Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
2.
J Invest Dermatol ; 143(4): 578-587.e3, 2023 04.
Article in English | MEDLINE | ID: mdl-36270546

ABSTRACT

Alopecia areata (AA) is an autoimmune hair loss disorder with no cure. Patients with sequence variation in AIRE are 15 times more likely to develop AA than the general population, yet the roles of AIRE in AA pathogenesis are unknown. In this study, we report that 62% of C57BL/6J female Aire‒/‒ mice spontaneously developed persistent AA-like lesions that displayed several hallmarks of human AA. Lesional Aire‒/‒ skin exhibited hair follicle (HF) dystrophy as determined by a reduced number of anagen HFs, decreased anagen HF proliferation, hair pigmentary changes, and decreased hair width and length. Inflammatory infiltrate comprising CD8+ T cells, CD4+ T cells, CD68+ macrophages, and mast cells was prominent in lesional Aire‒/‒ HFs. From gene expression analyses, we found lesional Aire‒/‒ skin to have significantly increased expression of human AA signature genes, including H2-Ab1, Ifnγ, IFN-γ‒induced chemokines (Ccl5, Cxcl9‒11), γc family cytokine receptor Il2RA, and JAK‒signal transducer and activator of transcription (STAT) signaling components (Stat1, Stat2, Stat4). By immunostaining, lesional Aire‒/‒ HFs also show upregulated major histocompatibility complex class I and downregulated α-melanocyte-stimulating hormone, signifying immune privilege collapse, and increased STAT1 activation in HF keratinocytes. Our study highlights a role for AIRE in HF biology and shows that Aire‒/‒ mice may serve as a valuable model system to study AA pathogenesis.


Subject(s)
Alopecia Areata , Polyendocrinopathies, Autoimmune , Humans , Female , Mice , Animals , CD8-Positive T-Lymphocytes , Interleukin Receptor Common gamma Subunit , Polyendocrinopathies, Autoimmune/pathology , Mice, Inbred C57BL , Hair Follicle/pathology
3.
J Cell Biol ; 220(8)2021 08 02.
Article in English | MEDLINE | ID: mdl-34037658

ABSTRACT

Oncogene-induced senescence (OIS) is a stable cell cycle arrest that occurs in normal cells upon oncogene activation. Cells undergoing OIS express a wide variety of secreted factors that affect the senescent microenvironment termed the senescence-associated secretory phenotype (SASP), which is beneficial or detrimental in a context-dependent manner. OIS cells are also characterized by marked epigenetic changes. We globally assessed histone modifications of OIS cells and discovered an increase in the active histone marks H3K79me2/3. The H3K79 methyltransferase disruptor of telomeric silencing 1-like (DOT1L) was necessary and sufficient for increased H3K79me2/3 occupancy at the IL1A gene locus, but not other SASP genes, and was downstream of STING. Modulating DOT1L expression did not affect the cell cycle arrest. Together, our studies establish DOT1L as an epigenetic regulator of the SASP, whose expression is uncoupled from the senescence-associated cell cycle arrest, providing a potential strategy to inhibit the negative side effects of senescence while maintaining the beneficial inhibition of proliferation.


Subject(s)
Cellular Senescence , DNA Methylation , Epigenesis, Genetic , Fibroblasts/enzymology , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Interleukin-1alpha/metabolism , 9,10-Dimethyl-1,2-benzanthracene , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Cycle Checkpoints , Cell Proliferation , Female , HEK293 Cells , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Humans , Interleukin-1alpha/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Microscopy, Fluorescence , Papilloma/chemically induced , Papilloma/genetics , Papilloma/metabolism , Papilloma/pathology , Phenotype , Secretory Pathway , Skin Neoplasms/chemically induced , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tetradecanoylphorbol Acetate
4.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Article in English | MEDLINE | ID: mdl-33762306

ABSTRACT

High levels of the intermediate filament protein keratin 17 (K17) are associated with poor prognoses for several human carcinomas. Studies in mouse models have shown that K17 expression is positively associated with growth, survival, and inflammation in skin and that lack of K17 delays onset of tumorigenesis. K17 occurs in the nucleus of human and mouse tumor keratinocytes where it impacts chromatin architecture, gene expression, and cell proliferation. We report here that K17 is induced following DNA damage and promotes keratinocyte survival. The presence of nuclear K17 is required at an early stage of the double-stranded break (DSB) arm of the DNA damage and repair (DDR) cascade, consistent with its ability to associate with key DDR effectors, including γ-H2A.X, 53BP1, and DNA-PKcs. Mice lacking K17 or with attenuated K17 nuclear import showed curtailed initiation in a two-step skin carcinogenesis paradigm. The impact of nuclear-localized K17 on DDR and cell survival provides a basis for the link between K17 induction and poor clinical outcomes for several human carcinomas.


Subject(s)
Carcinoma/genetics , DNA Repair , Keratin-17/metabolism , Keratins/metabolism , Neoplasms, Experimental/genetics , 9,10-Dimethyl-1,2-benzanthracene/administration & dosage , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Active Transport, Cell Nucleus , Animals , Carcinogenesis/chemically induced , Carcinogenesis/genetics , Carcinogenesis/pathology , Carcinoma/chemically induced , Carcinoma/pathology , Cell Nucleus/metabolism , Cell Survival/genetics , DNA Breaks, Double-Stranded/drug effects , Female , Gene Knockout Techniques , HeLa Cells , Humans , Intravital Microscopy , Keratin-17/genetics , Keratinocytes , Keratins/genetics , Male , Mice, Knockout , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/pathology , Time-Lapse Imaging
5.
J Cell Sci ; 133(20)2020 10 30.
Article in English | MEDLINE | ID: mdl-33008845

ABSTRACT

Keratin 17 (KRT17; K17), a non-lamin intermediate filament protein, was recently found to occur in the nucleus. We report here on K17-dependent differences in nuclear morphology, chromatin organization, and cell proliferation. Human tumor keratinocyte cell lines lacking K17 exhibit flatter nuclei relative to normal. Re-expression of wild-type K17, but not a mutant form lacking an intact nuclear localization signal (NLS), rescues nuclear morphology in KRT17-null cells. Analyses of primary cultures of skin keratinocytes from a mouse strain expressing K17 with a mutated NLS corroborated these findings. Proteomics screens identified K17-interacting nuclear proteins with known roles in gene expression, chromatin organization and RNA processing. Key histone modifications and LAP2ß (an isoform encoded by TMPO) localization within the nucleus are altered in the absence of K17, correlating with decreased cell proliferation and suppression of GLI1 target genes. Nuclear K17 thus impacts nuclear morphology with an associated impact on chromatin organization, gene expression, and proliferation in epithelial cells.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Keratin-17 , Keratinocytes , Animals , Cell Proliferation/genetics , Chromatin/genetics , Keratin-17/genetics , Mice , Skin
6.
PLoS Pathog ; 15(10): e1007778, 2019 10.
Article in English | MEDLINE | ID: mdl-31603920

ABSTRACT

Type I interferons (T1-IFN) are critical in the innate immune response, acting upon infected and uninfected cells to initiate an antiviral state by expressing genes that inhibit multiple stages of the lifecycle of many viruses. T1-IFN triggers the production of Interferon-Stimulated Genes (ISGs), activating an antiviral program that reduces virus replication. The importance of the T1-IFN response is highlighted by the evolution of viral evasion strategies to inhibit the production or action of T1-IFN in virus-infected cells. T1-IFN is produced via activation of pathogen sensors within infected cells, a process that is targeted by virus-encoded immunomodulatory molecules. This is probably best exemplified by the prototypic poxvirus, Vaccinia virus (VACV), which uses at least 6 different mechanisms to completely block the production of T1-IFN within infected cells in vitro. Yet, mice lacking aspects of T1-IFN signaling are often more susceptible to infection with many viruses, including VACV, than wild-type mice. How can these opposing findings be rationalized? The cytosolic DNA sensor cGAS has been implicated in immunity to VACV, but has yet to be linked to the production of T1-IFN in response to VACV infection. Indeed, there are two VACV-encoded proteins that effectively prevent cGAS-mediated activation of T1-IFN. We find that the majority of VACV-infected cells in vivo do not produce T1-IFN, but that a small subset of VACV-infected cells in vivo utilize cGAS to sense VACV and produce T1-IFN to protect infected mice. The protective effect of T1-IFN is not mediated via ISG-mediated control of virus replication. Rather, T1-IFN drives increased expression of CCL4, which recruits inflammatory monocytes that constrain the VACV lesion in a virus replication-independent manner by limiting spread within the tissue. Our findings have broad implications in our understanding of pathogen detection and viral evasion in vivo, and highlight a novel immune strategy to protect infected tissue.


Subject(s)
Chemokine CCL4/metabolism , Interferon Type I/pharmacology , Membrane Proteins/physiology , Nucleotidyltransferases/physiology , Vaccinia virus/drug effects , Vaccinia/prevention & control , Viral Load/drug effects , Animals , Antiviral Agents/pharmacology , Chemokine CCL4/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/drug effects , Monocytes/immunology , Monocytes/virology , Vaccinia/immunology , Vaccinia/metabolism , Vaccinia/virology , Vaccinia virus/immunology , Virus Replication
7.
Cancer Med ; 8(13): 6106-6113, 2019 10.
Article in English | MEDLINE | ID: mdl-31436046

ABSTRACT

Although targeting DNA repair signaling pathways has emerged as a promising therapeutic for skin cancer, the relevance of DNA damage responses (DDR) in the development and survival of nonmelanoma skin cancer (NMSC), the most common type of skin cancer, remains obscure. Here, we report that Src-associated substrate during mitosis of 68 kDa (Sam68), an early signaling molecule in DDR, is elevated in skin tumor tissues derived from NMSC patients and skin lesions from Gli2-transgenic mice. Downregulation of Sam68 impacts the growth and survival of human tumor keratinocytes and genetic ablation of Sam68 delays the onset of basal cell carcinomas (BCC) in Gli2-transgenic mice. Moreover, Sam68 plays a critical role in DNA damage-induced DNA repair and nuclear factor kappa B (NF-κB) signaling pathways in keratinocytes, hence conferring keratinocyte sensitivity to DNA damaging agents. Together, our data reveal a novel function of Sam68 in regulating DDR in keratinocytes that is crucial for the growth and survival of NMSC.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , RNA-Binding Proteins/genetics , Skin Neoplasms/pathology , Animals , Cell Line , DNA Damage , DNA Repair , Female , Humans , Male , Mice, Transgenic , NF-kappa B/metabolism , Signal Transduction , Skin Neoplasms/genetics , Zinc Finger Protein Gli2/genetics
8.
Metallomics ; 10(12): 1728-1742, 2018 12 12.
Article in English | MEDLINE | ID: mdl-30206620

ABSTRACT

Calprotectin is a potent antimicrobial that inhibits the growth of pathogens by tightly binding transition metals such as Mn and Zn, thereby preventing their uptake and utilization by invading microbes. At sites of infection, calprotectin is abundantly released from neutrophils, but calprotectin is also present in non-neutrophil cell types that may be relevant to infections. We show here that in patients infected with the Lyme disease pathogen Borreliella (Borrelia) burgdorferi, calprotectin is produced in neutrophil-free regions of the skin, in both epidermal keratinocytes and in immune cells infiltrating the dermis, including CD68 positive macrophages. In culture, B. burgdorferi's growth is inhibited by calprotectin, but surprisingly, the mechanism does not involve the classical withholding of metal nutrients. B. burgdorferi cells exposed to calprotectin cease growth with no reduction in intracellular Mn and no loss in activity of Mn enzymes including the SodA superoxide dismutase. Additionally, there is no obvious loss in intracellular Zn. Rather than metal depletion, we find that calprotectin inhibits B. burgdorferi growth through a mechanism that requires physical association of calprotectin with the bacteria. By comparison, calprotectin inhibited E. coli growth without physically interacting with the microbe, and calprotectin effectively depleted E. coli of intracellular Mn and Zn. Our studies with B. burgdorferi demonstrate that the antimicrobial capacity of calprotectin is complex and extends well beyond simple withholding of metal micronutrients.


Subject(s)
Anti-Bacterial Agents/pharmacology , Borrelia burgdorferi/drug effects , Glossitis, Benign Migratory/drug therapy , Leukocyte L1 Antigen Complex/pharmacology , Lyme Disease/complications , Manganese/metabolism , Zinc/metabolism , Escherichia coli/drug effects , Glossitis, Benign Migratory/metabolism , Glossitis, Benign Migratory/microbiology , Humans , Neutrophils/drug effects , Neutrophils/metabolism , Neutrophils/microbiology
9.
J Invest Dermatol ; 137(7): 1410-1412, 2017 07.
Article in English | MEDLINE | ID: mdl-28390815

ABSTRACT

Unchecked inflammation, impaired keratinocyte differentiation, and heightened host defense responses typify psoriasis. Lambert et al. make clever use of psoriasis patient genetics and whole transcriptome RNA-Seq analysis to implicate Act1 in these seemingly variegated processes by keeping IL-17 receptor signaling in check while supporting differentiation and limiting innate immune responses in human keratinocytes.


Subject(s)
Connexin 43/genetics , Genetic Predisposition to Disease , Keratinocytes/pathology , Peptide Fragments/genetics , Polymorphism, Genetic , Psoriasis/genetics , Cell Differentiation , Connexin 43/metabolism , Humans , Peptide Fragments/metabolism , Psoriasis/metabolism , Psoriasis/pathology , Signal Transduction
10.
Elife ; 52016 12 20.
Article in English | MEDLINE | ID: mdl-27996939

ABSTRACT

Previously we reported that Src-associated-substrate-during-mitosis-of-68kDa (Sam68/KHDRBS1) is pivotal for DNA damage-stimulated NF-κB transactivation of anti-apoptotic genes (Fu et al., 2016). Here we show that Sam68 is critical for genotoxic stress-induced NF-κB activation in the γ-irradiated colon and animal and that Sam68-dependent NF-κB activation provides radioprotection to colon epithelium in vivo. Sam68 deletion diminishes γ-irradiation-triggered PAR synthesis and NF-κB activation in colon epithelial cells (CECs), thus hampering the expression of anti-apoptotic molecules in situ and facilitating CECs to undergo apoptosis in mice post whole-body γ-irradiation (WBIR). Sam68 knockout mice suffer more severe damage in the colon and succumb more rapidly from acute radiotoxicity than the control mice following WBIR. Our results underscore the critical role of Sam68 in orchestrating genotoxic stress-initiated NF-κB activation signaling in the colon tissue and whole animal and reveal the pathophysiological relevance of Sam68-dependent NF-κB activation in colonic cell survival and recovery from extrinsic DNA damage.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Colon/radiation effects , Gamma Rays , Intestinal Mucosa/radiation effects , NF-kappa B p50 Subunit/metabolism , RNA-Binding Proteins/metabolism , Animals , Mice, Knockout
11.
Dev Cell ; 38(3): 227-33, 2016 08 08.
Article in English | MEDLINE | ID: mdl-27505414

ABSTRACT

Previously thought to reside exclusively in the cytoplasm, the cytoskeletal protein keratin 17 (K17) has been recently identified inside the nucleus of tumor epithelial cells with a direct impact on cell proliferation and gene expression. We comment on fundamental questions raised by this new finding and the associated significance.


Subject(s)
Cell Nucleus/metabolism , Cell Proliferation , Epithelial Cells/metabolism , Keratins/metabolism , Humans
12.
Nat Genet ; 47(8): 933-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26168014

ABSTRACT

Expression of the intermediate filament protein keratin 17 (K17) is robustly upregulated in inflammatory skin diseases and in many tumors originating in stratified and pseudostratified epithelia. We report that autoimmune regulator (Aire), a transcriptional regulator, is inducibly expressed in human and mouse tumor keratinocytes in a K17-dependent manner and is required for timely onset of Gli2-induced skin tumorigenesis in mice. The induction of Aire mRNA in keratinocytes depends on a functional interaction between K17 and the heterogeneous nuclear ribonucleoprotein hnRNP K. Further, K17 colocalizes with Aire protein in the nucleus of tumor-prone keratinocytes, and each factor is bound to a specific promoter region featuring an NF-κB consensus sequence in a relevant subset of K17- and Aire-dependent proinflammatory genes. These findings provide radically new insight into keratin intermediate filament and Aire function, along with a molecular basis for the K17-dependent amplification of inflammatory and immune responses in diseased epithelia.


Subject(s)
Gene Expression Regulation , Keratin-17/genetics , Keratinocytes/metabolism , Skin Neoplasms/genetics , Transcription Factors/genetics , Animals , Cell Line, Tumor , Cells, Cultured , HeLa Cells , Humans , Immunoblotting , In Situ Hybridization , Keratin-17/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Molecular Sequence Data , Protein Binding , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Transcription Factors/metabolism , AIRE Protein
13.
J Cell Sci ; 126(Pt 18): 4195-207, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23843618

ABSTRACT

Plectin is a versatile cytolinker of the plakin family conferring cell resilience to mechanical stress in stratified epithelia and muscles. It acts as a critical organizer of the cytoskeletal system by tethering various intermediate filament (IF) networks through its C-terminal IF-binding domain (IFBD). Mutations affecting the IFBD cause devastating human diseases. Here, we show that serine 4642, which is located in the extreme C-terminus of plectin, is phosphorylated in different cell lines. Phosphorylation of S4642 decreased the ability of plectin IFBD to associate with various IFs, as assessed by immunofluorescence microscopy and cell fractionation studies, as well as in yeast two-hybrid assays. Plectin phosphorylated at S4642 was reduced at sites of IF network anchorage along cell-substrate contacts in both skin and cultured keratinocytes. Treatment of SK-MEL-2 and HeLa cells with okadaic acid increased plectin S4642 phosphorylation, suggesting that protein phosphatase 2A dephosphorylates this residue. Moreover, plectin S4642 phosphorylation was enhanced after cell treatment with EGF, phorbol ester, sorbitol and 8-bromo-cyclic AMP, as well as during wound healing and protease-mediated cell detachment. Using selective protein kinase inhibitors, we identified two different kinases that modulate the phosphorylation of plectin S4642 in HeLa cells: MNK2, which is downstream of the ERK1/2-dependent MAPK cascade, and PKA. Our study indicates that phosphorylation of S4642 has an important regulatory role in the interaction of plectin with IFs and identifies a novel link between MNK2 and the cytoskeleton.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Plectin/metabolism , Protein Serine-Threonine Kinases/metabolism , Serine/metabolism , Cell Movement , Cytoskeleton/metabolism , Humans , Intermediate Filaments/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Phosphorylation , Plectin/genetics , Protein Binding , Protein Serine-Threonine Kinases/genetics , Serine/genetics , Transfection
14.
Curr Opin Cell Biol ; 25(1): 47-56, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23270662

ABSTRACT

Intermediate filaments are assembled from a diverse group of evolutionary conserved proteins and are specified in a tissue-dependent, cell type-dependent, and context-dependent fashion in the body. Genetic mutations in intermediate filament proteins account for a large number of diseases, ranging from skin fragility conditions to cardiomyopathies and premature aging. Keratins, the epithelial-specific intermediate filaments, are now recognized as multi-faceted effectors in their native context. In this review, we emphasize the recent progress made in defining the role of keratins towards the regulation of cytoarchitecture, cell growth and proliferation, apoptosis, and cell motility during embryonic development, in normal adult tissues, and in select diseases such as cancer.


Subject(s)
Epithelium/pathology , Epithelium/physiology , Intermediate Filaments/pathology , Intermediate Filaments/physiology , Keratins/metabolism , Animals , Apoptosis , Cell Movement , Cell Nucleus/metabolism , Embryonic Development , Homeostasis , Humans , Intermediate Filaments/chemistry , Neoplasms/metabolism , Neoplasms/pathology , Skin Diseases/metabolism , Skin Diseases/pathology , Stress, Physiological
15.
J Invest Dermatol ; 133(2): 578-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22931915
18.
FASEB J ; 25(3): 990-1001, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21156808

ABSTRACT

Darier's disease (DD) is an inherited autosomal-dominant skin disorder characterized histologically by loss of adhesion between keratinocytes. DD is typically caused by mutations in sarcoendoplasmic reticulum Ca(2+)-ATPase isoform 2 (SERCA2), a major regulator of intracellular Ca(2+) homeostasis in the skin. However, a defined role for SERCA2 in regulating intercellular adhesion remains poorly understood. We found that diminution of SERCA2 function by pharmacological inhibition or siRNA silencing in multiple human epidermal-derived cell lines was sufficient to disrupt desmosome assembly and weaken intercellular adhesive strength. Specifically, SERCA2-deficient cells exhibited up to a 60% reduction in border translocation of desmoplakin (DP), the desmosomal cytolinker protein necessary for intermediate filament (IF) anchorage to sites of robust cell-cell adhesion. In addition, loss of SERCA2 impaired the membrane translocation of protein kinase C α (PKCα), a known regulator of DP-IF association and desmosome assembly, to the plasma membrane by up to 70%. Exogenous activation of PKCα in SERCA2-deficient cells was sufficient to rescue the defective DP localization, desmosome assembly, and intercellular adhesive strength to levels comparable to controls. Our findings indicate that SERCA2-deficiency is sufficient to impede desmosome assembly and weaken intercellular adhesive strength via a PKCα-dependent mechanism, implicating SERCA2 as a novel regulator of PKCα signaling.


Subject(s)
Darier Disease/metabolism , Desmoplakins/metabolism , Protein Kinase C-alpha/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Signal Transduction/physiology , Calcium/metabolism , Carcinoma, Squamous Cell , Cell Adhesion/physiology , Cell Communication/physiology , Cell Line, Tumor , Darier Disease/pathology , Desmosomes/metabolism , Desmosomes/pathology , Humans , Intermediate Filaments/metabolism , Keratins/metabolism , Mouth Neoplasms , RNA, Small Interfering , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics
20.
Mol Biol Cell ; 21(16): 2844-59, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20554761

ABSTRACT

Plakophilin 2 (PKP2), an armadillo family member closely related to p120 catenin (p120ctn), is a constituent of the intercellular adhesive junction, the desmosome. We previously showed that PKP2 loss prevents the incorporation of desmosome precursors enriched in the plaque protein desmoplakin (DP) into newly forming desmosomes, in part by disrupting PKC-dependent regulation of DP assembly competence. On the basis of the observation that DP incorporation into junctions is cytochalasin D-sensitive, here we ask whether PKP2 may also contribute to actin-dependent regulation of desmosome assembly. We demonstrate that PKP2 knockdown impairs cortical actin remodeling after cadherin ligation, without affecting p120ctn expression or localization. Our data suggest that these defects result from the failure of activated RhoA to localize at intercellular interfaces after cell-cell contact and an elevation of cellular RhoA, stress fibers, and other indicators of contractile signaling in squamous cell lines and atrial cardiomyocytes. Consistent with these observations, RhoA activation accelerated DP redistribution to desmosomes during the first hour of junction assembly, whereas sustained RhoA activity compromised desmosome plaque maturation. Together with our previous findings, these data suggest that PKP2 may functionally link RhoA- and PKC-dependent pathways to drive actin reorganization and regulate DP-IF interactions required for normal desmosome assembly.


Subject(s)
Actomyosin/metabolism , Desmosomes/metabolism , Plakophilins/metabolism , rhoA GTP-Binding Protein/metabolism , Actins/metabolism , Animals , Cadherins/metabolism , Catenins/metabolism , Cell Communication , Cell Line , Cell Line, Tumor , Cytoskeleton/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Intercellular Junctions/metabolism , Microscopy, Fluorescence , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myosin Light Chains/metabolism , Plakophilins/genetics , Protein Binding , Protein Kinase C/metabolism , RNA Interference , Signal Transduction , Delta Catenin
SELECTION OF CITATIONS
SEARCH DETAIL
...