Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Int J Mol Sci ; 20(7)2019 Mar 27.
Article in English | MEDLINE | ID: mdl-30934690

ABSTRACT

Activation and/or upregulated expression of p38δ are demonstrated in human skin malignancies including cutaneous squamous cell carcinoma, suggesting a role for p38δ in skin carcinogenesis. We previously reported that mice with germline deletion of the p38δ gene are significantly protected from chemical skin carcinogenesis. Here, we investigated the effects of cell-selective targeted ablation of p38δ in keratinocytes and in immune (myeloid) cells on skin tumor development in a two-stage 7,12-dimethylbenz(a)anthracene (DMBA)/12-O-tetradecanoylphorbol-13-acetate (TPA) chemical mouse skin carcinogenesis model. Conditional keratinocyte-specific p38δ ablation (p38δ-cKO∆K) did not influence the latency, incidence, or multiplicity of chemically-induced skin tumors, but led to increased tumor volume in females during the TPA promotion stage, and reduced malignant progression in males and females relative to their wild-type counterparts. In contrast, conditional myeloid cell-specific p38δ deletion (p38δ-cKO∆M) inhibited DMBA/TPA-induced skin tumorigenesis in male but not female mice. Thus, tumor onset was delayed, and tumor incidence, multiplicity, and volume were reduced in p38δ-cKO∆M males compared with control wild-type males. Moreover, the percentage of male mice with malignant tumors was decreased in the p38δ-cKO∆M group relative to their wild-type counterparts. Collectively, these results reveal that cell-specific p38δ targeting modifies susceptibility to chemical skin carcinogenesis in a context-, stage-, and sex-specific manner.


Subject(s)
Mitogen-Activated Protein Kinase 13/metabolism , Sex Characteristics , Skin Neoplasms/enzymology , Skin Neoplasms/pathology , 9,10-Dimethyl-1,2-benzanthracene , Animals , Carcinogenesis/pathology , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Cytokines/metabolism , Disease Progression , Female , Gene Deletion , Inflammation Mediators/metabolism , Keratinocytes/enzymology , Male , Mice, Knockout , Myeloid Cells/metabolism , Neoplasm Staging , Phenotype , Skin/pathology , Tetradecanoylphorbol Acetate
2.
J Mol Cell Cardiol ; 96: 82-92, 2016 07.
Article in English | MEDLINE | ID: mdl-26093152

ABSTRACT

Heart failure (HF) claims 250,000 lives per year in the US, and nearly half of these deaths are sudden and presumably due to ventricular tachyarrhythmias. QT interval and action potential (AP) prolongation are hallmark proarrhythmic changes in the failing myocardium, which potentially result from alterations in repolarizing potassium currents. Thus, we aimed to examine whether decreased expression of the rapid delayed rectifier potassium current, IKr, contributes to repolarization abnormalities in human HF. To map functional IKr expression across the left ventricle (LV), we optically imaged coronary-perfused LV free wall from donor and end-stage failing human hearts. The LV wedge preparation was used to examine transmural AP durations at 80% repolarization (APD80), and treatment with the IKr-blocking drug, E-4031, was utilized to interrogate functional expression. We assessed the percent change in APD80 post-IKr blockade relative to baseline APD80 (∆APD80) and found that ∆APD80s are reduced in failing versus donor hearts in each transmural region, with 0.35-, 0.43-, and 0.41-fold reductions in endo-, mid-, and epicardium, respectively (p=0.008, 0.037, and 0.022). We then assessed hERG1 isoform gene and protein expression levels using qPCR and Western blot. While we did not observe differences in hERG1a or hERG1b gene expression between donor and failing hearts, we found a shift in the hERG1a:hERG1b isoform stoichiometry at the protein level. Computer simulations were then conducted to assess IKr block under E-4031 influence in failing and nonfailing conditions. Our results confirmed the experimental observations and E-4031-induced relative APD80 prolongation was greater in normal conditions than in failing conditions, provided that the cellular model of HF included a significant downregulation of IKr. In human HF, the response to IKr blockade is reduced, suggesting decreased functional IKr expression. This attenuated functional response is associated with altered hERG1a:hERG1b protein stoichiometry in the failing human LV, and failing cardiomyoctye simulations support the experimental findings. Thus, of IKr protein and functional expression may be important determinants of repolarization remodeling in the failing human LV.


Subject(s)
Action Potentials , ERG1 Potassium Channel/metabolism , Heart Failure/metabolism , Heart Failure/physiopathology , Myocardium/metabolism , Potassium/metabolism , Adolescent , Adult , Anti-Arrhythmia Agents/pharmacology , Computer Simulation , ERG1 Potassium Channel/antagonists & inhibitors , ERG1 Potassium Channel/chemistry , ERG1 Potassium Channel/genetics , Female , Gene Expression , Heart Failure/diagnosis , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Heart Ventricles/physiopathology , Humans , Male , Middle Aged , Models, Biological , Piperidines/pharmacology , Pyridines/pharmacology , Young Adult
3.
Mol Carcinog ; 55(5): 563-74, 2016 May.
Article in English | MEDLINE | ID: mdl-25753147

ABSTRACT

p38δ expression and/or activity are increased in human cutaneous malignancies, including invasive squamous cell carcinoma (SCC) and head and neck SCC, but the role of p38δ in cutaneous carcinogenesis has not been well-defined. We have reported that mice with germline loss of p38δ exhibited a reduced susceptibility to skin tumor development compared with wild-type mice in the two-stage 7,12-dimethylbenz(a)anthracene (DMBA)/12-O-tetradecanoylphorbol-13-acetate (TPA) chemical skin carcinogenesis model. Here, we report that p38δ gene ablation inhibited the growth of tumors generated from v-ras(Ha) -transformed keratinocytes in skin orthografts to nude mice, indicating that keratinocyte-intrinsic p38δ is required for Ras-induced tumorigenesis. Gene expression profiling of v-ras(Ha) -transformed p38δ-null keratinocytes revealed transcriptional changes associated with cellular responses linked to tumor suppression, such as reduced proliferation and increased differentiation, cell adhesion, and cell communications. Notably, a short-term DMBA/TPA challenge, modeling the initial stages of chemical skin carcinogenesis treatment, elicited an enhanced inflammation in p38δ-null skin compared with skin of wild-type mice, as assessed by measuring the expression of pro-inflammatory cytokines, including IL-1ß, IL-6, IL-17, and TNFα. Additionally, p38δ-null skin and p38δ-null keratinocytes exhibited increased p38α activation and signaling in response to acute inflammatory challenges, suggesting a role for p38α in stimulating the elevated inflammatory response in p38δ-null skin during the initial phases of the DMBA/TPA treatment compared with similarly treated p38δ(+/+) skin. Altogether, our results indicate that p38δ signaling regulates skin carcinogenesis not only by keratinocyte cell-autonomous mechanisms, but also by influencing the interaction between between the epithelial compartment of the developing skin tumor and its stromal microenvironment.


Subject(s)
Mitogen-Activated Protein Kinase 13/genetics , Skin Neoplasms/genetics , Skin/pathology , ras Proteins/genetics , Animals , Benz(a)Anthracenes/toxicity , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 13/metabolism , Skin/drug effects , Skin/metabolism , Skin Neoplasms/chemically induced , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tetradecanoylphorbol Acetate/toxicity , ras Proteins/pharmacology
4.
Am J Physiol Cell Physiol ; 306(10): C899-909, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24598361

ABSTRACT

Proline-rich protein tyrosine kinase 2 (Pyk2) is a member of the focal adhesion kinase family. We used Pyk2 knockout (Pyk2-KO) mice to study the role of Pyk2 in cutaneous wound repair. We report that the rate of wound closure was delayed in Pyk2-KO compared with control mice. To examine whether impaired wound healing of Pyk2-KO mice was caused by a keratinocyte cell-autonomous defect, the capacities of primary keratinocytes from Pyk2-KO and wild-type (WT) littermates to heal scratch wounds in vitro were compared. The rate of scratch wound repair was decreased in Pyk2-KO keratinocytes compared with WT cells. Moreover, cultured human epidermal keratinocytes overexpressing the dominant-negative mutant of Pyk2 failed to heal scratch wounds. Conversely, stimulation of Pyk2-dependent signaling via WT Pyk2 overexpression induced accelerated scratch wound closure and was associated with increased expression of matrix metalloproteinase (MMP)-1, MMP-9, and MMP-10. The Pyk2-stimulated increase in the rate of scratch wound repair was abolished by coexpression of the dominant-negative mutant of PKCδ and by GM-6001, a broad-spectrum inhibitor of MMP activity. These results suggest that Pyk2 is essential for skin wound reepithelialization in vivo and in vitro and that it regulates epidermal keratinocyte migration via a pathway that requires PKCδ and MMP functions.


Subject(s)
Focal Adhesion Kinase 2/genetics , Keratinocytes/metabolism , Protein Kinase C-delta/metabolism , Re-Epithelialization/genetics , Skin/metabolism , Animals , Cell Movement/drug effects , Cells, Cultured , Dipeptides/pharmacology , Focal Adhesion Kinase 2/deficiency , Gene Expression Regulation , Genes, Dominant , Humans , Keratinocytes/cytology , Keratinocytes/drug effects , Male , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 10/genetics , Matrix Metalloproteinase 10/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Mice , Mice, Knockout , Protein Kinase C-delta/genetics , Signal Transduction , Skin/drug effects , Skin/injuries
5.
Circ Arrhythm Electrophysiol ; 7(5): 875-82, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25114062

ABSTRACT

BACKGROUND: Ventricular arrhythmias occur more frequently in heart failure during episodes of ischemia-reperfusion although the mechanisms underlying this in humans are unclear. We assessed, in explanted human hearts, the remodeled electrophysiological response to acute ischemia-reperfusion in heart failure and its potential causes, including the remodeling of metabolic gene expression. METHODS AND RESULTS: We optically mapped coronary-perfused left ventricular wedge preparations from 6 human end-stage failing hearts (F) and 6 donor hearts rejected for transplantation (D). Preparations were subjected to 30 minutes of global ischemia, followed by 30 minutes of reperfusion. Failing hearts had exaggerated electrophysiological responses to ischemia-reperfusion, with greater action potential duration shortening (P<0.001 at 8-minute ischemia; P=0.001 at 12-minute ischemia) and greater conduction slowing during ischemia, delayed recovery of electric excitability after reperfusion (F, 4.8±1.8 versus D, 1.0±0 minutes; P<0.05), and incomplete restoration of action potential duration and conduction velocity early after reperfusion. Expression of 46 metabolic genes was probed using custom-designed TaqMan arrays, using extracted RNA from 15 failing and 9 donor hearts. Ten genes important in cardiac metabolism were downregulated in heart failure, with SLC27A4 and KCNJ11 significantly downregulated at a false discovery rate of 0%. CONCLUSIONS: We demonstrate, for the first time in human hearts, that the electrophysiological response to ischemia-reperfusion in heart failure is accelerated during ischemia with slower recovery after reperfusion. This can enhance spatial conduction and repolarization gradients across the ischemic border and increase arrhythmia susceptibility. This adverse response was associated with downregulation of expression of cardiac metabolic genes.


Subject(s)
Energy Metabolism/genetics , Heart Conduction System/physiopathology , Heart Failure/genetics , Heart Failure/physiopathology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/physiopathology , Action Potentials , Adult , Aged , Female , Gene Expression Profiling/methods , Gene Expression Regulation , Heart Conduction System/metabolism , Heart Failure/diagnosis , Heart Failure/metabolism , Humans , Kinetics , Male , Middle Aged , Myocardial Reperfusion Injury/diagnosis , Myocardial Reperfusion Injury/metabolism , Oligonucleotide Array Sequence Analysis , Recovery of Function
6.
J Invest Dermatol ; 133(3): 608-617, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23151848

ABSTRACT

Serum response factor (SRF) is a transcription factor that regulates the expression of growth-related immediate-early, cytoskeletal, and muscle-specific genes to control growth, differentiation, and cytoskeletal integrity in different cell types. To investigate the role for SRF in epidermal development and homeostasis, we conditionally knocked out SRF in epidermal keratinocytes. We report that SRF deletion disrupted epidermal barrier function leading to early postnatal lethality. Mice lacking SRF in epidermis displayed morphogenetic defects, including an eye-open-at-birth phenotype and lack of whiskers. SRF-null skin exhibited abnormal morphology, hyperplasia, aberrant expression of differentiation markers and transcriptional regulators, anomalous actin organization, enhanced inflammation, and retarded hair follicle (HF) development. Transcriptional profiling experiments uncovered profound molecular changes in SRF-null E17.5 epidermis and revealed that many previously identified SRF target CArG box-containing genes were markedly upregulated in SRF-null epidermis, indicating that SRF may function to repress transcription of a subset of its target genes in epidermis. Remarkably, when transplanted onto nude mice, engrafted SRF-null skin lacked hair but displayed normal epidermal architecture with proper expression of differentiation markers, suggesting that although keratinocyte SRF is essential for HF development, a cross-talk between SRF-null keratinocytes and the surrounding microenvironment is likely responsible for the barrier-deficient mutant epidermal phenotype.


Subject(s)
Epidermis/physiopathology , Hair Follicle/growth & development , Morphogenesis/physiology , Serum Response Factor/physiology , Transcription Factors/physiology , Transcription, Genetic/physiology , Animals , Cell Communication/physiology , Cell Proliferation , Epidermis/pathology , Female , Hair Follicle/physiology , Keratinocytes/pathology , Mice , Mice, Knockout , Mice, Nude , Models, Animal , Phenotype , Serum Response Factor/deficiency , Serum Response Factor/genetics , Signal Transduction/physiology , Transcription Factors/deficiency , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL