Mesenchymal Stromal Cell-Derived PTX3 Promotes Wound Healing via Fibrin Remodeling  Claudia Cappuzzello, Andrea Doni, Erica Dander, Fabio Pasqualini,

Slides:



Advertisements
Similar presentations
Volume 132, Issue 1, Pages (January 2007)
Advertisements

Monocyte/Macrophage MMP-14 Modulates Cell Infiltration and T-Cell Attraction in Contact Dermatitis But Not in Murine Wound Healing  Anke Klose, Paola.
Matrix Metalloproteinase-9 Is Required for Tumor Vasculogenesis but Not for Angiogenesis: Role of Bone Marrow-Derived Myelomonocytic Cells  G-One Ahn,
Volume 39, Issue 5, Pages (November 2013)
The Suppressor of Cytokine Signaling-3 Is Upregulated in Impaired Skin Repair: Implications for Keratinocyte Proliferation  Itamar Goren, Andreas Linke,
Antimicrobial Peptides Human β-Defensins Stimulate Epidermal Keratinocyte Migration, Proliferation and Production of Proinflammatory Cytokines and Chemokines 
Glycoprotein Nonmelanoma Clone B Regulates the Crosstalk between Macrophages and Mesenchymal Stem Cells toward Wound Repair  Bing Yu, Talib Alboslemy,
Interleukin-22 Promotes Wound Repair in Diabetes by Improving Keratinocyte Pro- Healing Functions  Simona Avitabile, Teresa Odorisio, Stefania Madonna,
Critical Role of 5-Lipoxygenase and Heme Oxygenase-1 in Wound Healing
Volume 44, Issue 3, Pages (March 2016)
Identification of Bone Marrow-Derived Soluble Factors Regulating Human Mesenchymal Stem Cells for Bone Regeneration  Tsung-Lin Tsai, Wan-Ju Li  Stem Cell.
Kynurenine Increases Matrix Metalloproteinase-1 and -3 Expression in Cultured Dermal Fibroblasts and Improves Scarring In Vivo  Yunyuan Li, Ruhangiz T.
IL-21 Reduces Immediate Hypersensitivity Reactions in Mouse Skin by Suppressing Mast Cell Activation or IgE Production  Risa Tamagawa-Mineoka, Tsunao.
Gregory D. Rak, Lisa C. Osborne, Mark C. Siracusa, Brian S
CD90+ Human Dermal Stromal Cells Are Potent Inducers of FoxP3+ Regulatory T Cells  Karin Pfisterer, Karoline M. Lipnik, Erhard Hofer, Adelheid Elbe-Bürger 
Mesenchymal Stem Cells (MSCs) Attenuate Cutaneous Sclerodermatous Graft-Versus- Host Disease (Scl-GVHD) through Inhibition of Immune Cell Infiltration.
Jackie R. Bickenbach, Matthew M. Stern, Katie L
Epstein–Barr Virus Infection Induces Aberrant TLR Activation Pathway and Fibroblast– Myofibroblast Conversion in Scleroderma  Antonella Farina, Mara Cirone,
TSG-6 Released from Intradermally Injected Mesenchymal Stem Cells Accelerates Wound Healing and Reduces Tissue Fibrosis in Murine Full-Thickness Skin.
Volume 132, Issue 1, Pages (January 2007)
Kruppel-Like Factor KLF4 Facilitates Cutaneous Wound Healing by Promoting Fibrocyte Generation from Myeloid-Derived Suppressor Cells  Lingling Ou, Ying.
Human renal epithelial cells produce the long pentraxin PTX3
Inhibition of UVB-Induced Skin Tumor Development by Drinking Green Tea Polyphenols Is Mediated Through DNA Repair and Subsequent Inhibition of Inflammation 
Regulation and Function of the Caspase-1 in an Inflammatory Microenvironment  Dai-Jen Lee, Fei Du, Shih-Wei Chen, Manando Nakasaki, Isha Rana, Vincent.
Stromal Fibroblast–Specific Expression of ADAM-9 Modulates Proliferation and Apoptosis in Melanoma Cells In Vitro and In Vivo  Anna N. Abety, Jay W. Fox,
Accelerated Wound Repair in ADAM-9 Knockout Animals
Volume 39, Issue 5, Pages (November 2013)
Toshiyuki Ozawa, Daisuke Tsuruta, Jonathan C. R
Manuela Schmidt, Danny Gutknecht, Jan C
Volume 24, Issue 13, Pages e5 (September 2018)
Spleen Tyrosine Kinase Mediates EGFR Signaling to Regulate Keratinocyte Terminal Differentiation  Nan-Lin Wu, Duen-Yi Huang, Li-Fang Wang, Reiji Kannagi,
Volume 134, Issue 3, Pages (March 2008)
Reversal of CD8 T-Cell–Mediated Mucocutaneous Graft-Versus-Host-Like Disease by the JAK Inhibitor Tofacitinib  Naoko Okiyama, Yasuko Furumoto, Vadim A.
Impaired Skin Regeneration and Remodeling after Cutaneous Injury and Chemically Induced Hyperplasia in Taps-Transgenic Mice  Maike Hildenbrand, Verena.
Toll-Like Receptor 4 Has an Essential Role in Early Skin Wound Healing
Natural Killer T Cells Are Essential for the Development of Contact Hypersensitivity in BALB/c Mice  Chihiro Shimizuhira, Atsushi Otsuka, Tetsuya Honda,
Volume 7, Issue 2, Pages (February 2010)
Capsiate Inhibits DNFB-Induced Atopic Dermatitis in NC/Nga Mice through Mast Cell and CD4+ T-Cell Inactivation  Ji H. Lee, Yun S. Lee, Eun-Jung Lee, Ji.
Georgios Theocharidis, Zoe Drymoussi, Alexander P. Kao, Asa H
Mohammad Rashel, Ninche Alston, Soosan Ghazizadeh 
Min Qin, Aslan Pirouz, Myung-Hwa Kim, Stephan R. Krutzik, Hermes J
C/EBPγ Regulates Wound Repair and EGF Receptor Signaling
Prolonged Activation of ERK Contributes to the Photorejuvenation Effect in Photodynamic Therapy in Human Dermal Fibroblasts  Yong Hyun Jang, Gi-Bang Koo,
Anne T. Funding, Claus Johansen, Matthias Gaestel, Bo M
IL-22 Promotes Fibroblast-Mediated Wound Repair in the Skin
Min Qin, Aslan Pirouz, Myung-Hwa Kim, Stephan R. Krutzik, Hermes J
Mathieu P. Rodero, Samantha S
Matrix Metalloproteinase-9 Is Required for Tumor Vasculogenesis but Not for Angiogenesis: Role of Bone Marrow-Derived Myelomonocytic Cells  G-One Ahn,
Adam Mor, Roni Haklai, Ofer Ben-Moshe, Yoseph A. Mekori, Yoel Kloog 
John T. Walker, Christopher G. Elliott, Thomas L. Forbes, Douglas W
The Suppressor of Cytokine Signaling (SOCS)-3 Determines Keratinocyte Proliferative and Migratory Potential during Skin Repair  Andreas Linke, Itamar.
Nrf2 Promotes Keratinocyte Proliferation in Psoriasis through Up-Regulation of Keratin 6, Keratin 16, and Keratin 17  Luting Yang, Xueli Fan, Tingting.
Volume 44, Issue 3, Pages (March 2016)
A Role for Estrogen Receptor-α and Estrogen Receptor-β in Collagen Biosynthesis in Mouse Skin  Margaret Markiewicz, Sergey Znoyko, Lukasz Stawski, Angela.
Maria C. Lebre, Angelic M. G. van der Aar, Lisa van Baarsen, Toni M. M
Green Tea Polyphenol Epigallocatechin-3-Gallate Suppresses Collagen Production and Proliferation in Keloid Fibroblasts via Inhibition of the STAT3-Signaling.
Increased Expression of Wnt2 and SFRP4 in Tsk Mouse Skin: Role of Wnt Signaling in Altered Dermal Fibrillin Deposition and Systemic Sclerosis  Julie Bayle,
TSLP Is a Potential Initiator of Collagen Synthesis and an Activator of CXCR4/SDF-1 Axis in Keloid Pathogenesis  Jung U Shin, Seo Hyeong Kim, Hyeran Kim,
Syed M. Meeran, Thejass Punathil, Santosh K. Katiyar 
Kim L. Kroeze, Wouter J. Jurgens, Behrouz Z. Doulabi, Florine J
Human Beta-Defensin 3 Induces Maturation of Human Langerhans Cell–Like Dendritic Cells: An Antimicrobial Peptide that Functions as an Endogenous Adjuvant 
HGF-Promoted Motility in Primary Human Melanocytes Depends on CD44v6 Regulated via NF-kappa B, Egr-1, and C/EBP-beta  Sabine Damm, Petra Koefinger, Martina.
High-Mobility Group Box 1 Protein in Human and Murine Skin: Involvement in Wound Healing  Stefania Straino, Anna Di Carlo, Antonella Mangoni, Roberta.
TAK1 Is Required for Dermal Wound Healing and Homeostasis
All-Trans Retinoic Acid Antagonizes UV-Induced VEGF Production and Angiogenesis via the Inhibition of ERK Activation in Human Skin Keratinocytes  Mi-Sun.
The Angiogenesis Inhibitor Vasostatin does not Impair Wound Healing at Tumor- Inhibiting Doses  Bernhard Lange-Asschenfeldt, Paula Velasco, Michael Streit,
Jackie R. Bickenbach, Matthew M. Stern, Katie L
Volume 25, Issue 6, Pages (June 2017)
by Gonghua Huang, Yanyan Wang, Peter Vogel, and Hongbo Chi
Interleukin-17 is Produced by Both Th1 and Th2 Lymphocytes, and Modulates Interferon-γ- and Interleukin-4-Induced Activation of Human Keratinocytes  Cristina.
Presentation transcript:

Mesenchymal Stromal Cell-Derived PTX3 Promotes Wound Healing via Fibrin Remodeling  Claudia Cappuzzello, Andrea Doni, Erica Dander, Fabio Pasqualini, Manuela Nebuloni, Barbara Bottazzi, Alberto Mantovani, Andrea Biondi, Cecilia Garlanda, Giovanna D’Amico  Journal of Investigative Dermatology  Volume 136, Issue 1, Pages 293-300 (January 2016) DOI: 10.1038/JID.2015.346 Copyright © 2015 The Authors Terms and Conditions

Figure 1 In vitro characterization of BM-derived Ptx3−/− and wt MSCs. (a) Box plot showing the PTX3 content in supernatants (n = 4) of cytokine-stimulated (st) wt MSCs. *P ≤ 0.05, versus unexposed MSCs (un). (b) PTX3 deficiency in Ptx3−/− MSCs and increased expression in the wt MSCs on cytokine exposure. (c) Representative FACS analysis (n = 3) of Ptx3−/− and wt MSCs (dotted line, control; black line, cells with antibody). (d) Microscopic view showing the lipid drop accumulation (left panel; Oil Red-O staining) and mineralization (right panel; Alizarin Red-S staining) after 3 weeks of culture in control (−) and induction medium (+). (e) Unstimulated MSC proliferation. (f) Expression of CD106, CD54, and CD274 after 24 hours of stimulation with IFN-γ and TNF-α. Black or red line: antigen expression of unstimulated or stimulated MSC, respectively; black-dotted line, isotype Ab. (g) Inhibition of lymphocyte proliferation by MSCs. BM, bone marrow; MSC, mesenchymal stromal cell; PTX3, pentraxin 3; Ptx3−/−, Ptx3-deficient; TNF-α, tumor necrosis factor-α. Journal of Investigative Dermatology 2016 136, 293-300DOI: (10.1038/JID.2015.346) Copyright © 2015 The Authors Terms and Conditions

Figure 2 Effect of Ptx3−/− MSCs on wound healing. (a) Kinetics of excisional wound closure in mice treated with Ptx3−/− (n = 33), wt MSCs (n = 45) and vehicle (sham) (n = 15), respectively. Values represent the mean percentage of wound closure ± SEM (n = 5 experiments), **P ≤ 0.01, and *P ≤ 0.05, Ptx3−/− versus wt MSCs. $P ≤ 0.05, Ptx3−/− MSCs versus sham. (b) Representative digital photographs of full-thickness excisional wounds taken immediately or at different time points after wounding. (c) Representative photographs of histology at day 7 (×20 magnification). c, clot; gt, granulation tissue. Bar = 1,000 μm. (d) Graph showing computer-assisted morphometrical analysis of the granulation tissue area (from 7 to 12 samples analyzed) at day 7. *P ≤ 0.05, Ptx3−/− MSCs versus wt MSCs or wt MSCs versus sham. MSC, mesenchymal stromal cell; Ptx3−/−, Ptx3-deficient. Journal of Investigative Dermatology 2016 136, 293-300DOI: (10.1038/JID.2015.346) Copyright © 2015 The Authors Terms and Conditions

Figure 3 Analysis of Ptx3−/− MSC recruitment to the wounded skin and colocalization of PTX3 expression with the pericellular matrix of MSCs. (a, b) Confocal images of wt (n = 5) and Ptx3−/− (n = 5) MSCs entering the clot (c) and wound bed (wb) (day 2). (a) ×10; (b) ×40. (a, b) left panels: merged images; right panels: staining of nucleus (blue, upper and lower panels) and PTX3 (cyan, upper panel), or nucleus and GFP (green, lower panel). Yellow arrowheads: localization of PTX3 at the pericellular matrix of MSCs invading the wound bed. (b) right: close-up of squared areas in (b) left. Bar = 200 μm (a) or 50 μm (b). (c) Western blotting and (d) densitometry of GFP and β-actin expression in wound extracts (day 2). Data are fold change mean ± SEM of four independent samples. Lysates from 1 sham wound and GFP+ cells (c) were loaded as controls. **P ≤ 0.01, versus wt MSCs. GFP, green fluorescent protein; MSC, mesenchymal stromal cell; PTX3, pentraxin 3; Ptx3−/−, Ptx3-deficient. Journal of Investigative Dermatology 2016 136, 293-300DOI: (10.1038/JID.2015.346) Copyright © 2015 The Authors Terms and Conditions

Figure 4 Fibrin-rich matrix remodeling in wounds treated with Ptx3−/− MSCs. (a) Representative confocal images (×10) of day 2 skin specimens (n = 5), showing fibrin immunostaining (red) in the wound bed (wb), and colocalization of PTX3 (cyan) with fibrin fibers. Upper panel: bundles of PTX3-expressing cells invading the loose fibrin matrix of wounded skins treated with wt MSCs. Bottom panel: confluent fibrin staining in the matrix of Ptx3−/− MSC-treated wounds. Left panels: merged images; right panels: single markers with nucleus (blue) are shown. C, clot; p, perilesion area. Bar = 200 μm. (b) Fibrin staining quantification in the wound bed of Ptx3−/− and wt MSC-treated skins. **P ≤ 0.01, Ptx3−/− MSC- versus wt MSC-treated wounds. (c) ELISA of D-dimer content in wound extracts at day 2. *P ≤ 0.05, Ptx3−/− MSC versus wt MSC-treated wounds. MSC, mesenchymal stromal cell; PTX3, pentraxin 3; Ptx3−/−, Ptx3-deficient. Journal of Investigative Dermatology 2016 136, 293-300DOI: (10.1038/JID.2015.346) Copyright © 2015 The Authors Terms and Conditions

Figure 5 In vitro pericellular fibrinolysis of Ptx3−/− MSCs. (a) ELISA of soluble fibrinolysis products generated by Ptx3−/− and wt MSCs embedded in fibrin gel with or without APR or PAI-1. Values are expressed as O.D. mean ± SEM of triplicates. Gray column represents medium without cells. **P ≤ 0.01. (b) Western blot analysis showing the decreased amount of fibrinolysis products in culture of Ptx3−/− MSCs and the fibrinolysis abolishment by plasminogen-conversion inhibitors. Mouse serum (1 μl/lane) was also loaded. α, β, and γ chains of mouse fibrinogen are shown. APR, aprotinin; MSC, mesenchymal stromal cell; PAI-1, plasminogen activator inhibitor-1; PTX3, pentraxin 3; Ptx3−/−, Ptx3-deficient. Journal of Investigative Dermatology 2016 136, 293-300DOI: (10.1038/JID.2015.346) Copyright © 2015 The Authors Terms and Conditions

Figure 6 Migration of Ptx3−/− MSCs through fibrin. Migration ability of Ptx3−/− and wt MSCs (n = 2) through uncoated (−) or fibrin-coated filters in response to CXCL12 (200 ng/ml). Results are expressed as migration index ± SEM of stimulated versus unstimulated cells. The migration index was calculated as the ratio of the number of cells migrating toward the CXCL12 gradient to the number of cells migrating toward medium alone. Five-six randomly selected fields/insert were evaluated in three independent experiments. **P ≤ 0.01; *P ≤ 0.05. MSC, mesenchymal stromal cell; Ptx3−/−, Ptx3-deficient. Journal of Investigative Dermatology 2016 136, 293-300DOI: (10.1038/JID.2015.346) Copyright © 2015 The Authors Terms and Conditions