Role for Protein Kinase C-α in Keratinocyte Growth Arrest

Slides:



Advertisements
Similar presentations
Nan-Hyung Kim, Ai-Young Lee  Journal of Investigative Dermatology 
Advertisements

CD271 Mediates Stem Cells to Early Progeny Transition in Human Epidermis  Francesca Truzzi, Annalisa Saltari, Elisabetta Palazzo, Roberta Lotti, Tiziana.
The δ-Opioid Receptor Affects Epidermal Homeostasis via ERK-Dependent Inhibition of Transcription Factor POU2F3  Christine Neumann, Mei Bigliardi-Qi,
Differential Expression of D-Type Cyclins in HaCaT Keratinocytes and in Psoriasis  Nóra Belsõ, Márta Széll, Andor Pivarcsi, Kornélia Kis, Bernadett Kormos,
Amanda M. Nelson, Kathryn L. Gilliland, Zhaoyuan Cong, Diane M
PFKFB3, a Direct Target of p63, Is Required for Proliferation and Inhibits Differentiation in Epidermal Keratinocytes  Robert B. Hamanaka, Gökhan M. Mutlu 
MicroRNA-31 Promotes Skin Wound Healing by Enhancing Keratinocyte Proliferation and Migration  Dongqing Li, X.I. Li, Aoxue Wang, Florian Meisgen, Andor.
The TCL1 oncoprotein inhibits activation-induced cell death by impairing PKCθ and ERK pathways by Gilles Despouy, Marjorie Joiner, Emilie Le Toriellec,
Downregulation of STRA6 Expression in Epidermal Keratinocytes Leads to Hyperproliferation-Associated Differentiation in Both In Vitro and In Vivo Skin.
Yongping Shao, Kaitlyn Le, Hanyin Cheng, Andrew E. Aplin 
Verena N. Lorenz, Michael P. Schön, Cornelia S. Seitz 
Effects of Betulinic Acid Alone and in Combination with Irradiation in Human Melanoma Cells  Edgar Selzer, Emilio Pimentel, Volker Wacheck, Werner Schlegel,
Sirpa Aho, Clive R. Harding, Jian-Ming Lee, Helen Meldrum, Carol A
Malignant T Cells Secrete Galectins and Induce Epidermal Hyperproliferation and Disorganized Stratification in a Skin Model of Cutaneous T-Cell Lymphoma 
Epidermal Growth Factor Induces Fibronectin Expression in Human Dermal Fibroblasts via Protein Kinase C δ Signaling Pathway  Yoshihiro Mimura, Hironobu.
Daisuke Suzuki, Makoto Senoo  Journal of Investigative Dermatology 
Andreea M. Bujor, Jaspreet Pannu, Shizhong Bu, Edwin A. Smith, Robin C
Stefan W. Stoll, Jessica L. Johnson, Yong Li, Laure Rittié, James T
CD271 Mediates Stem Cells to Early Progeny Transition in Human Epidermis  Francesca Truzzi, Annalisa Saltari, Elisabetta Palazzo, Roberta Lotti, Tiziana.
Decreased Growth Inhibitory Responses of Squamous Carcinoma Cells to Interferon-γ Involve Failure to Recruit cki Proteins into cdk2 Complexes  Beth L.
c-Jun Promotes whereas JunB Inhibits Epidermal Neoplasia
Matrix Metalloproteinase-21 Expression Is Associated with Keratinocyte Differentiation and Upregulated by Retinoic Acid in HaCaT Cells  Tiina Skoog, Outi.
Isolation (From a Basal Cell Carcinoma) of a Functionally Distinct Fibroblast-Like Cell Type that Overexpresses Ptch  Anthony J. Dicker, Magdalena M.
Enhanced Death Ligand-Induced Apoptosis in Cutaneous SCC Cells by Treatment with Diclofenac/Hyaluronic Acid Correlates with Downregulation of c-FLIP 
Integrin β6-Deficient Mice Show Enhanced Keratinocyte Proliferation and Retarded Hair Follicle Regression after Depilation  Yanshuang Xie, Kevin J. McElwee,
Brian Poligone, Elaine S. Gilmore, Carolina V
Characterization of Glucose Transport System in Keratinocytes: Insulin and IGF-1 Differentially Affect Specific Transporters  Shlomzion Shen, Sanford.
AKT Has an Anti-Apoptotic Role in ABCA12-Deficient Keratinocytes
Epidermal RelA Specifically Restricts Contact Allergen–Induced Inflammation and Apoptosis in Skin  Snehlata Kumari, Benjamin Herzberg, Ruth Pofahl, Thomas.
Shalini S. Tibudan, Yihua Wang, Mitchell F. Denning 
The TRAF-Interacting Protein (TRIP) Is a Regulator of Keratinocyte Proliferation  Stéphanie Almeida, Stephan Ryser, Magdalena Obarzanek-Fojt, Daniel Hohl,
Calmodulin-Like Protein Upregulates Myosin-10 in Human Keratinocytes and Is Regulated during Epidermal Wound Healing In Vivo  Richard D. Bennett, Amy.
K6PC-5, a Direct Activator of Sphingosine Kinase 1, Promotes Epidermal Differentiation Through Intracellular Ca2+ Signaling  Jeong Hee Hong, Jong-Kyung.
Role of p38 MAPK in UVB-Induced Inflammatory Responses in the Skin of SKH-1 Hairless Mice  Arianna L. Kim, Jeffrey M. Labasi, Yucui Zhu, Xiuwei Tang,
Bax Activation and Induction of Apoptosis in Human Keratinocytes by the Protein Kinase C δ Catalytic Domain  Leonid A. Sitailo, Shalini S. Tibudan, Mitchell.
Electrical Stimulation Enhances Epidermal Proliferation in Human Cutaneous Wounds by Modulating p53–SIVA1 Interaction  Anil Sebastian, Syed A. Iqbal,
Genetic and Pharmacological Analysis Identifies a Physiological Role for the AHR in Epidermal Differentiation  Ellen H. van den Bogaard, Michael A. Podolsky,
14-3-3σ Regulates Keratinocyte Proliferation and Differentiation by Modulating Yap1 Cellular Localization  Sumitha A.T. Sambandam, Ramesh B. Kasetti,
Suppression of E-Cadherin Function Drives the Early Stages of Ras-Induced Squamous Cell Carcinoma through Upregulation of FAK and Src  Addy Alt-Holland,
Leah C. Biggs, Lindsey Rhea, Brian C. Schutte, Martine Dunnwald 
Dual Role of the Anaphase Promoting Complex/Cyclosome in Regulating Stemness and Differentiation in Human Primary Keratinocytes  Ling Shih Quek, Nicolas.
Wound Healing Is Defective in Mice Lacking Tetraspanin CD151
Pituitary Tumor-Transforming Gene 1 Enhances Proliferation and Suppresses Early Differentiation of Keratinocytes  Yosuke Ishitsuka, Yasuhiro Kawachi,
Nrf2 Promotes Keratinocyte Proliferation in Psoriasis through Up-Regulation of Keratin 6, Keratin 16, and Keratin 17  Luting Yang, Xueli Fan, Tingting.
Overexpression of the Transcription Factor Yin-Yang-1 Suppresses Differentiation of HaCaT Cells in Three-Dimensional Cell Culture  Shijima Taguchi, Yasuhiro.
Denis Mehic, Latifa Bakiri, Minoo Ghannadan, Erwin F
Protease-Activated Receptor 2, a Receptor Involved in Melanosome Transfer, is Upregulated in Human Skin by Ultraviolet Irradiation  Glynis Scott, Cristina.
PPARδ Is a Type 1 IFN Target Gene and Inhibits Apoptosis in T Cells
Insulin-Like Growth Factor-Binding Protein 7 Regulates Keratinocyte Proliferation, Differentiation and Apoptosis  Janna Nousbeck, Ofer Sarig, Nili Avidan,
Mark A. Rovedo, Nancy L. Krett, Steven T. Rosen 
Retinoid-Induced Epidermal Hyperplasia Is Mediated by Epidermal Growth Factor Receptor Activation Via Specific Induction of its Ligands Heparin-Binding.
Suprabasal Spongiosis in Acute Eczematous Dermatitis: cFLIP Maintains Resistance of Basal Keratinocytes to T-Cell-Mediated Apoptosis  Nicole Armbruster,
Hydroxychloroquine Modulates Metabolic Activity and Proliferation and Induces Autophagic Cell Death of Human Dermal Fibroblasts  Bettina Ramser, Agatha.
Jaana Mannik, Kamil Alzayady, Soosan Ghazizadeh 
Activation of Dual Apoptotic Pathways in Human Melanocytes and Protection by Survivin  Tong Liu, Diana Biddle, Adrianne N. Hanks, Brook Brouha, Hui Yan,
Regulation of Human Epidermal Keratinocyte Differentiation by the Vitamin D Receptor and its Coactivators DRIP205, SRC2, and SRC3  Nathaniel P. Hawker,
Epidermal CCL27 Expression Is Regulated during Skin Development and Keratinocyte Differentiation  Michael Mildner, Marion Prior, Maria Gschwandtner, Christopher.
Targeted Depletion of Polo-Like Kinase (Plk) 1 Through Lentiviral shRNA or a Small- Molecule Inhibitor Causes Mitotic Catastrophe and Induction of Apoptosis.
DNA Damage Foci at Dysfunctional Telomeres
Nan-Hyung Kim, Ai-Young Lee  Journal of Investigative Dermatology 
TAK1 Is Required for Dermal Wound Healing and Homeostasis
IL-17A Upregulates Keratin 17 Expression in Keratinocytes through STAT1- and STAT3- Dependent Mechanisms  Xiaowei Shi, Liang Jin, Erle Dang, Ting Chang,
Piyush Koria, Stelios T. Andreadis 
ATR–Chk1 Pathway Inhibition Promotes Apoptosis after UV Treatment in Primary Human Keratinocytes: Potential Basis for the UV Protective Effects of Caffeine 
Myeloid Differentiation Factor 88 Regulates Basal and UV-Induced Expressions of IL-6 and MMP-1 in Human Epidermal Keratinocytes  Youngae Lee, Hyunjung.
Activation of Keratinocyte Protein Kinase Cζ in Psoriasis Plaques
All-Trans Retinoic Acid Antagonizes UV-Induced VEGF Production and Angiogenesis via the Inhibition of ERK Activation in Human Skin Keratinocytes  Mi-Sun.
Expression of Matrix Metalloproteinase-13 Is Controlled by IL-13 via PI3K/Akt3 and PKC-δ in Normal Human Dermal Fibroblasts  Chikako Moriya, Masatoshi.
Roland Houben, Sonja Ortmann, Astrid Drasche, Jakob Troppmair, Marco J
Role and Regulation of STAT3 Phosphorylation at Ser727 in Melanocytes and Melanoma Cells  Masanobu Sakaguchi, Masahiro Oka, Tetsushi Iwasaki, Yasuo Fukami,
Presentation transcript:

Role for Protein Kinase C-α in Keratinocyte Growth Arrest Anita Jerome-Morais, Heidi R. Rahn, Shalini S. Tibudan, Mitchell F. Denning  Journal of Investigative Dermatology  Volume 129, Issue 10, Pages 2365-2375 (October 2009) DOI: 10.1038/jid.2009.74 Copyright © 2009 The Society for Investigative Dermatology, Inc Terms and Conditions

Figure 1 PKCα is expressed and activated in the suprabasal layers of the epidermis. Immunoperoxidase staining of normal human foreskin was performed to visualize localization of total PKCα and phosphorylated PKCα substrates (P∼PKC substrate). Note the membrane localization of PKCα, and the increased cytoplasmic P∼PKC substrate staining in suprabasal KCs. Staining was performed on five skin samples, and representative staining is shown. Scale bar represents 10μm. Journal of Investigative Dermatology 2009 129, 2365-2375DOI: (10.1038/jid.2009.74) Copyright © 2009 The Society for Investigative Dermatology, Inc Terms and Conditions

Figure 2 PKCα Δ22–28 is constitutively active. (a) Construction of the constitutively active PKCα Δ22–28 mutant. The pseudosubstrate domain of human PKCα (amino acids 22–28) was deleted to generate the constitutively active PKCα mutant, PKCα Δ22–28. (b) PKCα translocation: HaCaT KCs were transduced with either the empty vector LZRS-Linker (Control) or LZRS-PKCα Δ22–28 retroviruses and treated with 100nM TPA for 30minutes as indicated. Membrane and cytosolic fractionation was performed, and PKCα (arrows) and P∼PKCα (Ser 657) were detected by western blotting. Similar results were obtained in one additional experiment. (c) Phosphorylated PKC substrate levels: KCs were transduced with LZRS-Linker (Control) or LZRS-PKCα Δ22–28 retroviruses, protein lysates after 18hours were analyzed for total PKCα (arrow), the phosphorylation of PKCα substrates using the P∼PKC substrate antibody and the autophosphorylation of P∼PKCα at Thr 638. Similar results were obtained in two additional experiments. Journal of Investigative Dermatology 2009 129, 2365-2375DOI: (10.1038/jid.2009.74) Copyright © 2009 The Society for Investigative Dermatology, Inc Terms and Conditions

Figure 3 Active PKCα inhibits KC proliferation. (a) Normal human KCs (105) were transduced with LZRS-Linker (Control) or the active PKCα Δ22–28 PKCα retrovirus. Cell counts were made 4 days after infection. Results are shown from three independent experiments (mean±SEM). (b) Normal human KCs (105) were infected in quadruplicate with the control vector or active PKCα Δ22–28. After 2 days, 103 cells were then replated at a low density, cultured for 10 days, and colonies counted. Similar results were obtained in one additional experiment. (c–d) 3H Thymidine incorporation was used to measure DNA synthesis in normal human KCs retrovirally transduced with the indicated viruses, or treated with TPA (10nM) for 4hours. (c) Shows thymidine incorporation of cells that were not replated. The inhibition of DNA synthesis by PKCα Δ22–28 without replating was significantly different than the control (* denotes P<0.01) at 24, 48, and 72hours. (d) Shows thymidine incorporation after replating at low density. The inhibition of DNA synthesis by PKCα Δ22–28 after replating was significantly different from control (# denotes P<0.001) at 72 and 96hours. Results are shown from seven independently infected/treated wells (mean±SD). Similar results were obtained in three additional experiments. Journal of Investigative Dermatology 2009 129, 2365-2375DOI: (10.1038/jid.2009.74) Copyright © 2009 The Society for Investigative Dermatology, Inc Terms and Conditions

Figure 4 Active PKCα induces G1 accumulation without increased apoptosis. (a) Normal human KCs were infected in duplicate with LZRS-Linker (Control), active PKCα Δ22–28, or PKCα retroviruses and harvested after 1 and 2 days. Propidium iodide staining and flow cytometry to analyze cell cycle distribution showed that PKCα activation induced a G1 arrest and a corresponding decrease in S and G2/M with no significant induction of sub-G1 apoptotic cell population. Similar results were obtained in two additional experiments. (b) Calculation of the G1/S ratio confirms an increased proportion of cells in the G1 phase. Shown is the mean±range. (c) Annexin V staining and flow cytometry of KCs confirmed low levels of apoptosis in PKCα Δ22–28-infected KCs after 3 days. Shown is the mean±range (N=2) from one experiment. Similar results obtained in one additional experiment. Journal of Investigative Dermatology 2009 129, 2365-2375DOI: (10.1038/jid.2009.74) Copyright © 2009 The Society for Investigative Dermatology, Inc Terms and Conditions

Figure 5 Induction of p21 is required for inhibition of DNA synthesis by PKCα Δ22–28. (a) Normal KCs were infected with either the LZRS-Linker (Control) or LZRS-PKCα Δ22–28 retrovirus. Protein was isolated for western blotting at both the 24- and 48-hour time points. p21 was transiently upregulated at the protein level at the 24hour time point. The arrow denotes PKCα. Similar results were obtained in two additional experiments. (b) Control or p21 siRNA oligonucleotides were used to transfect normal human KCs. After 24hours, KCs were infected with either LZRS-Linker (Control) or LZRS-PKCα Δ22–28 retrovirus. Cells were treated with 100nM TPA for 4hours the next day, after which protein was isolated to be analyzed by western blotting. Note that p21 siRNA oligos downregulated p21 in KCs. Similar results were obtained in two additional experiments. (c) KCs were transfected with the control and p21 siRNA oligos and then retrovirally transduced with either LZRS-Linker (Control) or PKCα Δ22–28 retrovirus. Twenty-four hours after infection, DNA synthesis was measured using 3H thymidine incorporation. Note that PKCα activation caused an inhibition of DNA synthesis in the control siRNA cells, but not in cells where p21 is downregulated. The KCs treated with 10nM TPA for 1hour was used as a positive control for inhibition of DNA synthesis. Shown are the mean±SD. Similar results were obtained in two additional experiments. Journal of Investigative Dermatology 2009 129, 2365-2375DOI: (10.1038/jid.2009.74) Copyright © 2009 The Society for Investigative Dermatology, Inc Terms and Conditions

Figure 6 Knock-down of PKCα in normal human KCs. KCs infected with control or PKCα shRNA virus were cultured submerged in monolayer, or plated on collagen/fibroblast dermal equivalents and lifted to the air–liquid interface for 7 days to generate organotypic raft cultures. (a) Western blot analysis showed that PKCα was downregulated in monolayer and maintained downregulated in the organotypic raft epidermis. The arrow denotes PKCα. (b) Photograph of the raft culture after 7 days at air–liquid interface. The epidermal layer has shrunk and sits on the larger collagen/fibroblast dermis. Note that when PKCα is downregulated, the organotypic epidermis occupies a larger surface area compared with the control shRNA organotypic culture. Similar results were obtained in four additional experiments. Journal of Investigative Dermatology 2009 129, 2365-2375DOI: (10.1038/jid.2009.74) Copyright © 2009 The Society for Investigative Dermatology, Inc Terms and Conditions

Figure 7 Downregulation of PKCα resulted in decreased epidermal thickness and increased basal/suprabasal proliferation. (a) Organotypic cultures of KCs transduced with control shRNA or PKCα shRNA viruses were generated. Hematoxylin and eosin staining of the organotypic raft epidermis showed decreased epidermal thickness when PKCα was downregulated. Immunofluorescent staining showed increased basal and suprabasal expression of the proliferation markers Ki67 and PCNA in the PKCα shRNA cultures. Scale bar represents 10μm. (b) Quantification of the basal and suprabasal expression of Ki67 and PCNA. Note increased basal and suprabasal proliferation markers in PKCα shRNA cultures. Similar results were obtained in three additional experiments. (c) Quantification of the epidermal thickness in control and PKCα downregulated organotypic raft epidermises. Data are average of three independent experiments, with error bars denoting SD. (d) Analysis of the spinous differentiation marker expression (K1) and granular layer differentiation marker expression (Loricrin) by immunofluorescence staining of the raft epidermis. Note reduced K1 and increased loricrin expression in shPKCα organotypic culture. Similar results were obtained in four additional experiments. Scale bar represents 10μm. Journal of Investigative Dermatology 2009 129, 2365-2375DOI: (10.1038/jid.2009.74) Copyright © 2009 The Society for Investigative Dermatology, Inc Terms and Conditions