Reduced FOXO1 Expression Accelerates Skin Wound Healing and Attenuates Scarring  Ryoichi Mori, Katsuya Tanaka, Maiko de Kerckhove, Momoko Okamoto, Kazuya.

Slides:



Advertisements
Similar presentations
Christina K. Marko, Balaraj B. Menon, Gang Chen, Jeffrey A
Advertisements

Lack of Platelet Endothelial Cell Adhesion Molecule-1 Attenuates Foreign Body Inflammation because of Decreased Angiogenesis  Anna Solowiej, Purba Biswas,
TRPV1 Involvement in Inflammatory Tissue Fibrosis in Mice
Monocyte/Macrophage MMP-14 Modulates Cell Infiltration and T-Cell Attraction in Contact Dermatitis But Not in Murine Wound Healing  Anke Klose, Paola.
Gurmel S. Sidhu, Anoop K. Singh, Krishna K. Banaudha, Jaya P
Interleukin-22 Promotes Wound Repair in Diabetes by Improving Keratinocyte Pro- Healing Functions  Simona Avitabile, Teresa Odorisio, Stefania Madonna,
MicroRNA-31 Promotes Skin Wound Healing by Enhancing Keratinocyte Proliferation and Migration  Dongqing Li, X.I. Li, Aoxue Wang, Florian Meisgen, Andor.
Pharmacological Mobilization of Endogenous Stem Cells Significantly Promotes Skin Regeneration after Full-Thickness Excision: The Synergistic Activity.
Integrin α2β1 Is Required for Regulation of Murine Wound Angiogenesis but Is Dispensable for Reepithelialization  Manon C. Zweers, Jeffrey M. Davidson,
S100A12 Induced in the Epidermis by Reduced Hydration Activates Dermal Fibroblasts and Causes Dermal Fibrosis  Jingling Zhao, Aimei Zhong, Emily E. Friedrich,
Athena Kalyvas, Samuel David  Neuron 
Development of Cell-Penetrating Asymmetric Interfering RNA Targeting Connective Tissue Growth Factor  Jihye Hwang, Chanil Chang, Ji Hyun Kim, Chang Taek.
Latency-Associated Peptide Prevents Skin Fibrosis in Murine Sclerodermatous Graft- Versus-Host Disease, a Model for Human Scleroderma  Yan Zhang, Laura.
Counterregulation of Interleukin-18 mRNA and Protein Expression During Cutaneous Wound Repair in Mice  Heiko Kämpfer, Heiko Mühl, Josef Pfeilschifter,
Gregory D. Rak, Lisa C. Osborne, Mark C. Siracusa, Brian S
MiR-142 Is Required for Staphylococcus aureus Clearance at Skin Wound Sites via Small GTPase-Mediated Regulation of the Neutrophil Actin Cytoskeleton 
Volume 133, Issue 6, Pages (December 2007)
Enhanced Inflammation and Accelerated Wound Closure Following Tetraphorbol Ester Application or Full-Thickness Wounding in Mice Lacking Hyaluronan Synthases.
Regulation and Function of the Caspase-1 in an Inflammatory Microenvironment  Dai-Jen Lee, Fei Du, Shih-Wei Chen, Manando Nakasaki, Isha Rana, Vincent.
Galectin-1 Accelerates Wound Healing by Regulating the Neuropilin-1/Smad3/NOX4 Pathway and ROS Production in Myofibroblasts  Yueh-Te Lin, Jhih-Sian Chen,
Ectodysplasin A Pathway Contributes to Human and Murine Skin Repair
Accelerated Wound Repair in ADAM-9 Knockout Animals
Tumor Necrosis Factor-α-Activated Human Adipose Tissue–Derived Mesenchymal Stem Cells Accelerate Cutaneous Wound Healing through Paracrine Mechanisms 
Volume 18, Issue 10, Pages (October 2010)
Manuela Schmidt, Danny Gutknecht, Jan C
Exploring the “Hair Growth–Wound Healing Connection”: Anagen Phase Promotes Wound Re-Epithelialization  David M. Ansell, Jennifer E. Kloepper, Helen A.
Volume 24, Issue 13, Pages e5 (September 2018)
The Cathelicidin Anti-Microbial Peptide LL-37 is Involved in Re-Epithelialization of Human Skin Wounds and is Lacking in Chronic Ulcer Epithelium  Johan.
Volume 25, Issue 6, Pages (June 2017)
Raija Tammi  Journal of Investigative Dermatology 
Impaired Skin Regeneration and Remodeling after Cutaneous Injury and Chemically Induced Hyperplasia in Taps-Transgenic Mice  Maike Hildenbrand, Verena.
Volume 24, Issue 12, Pages (June 2014)
Toll-Like Receptor 4 Has an Essential Role in Early Skin Wound Healing
Delayed Wound Healing and Epidermal Hyperproliferation in Mice Lacking JunB in the Skin  Lore Florin, Julia Knebel, Paola Zigrino, Birgitta Vonderstrass,
Georgios Theocharidis, Zoe Drymoussi, Alexander P. Kao, Asa H
Mohammad Rashel, Ninche Alston, Soosan Ghazizadeh 
Calmodulin-Like Protein Upregulates Myosin-10 in Human Keratinocytes and Is Regulated during Epidermal Wound Healing In Vivo  Richard D. Bennett, Amy.
Sustained Activation of Fibroblast Transforming Growth Factor-β/Smad Signaling in a Murine Model of Scleroderma  Shinsuke Takagawa, Gabriella Lakos, Yasuji.
Lack of Collagen VI Promotes Wound-Induced Hair Growth
The Neurofibromatosis Type 1 (Nf1) Tumor Suppressor is a Modifier of Carcinogen- Induced Pigmentation and Papilloma Formation in C57BL/6 Mice  Radhika.
Overexpression of CD109 in the Epidermis Differentially Regulates ALK1 Versus ALK5 Signaling and Modulates Extracellular Matrix Synthesis in the Skin 
IL-22 Promotes Fibroblast-Mediated Wound Repair in the Skin
Collagen VII Half-Life at the Dermal-Epidermal Junction Zone: Implications for Mechanisms and Therapy of Genodermatoses  Tobias Kühl, Markus Mezger, Ingrid.
Tropomyosin Regulates Cell Migration during Skin Wound Healing
Mathieu P. Rodero, Samantha S
Molecular Therapy - Nucleic Acids
Epithelial Cells in the Hair Follicle Bulge do not Contribute to Epidermal Regeneration after Glucocorticoid-Induced Cutaneous Atrophy  Dmitry V. Chebotaev,
Wound Healing Is Defective in Mice Lacking Tetraspanin CD151
John T. Walker, Christopher G. Elliott, Thomas L. Forbes, Douglas W
Increased Severity of Bleomycin-Induced Skin Fibrosis in Mice with Leukocyte-Specific Protein 1 Deficiency  JianFei Wang, Haiyan Jiao, Tara L. Stewart,
The Suppressor of Cytokine Signaling (SOCS)-3 Determines Keratinocyte Proliferative and Migratory Potential during Skin Repair  Andreas Linke, Itamar.
Volume 7, Issue 1, Pages (January 2008)
Epidermal Inactivation of the Glucocorticoid Receptor Triggers Skin Barrier Defects and Cutaneous Inflammation  Lisa M. Sevilla, Víctor Latorre, Ana Sanchis,
The Nf1 Tumor Suppressor Regulates Mouse Skin Wound Healing, Fibroblast Proliferation, and Collagen Deposited by Fibroblasts  Radhika P. Atit, Maria J.
YAP and TAZ Regulate Skin Wound Healing
SPARC-Null Mice Display Abnormalities in the Dermis Characterized by Decreased Collagen Fibril Diameter and Reduced Tensile Strength  Amy D. Bradshaw,
Age-Related Alterations in the Inflammatory Response to Dermal Injury
Increased Expression of Wnt2 and SFRP4 in Tsk Mouse Skin: Role of Wnt Signaling in Altered Dermal Fibrillin Deposition and Systemic Sclerosis  Julie Bayle,
Normal Wound Healing in Mice Deficient for Fibulin-5, an Elastin Binding Protein Essential for Dermal Elastic Fiber Assembly  Qian Zheng, Jiwon Choi,
High-Mobility Group Box 1 Protein in Human and Murine Skin: Involvement in Wound Healing  Stefania Straino, Anna Di Carlo, Antonella Mangoni, Roberta.
Jun Asai, Hideya Takenaka, Norito Katoh, Saburo Kishimoto 
Zhong Zheng, Calvin Nguyen, Xinli Zhang, Hooman Khorasani, Joyce Z
Holly N. Wilkinson, Christopher Clowes, Kayleigh L
A Collagen-based Scaffold Delivering Exogenous MicroRNA-29B to Modulate Extracellular Matrix Remodeling  Michael Monaghan, Shane Browne, Katja Schenke-Layland,
Kallikrein-Related Peptidase 8–Dependent Skin Wound Healing Is Associated with Upregulation of Kallikrein-Related Peptidase 6 and PAR2  Mari Kishibe,
Comparison of Mouse Matrix Metalloproteinase 13 Expression in Free-Electron Laser and Scalpel Incisions During Wound Healing  Nanjun Wu, E. Duco Jansen,
Keratinocyte-Derived Granulocyte-Macrophage Colony Stimulating Factor Accelerates Wound Healing: Stimulation of Keratinocyte Proliferation, Granulation.
Delayed Wound Healing in CXCR2 Knockout Mice
Role of TGFβ-Mediated Inflammation in Cutaneous Wound Healing
Matrix Metalloproteinase Inhibitor BB-3103 Unlike the Serine Proteinase Inhibitor Aprotinin Abrogates Epidermal Healing of Human Skin Wounds Ex Vivo1 
Presentation transcript:

Reduced FOXO1 Expression Accelerates Skin Wound Healing and Attenuates Scarring  Ryoichi Mori, Katsuya Tanaka, Maiko de Kerckhove, Momoko Okamoto, Kazuya Kashiyama, Katsumi Tanaka, Sangeun Kim, Takuya Kawata, Toshimitsu Komatsu, Seongjoon Park, Kazuya Ikematsu, Akiyoshi Hirano, Paul Martin, Isao Shimokawa  The American Journal of Pathology  Volume 184, Issue 9, Pages 2465-2479 (September 2014) DOI: 10.1016/j.ajpath.2014.05.012 Copyright © 2014 American Society for Investigative Pathology Terms and Conditions

Figure 1 Skin wound healing is accelerated in Foxo1+/− mice. A: Gene expression of murine Foxo family in skin wound healing measured by quantitative PCR relative to 18S ribosomal RNA (n = 4 to 6). B: Representative images for gross appearances of excisional wound in WT (white bars) and Foxo1+/− (black bars) mice. C: The proportion of the wound remaining open relative to the initial wound area at each time point (n = 12). D: IHC for FOXO1 and neutrophils shows neutrophils, epithelium, and hair follicle expressing FOXO1 1 day after injury in WT mice. Nuclei were counterstained with DAPI [wound margin (white arrowhead), leading edge of epithelia (arrow), and FOXO1-expressing neutrophils (open arrowheads)]. IHC of FOXO1 and F4/80 or CD31 shows wound-infiltrated macrophages (E), fibroblasts (open arrowhead, E), and endothelial cells (F) at 7 days after injury in WT mice. G: H&E staining of re-epithelialization [wound margin (arrowheads) and the leading edge of epithelia (arrows)]. Top panels: The higher-power field of the bottom panels. H: Measurement of epithelial tongue at 3 days after injury (n = 13). I: Middle of wound tissue at 14 days after injury stained with Masson’s trichrome, and the extent (cross-sectional area) of granulation tissue visualized and quantified at the midpoint of the wound (dotted lines) [wound margin (arrowheads)]. J: Quantification of the area of granulation tissue 14 days after injury (n = 6). All values represent means ± SEM. ∗P < 0.05, ∗∗P < 0.01, and ∗∗∗P < 0.001. Scale bars: 100 μm (D and bottom panels in G); 10 μm (E and F); 500 μm (high-power fields in G and I). The American Journal of Pathology 2014 184, 2465-2479DOI: (10.1016/j.ajpath.2014.05.012) Copyright © 2014 American Society for Investigative Pathology Terms and Conditions

Figure 2 Attenuation of neutrophil and macrophage infiltration and inflammatory responses at wound sites of Foxo1+/− mice. A: IHC for neutrophils (red) showing the number of wound-infiltrated neutrophils at wound sites 1 day after injury, which is reduced in Foxo1+/− mice compared with WT mice (nuclei were counterstained with DAPI). B: MPO concentration using ELISA reveals that MPO levels at wound sites of Foxo1+/− mice are significantly reduced compared with WT mice (n = 8 per group). C: IHC for macrophages using F4/80 at the middle area of wound sites 7 days after injury. D: Quantification of F4/80-positive macrophages at wound sites of WT mice (n = 6 per group for 3 days, and n = 5 per group for 7 days) and Foxo1+/− mice (n = 6 per group for both 3 and 7 days). E: Western immunoblot analysis of pNF-κB p65 and total NF-κB. Full scans for Western immunoblots in Supplemental Figure S5A. F: Densitometric analysis for pNF-κB activity over total NF-κB (n = 4 to 5). All values represent means ± SEM. ∗P < 0.05. Scale bar = 10 μm (A and C). ND, not detected. The American Journal of Pathology 2014 184, 2465-2479DOI: (10.1016/j.ajpath.2014.05.012) Copyright © 2014 American Society for Investigative Pathology Terms and Conditions

Figure 3 ERK1/2 activation and myosin IIb expression are enhanced at wound sites of Foxo1+/− mice. A: FOXO1 binding activity in the nuclear wound tissue extract was measured by immobilized oligonucleotide ELISA. FOXO1 consensus oligonucleotide-treated extracts were used as negative control (n = 2 to 4). B: Western immunoblot analysis of wound tissue at 3 days shows weak expression of total FOXO1 and pFOXO1 (Thr24) at wound sites of Foxo1+/− mice. Bands for total FOXO3A, pFOXO3A (Ser318/321), and FOXO4 remain unchanged in both groups. Full scans for Western immunoblot analysis in Supplemental Figure S5B. C: Western immunoblot analysis of pERK1/2 (Thr202/Tyr204), total ERK1/2, pAKT (Ser473), total AKT, and myosin IIb. Full scans for Western immunoblots in Supplemental Figure S5C. D: Densitometric analysis of pERK1/2 (Thr202/Tyr204) and pAKT (Ser473) over total ERK1/2 and AKT, respectively, and myosin IIb (n = 4 to 5). E: Wound scratch assay 24 hours after treatment of mFGF2 in PKs with control (left panel) or Foxo1 (right panel) siRNAs. F: Wound closure ratio (n = 4). G: Confocal images of pERK in mFGF2-treated control PKs (left panel) and Foxo1 siRNA-treated PKs (right panel) 24 hours after scratching (n = 3). H: Fluorescence intensity of pERK by mFGF2 24 hours after scratching in control PKs and Foxo1 siRNA-treated PKs (n = 3). All values represent means ± SEM. ∗P < 0.05. Scale bars: 100 μm (E); 50 μm (G). The American Journal of Pathology 2014 184, 2465-2479DOI: (10.1016/j.ajpath.2014.05.012) Copyright © 2014 American Society for Investigative Pathology Terms and Conditions

Figure 4 Scarring at wound sites is attenuated in Foxo1+/− mice. A: Gross appearance of scarring at the incisional wound 21 days after injury (asterisk indicates wound edge). Images shown are representative of six independent experiments. B: Picrosirius red–stained sections of incisional wound sites at 21 days after injury for analysis of collagen fibers and alignments [type I collagen (red and yellow), type III collagen (green), and wound edge (arrowheads)]. Granulation tissue was visualized at the midpoint of the wound (dotted lines). Images shown are representative of eight independent experiments. Low magnifications were taken as nonpolarized images. High-magnification details from boxed areas indicated are differential interference contract images using polarized light microscopy. C: TEM images of connective tissue from midwound sites 21 days after injury. High-magnification insets illustrate differing collagen fibril diameters in this tissue. D: Histogram of total range of fibril diameters in the wound site 21 days after injury (n = 1090 fibrils from three WT mice, and n = 1354 fibrils from three Foxo1+/− mice). Fibril diameters at wound sites of Foxo1+/− mice tend to be smaller than in WT mice. E: Quantification of vacant extracellular spaces in wound sites at 21 days (n = 3 to 4). F: Quantification of gene expression of Col1α1 7 days after injury at wound sites (measured by quantitative PCR), relative to 18S ribosomal RNA (n = 8). All values represent means ± SEM. ∗P < 0.05. Scale bars: 50 μm (B); 1 μm (C); 100 nm (inset, C). The American Journal of Pathology 2014 184, 2465-2479DOI: (10.1016/j.ajpath.2014.05.012) Copyright © 2014 American Society for Investigative Pathology Terms and Conditions

Figure 5 Skin wound healing is accelerated in Foxo1 AS ODN–treated wounds. A: Optimization of Foxo1 AS ODNs in vitro. Cleavage was visualized when transcribed Foxo1 mRNA was incubated with control and AS ODNs in vitro (n = 2 independent experiments) (M1 and M2 = RNA loading markers). Sequences are shown in Table 2. Foxo1 AS ODN (1717) is more efficient than other Foxo1 AS ODNs. B: Western immunoblot analysis from wound sites (6 hours after injury) to detect the effective dose of Foxo1 AS ODN (1717) to decrease FOXO1 protein expression in wounds in vivo. Full scans for Western immunoblots in Supplemental Figure S5D. C: Quantification of FOXO1 protein expression in wound sites exposed to 10 μmol/L Foxo1 AS ODN (1717) (n = 6 per group) reveals that FOXO1 is significantly reduced. Data are presented as the means ± SEM and analyzed by Tukey’s multiple-comparison tests. D: Representative photomicrographs for gross appearances of excisional control and Foxo1 AS ODN–treated wounds at various time points after wounding (n = 12). E: The proportion of the wound remaining open relative to the initial wound area at each time point after the injury in control versus Foxo1 AS ODN–treated wounds (n = 12 per group). All values represent means ± SEM. ∗P < 0.05, ∗∗P < 0.01. The American Journal of Pathology 2014 184, 2465-2479DOI: (10.1016/j.ajpath.2014.05.012) Copyright © 2014 American Society for Investigative Pathology Terms and Conditions

Figure 6 Expression of FOXO1 at human keloids. A–C: IHC for FOXO1 shows that FOXO1 (brown) is highly present in basal lamina keratinocytes at keloid sites in the Japanese population [nuclei are stained with hematoxylin (violet)] [FOXO1-expressing cells (arrowheads)]. D and E: FOXO1 is present in normal epidermis and dermis in the immediate vicinity of keloid sites compared with mature keloid sites. F: IHC for FOXO1 shows highly prominent FOXO1-positive cells at the surface and deep in keloid sites of African Americans compared with Japanese [mature deep keloid sites are shown (inset)]. Micrographs are representative of three and six sections for D–F and A–C, respectively. G: Percentage of FOXO1-positive cells at the intact skin in the immediate vicinity of keloid sites (three cases) and in keloid sites (six cases). H: Percentage of FOXO1-positive cells at mature keloid sites of an African American (three cases) and two Japanese (six cases) patients. I: A proposed model of the exacerbation of keloids by FOXO1. Elevated expression of FOXO1 at the epidermis may cause hyperplasia of keloids. In contrast, numbers of FOXO1-expressing cells at deeply mature keloid sites are markedly reduced. Intact skin in the vicinity of keloids appears normal. However, expression of FOXO1 is markedly increased compared with mature keloid, suggesting that FOXO1-positive cells are associated with keloid expansion. Collectively, many FOXO1-expressing cells produce collagen and enhance the inflammatory response, leading to the exacerbation of keloid scarring. All values represent means ± SEM. ∗P < 0.05. Scale bars: 500 μm (A and D); 100 μm (B, C, E, and F). The American Journal of Pathology 2014 184, 2465-2479DOI: (10.1016/j.ajpath.2014.05.012) Copyright © 2014 American Society for Investigative Pathology Terms and Conditions