Absence of γ-Chain in Keratinocytes Alters Chemokine Secretion, Resulting in Reduced Immune Cell Recruitment  Karolin Nowak, Daniela Linzner, Adrian J.

Slides:



Advertisements
Similar presentations
Volume 94, Issue 2, Pages (March 2014)
Advertisements

IL-18 Downregulates Collagen Production in Human Dermal Fibroblasts via the ERK Pathway  Hee Jung Kim, Seok Bean Song, Jung Min Choi, Kyung Moon Kim,
Platelet-Derived Growth Factor-BB Mediates Cell Migration through Induction of Activating Transcription Factor 4 and Tenascin-C  Kristine P. Malabanan,
Ganglioside GM3 Mediates Glucose-Induced Suppression of IGF-1 Receptor–Rac1 Activation to Inhibit Keratinocyte Motility  Duncan Hieu M. Dam, Xiao-Qi Wang,
MicroRNA-21 Expression in CD4+ T Cells Is Regulated by STAT3 and Is Pathologically Involved in Sézary Syndrome  Leslie van der Fits, Marloes S. van Kester,
IFN-γ Primes Keratinocytes for HSV-1–Induced Inflammasome Activation
Ganglioside GM3 Mediates Glucose-Induced Suppression of IGF-1 Receptor–Rac1 Activation to Inhibit Keratinocyte Motility  Duncan Hieu M. Dam, Xiao-Qi Wang,
Nickel Sulfate Promotes IL-17A Producing CD4+ T Cells by an IL-23-Dependent Mechanism Regulated by TLR4 and Jak-STAT Pathways  Rami Bechara, Diane Antonios,
PFKFB3, a Direct Target of p63, Is Required for Proliferation and Inhibits Differentiation in Epidermal Keratinocytes  Robert B. Hamanaka, Gökhan M. Mutlu 
Nickel Sulfate Promotes IL-17A Producing CD4+ T Cells by an IL-23-Dependent Mechanism Regulated by TLR4 and Jak-STAT Pathways  Rami Bechara, Diane Antonios,
Membrane-Tethered Intracellular Domain of Amphiregulin Promotes Keratinocyte Proliferation  Stefan W. Stoll, Philip E. Stuart, Sylviane Lambert, Alberto.
Cdc42 Inhibits ERK-Mediated Collagenase-1 (MMP-1) Expression in Collagen-Activated Human Keratinocytes  Maryam G. Rohani, Brian K. Pilcher, Peter Chen,
TWEAK/Fn14 Activation Contributes to the Pathogenesis of Bullous Pemphigoid  Yale Liu, Lingling Peng, Liang Li, Chengfei Liu, Xiao Hu, Shengxiang Xiao,
Requirement of Gαi1/3–Gab1 Signaling Complex for Keratinocyte Growth Factor– Induced PI3K–AKT–mTORC1 Activation  Yi-ming Zhang, Zhi-qing Zhang, Yuan-yuan.
IL-21 May Promote Granzyme B-Dependent NK/Plasmacytoid Dendritic Cell Functional Interaction in Cutaneous Lupus Erythematosus  Valentina Salvi, William.
IL-2–mediated apoptosis of kidney tubular epithelial cells is regulated by the caspase-8 inhibitor c-FLIP  Caigan Du, Qiunong Guan, Ziqin Yin, Robert.
Staphylococcus aureus Stimulates Neutrophil Targeting Chemokine Expression in Keratinocytes through an Autocrine IL-1α Signaling Loop  Florina Olaru,
Human renal epithelial cells produce the long pentraxin PTX3
Interferon-γ Protects from Staphylococcal Alpha Toxin-Induced Keratinocyte Death through Apolipoprotein L1  Anne M. Brauweiler, Elena Goleva, Donald Y.M.
IL-13-Stimulated Human Keratinocytes Preferentially Attract CD4+CCR4+ T cells: Possible Role in Atopic Dermatitis  Rahul Purwar, Thomas Werfel, Miriam.
Cytokine Effects Induced by the Human Autoallergen α-NAC
Nicastrin/miR-30a-3p/RAB31 Axis Regulates Keratinocyte Differentiation by Impairing EGFR Signaling in Familial Acne Inversa  Yanyan He, Haoxiang Xu, Chengrang.
Vγ4 γδ T Cells Provide an Early Source of IL-17A and Accelerate Skin Graft Rejection  Yashu Li, Zhenggen Huang, Rongshuai Yan, Meixi Liu, Yang Bai, Guangping.
Increased Lipocalin-2 Contributes to the Pathogenesis of Psoriasis by Modulating Neutrophil Chemotaxis and Cytokine Secretion  Shuai Shao, Tianyu Cao,
Fetal Human Keratinocytes Produce Large Amounts of Antimicrobial Peptides: Involvement of Histone-Methylation Processes  Maria Gschwandtner, Shaomin Zhong,
Toll-Like Receptor-Mediated Upregulation of CXCL16 in Psoriasis Orchestrates Neutrophil Activation  Sabine Steffen, Susanne Abraham, Maik Herbig, Franziska.
Ha Linh Vu, Sheera Rosenbaum, Claudia Capparelli, Timothy J
Stefan W. Stoll, Jessica L. Johnson, Yong Li, Laure Rittié, James T
Hic-5 Promotes the Hypertrophic Scar Myofibroblast Phenotype by Regulating the TGF- β1 Autocrine Loop  Ganary Dabiri, David A. Tumbarello, Christopher.
A Varp-Binding Protein, RACK1, Regulates Dendrite Outgrowth through Stabilization of Varp Protein in Mouse Melanocytes  Soujiro Marubashi, Norihiko Ohbayashi,
MicroRNA Expression Profiling Identifies miR-31 and miR-485-3p as Regulators in the Pathogenesis of Discoid Cutaneous Lupus  Cristina Solé, Sandra Domingo,
BPAG1-e Restricts Keratinocyte Migration through Control of Adhesion Stability  Magdalene Michael, Rumena Begum, Kenneth Fong, Celine Pourreyrone, Andrew.
Regulation of IL-33 Expression by IFN-γ and Tumor Necrosis Factor-α in Normal Human Epidermal Keratinocytes  Jitlada Meephansan, Hidetoshi Tsuda, Mayumi.
BCL11B-Mediated Epigenetic Repression Is a Crucial Target for Histone Deacetylase Inhibitors in Cutaneous T-Cell Lymphoma  Wenjing Fu, Shengguo Yi, Lei.
IL-31 Induces Chemotaxis, Calcium Mobilization, Release of Reactive Oxygen Species, and CCL26 in Eosinophils, Which Are Capable to Release IL-31  Nikola.
Anti-Inflammatory Activity of Sertaconazole Nitrate Is Mediated via Activation of a p38– COX-2–PGE2 Pathway  Runa Sur, Jeffrey M. Babad, Michelle Garay,
Min Qin, Aslan Pirouz, Myung-Hwa Kim, Stephan R. Krutzik, Hermes J
The Human Skin–Associated Autoantigen α-NAC Activates Monocytes and Dendritic Cells via TLR-2 and Primes an IL-12-Dependent Th1 Response  Susanne Hradetzky,
Phenotype and Antimicrobial Activity of Th17 Cells Induced by Propionibacterium acnes Strains Associated with Healthy and Acne Skin  George W. Agak, Stephanie.
MALT1 Protease Activity Controls the Expression of Inflammatory Genes in Keratinocytes upon Zymosan Stimulation  Anja Schmitt, Paula Grondona, Tabea Maier,
Volume 132, Issue 5, Pages (May 2007)
Profiling Motility Signal-Specific Genes in Primary Human Keratinocytes  Chieh-Fang Cheng, Jianhua Fan, Balaji Bandyopahdhay, Dennis Mock, Shengxi Guan,
Min Qin, Aslan Pirouz, Myung-Hwa Kim, Stephan R. Krutzik, Hermes J
Histamine Enhances the Production of Granulocyte-Macrophage Colony-Stimulating Factor via Protein Kinase Cα and Extracellular Signal-Regulated Kinase.
Naoko Kanda, Shinichi Watanabe  Journal of Investigative Dermatology 
Suppressor of Cytokine Signaling 1/JAB and Suppressor of Cytokine Signaling 3/Cytokine-Inducible SH2 Containing Protein 3 Negatively Regulate the Signal.
P38 Mitogen-activated Protein Kinase and Extracellular Signal-regulated Kinases Play Distinct Roles in the Activation of Dendritic Cells by Two Representative.
All-Trans-Retinoic Acid Induces Interleukin-8 via the Nuclear Factor-κB and p38 Mitogen-Activated Protein Kinase Pathways in Normal Human Keratinocytes 
Positive Selection of Dendritic Epidermal γδ T Cell Precursors in the Fetal Thymus Determines Expression of Skin-Homing Receptors  Na Xiong, Chuhlo Kang,
17β-estradiol Inhibits the Production of RANTES in Human Keratinocytes
Nrf2 Promotes Keratinocyte Proliferation in Psoriasis through Up-Regulation of Keratin 6, Keratin 16, and Keratin 17  Luting Yang, Xueli Fan, Tingting.
The IL-6 Trans-Signaling-STAT3 Pathway Mediates ECM and Cellular Proliferation in Fibroblasts from Hypertrophic Scar  Sutapa Ray, Xiaoxi Ju, Hong Sun,
UVB and Proinflammatory Cytokines Synergistically Activate TNF-α Production in Keratinocytes through Enhanced Gene Transcription  Muhammad M. Bashir,
Maria C. Lebre, Angelic M. G. van der Aar, Lisa van Baarsen, Toni M. M
Anti-Mycotics Suppress Interleukin-4 and Interleukin-5 Production in Anti-CD3 Plus Anti- CD28-Stimulated T Cells from Patients with Atopic Dermatitis 
PPARδ Is a Type 1 IFN Target Gene and Inhibits Apoptosis in T Cells
Gottron's Papules Exhibit Dermal Accumulation of CD44 Variant 7 (CD44v7) and Its Binding Partner Osteopontin: A Unique Molecular Signature  Jessica S.
IL-18 Downregulates Collagen Production in Human Dermal Fibroblasts via the ERK Pathway  Hee Jung Kim, Seok Bean Song, Jung Min Choi, Kyung Moon Kim,
TWEAK/Fn14 Activation Contributes to the Pathogenesis of Bullous Pemphigoid  Yale Liu, Lingling Peng, Liang Li, Chengfei Liu, Xiao Hu, Shengxiang Xiao,
Deficient SOCS3 and SHP-1 Expression in Psoriatic T Cells
IFN-α Enhances IL-22 Receptor Expression in Keratinocytes: A Possible Role in the Development of Psoriasis  Mikiko Tohyama, Lujun Yang, Yasushi Hanakawa,
Collagen Synthesis Is Suppressed in Dermal Fibroblasts by the Human Antimicrobial Peptide LL-37  Hyun Jeong Park, Dae Ho Cho, Hee Jung Kim, Jun Young.
Human Beta-Defensin 3 Induces Maturation of Human Langerhans Cell–Like Dendritic Cells: An Antimicrobial Peptide that Functions as an Endogenous Adjuvant 
IL-4 and IL-13 Alter Plasmacytoid Dendritic Cell Responsiveness to CpG DNA and Herpes Simplex Virus-1  Jurjen Tel, Ruurd Torensma, Carl G. Figdor, I.
BCL11B-Mediated Epigenetic Repression Is a Crucial Target for Histone Deacetylase Inhibitors in Cutaneous T-Cell Lymphoma  Wenjing Fu, Shengguo Yi, Lei.
Volume 6, Issue 4, Pages (April 2016)
Naoko Kanda, Shinichi Watanabe  Journal of Investigative Dermatology 
PU.1 Expression Delineates Heterogeneity in Primary Th2 Cells
Hyun Jeong Park, Hee Jung Kim, Jun Young Lee, Baik Kee Cho, Richard L
Regulation of IL-13 Receptors in Human Keratinocytes
Presentation transcript:

Absence of γ-Chain in Keratinocytes Alters Chemokine Secretion, Resulting in Reduced Immune Cell Recruitment  Karolin Nowak, Daniela Linzner, Adrian J. Thrasher, Paul F. Lambert, Wei-Li Di, Siobhan O. Burns  Journal of Investigative Dermatology  Volume 137, Issue 10, Pages 2120-2130 (October 2017) DOI: 10.1016/j.jid.2017.05.024 Copyright © 2017 The Authors Terms and Conditions

Figure 1 Keratinocytes express functional γc and its co-receptors. (a) RNA of ED-7R cell lines, NIKS, and primary keratinocytes (KCs) was isolated, and reverse transcription PCR was performed with primers specific for γc, its co-receptors, and GAPDH. (b) KC, NIKS, CD8+ T cells, and ED-7R cell lines were stained with anti-γc antibody and analyzed by flow cytometry. (c) NIKS and KCs were lysed and analyzed for their expression of IL-7Rα and IL-2Rβ by Western blot. (d) NIKS were stained with antibodies for γc–co-receptors and analyzed by flow cytometry. (e) NIKS were cultured in serum-free medium overnight, stimulated with 100 ng/ml of the indicated cytokines for 10 minutes, and analyzed by immunoblot for the expression of pAKT. γc, common gamma chain; APC, allophycocyanin; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; KC, keratinocyte; NIKS, normal immortalized keratinocyte cell line; pAKT, phosphorylated protein kinase B. Journal of Investigative Dermatology 2017 137, 2120-2130DOI: (10.1016/j.jid.2017.05.024) Copyright © 2017 The Authors Terms and Conditions

Figure 2 Generation of a γc-knockdown cell line. NIKS were transduced with short hairpin RNA against γc or with scrambled control (scr) short hairpin RNA. (a) RNA was isolated from scr and γc-knockdown (KD) NIKS cell lines, and reverse transcription PCR was performed using γc- and GAPDH-specific primers; bar charts showing the band intensities compared with untransduced, mean ± standard error of the mean. ∗P < 0.02, n = 4. (b) γc-expression in scr and KD cells was analyzed by flow cytometry. (c) NIKS cell lines were cultured in serum-free medium overnight, stimulated with 10 ng/ml IL-15 for 10 minutes, lysed, and analyzed for the expression of pAKT. Bar chart shows comparison of intensities of the Western blot bands, mean ± standard error of the mean. ∗P < 0.05, n = 5. GAPDH, glyceraldehyde-3-phosphate dehydrogenase; APC, allophycocyanin; KD, γc-knockdown; NIKS, normal immortalized keratinocyte cell line; pAKT, phosphorylated protein kinase B; scr, scrambled control. Journal of Investigative Dermatology 2017 137, 2120-2130DOI: (10.1016/j.jid.2017.05.024) Copyright © 2017 The Authors Terms and Conditions

Figure 3 Secretion of chemokines after IL-15 stimulation. Scrambled control (scr) and γc-knock-down (KD) NIKS were serum-depleted for 6 hours and then stimulated for 24 hours with 100 ng/ml IL-15 at 37 °C. Supernatants were collected and analyzed for secretion of (a) CXCL1, (b) CXCL8, (c) CCL20, (d) CCL5, (e) IL-1α, and (f) CXCL10 using Luminex bead assays, mean ± standard error of the mean. (a–c) n = 6, ∗P < 0.05. (d–f) n = 3. KD, γc-knockdown; NIKS, normal immortalized keratinocyte cell line; scr, scrambled control. Journal of Investigative Dermatology 2017 137, 2120-2130DOI: (10.1016/j.jid.2017.05.024) Copyright © 2017 The Authors Terms and Conditions

Figure 4 Migration of neutrophils. Neutrophils were isolated from whole blood and used for migration experiments using Dunn chambers, with the cells imaged every minute for 1 hour. (a) Cytospin of neutrophils before plating them on coverslips for migration. Scale bar = 25 μm. (b, c) Quantification for migration of one representative donor, where each dot represents one tracked cell. (d, e) Quantification for multiple migration experiments, where each dot represents the mean value for all cells analyzed from one donor. (b–e) “None” contains no chemoattractant; “chemokine cocktail” represents a cocktail of 10 ng/ml CXCL1, 3 ng/ml CXCL8, and 1 ng/ml CCL20. fMLP at 100 nmol/L was used as a positive control. Mean ± standard error of the mean; ∗P < 0.05, ∗∗∗P < 0.001; n = 4. Shown are (b, d) migratory speed and (c, d) directionality. fMLP, N-formylmethionyl-leucyl-phenylalanine; ns, not significant. Journal of Investigative Dermatology 2017 137, 2120-2130DOI: (10.1016/j.jid.2017.05.024) Copyright © 2017 The Authors Terms and Conditions

Figure 5 Migration of dendritic cells and CD4+ T cells. (a–c) CD14+-derived dendritic cells (DCs) were cultured on μ-Slides Chemotaxis 3D (Ibidi, Martinsreid, Germany; collagen IV coated) for migration assays. (a) Microscopic image of adherent DCs. Scale bar = 10 μm. (b) Quantification of an individual migration experiment, where each dot represents one cell tracked. (c) Quantification for multiple experiments, where each dot represents the mean value for all cells analyzed from one donor. (d, e) CD4+ T cells were isolated from whole blood and used for Transwell migration assays; values were normalized to the sample containing no chemoattractant. Assays were run in triplicate. (a–d) “None” contains no chemoattractant; “scr + IL-15” is a cocktail of 10 ng/ml CXCL1, 3 ng/ml CXCL8, and 1 ng/ml CCL20; and “KD + IL-15” the same chemokines at half the concentration. (e) Migration toward CX3CL1 at 40 ng/ml, n = 4, mean ± standard error of the mean. ∗P < 0.05. KD, γc-knockdown; min, minute; ns, not significant; scr, scrambled control. Journal of Investigative Dermatology 2017 137, 2120-2130DOI: (10.1016/j.jid.2017.05.024) Copyright © 2017 The Authors Terms and Conditions

Figure 6 Chemokine secretion from HPV18-positive cells. (a) Scrambled control (scr) and γc-knockdown (KD) NIKS were transfected with HPV18 plasmid. DNA was isolated and used for PCR using HPV18-specific primers. (b) CD4+ T cells were isolated from whole blood and used for Transwell migration assays; migration was measured toward supernatants harvested from scr control and KD NIKS with and without HPV18. (c) CD8+ T cells were isolated from whole blood and used for Transwell migration assays as with CD4+ T cells. (b, c) Values shown represent the number of migrating cells normalized to experiments using supernatant from HPV18-negative samples; each dot represents the mean of duplicate wells analyzed for a single supernatant. Four different sets of supernatants and two different blood donors were used; mean ± standard error of the mean are shown. *P < 0.05. HPV, human papilloma virus; NIKS, normal immortalized keratinocyte cell line; scr, scrambled control. Journal of Investigative Dermatology 2017 137, 2120-2130DOI: (10.1016/j.jid.2017.05.024) Copyright © 2017 The Authors Terms and Conditions