissue in Clinical Applications for Dermatological Indications and Skin Aging. Int. J. Mol. Sci., 18.

Nybo, 2010, Injury is a major inducer of epidermal innate immune responses during wound healing, J. Investig. Dermatol., 130, 910, 10.1038/jid.2009.414

Klar, 2014, Tissue-engineered dermo-epidermal skin grafts prevascularized with adipose-derived cells, Biomaterials, 35, 5065, 10.1016/j.biomaterials.2014.02.049

Klar, 2013, “Trooping the color”: Restoring the original donor skin color by addition of melanocytes to bioengineered skin analogs, Pediatr. Surg. Int., 29, 239, 10.1007/s00383-012-3217-0

Klar, 2014, Analysis of blood and lymph vascularization patterns in tissue-engineered human dermo-epidermal skin analogs of different pigmentation, Pediatr. Surg. Int., 30, 223, 10.1007/s00383-013-3451-0

Tavakoli, S., and Klar, A.S. (2021). Bioengineered Skin Substitutes: Advances and Future Trends. Appl. Sci., 11.

Klar, 2016, Characterization of vasculogenic potential of human adipose-derived endothelial cells in a three-dimensional vascularized skin substitute, Pediatr. Surg. Int., 32, 17, 10.1007/s00383-015-3808-7

Klar, 2014, Differential expression of granulocyte, macrophage, and hypoxia markers during early and late wound healing stages following transplantation of tissue-engineered skin substitutes of human origin, Pediatr. Surg. Int., 30, 1257, 10.1007/s00383-014-3616-5

Zimoch, 2018, Polyisocyanopeptide hydrogels: A novel thermo-responsive hydrogel supporting pre-vascularization and the development of organotypic structures, Acta Biomater., 70, 129, 10.1016/j.actbio.2018.01.042

Halim, 2010, Biologic and synthetic skin substitutes: An overview, Indian J. Plast Surg., 43, S23, 10.4103/0970-0358.70712

Klar, 2017, The Use of Adipose Derived Cells for Skin Nerve Regeneration-Short Review of Experimental Research, J. Tissue Sci. Eng., 8, 2, 10.4172/2157-7552.1000191

Shevchenko, 2010, A review of tissue-engineered skin bioconstructs available for skin reconstruction, J. R. Soc. Interface, 7, 229, 10.1098/rsif.2009.0403

Vacanti, 1999, Tissue engineering: The design and fabrication of living replacement devices for surgical reconstruction and transplantation, Lancet, 354, 32, 10.1016/S0140-6736(99)90247-7

Chen, 2009, Stem cells for skin tissue engineering and wound healing, Crit. Rev. Biomed. Eng., 37, 399, 10.1615/CritRevBiomedEng.v37.i4-5.50

Klar, 2017, Skin tissue engineering: Application of adipose-derived stem cells, Biomed Res. Int., 2017, 9747010, 10.1155/2017/9747010

Tavakoli, S., and Klar, A.S. (2020). Advanced Hydrogels as Wound Dressings. Biomolecules, 10.

Metcalfe, 2007, Bioengineering skin using mechanisms of regeneration and repair, Biomaterials, 28, 5100, 10.1016/j.biomaterials.2007.07.031

Metcalfe, 2007, Tissue engineering of replacement skin: The crossroads of biomaterials, wound healing, embryonic development, stem cells and regeneration, J. R. Soc. Interface, 4, 413, 10.1098/rsif.2006.0179

Clayton, 2017, Langerhans cells-programmed by the epidermis, Front. Immunol., 8, 1676, 10.3389/fimmu.2017.01676

(2017). MA Nilforoushzadeh Dermal Fibroblast Cells: Biology and Function in Skin Regeneration. J. Ski., Available online: https://sites.kowsarpub.com/jssc/articles/69080.html.

Dhivya, 2015, Wound dressings–a review, Biomedicine, 5, 22, 10.7603/s40681-015-0022-9

Moore, 2006, Prediction and monitoring the therapeutic response of chronic dermal wounds, Int. Wound J., 3, 89, 10.1111/j.1742-4801.2006.00212.x

Lazarus, 1994, Definitions and guidelines for assessment of wounds and evaluation of healing, Wound Repair Regen., 2, 165, 10.1046/j.1524-475X.1994.20305.x

Boateng, 2008, Wound healing dressings and drug delivery systems: A review, J. Pharm. Sci., 97, 2892, 10.1002/jps.21210

Percival, 2002, Classification of Wounds and their Management, Surgery, 20, 114

Singer, 1999, Cutaneous Wound Healing, N. Engl. J. Med., 341, 738, 10.1056/NEJM199909023411006

Larson, 2010, Scarless fetal wound healing: A basic science review, Plast. Reconstr. Surg., 126, 1172, 10.1097/PRS.0b013e3181eae781

Guo, 2010, Factors Affecting Wound Healing, J. Dent. Res., 89, 219, 10.1177/0022034509359125

Zengaffinen, 2014, Primary wound closure with a Limberg flap vs. secondary wound healing after excision of a pilonidal sinus: A multicentre randomised controlled study, Int. J. Colorectal Dis., 30, 97

Burns, 2001, Burn wound healing and skin substitutes, Burns, 27, 517, 10.1016/S0305-4179(01)00017-1

Martin, 1997, Wound Healing-Aiming for Perfect Skin Regeneration, Science, 276, 75, 10.1126/science.276.5309.75

Wallace, H., Basehore, B., and Zito, P. (2020, December 08). Wound Healing Phases; StatPearls Publishing, Treasure Island (FL) 2020. Available online: https://europepmc.org/books/n/statpearls/article-34001/.

Kloc, 2019, Macrophage functions in wound healing, J. Tissue Eng. Regen. Med., 13, 99

Gilmore, 1991, Phases of wound healing, Dimens Oncol. Nurs., 5, 32

Wilgus, 2013, Neutrophils and Wound Repair: Positive Actions and Negative Reactions, Adv. Wound Care, 2, 379, 10.1089/wound.2012.0383

Iacob, A.T., Drăgan, M., Ionescu, O.M., Profire, L., Ficai, A., Andronescu, E., Confederat, L.G., and Lupașcu, D. (2020). An Overview of Biopolymeric Electrospun Nanofibers Based on Polysaccharides for Wound Healing Management. Pharmaceutics, 12.

McCartney-Francis, N.L., and Wahl, S.M. (2001). TGF-β and macrophages in the rise and fall of inflammation. TGF-β and Related Cytokines in Inflammation, Birkhäuser.

Turner, 2014, Cytokines and chemokines: At the crossroads of cell signalling and inflammatory disease, Biochim. Biophys. Acta, 1843, 2563, 10.1016/j.bbamcr.2014.05.014

Werner, 2007, Keratinocyte–fibroblast interactions in wound healing, Keratinocyte–Fibroblast Interact. Wound Health, 127, 998

Nelson, 2003, Nutrition for optimum wound healing, Nurs. Stand., 18, 55

Gordillo, 2003, Revisiting the essential role of oxygen in wound healing, Am. J. Surg., 186, 259, 10.1016/S0002-9610(03)00211-3

Branton, 1999, TGF-β and fibrosis, Microbes. Infect., 1, 1349, 10.1016/S1286-4579(99)00250-6

Phan, 2008, Biology of Fibroblasts and Myofibroblasts, Proc. Am. Thorac. Soc., 5, 334, 10.1513/pats.200708-146DR

Varney, 2013, Mechanoregulation of the Myofibroblast in Wound Contraction, Scarring, and Fibrosis: Opportunities for New Therapeutic Intervention, Adv. Wound Care, 2, 122, 10.1089/wound.2012.0393

Darby, 2014, Fibroblasts and myofibroblasts in wound healing, Clin. Cosmet. Investig. Dermatol., 7, 301

Basu, A., Kligman, L.H., Samulewicz, S.J., and Howe, C.C. (2001). Impaired wound healing in mice deficient in a matricellular protein SPARC (osteonectin, BM-40). BMC Cell Biol., 2.

Chen, 2016, Insight into reepithelialization: How do mesenchymal stem cells perform?, Stem Cells Int., 3, 1

Santoro, 2005, Cellular and molecular facets of keratinocyte reepithelization during wound healing, Exp. Cell Res., 304, 274, 10.1016/j.yexcr.2004.10.033

Martins, 2013, Matrix metalloproteinases and epidermal wound repair, Cell Tissue Res., 351, 255, 10.1007/s00441-012-1410-z

Nguyen, T.T., Mobashery, S., and Chang, M. (2016). Roles of Matrix Metalloproteinases in Cutaneous Wound Healing. Wound Health New Insights Into Anc. Chall., 37–71.

Michopoulou, 2015, How do epidermal matrix metalloproteinases support re-epithelialization during skin healing?, Artic. Eur. J. Dermatol., 25, 33, 10.1684/ejd.2015.2553

Pastar, 2014, Epithelialization in Wound Healing: A Comprehensive Review, Adv. Wound Care, 3, 445, 10.1089/wound.2013.0473

Krzyszczyk, 2018, The Role of Macrophages in Acute and Chronic Wound Healing and Interventions to Promote Pro-wound Healing Phenotypes, Front. Physiol., 9, 419, 10.3389/fphys.2018.00419

Wynn, 2016, Macrophages in Tissue Repair, Regeneration, and Fibrosis, Immunity, 44, 450, 10.1016/j.immuni.2016.02.015

Martinez, F.O., and Gordon, S. (2014). The M1 and M2 paradigm of macrophage activation: Time for reassessment. F1000Prime Rep., 6.

Schnoor, 2008, Production of type VI collagen by human macrophages: A new dimension in macrophage functional heterogeneity, J. Immunol., 180, 5707, 10.4049/jimmunol.180.8.5707

Weitkamp, 1999, Human macrophages synthesize type VIII collagen in vitro and in the atherosclerotic plaque, FASEB J., 13, 1445, 10.1096/fasebj.13.11.1445

Ogle, 2016, Monocytes and macrophages in tissue repair: Implications for immunoregenerative biomaterial design, Exp. Biol. Med., 241, 1084, 10.1177/1535370216650293

Recalcati, 2010, Differential regulation of iron homeostasis during human macrophage polarized activation, Eur. J. Immunol., 40, 824, 10.1002/eji.200939889

Mantovani, 2004, The chemokine system in diverse forms of macrophage activation and polarization, Trends Immonology, 25, 677, 10.1016/j.it.2004.09.015

Murray, 2011, Protective and pathogenic functions of macrophage subsets, Nat. Rev. Immunol., 11, 723, 10.1038/nri3073

Zhu, 2016, The pentacyclic triterpene Lupeol switches M1 macrophages to M2 and ameliorates experimental inflammatory bowel disease, Int. Immunopharmacol., 30, 74, 10.1016/j.intimp.2015.11.031

Ferrante, 2012, Regulation of Macrophage Polarization and Wound Healing, Adv. Wound Care, 1, 10, 10.1089/wound.2011.0307

Martinez, 2008, Alternative Activation of Macrophages: An Immunologic Functional Perspective, Annu. Rev. Immunol., 27, 451, 10.1146/annurev.immunol.021908.132532

Wang, 2019, M2b macrophage polarization and its roles in diseases, J. Leukoc. Biol., 106, 345, 10.1002/JLB.3RU1018-378RR

Das, 2015, Monocyte and Macrophage Plasticity in Tissue Repair and Regeneration, Am. J. Pathol., 185, 2596, 10.1016/j.ajpath.2015.06.001

Larouche, 2018, Immune regulation of skin wound healing: Mechanisms and novel therapeutic targets, Adv. Wound Care, 7, 209, 10.1089/wound.2017.0761

Zhao, R., Liang, H., Clarke, E., Jackson, C., and Xue, M. (2016). Inflammation in Chronic Wounds. Int. J. Mol. Sci., 17.

Dinarello, 2018, Introduction to the interleukin-1 family of cytokines and receptors: Drivers of innate inflammation and acquired immunity, Immunol. Rev., 281, 5, 10.1111/imr.12624

Frykberg, 2015, Challenges in the Treatment of Chronic Wounds, Adv. Wound Care, 4, 560, 10.1089/wound.2015.0635

Wysocki, 1993, Wound fluid from chronic leg ulcers contains elevated levels of metalloproteinases MMP-2 and MMP-9, J. Investig. Dermatol., 10, 64, 10.1111/1523-1747.ep12359590

Wallace, 1998, Levels of tumor necrosis factor-α (TNF-α) and soluble TNF receptors in chronic venous leg ulcers–correlations to healing status, J. Investig. Dermatol., 110, 292, 10.1046/j.1523-1747.1998.00113.x

Takeo, 2015, Wound Healing and Skin Regeneration, Cold Spring Harb. Perspect Med., 5, a023267, 10.1101/cshperspect.a023267

Jin, 2018, Macrophages in keloid are potent at promoting the differentiation and function of regulatory T cells, Exp. Cell Res., 362, 472, 10.1016/j.yexcr.2017.12.011

Li, 2017, Status of M1 and M2 type macrophages in keloid, Int. J. Clin. Exp. Pathol., 10, 11098

Ellis, 2018, Immunology of Wound Healing, Curr. Dermatol. Rep., 7, 350, 10.1007/s13671-018-0234-9

Ostuni, 2015, Macrophages and cancer: From mechanisms to therapeutic implications, Trends Immunol., 36, 229, 10.1016/j.it.2015.02.004

Cromack, 1987, Transforming growth factor beta levels in rat wound chambers, J. Surg. Res., 42, 622, 10.1016/0022-4804(87)90005-9

Yamakawa, 2019, Advances in surgical applications of growth factors for wound healing, Burn. Trauma, 7, 10, 10.1186/s41038-019-0148-1

Yoshida, 2003, Neutralization of hepatocyte growth factor leads to retarded cutaneous wound healing associated with decreased neovascularization and granulation tissue formation, J. Investig. Dermatol., 120, 335, 10.1046/j.1523-1747.2003.12039.x

Wang, 2009, MEK, p38, and PI-3K mediate cross talk between EGFR and TNFR in enhancing hepatocyte growth factor production from human mesenchymal stem cells, Am. J. Physiol. Cell Physiol., 297, C1284, 10.1152/ajpcell.00183.2009

Joseph, P., and Christopher, C. (2020, December 08). Skin Grafting -StatPearls -NCBI Bookshelf, Available online: https://www.ncbi.nlm.nih.gov/books/NBK532874/.

(2020, December 08). Immunobiology-NCBI Bookshelf, (n.d.), Available online: https://www.ncbi.nlm.nih.gov/books/NBK10757/.

Middelkoop, E. (2018). Skin substitutes and “the next level”. Total Burn Care, Elsevier. Available online: https://www.sciencedirect.com/science/article/pii/B9780323476614000150.

Hardin-Young, J., Teumer, J., and Ross, R.N. (2020). Approaches to transplanting engineered cells and tissues. Princ. Tissue Eng., 281–291. Available online: http://www.academia.edu/download/61780013/Principles_of_Tissue_Engineering20200114-84151-1u473co.pdf#page=324.

Buchbinder, 2007, Wound healing: Adjuvant therapy and treatment adherence, Venous Ulcers, 8, 91, 10.1016/B978-012373565-2.50012-9

Carter, J.E., and Holmes, J.H. (2016). The Surgical Management of Burn Wounds. Skin Tissue Engineering and Regenerative Medicine, Elsevier Inc.

Cascalho, M. (2008). Challenges and potentials of xenotransplantation. Clin. Immunol., 1215–1222. Available online: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7152187/.

Kuo, 2018, Comparison of two decellularized dermal equivalents, J. Tissue Eng. Regen. Med., 12, 983, 10.1002/term.2530

Bello, 2001, Tissue-engineered skin: Current status in wound healing, Am. J. Clin. Dermatol., 2, 305, 10.2165/00128071-200102050-00005

Pourmoussa, 2016, An update and review of cell-based wound dressings and their integration into clinical practice, Ann. Transl. Med., 4, 457, 10.21037/atm.2016.12.44

Becker, 1963, Cytological Demonstration of the Clonal Nature of Spleen Colonies Derived from Transplanted Mouse Marrow Cells, Nature, 197, 452, 10.1038/197452a0

Siminovitch, 1963, The Distribution of Colony-Forming Cells among Spleen Colonies, J. Cell. Comp. Physiol., 62, 327, 10.1002/jcp.1030620313

Duscher, 2016, Stem cells in wound healing: The future of regenerative medicine? A mini-review, Gerontology, 62, 216, 10.1159/000381877

Gorecka, 2019, The potential and limitations of induced pluripotent stem cells to achieve wound healing, Stem Cell Res. Ther., 10, 1, 10.1186/s13287-019-1185-1

Dash, B., Xu, Z., Lin, L., Koo, A., Ndon, S., Berthiaume, F., Dardik, A., and Hsia, H. (2018). Stem Cells and Engineered Scaffolds for Regenerative Wound Healing. Bioengineering, 5.

Smith, 2001, Embryo-derived stem cells: Of mice and men, Annu. Rev. Cell Dev. Biol., 17, 435, 10.1146/annurev.cellbio.17.1.435

Martin, 1981, Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells, Proc. Natl. Acad Sci. USA, 78, 7634, 10.1073/pnas.78.12.7634

Medvedev, 2010, Induced pluripotent stem cells: Problems and advantages when applying them in regenerative medicine, Acta Nat., 2, 18, 10.32607/20758251-2010-2-2-18-27

Aoi, 2008, Generation of pluripotent stem cells from adult mouse liver and stomach cells, Science, 321, 699, 10.1126/science.1154884

Hanna, 2008, Direct reprogramming of terminally differentiated mature B lymphocytes to pluripotency, Cell, 133, 250, 10.1016/j.cell.2008.03.028

Kim, 2014, Pluripotent stem cells induced from adult neural stem cells by reprogramming with two factors, nature.com, Nature, 454, 646, 10.1038/nature07061

Eminli, 2008, Reprogramming of Neural Progenitor Cells into Induced Pluripotent Stem Cells in the Absence of Exogenous Sox2 Expression, Stem Cells, 26, 2467, 10.1634/stemcells.2008-0317

Takahashi, 2007, Induction of pluripotent stem cells from adult human fibroblasts by defined factors, Cell, 131, 861, 10.1016/j.cell.2007.11.019

Yu, 2007, Induced Pluripotent Stem Cell Lines Derived from Human Somatic Cells, Science, 21, 1917, 10.1126/science.1151526

Lian, 2014, Paracrine Mechanisms of Mesenchymal Stem Cell-Based Therapy: Current Status and Perspectives, Cell Transplant., 23, 1045, 10.3727/096368913X667709

Baraniak, 2010, Stem cell paracrine actions and tissue regeneration, Regen. Med., 5, 121, 10.2217/rme.09.74

Devrim, 2004, Comparison of Keratinocyte Proliferation in Diabetic and Non-Diabetic Inflamed Gingiva, J. Periodontol., 75, 989, 10.1902/jop.2004.75.7.989

Kim, 2013, Cooperation of endothelial and smooth muscle cells derived from human induced pluripotent stem cells enhances neovascularization in dermal wounds, Proceedings of the Tissue Engineering—Part A, Volume 19, 2478, 10.1089/ten.tea.2012.0768

Clayton, 2018, Induced pluripotent stem cell-derived endothelial cells promote angiogenesis and accelerate wound closure in a murine excisional wound healing model, Biosci. Rep., 38, BSR20180563, 10.1042/BSR20180563

Casqueiro, 2012, Infections in patients with diabetes mellitus: A review of pathogenesis, Indian J. Endocrinol. Metab., 16, S27, 10.4103/2230-8210.94253

Zhang, 2015, Exosomes released from human induced pluripotent stem cells-derived MSCs facilitate cutaneous wound healing by promoting collagen synthesis and angiogenesis, J. Transl. Med., 13, 1, 10.1186/s12967-015-0417-0

Itoh, M., Umegaki-Arao, N., Guo, Z., Liu, L., Higgins, C.A., and Christiano, A.M. (2013). Generation of 3D Skin Equivalents Fully Reconstituted from Human Induced Pluripotent Stem Cells (iPSCs). PLoS ONE, 8.

Kuzuya, 1995, Induction of angiogenesis by smooth muscle cell-derived factor: Possible role in neovascularization in atherosclerotic plaque, J. Cell. Physiol., 164, 658, 10.1002/jcp.1041640324

Shen, 2016, Engineered human vascularized constructs accelerate diabetic wound healing, Biomaterials, 102, 107, 10.1016/j.biomaterials.2016.06.009

Tan, 2018, Integration of induced pluripotent stem cell-derived endothelial cells with polycaprolactone/gelatin-based electrospun scaffolds for enhanced therapeutic angiogenesis, Stem Cell Res. Ther., 9, 1, 10.1186/s13287-018-0824-2

Kashpur, 2019, Differentiation of diabetic foot ulcer-derived induced pluripotent stem cells reveals distinct cellular and tissue phenotypes, FASEB J., 33, 1262, 10.1096/fj.201801059

Nakayama, 2018, The development of induced pluripotent stem cell-derived mesenchymal stem/stromal cells from normal human and RDEB epidermal keratinocytes, J. Dermatol. Sci., 91, 301, 10.1016/j.jdermsci.2018.06.004

Kobayashi, H. (2018). Effects of Exosomes Derived from the Induced Pluripotent Stem Cells on Skin Wound Healing, Nagoya University. Available online: https://ci.nii.ac.jp/naid/500001336521/.

Liubaviciute, 2020, Modulated mesenchymal stromal cells improve skin wound healing, Biologicals, 67, 1, 10.1016/j.biologicals.2020.08.003

Tencerova, 2016, The bone marrow-derived stromal cells: Commitment and regulation of adipogenesis, Front. Endocrinol., 7, 127, 10.3389/fendo.2016.00127

Rosen, 2015, Energy Excess, Glucose Utilization, and Skeletal Remodeling: New Insights, J. Bone Miner. Res., 30, 1356, 10.1002/jbmr.2574

Lindner, 2010, Mesenchymal Stem or Stromal Cells: Toward a Better Understanding of Their Biology?, Transfus Med. Hemother., 37, 75, 10.1159/000290897

Abboud, 1993, A bone marrow stromal cell line is a source and target for platelet-derived growth factor, Blood, 81, 2547, 10.1182/blood.V81.10.2547.2547

Rocha, 2012, Metabolic labeling of human bone marrow mesenchymal stem cells for the quantitative analysis of their chondrogenic differentiation, J. Proteome Res., 11, 5350, 10.1021/pr300572r

Wu, 2007, Bone marrow-derived stem cells in wound healing: A review, Wound Repair Regen., 15, S18, 10.1111/j.1524-475X.2007.00221.x

Hao, 2009, Transplantation of BMSCs expressing hPDGF-A/hBD2 promotes wound healing in rats with combined radiation-wound injury, Gene Ther., 16, 34, 10.1038/gt.2008.133

Basiouny, 2013, Effect of bone marrow derived mesenchymal stem cells on healing of induced full-thickness skin wounds in albino rat, Int. J. Stem Cells, 6, 12, 10.15283/ijsc.2013.6.1.12

Wu, 2007, Mesenchymal Stem Cells Enhance Wound Healing Through Differentiation and Angiogenesis, Stem Cells, 25, 2648, 10.1634/stemcells.2007-0226

Dash, 2009, Targeting nonhealing ulcers of lower extremity in human through autologous bone marrow-derived mesenchymal stem cells, Rejuvenation Res., 12, 359, 10.1089/rej.2009.0872

Sorrell, 2010, Topical delivery of mesenchymal stem cells and their function in wounds, Stem Cell Res. Ther., 1, 1, 10.1186/scrt30

Orlic, 2001, Bone marrow cells regenerate infarcted myocardium, Nature, 5, 701, 10.1038/35070587

Horwitz, 1999, Transplantability and therapeutic effects of bone marrow-derived mesenchymal cells in children with osteogenesis imperfecta, Nat. Med., 5, 309, 10.1038/6529

Salinas, 2009, Mesenchymal stem cells for craniofacial tissue regeneration: Designing hydrogel delivery vehicles, J. Dent. Res., 88, 681, 10.1177/0022034509341553

Lei, 2018, Bone marrow-derived mesenchymal stem cells laden novel thermo-sensitive hydrogel for the management of severe skin wound healing, Mater. Sci. Eng. C Mater. Biol. Appl., 1, 159, 10.1016/j.msec.2018.04.045

Viezzer, 2020, A new waterborne chitosan-based polyurethane hydrogel as a vehicle to transplant bone marrow mesenchymal cells improved wound healing of ulcers in a diabetic, Carbohydr. P, 231, 115734, 10.1016/j.carbpol.2019.115734

Bharti, 2020, Effect of cryopreservation on therapeutic potential of canine bone marrow derived mesenchymal stem cells augmented mesh scaffold for wound healing in guinea pig, Biomed. Pharmacother., 121, 109573, 10.1016/j.biopha.2019.109573

Erben, 1997, Short-Term Treatment of Rats with High Dose 1,25-Dihydroxyvitamin D3 Stimulates Bone Formation and Increases the Number of Osteoblast Precursor Cells in Bone, Endocrinology, 138, 4629, 10.1210/endo.138.11.5511

Yoshimura, 2006, Characterization of freshly isolated and cultured cells derived from the fatty and fluid portions of liposuction aspirates, J. Cell. Physiol., 208, 64, 10.1002/jcp.20636

Conese, 2020, The Role of Adipose-Derived Stem Cells, Dermal Regenerative Templates, and Platelet-Rich Plasma in Tissue Engineering-Based Treatments of Chronic Skin Wounds, Stem Cells Int., 2020, 7056261, 10.1155/2020/7056261

Patrikoski, 2019, Perspectives for clinical translation of adipose stromal/stem cells, Stem Cells Int., 2019, 1, 10.1155/2019/5858247

Ntege, 2020, Advances in regenerative therapy: A review of the literature and future directions, Regen. Ther., 14, 136, 10.1016/j.reth.2020.01.004

Peng, Q., Alipour, H., Porsborg, S., Fink, T., and Zachar, V. (2020). Evolution of ASC Immunophenotypical Subsets During Expansion In Vitro. Int. J. Mol. Sci. Artic., 21.

Fristad, 2018, Adipose-derived and bone marrow mesenchymal stem cells: A donor-matched comparison, Stem Cell Res. Ther., 9, 1

Yu, 2010, Yield and characterization of subcutaneous human adipose-derived stem cells by flow cytometric and adipogenic mRNA analyzes, Cytotherapy, 12, 538, 10.3109/14653241003649528

Gimble, 2007, Adipose-derived stem cells for regenerative medicine, Circ. Res., 100, 1249, 10.1161/01.RES.0000265074.83288.09

Gimble, 2011, Adipose-derived stromal/stem cells (ASC) in regenerative medicine: Pharmaceutical applications, Curr. Pharm. Des., 17, 332, 10.2174/138161211795164220

Rodriguez, J., Pratta, A., Abbassi, N., and Fabre, H. (2017). Evaluation of three devices for the isolation of the stromal vascular fraction from adipose tissue and for ASC culture: A comparative study. Stem Cells Int., 2017.

Hur, W., Lee, H.Y., Min, H.S., Wufuer, M., Lee, C.W., Hur, J.A., Kim, S.H., Kim, B.K., and Choi, T.H. (2017). Regeneration of full-thickness skin defects by differentiated adipose-derived stem cells into fibroblast-like cells by fibroblast-conditioned medium. Stem Cell Res. Ther., 8.

Kim, 2007, Wound healing effect of adipose-derived stem cells: A critical role of secretory factors on human dermal fibroblasts, J. Dermatol. Sci., 48, 15, 10.1016/j.jdermsci.2007.05.018

Lee, 2012, Safety and Effect of Adipose Tissue-Derived Stem Cell Implantation in Patients With Critical Limb Ischemia, Circ. J., 76, 1750, 10.1253/circj.CJ-11-1135

Bura, 2014, Phase I trial: The use of autologous cultured adipose-derived stroma/stem cells to treat patients with non-revascularizable critical limb ischemia, Cytotherapy, 16, 245, 10.1016/j.jcyt.2013.11.011

Parvizi, 2015, Therapeutic Prospect of Adipose-Derived Stromal Cells for the Treatment of Abdominal Aortic Aneurysm, Stem Cells Dev., 24, 1493, 10.1089/scd.2014.0517

Rennert, 2014, Diabetes impairs the angiogenic potential of adipose-derived stem cells by selectively depleting cellular subpopulations, Stem Cell Res. Ther., 5, 1, 10.1186/scrt468

Siennicka, K., Zolocinska, A., Stepien, K., Lubina-Dabrowska, N., Maciagowska, M., Zolocinska, E., Slysz, A., Piusinska-Macoch, R., Mazur, S., and Zdanowicz, U. (2016). Adipose-Derived Cells (Stromal Vascular Fraction) Transplanted for Orthopedical or Neurological Purposes: Are They Safe Enough?. Stem Cells Int., 2016.

Kim, 2018, Effects of donor age on human adipose-derived adherent stromal cells under oxidative stress conditions, J. Int. Med. Res., 46, 951, 10.1177/0300060517731684

Klar, 2018, Characterization of M1 and M2 polarization of macrophages in vascularized human dermo-epidermal skin substitutes in vivo, Pediatr. Surg. Int., 34, 129, 10.1007/s00383-017-4179-z

Barsotti, 2011, Fibrin acts as biomimetic niche inducing both differentiation and stem cell marker expression of early human endothelial progenitor cells, Cell Prolif., 44, 33, 10.1111/j.1365-2184.2010.00715.x

Davis, 2011, Supplementation of fibrin gels with sodium chloride enhances physical properties and ensuing osteogenic response, Acta Biomater., 7, 691, 10.1016/j.actbio.2010.09.007

Murphy, 2017, Engineering fibrin hydrogels to promote the wound healing potential of mesenchymal stem cell spheroids, Acta Biomater., 64, 176, 10.1016/j.actbio.2017.10.007

Chae, 2017, Stromal vascular fraction shows robust wound healing through high chemotactic and epithelialization property, Cytotherapy, 19, 543, 10.1016/j.jcyt.2017.01.006

Gobin, 2003, Effects of Epidermal Growth Factor on Fibroblast Migration through Biomimetic Hydrogels, Biotechnol. Prog., 19, 1781, 10.1021/bp0341390

Blay, 1985, Epidermal growth factor promotes the chemotactic migration of cultured rat intestinal epithelial cells, J. Cell. Physiol., 124, 107, 10.1002/jcp.1041240117

Matthay, 1993, Transient effect of epidermal growth factor on the motility of an immortalized mammary epithelial cell line, J. Cell. Sci., 106, 869, 10.1242/jcs.106.3.869

Nilforoushzadeh, 2020, Engineered skin graft with stromal vascular fraction cells encapsulated in fibrin–collagen hydrogel: A clinical study for diabetic wound healing, J. Tissue Eng. Regen. Med., 14, 424, 10.1002/term.3003

Guo, 2018, Adipose-derived mesenchymal stem cells accelerate diabetic wound healing in a similar fashion as bone marrow-derived cells, Am. J. Physiol.Cell Physiol., 315, C885, 10.1152/ajpcell.00120.2018

Morizono, 2003, Comparison of multi-lineage cells from human adipose tissue and bone marrow, Cells Tissues Organs, 174, 101, 10.1159/000071150

Koenen, 2013, Adipose-Derived Stem Cells in Wound Healing: Recent Results In Vitro and In Vivo, OA Mol. Cell Biol., 1, 8

Aggarwal, 2004, Human mesenchymal stem cells modulate allogeneic immune cell responses, Blood, 105, 1815, 10.1182/blood-2004-04-1559

Magni, 2002, Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli, Blood, 99, 3838, 10.1182/blood.V99.10.3838

Montesinos, 2015, Immunoregulation by Mesenchymal Stem Cells: Biological Aspects and Clinical Applications, J. Immunol. Res., 2015, 394917

Krampera, 2003, Bone marrow mesenchymal stem cells inhibit the response of naive and memory antigen-specific T cells to their cognate peptide, Blood, 101, 3722, 10.1182/blood-2002-07-2104

Meisel, 2004, Human bone marrow stromal cells inhibit allogeneic T-cell responses by indoleamine 2,3-dioxygenase-mediated tryptophan degradation, Blood, 103, 4619, 10.1182/blood-2003-11-3909

Rhijn, 2012, Mesenchymal stem cells derived from adipose tissue are not affected by renal disease, Kidney Int., 82, 748, 10.1038/ki.2012.187

2010, Mesenchymal stem cells: A new therapeutic tool for AKI, Nat. Rev. Nephrol., 6, 179, 10.1038/nrneph.2009.229

Rahimnejad, 2017, Biomaterials and tissue engineering for scar management in wound care, Burn. Trauma, 5, 4, 10.1186/s41038-017-0069-9

Li, 2015, Three-dimensional graphene foams loaded with bone marrow derived mesenchymal stem cells promote skin wound healing with reduced scarring, Mater. Sci. Eng. C Mater. Biol. Appl., 1, 181, 10.1016/j.msec.2015.07.062

Maggini, J., Mirkin, G., Bognanni, I., Holmberg, J., Piazzón, I.M., Nepomnaschy, I., Costa, H., Cañones, C., Raiden, S., and Vermeulen, M. (2010). Mouse bone marrow-derived mesenchymal stromal cells turn activated macrophages into a regulatory-like profile. PLoS ONE, 5.

Jackson, 2012, Mesenchymal stem cell therapy for attenuation of scar formation during wound healing, Stem Cell Res. Ther., 3, 20, 10.1186/scrt111

Moore, 2001, Interleukin-10 and the interleukin-10 receptor, Annu. Rev. Immunol., 19, 683, 10.1146/annurev.immunol.19.1.683

Liechty, 2000, Fetal wound repair results in scar formation in interleukin-10–deficient mice in a syngeneic murine model of scarless fetal wound repair, J. Pediatr. Surg., 35, 866, 10.1053/jpsu.2000.6868

Peranteau, 2008, IL-10 overexpression decreases inflammatory mediators and promotes regenerative healing in an adult model of scar formation, J. Investig. Dermatol, 128, 1852, 10.1038/sj.jid.5701232

Gordon, 2008, Permissive environment in postnatal wounds induced by adenoviral-mediated overexpression of the anti-inflammatory cytokine interleukin-10 prevents scar formation, Wound Repair Regen., 16, 70, 10.1111/j.1524-475X.2007.00326.x

Mattar, 2015, Comparing the immunomodulatory properties of bone marrow, adipose tissue, and birth-associated tissue mesenchymal stromal cells, Front. Immunol., 6, 560, 10.3389/fimmu.2015.00560

Ceccarelli, 2020, Immunomodulatory Effect of Adipose-Derived Stem Cells: The Cutting Edge of Clinical Application, Front. Cell Dev. Biol., 8, 236, 10.3389/fcell.2020.00236

Cao, 2020, Characterization of the immunomodulatory properties of alveolar bone-derived mesenchymal stem cells, Stem Cell Res. Ther., 11, 120, 10.1186/s13287-020-01605-x

Rostami, 2020, Immunoregulatory properties of mesenchymal stem cells: Micro-RNAs, Immunol. Lett., 219, 34, 10.1016/j.imlet.2019.12.011

Wang, 2020, Immunomodulatory Properties of Stem Cells in Periodontitis: Current Status and Future Prospective, Stem Cell Int., 2020, 9836518

Anderson, 2009, Human adult stem cells derived from adipose tissue protect against experimental colitis and sepsis, Gut, 57, 929

Hong, 2010, Therapeutic potential of adipose-derived stem cells in vascular growth and tissue repair, Curr. Opin. Organ. Transplant., 15, 86, 10.1097/MOT.0b013e328334f074

Mitchell, 2006, Immunophenotype of Human Adipose-Derived Cells: Temporal Changes in Stromal-Associated and Stem Cell-Associated Markers, Stem Cells, 24, 376, 10.1634/stemcells.2005-0234

Puissant, 2005, Immunomodulatory effect of human adipose tissue-derived adult stem cells: Comparison with bone marrow mesenchymal stem cells, Br. J. Haematol., 129, 118, 10.1111/j.1365-2141.2005.05409.x

Wolbank, 2007, Dose-dependent immunomodulatory effect of human stem cells from amniotic membrane: A comparison with human mesenchymal stem cells from adipose tissue, Tissue Eng., 13, 1173, 10.1089/ten.2006.0313

Yoo, 2009, Comparison of immunomodulatory properties of mesenchymal stem cells derived from adult human tissues, Cell Immunol., 259, 150, 10.1016/j.cellimm.2009.06.010

Lindroos, 2011, The Potential of Adipose Stem Cells in Regenerative Medicine, Stem Cell Rev. Reports, 7, 269, 10.1007/s12015-010-9193-7

Kucerova, 2007, Adipose Tissue-Derived Human Mesenchymal Stem Cells Mediated Prodrug Cancer Gene Therapy, Cancer Res., 67, 6304, 10.1158/0008-5472.CAN-06-4024

Yu, 2008, Mesenchymal stem cells derived from human adipose tissues favor tumor cell growth in vivo, Stem Cells Dev., 17, 463, 10.1089/scd.2007.0181

Barone, 2013, Immunomodulatory effects of adipose-derived stem cells: Fact or fiction?, Biomed. Res. Int., 2013, 383685

Cui, 2007, Expanded adipose-derived stem cells suppress mixed lymphocyte reaction by secretion of prostaglandin E2, Tissue Eng., 13, 1185, 10.1089/ten.2006.0315

Lin, 2012, Allogeneic and xenogeneic transplantation of adipose-derived stem cells in immunocompetent recipients without immunosuppressants, Stem Cells Dev., 21, 2770, 10.1089/scd.2012.0176

Gonzalez, 2010, Human adipose-derived mesenchymal stem cells reduce inflammatory and T cell responses and induce regulatory T cells in vitro in rheumatoid arthritis, Ann. Rheum. Dis., 69, 241, 10.1136/ard.2008.101881

Fang, 2007, Favorable response to human adipose tissue-derived mesenchymal stem cells in steroid-refractory acute graft-versus-host disease, Transpl. Proc., 39, 3358, 10.1016/j.transproceed.2007.08.103

Fang, 2007, Human adipose tissue-derived mesenchymal stromal cells as salvage therapy for treatment of severe refractory acute graft-vs.-host disease in two children, Pediatr. Transplant., 11, 814, 10.1111/j.1399-3046.2007.00780.x

Fang, 2007, Using human adipose tissue-derived mesenchymal stem cells as salvage therapy for hepatic graft-versus-host disease resembling acute hepatitis, Transpl. Proc., 39, 1710, 10.1016/j.transproceed.2007.02.091

Park, 2011, Cell cycle regulators are critical for maintaining the differentiation potential and immaturity in adipogenesis of adipose-derived stem cells, Differentiation, 82, 136, 10.1016/j.diff.2011.06.002

Jiang, 2020, Immune modulation by mesenchymal stem cells, Cell Prolif., 53, e12712, 10.1111/cpr.12712

Bartholomew, 2002, Mesenchymal stem cells suppress lymphocyte proliferation in vitro and prolong skin graft survival in vivo, Exp. Hematol., 30, 42, 10.1016/S0301-472X(01)00769-X

Koppula, 2009, Histocompatibility testing of cultivated human bone marrow stromal cells—A promising step towards pre-clinical screening for allogeneic stem cell therapy, Cell. Immunol., 259, 61, 10.1016/j.cellimm.2009.05.014

Mohanty, 2020, Immunomodulatory properties of bone marrow mesenchymal stem cells, J. Biosci., 45, 1, 10.1007/s12038-020-00068-9

Bertozzi, 2017, The biological and clinical basis for the use of adipose-derived stem cells in the field of wound healing, Ann. Med. Surg, 20, 41, 10.1016/j.amsu.2017.06.058

Franz, 2011, Immune responses to implants—A review of the implications for the design of immunomodulatory biomaterials, Biomaterials, 32, 6692, 10.1016/j.biomaterials.2011.05.078

Sun, 2018, Engineering Pro-Regenerative Hydrogels for Scarless Wound Healing, Adv. Healthc. Mater., 7, 1800016, 10.1002/adhm.201800016

Vishwakarma, 2016, Engineering Immunomodulatory Biomaterials To Tune the Inflammatory Response, Trends Biotechnol., 34, 470, 10.1016/j.tibtech.2016.03.009

Mantovani, 2013, Macrophage plasticity and polarization in tissue repair and remodelling, J. Pathol., 229, 176, 10.1002/path.4133

Brown, 2012, Macrophage polarization: An opportunity for improved outcomes in biomaterials and regenerative medicine, Biomaterials, 33, 3792, 10.1016/j.biomaterials.2012.02.034

Williams, 2008, On the mechanisms of biocompatibility, Biomaterials, 29, 2941, 10.1016/j.biomaterials.2008.04.023

Li, J., and Hastings, G.W. (2016). Oxide bioceramics: Inert ceramic materials in medicine and dentistry. Handbook of Biomaterial Properties, Springer. [2nd ed.].

Hayashi, 1993, Bone-implant interface mechanics of in vivo bio-inert ceramics, Biomaterials, 14, 1173, 10.1016/0142-9612(93)90163-V

Ehashi, T., Takemura, T., Hanagata, N., Minowa, T., Kobayashi, H., Ishihara, K., and Yamaoka, T. (2014). Comprehensive genetic analysis of early host body reactions to the bioactive and bio-inert porous scaffolds. PLoS ONE, 9.

Hench, L.L., and Wilson, J. (1993). An Introduction to Bioceramics, World Scientific.

Desai, 2017, Advances in islet encapsulation technologies, Nat. Rev. Drug Discov., 16, 338, 10.1038/nrd.2016.232

Onuki, 2008, A review of the biocompatibility of implantable devices: Current challenges to overcome foreign body response, J. Diabetes Sci. Technol., 2, 1003, 10.1177/193229680800200610

Mohammadi, 2020, Silk based scaffolds with immunomodulatory capacity: Anti-inflammatory effects of nicotinic acid, Biomater. Sci., 8, 148, 10.1039/C9BM00814D

Hench, 2010, Twenty-first century challenges for biomaterials, Royalsocietypublishing.Org, J. R. Soc. Interface, 7, S379, 10.1098/rsif.2010.0151.focus

Dziki, 2017, Extracellular Matrix Bioscaffolds as Immunomodulatory Biomaterials, Tissue Eng. Part. A, 23, 1152, 10.1089/ten.tea.2016.0538

Anderson, 2008, Foreign body reaction to biomaterials, Semin. Immunol., 20, 86, 10.1016/j.smim.2007.11.004

Bensiamar, 2015, Topographical cues regulate the crosstalk between MSCs and macrophages, Biomaterials, 37, 124, 10.1016/j.biomaterials.2014.10.028

Broughton, 2006, The Basic Science of Wound Healing, Plast Reconstr. Surg., 117, 12S, 10.1097/01.prs.0000225430.42531.c2

Badylak, 2008, Macrophage phenotype as a determinant of biologic scaffold remodeling, Tissue Eng. Part. A., 14, 1835, 10.1089/ten.tea.2007.0264

Brown, 2009, Macrophage phenotype and remodeling outcomes in response to biologic scaffolds with and without a cellular component, Biomaterials, 30, 1482, 10.1016/j.biomaterials.2008.11.040

Koh, 2011, Inflammation and wound healing: The role of the macrophage, Expert Rev. Mol., 13, e23, 10.1017/S1462399411001943

Gordon, 2003, Alternative activation of macrophages, Nat. Rev. Immunol., 3, 23, 10.1038/nri978

Sridharan, 2015, Biomaterial based modulation of macrophage polarization: A review and suggested design principles, Materialstoday, 18, 313

Singh, 2017, Unbiased Analysis of the Impact of Micropatterned Biomaterials on Macrophage Behavior Provides Insights beyond Predefined Polarization States, ACS Biomater. Sci. Eng., 3, 969, 10.1021/acsbiomaterials.7b00104

Corliss, 2016, Macrophages: An Inflammatory Link Between Angiogenesis and Lymphangiogenesis, Microcirculation, 23, 95, 10.1111/micc.12259

Ashouri, 2019, Macrophage polarization in wound healing: Role of aloe vera/chitosan nanohydrogel, Drug Deliv. Transl. Res., 9, 1027, 10.1007/s13346-019-00643-0

Kumar, 2018, Immunomodulatory injectable silk hydrogels maintaining functional islets and promoting anti-inflammatory M2 macrophage polarization, Biomaterials, 187, 1, 10.1016/j.biomaterials.2018.09.037

Li, 2016, Transition from inflammation to proliferation: A critical step during wound healing, Cell. Mol. Life Sci., 73, 3861, 10.1007/s00018-016-2268-0

Lucas, 2010, Differential roles of macrophages in diverse phases of skin repair, J. Immunol., 184, 3964, 10.4049/jimmunol.0903356

Klar, 2017, Comparison of in vivo immune responses following transplantation of vascularized and non-vascularized human dermo-epidermal skin substitutes, Pediatr. Surg., 33, 377, 10.1007/s00383-016-4031-x

Brown, 2012, Macrophage phenotype as a predictor of constructive remodeling following the implantation of biologically derived surgical mesh materials, Acta Biomater., 8, 978, 10.1016/j.actbio.2011.11.031

Meng, 2015, Solubilized extracellular matrix from brain and urinary bladder elicits distinct functional and phenotypic responses in macrophages, Biomaterials, 46, 131, 10.1016/j.biomaterials.2014.12.044

Chakraborty, 2020, Regulation of decellularized matrix mediated immune response, Biomater. Sci., 8, 1194, 10.1039/C9BM01780A

Kharaziha, 2021, Rational Design of Immunomodulatory Hydrogels for Chronic Wound Healing, Adv. Mater, 33, 2100176, 10.1002/adma.202100176

Raynal, 2009, Identification of multiple potent binding sites for human leukocyte associated Ig-like receptor LAIR on collagens II and III, Matrix Biol., 28, 202, 10.1016/j.matbio.2009.03.005

Chaffee, 2019, Stabilized collagen matrix dressing improves wound macrophage function and epithelialization, FASEB J., 33, 2144, 10.1096/fj.201800352R

Inv, 1993, Fibroblast Migration in Fibrin Gel Matrices, Am. J. Pathol., 142, 273

Clark, 2001, Fibrin and wound healing, Ann. N. Y. Acad. Sci., 936, 355, 10.1111/j.1749-6632.2001.tb03522.x

Stone, 2018, Advancements in regenerative strategies through the continuum of burn care, Front. Pharmacol., 9, 672, 10.3389/fphar.2018.00672

Hsieh, 2017, Differential regulation of macrophage inflammatory activation by fibrin and fibrinogen, Acta Biomater., 1, 14, 10.1016/j.actbio.2016.09.024

Qu, 2010, Interface between hemostasis and adaptive immunity, Curr. Opin. Immunol., 22, 634, 10.1016/j.coi.2010.08.017

Pravda, 2016, Hyaluronic Acid and Its Derivatives in Coating and Delivery Systems: Applications in Tissue Engineering, Regenerative Medicine and Immunomodulation, Adv. Healthc. Mater., 5, 2841, 10.1002/adhm.201600316

Rayahin, 2015, High and Low Molecular Weight Hyaluronic Acid Differentially Influence Macrophage Activation, ACS Biomater. Sci. Eng., 1, 481, 10.1021/acsbiomaterials.5b00181

Fong, 2018, Chitosan immunomodulatory properties: Perspectives on the impact of structural properties and dosage, Futur. Sci. OA, 4, FSO225, 10.4155/fsoa-2017-0064

Takei, 2012, Synthesis of a chitosan derivative soluble at neutral pH and gellable by freeze–thawing, and its application in wound care, Acta Biomater., 8, 686, 10.1016/j.actbio.2011.10.005

Porporatto, 2003, Chitosan induces different L L-arginine metabolic pathways in resting and inflammatory macrophages, Biochem. Biophys. Res. Commun., 304, 266, 10.1016/S0006-291X(03)00579-5

Moura, 2013, Chitosan-based dressings loaded with neurotensin-an efficient strategy to im-prove early diabetic wound healing, Acta Biomater., 10, 843, 10.1016/j.actbio.2013.09.040

Fitton, 2015, Therapies from Fucoidan: An Update, Mar. Drugs, 13, 5920, 10.3390/md13095920

Wijesekara, 2011, Biological activities and potential health benefits of sulfated polysaccharides derived from marine algae, Carbohydr. Polym., 84, 14, 10.1016/j.carbpol.2010.10.062

Kalitnik, 2015, Oligosaccharides of κ/β-carrageenan from the red alga Tichocarpus crinitus and their ability to induce interleukin 10, J. Appl. Phycol., 1, 545

He, 2014, Arginine-based polyester amide/polysaccharide hydrogels and their biological response, Acta Biomater., 10, 2482, 10.1016/j.actbio.2014.02.011

Salabas, 2013, The pathophysiology of Peyronie’s disease, Arab J. Urol., 11, 272, 10.1016/j.aju.2013.06.006

He, 2019, Biodegradable amino acid-based poly(ester amine) with tunable immunomodulating properties and their in vitro and in vivo wound healing studies in diabetic rats’ wounds, Acta Biomater., 84, 114, 10.1016/j.actbio.2018.11.053

Singh, 2014, Hydrogels and scaffolds for immunomodulation, Adv. Mater., 26, 6530, 10.1002/adma.201402105

Vaday, 2000, Extracellular matrix moieties, cytokines, and enzymes: Dynamic effects on immune cell behavior and inflammation, J. Leukoc. Biol., 67, 149, 10.1002/jlb.67.2.149

Rowley, 2019, Extracellular Matrix-Based Strategies for Immunomodulatory Biomaterials Engineering, Adv. Healthc. Mater., 8, e1801578, 10.1002/adhm.201801578

Badylak, 2008, Immune response to biologic scaffold materials, Semin. Immunol., 20, 109, 10.1016/j.smim.2007.11.003

Taraballi, 2018, Biomimetic Tissue Engineering: Tuning the Immune and Inflammatory Response to Implantable Biomaterials, Adv. Healthc. Mater., 7, 1800490, 10.1002/adhm.201800490

Cramer, 2019, Extracellular matrix-based biomaterials and their influence upon cell behavior, Biomater. Eng. Cell Behav., 48, 2132

Huleihel, 2017, Macrophage phenotype in response to ECM bioscaffolds, Semin. Immunol, 29, 2, 10.1016/j.smim.2017.04.004

Abraham, 2000, Evaluation of the porcine intestinal collagen layer as a biomaterial, J. Biomed. Mater. Res., 51, 442, 10.1002/1097-4636(20000905)51:3<442::AID-JBM19>3.0.CO;2-4

Parmaksiz, 2016, Clinical applications of decellularized extracellular matrices for tissue engineering and regenerative medicine, Biomed. Mater., 11, 022003, 10.1088/1748-6041/11/2/022003

Mokhtari, 2019, An injectable mechanically robust hydrogel of Kappa-carrageenan-dopamine functionalized graphene oxide for promoting cell growth, Carbohydr. Polym., 214, 234, 10.1016/j.carbpol.2019.03.030

Tavakoli, 2021, Nanocomposite hydrogel based on carrageenan-coated starch/cellulose nanofibers as a hemorrhage control material, Carbohydr. Polym., 251, 117013, 10.1016/j.carbpol.2020.117013

Tavakoli, 2019, Sprayable and injectable visible-light Kappa-carrageenan hydrogel for in-situ soft tissue engineering, Int. J. Biol. Macromol., 138, 590, 10.1016/j.ijbiomac.2019.07.126

Tavakoli, 2020, A multifunctional nanocomposite spray dressing of Kappa-carrageenan-polydopamine modified ZnO/L-glutamic acid for diabetic wounds, Mater. Sci. Eng. C, 111, 110837, 10.1016/j.msec.2020.110837

Yu, 2018, Injectable Bioresponsive Gel Depot for Enhanced Immune Checkpoint Blockade, Adv. Mater., 30, e1801527, 10.1002/adma.201801527

Yang, 2018, Engineering Dendritic-Cell-Based Vaccines and PD-1 Blockade in Self-Assembled Peptide Nanofibrous Hydrogel to Amplify Antitumor T-Cell Immunity, ACS Publ., 18, 4377

Sun, 2018, Injectable Hydrogels Coencapsulating Granulocyte-Macrophage Colony-Stimulating Factor and Ovalbumin Nanoparticles to Enhance Antigen Uptake Efficiency, ACS Appl. Mater. Interfaces, 10, 20315, 10.1021/acsami.8b04312

Zaveri, 2014, Integrin-directed modulation of macrophage responses to biomaterials, Biomaterials, 35, 3504, 10.1016/j.biomaterials.2014.01.007

Wilgus, 2008, Immune cells in the healing skin wound: Influential players at each stage of repair, Pharmacol. Res., 58, 112, 10.1016/j.phrs.2008.07.009

Jones, 2007, Proteomic analysis and quantification of cytokines and chemokines from biomaterial surface-adherent macrophages and foreign body giant cells, J. Biomed. Mater. Res. Part. A, 83, 585, 10.1002/jbm.a.31221

Curtis, 1997, Topographical control of cells, Biomaterials, 18, 1573, 10.1016/S0142-9612(97)00144-0

Akira, 2004, Toll-like receptor signalling, Nat. Rev. Immunol., 4, 499, 10.1038/nri1391

Zakrzewski, 2014, Overcoming immunological barriers in regenerative medicine, Nat. Biotechnol., 32, 786, 10.1038/nbt.2960

Babensee, 2005, Differential levels of dendritic cell maturation on different biomaterials used in combination products, J. Biomed. Mater. Res. Part A, 74, 503, 10.1002/jbm.a.30429

Zhou, 1996, CD14+ blood monocytes can differentiate into functionally mature CD83+ dendritic cells, Proc. Natl. Acad. Sci. USA, 93, 2588, 10.1073/pnas.93.6.2588

Banchereau, 1998, Dendritic cells and the control of immunity, Nature, 392, 245, 10.1038/32588

Adelmeijer, 2006, Collagens are functional, high affinity ligands for the inhibitory immune receptor LAIR-1, J. Exp. Med., 203, 1419, 10.1084/jem.20052554

Chattopadhyay, 2014, Review collagen-based biomaterials for wound healing, Biopolymers, 101, 821, 10.1002/bip.22486

Witherel, 2016, Response of human macrophages to wound matrices in vitro, Wound Repair Regen., 24, 514, 10.1111/wrr.12423

Orlandi, 2018, Long-term follow-up comparison of two different bi-layer dermal substitutes in tissue regeneration: Clinical outcomes and histological findings, Int. Wound J., 15, 695, 10.1111/iwj.12912

Agrawal, 2012, Macrophage phenotypes correspond with remodeling outcomes of various acellular dermal matrices, Open J. Regen. Med., 1, 25919

Podolnikova, 2003, Identification of a Novel Binding Site for Platelet Integrins IIb 3 (GPIIbIIIa) and 5 1 in the C-domain of Fibrinogen, J. Biol. Chem., 278, 32251, 10.1074/jbc.M300410200

Clark, R.A.F. (1988). Wound Repair. the Molecular and Cellular Biology of Wound Repair, Springer.

(2017). Local induction of lymphangiogenesis with engineered fibrin-binding VEGF-C promotes wound healing by increasing immune cell trafficking and matrix. Biomaterials, 131, 160–175. Available online: https://www.sciencedirect.com/science/article/pii/S0142961217301825.

Mokarram, 2014, A perspective on immunomodulation and tissue repair, Ann. Biomed. Eng., 42, 338, 10.1007/s10439-013-0941-0

Hashimoto, 2020, Gene expression advances skin reconstruction and wound repair better on silk fibroin-based materials than on collagen-based materials, Materialia, 9, 100519, 10.1016/j.mtla.2019.100519

Burdick, 2011, Hyaluronic acid hydrogels for biomedical applications, Adv. Mater., 23, 41, 10.1002/adma.201003963

Wang, H., Morales, R.T.T., Cui, X., Huang, J., Qian, W., Tong, J., and Chen, W. (2019). A Photoresponsive Hyaluronan Hydrogel Nanocomposite for Dynamic Macrophage Immunomodulation. Adv. Healthc. Mater., 8.

Zamboni, 2018, The potential of hyaluronic acid in immunoprotection and immunomodulation: Chemistry, processing and function, Prog. Mater., 97, 97, 10.1016/j.pmatsci.2018.04.003

Ruppert, 2014, Tissue integrity signals communicated by high-molecular weight hyaluronan and the resolution of inflammation, Immunol Res., 58, 186, 10.1007/s12026-014-8495-2

Saini, 2020, Immunomodulatory Properties of Chitosan: Impact on Wound Healing and Tissue Repair, Endocr Metab. Immune. Disord Drug. Targets, 20, 1611, 10.2174/1871530320666200503054605

Dai, 2011, Chitosan preparations for wounds and burns: Antimicrobial and wound-healing effects, Expert Rev. Anti. Infect. Ther., 9, 857, 10.1586/eri.11.59