RESUMEN
Regarding that the chronic use of commonly available non-steroidal and anti-inflammatory drugs (NSAIDs) is often restricted by their adverse effects, there is still a current need to search for and develop new, safe and effective anti-inflammatory agents. As a continuation of our previous work, we designed and synthesized a series of 18 novel N-substituted-1,2,4-triazole-based derivatives of pyrrolo[3,4-d]pyridazinone 4a-c-9a-c. The target compounds were afforded via a convenient way of synthesis, with good yields. The executed cell viability assay revealed that molecules 4a-7a, 9a, 4b-7b, 4c-7c do not exert a cytotoxic effect and were qualified for further investigations. According to the performed in vitro test, compounds 4a-7a, 9a, 4b, 7b, 4c show significant cyclooxygenase-2 (COX-2) inhibitory activity and a promising COX-2/COX-1 selectivity ratio. These findings are supported by a molecular docking study which demonstrates that new derivatives take position in the active site of COX-2 very similar to Meloxicam. Moreover, in the carried out in vitro evaluation within cells, the title molecules increase the viability of cells pre-incubated with the pro-inflammatory lipopolysaccharide and reduce the level of reactive oxygen and nitrogen species (RONS) in induced oxidative stress. The spectroscopic and molecular modeling study discloses that new compounds bind favorably to site II(m) of bovine serum albumin. Finally, we have also performed some in silico pharmacokinetic and drug-likeness predictions. Taking all of the results into consideration, the molecules belonging to series a (4a-7a, 9a) show the most promising biological profile.
Asunto(s)
Antiinflamatorios/farmacología , Inhibidores de la Ciclooxigenasa/farmacología , Dermis/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Piridazinas/química , Pirroles/química , Triazoles/química , Antiinflamatorios/química , Supervivencia Celular , Ciclooxigenasa 1/química , Ciclooxigenasa 2/química , Inhibidores de la Ciclooxigenasa/química , Dermis/citología , Dermis/enzimología , Diseño de Fármacos , Fibroblastos/citología , Fibroblastos/enzimología , Humanos , Técnicas In Vitro , Modelos Moleculares , Estructura Molecular , Relación Estructura-ActividadRESUMEN
Staphylococcus aureus is one of the main causative agent of infections acquired in both community and hospital environment. In this context, photodynamic therapy (PDT) consists in using a photosensitizer that, activated by light, evokes the formation of reactive oxygen species (ROS), which lead to the death of microorganisms due to oxidative damage; it is useful tool since this action, harmful to pathogens, does not significantly injure human cells. In view of this, this work proposes a more in-depth study on the use of resveratrol (RSV) as a possible photosensitizer. It was observed, in the intradermal infection model in animals' ear dermis, that photoactivated resveratrol promotes an increase in myeloperoxidase expression with reduced bacterial load in the draining lymph node. Besides that, the draining lymph node of the animals treated with photoactivated RSV controls inflammation through IL-10 production. These are pioneers data and this work being a pilot study; then, other works must be conducted with the objective of elucidate the photoactivated resveratrol mechanism of action.
Asunto(s)
Luz , Resveratrol/efectos de la radiación , Resveratrol/uso terapéutico , Infecciones Estafilocócicas/tratamiento farmacológico , Animales , Carga Bacteriana/efectos de los fármacos , Cadherinas/metabolismo , Dermis/efectos de los fármacos , Dermis/enzimología , Oído/patología , Humanos , Inflamación/patología , Interleucina-10/biosíntesis , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/patología , Ratones Endogámicos BALB C , Peroxidasa/metabolismo , Proyectos Piloto , Resveratrol/farmacología , Staphylococcus aureus/efectos de los fármacosRESUMEN
Fragmentation of collagen fibrils and aberrant elastic material (solar elastosis) in the dermal extracellular matrix (ECM) is among the most prominent features of photodamaged human skin. These alterations impair the structural integrity and create a dermal microenvironment prone to skin disorders. The objective of this study was to determine the physical properties (surface roughness, stiffness and hardness) of the dermal ECM in photodamaged and subject-matched sun-protected human skin. Skin samples were sectioned and analysed by histology, atomic force microscopy and nanoindentation. Dermal ECM collagen fibrils were more disorganized (ie, rougher surface), and the dermal ECM was stiffer and harder, in photodamaged forearm, compared to sun-protected underarm skin. Cleavage of collagen fibrils in sun-protected underarm dermis by recombinant human matrix metalloproteinase-1 resulted in rougher collagen fibril surface and reduced dermal stiffness and hardness. Degradation of elastotic material in photodamaged skin by treatment with purified neutrophil elastase reduced stiffness and hardness, without altering collagen fibril surface roughness. Additionally, expression of two members of the lysyl oxidase gene family, which insert cross-links that stiffen and harden collagen fibrils, was elevated in photodamaged forearm dermis. These data elucidate the contributions of fragmented collagen fibrils, solar elastosis and elevated collagen cross-linking to the physical properties of the dermal ECM in photodamaged human skin. This new knowledge extends current understanding of the impact of photodamage on the dermal ECM microenvironment.
Asunto(s)
Colágeno , Dermis/patología , Envejecimiento de la Piel/patología , Estudios de Casos y Controles , Dermis/enzimología , Matriz Extracelular/patología , Dureza , Humanos , Persona de Mediana Edad , Proteína-Lisina 6-Oxidasa/metabolismo , Luz Solar/efectos adversosRESUMEN
Atypical fibroxanthoma (AFX) is a histologic mimicker of a variety of spindle cell neoplasms, and careful microscopic and immunohistochemical evaluation is critical in establishing the correct diagnosis. Here we report the histologic and immunohistochemical work up of a 1 cm nodule involving the left dorsal hand of a 66-year-old patient. Light microscopy revealed fascicles of spindled and pleomorphic cells within the dermis showing increased mitotic activity occurring in the background of sun-damaged skin. There were numerous multinucleated cells with hyperchromatic nuclei and ample finely vacuolated or foamy cytoplasms. There was strong and diffuse CD10 and patchy CD68 expression among the spindled cells and multinucleated cells. The neoplastic cells did not show immunoreactivity against S100, p75-NGFR, HMB-45 or a panel of keratinocytic, vascular and smooth muscle markers. Tyrosinase and Melan-A were not expressed within the spindle cell component of this neoplasm; however, there was tyrosinase expression among numerous multinucleated giant cells. Melan-A expression was also observed among rare multinucleated giant cells. Tyrosinase expression has not previously been reported in AFX.
Asunto(s)
Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Monofenol Monooxigenasa/biosíntesis , Proteínas de Neoplasias/biosíntesis , Neoplasias Cutáneas , Xantomatosis , Anciano , Dermis/enzimología , Dermis/patología , Humanos , Masculino , Neoplasias Cutáneas/enzimología , Neoplasias Cutáneas/patología , Xantomatosis/enzimología , Xantomatosis/patologíaRESUMEN
Proteoglycan (PG) is a heavily glycosylated protein, localized to cell surface and extracellular matrix, and has various functions. Recently, it has been gradually revealed that PG interacts with various growth factors and morphogens and regulates cellular functions. Although salmon nasal cartilage PG (Salmon-PG) increases proliferation of immortalized cells, its mechanism remains unclear. In this study, we confirmed the effect of Salmon-PG on normal human dermal fibroblast (NHDF) and investigated the mechanism of PG action on NHDF. Salmon-PG dose- and time-dependently increased NHDF proliferation. Receptor tyrosine kinase array revealed that Salmon-PG increased only Erk1/2 signaling. Erk1/2 phosphorylation was significantly increased by Salmon-PG in a time-(10 min) and dose-(400 or 800 µg/mL) dependent manner. MEK inhibitor suppressed the enhancement of NHDF proliferation by Salmon-PG. The overall findings indicate that Salmon-PG plays a role as a growth factor in NHDF via Erk1/2 activation, suggesting that Salmon-PG contributes to the maintenance of skin homeostasis.
Asunto(s)
Dermis/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/genética , Cartílagos Nasales/química , Proteoglicanos/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Dermis/citología , Dermis/enzimología , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/enzimología , Flavonoides/farmacología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Análisis por Micromatrices , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteoglicanos/aislamiento & purificación , Salmón , Transducción de SeñalRESUMEN
The aim of our work was to examine content of serine-arginine protein kinase 1 (SRPK1) in human dermis at different ages (from 20 weeks of pregnancy to 85 years old). SRPK1, proliferating cells nuclear antigen (PCNA ), endothelial marker CD31 were detected in sections of the skin by indirect immunohistochemistry. Results showed, that content of SRPK1 in dermal fibroblasts was increased form antenatal period to 20 years of life followed by a decrease until 61-85 years period. SRPK1 content in dermal blood vessels is slowly gradually increased from antenatal period to 61-85 age interval. The number of fibroblasts and their proliferative activity, the number of CD31 positive blood vessels in dermis were decreased from antenatal period to 61-85 years period of life. Age-dependent decrease in SRPK1 in dermal fibroblasts from 20 years is associated with a reduction in the number and proliferative activity of fibroblasts. Age-related increase in SRPK1 content in dermal blood vessels is associated with a diminishing of the number of blood vessels. Hence, it can be supposed that SRPK1 has different actions on proliferation of differ components of dermis during aging.
Asunto(s)
Dermis/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Adolescente , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Envejecimiento/metabolismo , Niño , Preescolar , Femenino , Feto/enzimología , Fibroblastos/enzimología , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/análisis , Embarazo , Antígeno Nuclear de Célula en Proliferación/análisis , Adulto JovenRESUMEN
Identification of molecular mechanisms that regulate cellular replicative lifespan is needed to better understand the transition between a normal and a neoplastic cell phenotype. We have previously reported that low oxygen-mediated activity of FGF2 leads to an increase in cellular lifespan and acquisition of regeneration competence in human dermal fibroblasts (iRC cells). Though cells display a more plastic developmental phenotype, they remain non-tumorigenic when injected into SCID mice (Page et al. [2009] Cloning Stem Cells 11:417-426; Page et al. [2011] Eng Part A 17:2629-2640) allowing for investigation of mechanisms that regulate increased cellular lifespan in a non-tumorigenic system. Analysis of chromatin modification enzymes by qRT-PCR revealed a 13.3-fold upregulation of the arginine methyltransferase PRMT8 in iRC cells. Increased protein expression was confirmed in both iRC and human embryonic stem cells-the first demonstration of endogenous human PRMT8 expression outside the brain. Furthermore, iRC cells express a novel PRMT8 mRNA variant. Using siRNA-mediated knockdown we demonstrated that this novel variant was required for proliferation of human dermal fibroblasts (hDFs) and grade IV glioblastomas. PRMT8 upregulation in a non-tumorigenic system may offer a potential diagnostic biomarker and a therapeutic target for cells in pre-cancerous and cancerous states. J. Cell. Biochem. 117: 2056-2066, 2016. © 2016 Wiley Periodicals, Inc.
Asunto(s)
Proliferación Celular/fisiología , Dermis/enzimología , Fibroblastos/enzimología , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas de la Membrana , Proteína-Arginina N-Metiltransferasas , Regulación hacia Arriba/fisiología , Animales , Línea Celular , Fibroblastos/trasplante , Xenoinjertos , Humanos , Isoenzimas/biosíntesis , Isoenzimas/genética , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Ratones , Ratones SCID , Proteína-Arginina N-Metiltransferasas/biosíntesis , Proteína-Arginina N-Metiltransferasas/genéticaRESUMEN
Cathepsins (CTSs) are lysosomal cysteine proteases that play an important role in the turnover of intracellular proteins and extracellular proteins, such as the degradation of extracellular matrices and the processing of antigenic proteins. A CTS inhibitor, NC-2300, not only suppresses bone erosion by inhibition of cathepsin K (CTSK), but also ameliorates paw swelling at inflamed joints in adjuvant-induced arthritis in rats. It has been demonstrated that the amelioration of joint inflammation by NC-2300 is mediated by the downregulation of cytokine expression in dendritic cells, which are essential for Th17 activation. In this work, we studied the role for CTSs in the pathogenesis of psoriasis-like lesion in K5.Stat3C mice, a mouse model of psoriasis, in which Th17 contributes to lesion development similar to psoriasis. Psoriatic lesions expressed increased levels of Ctsk and Ctss mRNA compared with uninvolved skin and normal control skin. Similarly, the epidermis and dermis in K5.Stat3C mice demonstrated increased CTSK activities, which were sensitive to NC-2300. Topical treatment with NC-2300 significantly ameliorated 12-O-tetradecanoylphorbol-13-acetate-induced psoriasis-like lesions in K5.Stat3C mice, and downregulated the expression of IL-12, IL-23, and Th17 cytokines. In vitro experiments revealed that TLR7 activation of bone marrow-derived myeloid dendritic cells led to increase in IL-23 at mRNA and protein levels, which were downregulated by NC-2300. These results suggest that CTSK plays a role in development of psoriatic lesions through TLR7-dependent Th17 polarization.
Asunto(s)
Catepsina K/metabolismo , Dermis/enzimología , Psoriasis/enzimología , Células Th17/enzimología , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 8/metabolismo , Animales , Médula Ósea/inmunología , Médula Ósea/metabolismo , Médula Ósea/patología , Catepsina K/genética , Catepsina K/inmunología , Dermis/inmunología , Dermis/patología , Regulación hacia Abajo , Epidermis/enzimología , Epidermis/inmunología , Epidermis/patología , Femenino , Humanos , Interleucina-12/genética , Interleucina-12/inmunología , Interleucina-12/metabolismo , Interleucina-23/genética , Interleucina-23/inmunología , Interleucina-23/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Células Mieloides/inmunología , Células Mieloides/metabolismo , Células Mieloides/patología , Psoriasis/genética , Psoriasis/inmunología , Psoriasis/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Células Th17/inmunología , Células Th17/patología , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 8/genética , Receptor Toll-Like 8/inmunologíaRESUMEN
This study outlines the potential of a novel therapeutic dressing for the management of chronic wounds. The dressing incorporates polyphosphate, a non toxic compound with a number of beneficial characteristics in terms of wound healing, in a foam matrix. The aim of this study was to identify the potential of polyphosphate incorporated in the foam dressing to sequester the activity of matrix metalloproteinases (MMPs) and proteases derived from Pseudomonas aeruginosa. Methods used included gelatin zymography and milk-casein agar plate analysis. Results have shown that this dressing is effectively capable of reducing the levels of MMP-2 and MMP-9 in both their active and latent forms using an in vitro model. The dressing also demonstrated the compound's potential in the regulation of P. aeruginosa derived proteases.
Asunto(s)
Vendajes , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Polifosfatos , Cicatrización de Heridas/fisiología , Heridas y Lesiones/enzimología , Animales , Dermis/efectos de los fármacos , Dermis/enzimología , Dermis/patología , Caballos , Pseudomonas aeruginosa/fisiología , Técnicas de Cultivo de Tejidos , Heridas y Lesiones/microbiología , Heridas y Lesiones/patologíaRESUMEN
Cutaneous squamous cell carcinomas (SCCs) are commonly diagnosed skin cancers that may progress to invasiveness in the absence of early intervention. Using a murine model of SCC, we have previously demonstrated that activation of the Rho-associated kinase (ROCK) signaling pathway promotes rapid progression of pre-neoplastic lesions to invasive SCC. Herein we demonstrate that in human cutaneous SCC, ROCK signaling is increasingly up-regulated with tumor progression in both tumor cells and cells of the tumor microenvironment and is accompanied by key tumor-promoting changes in the extracellular matrix protein composition. The mechanotransduction pathway mediated by integrin signaling through FAK, GSK3ß, and the transcription coactivator ß-catenin is also progressively activated in human cutaneous SCC. Our observations indicate that ROCK activation is a tumor promoter in human cutaneous SCC and acts via mechanotransduction of signals to ß-catenin. Our experiments raise the possibility that inhibition of ROCK signaling could be a useful therapeutic approach to halt cutaneous SCC progression by reducing the signal flux through this pathway to physiologic levels, thereby normalizing the extracellular matrix composition.
Asunto(s)
Carcinoma de Células Escamosas/patología , Progresión de la Enfermedad , Mecanotransducción Celular , Neoplasias Cutáneas/patología , Animales , Carcinoma de Células Escamosas/enzimología , Moléculas de Adhesión Celular/metabolismo , Colágeno/metabolismo , Dermis/enzimología , Dermis/patología , Modelos Animales de Enfermedad , Activación Enzimática , Fibronectinas/metabolismo , Técnica del Anticuerpo Fluorescente , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Integrinas/metabolismo , Ratones , Invasividad Neoplásica , Neoplasias Cutáneas/enzimología , beta Catenina/metabolismo , Quinasas Asociadas a rho/metabolismoRESUMEN
TGFß binding to the TGFß receptor (TßR) activates R-Smad-dependent pathways, such as Smad2/3, and R-Smad-independent pathways, such as ERK1/2. The mechanism of the TGFß-TßRII-TßRI-Smad2/3 pathway is established; however, it is not known how TGFß activates ERK1/2. We show here that although TGFß equally activated Smad2/3 in all cells, it selectively activated ERK1/2 in dermal cells and inhibited ERK1/2 in epidermal cells. These opposite effects correlated with the distinct expression levels of TßRII, which are 7- to 18-fold higher in dermal cells than in epidermal cells. Reduction of TßRII expression in dermal cells abolished TGFß-stimulated ERK1/2 activation. Upregulation of TßRII expression in epidermal cells to a similar level as that in dermal cells switched TGFß-induced ERK1/2 inhibition to ERK1/2 activation. More intriguingly, in contrast to the equal importance of TßRII in mediating TGFß signaling to both Smad2/3 and ERK1/2, knockdown of TßRI/Alk5 blocked activation of only Smad2/3, not ERK1/2, in dermal cells. Similarly, expression of the constitutively activated TßRI-TD kinase activated only Smad2/3 and not ERK1/2 in epidermal cells. This study provides an explanation for why TGFß selectively activates ERK1/2 in certain cell types and direct evidence for TßRI-independent TßRII signaling to a R-Smad-independent pathway.
Asunto(s)
Dermis/metabolismo , Epidermis/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal , Línea Celular , Células Cultivadas , Dermis/citología , Dermis/enzimología , Células Epidérmicas , Epidermis/enzimología , Expresión Génica , Humanos , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteínas Serina-Treonina Quinasas/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Proteína Smad2/genética , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta/metabolismoAsunto(s)
Dermatitis Herpetiforme/dietoterapia , Dieta Sin Gluten , Inmunoglobulina A/metabolismo , Transglutaminasas/metabolismo , Adulto , Anciano , Niño , Dermatitis Herpetiforme/enzimología , Dermis/enzimología , Dermis/metabolismo , Femenino , Humanos , Cuidados a Largo Plazo , Masculino , Microscopía Fluorescente , Persona de Mediana Edad , Adulto JovenRESUMEN
BACKGROUND: Idiopathic and inflammation-dependent fibrotic diseases such systemic sclerosis (SSc) impose a major burden on modern societies. Understanding endogenous mechanisms, which counteract fibrosis, may yield new therapeutic approaches. Lipoxins are highly potent lipid mediators, which have recently been found to be decreased in SSc. OBJECTIVES: To determine the potential role of 12/15-lipoxygenase (12/15-LO), the key enzyme for the synthesis of lipoxins, in fibrosis. METHODS: Two mouse models for experimental dermal fibrosis (bleomycin-induced dermal fibrosis and tight-skin 1 mouse model) together with bone marrow transfers were used in wildtype and 12/15-LO(-/-) mice to elucidate the role of this enzyme during dermal fibrosis. Primary dermal fibroblasts of wildtype and 12/15-LO(-/-) mice, and 12/15-LO-derived eicosanoids, were used to identify underlying molecular mechanisms RESULTS: In both models, 12/15-LO(-/-) mice exhibited a significant exacerbation of the fibrotic tissue response. Bone marrow transfer experiments disclosed a predominant role of mesenchymal cell-derived 12/15-LO in these antifibrotic effects. Indeed, 12/15-LO(-/-) fibroblasts showed an enhanced activation of the mitogen-activated protein-kinase pathway and an increased col 1a2 mRNA expression in response to stimulation with transforming growth factor ß (TGFß), whereas 12/15-LO-derived eicosanoids blocked these TGFß-induced effects. CONCLUSIONS: These data indicate that 12/15-LO and its metabolites have a prominent antifibrotic role during dermal fibrosis. This opens new opportunities for therapeutic approaches in the treatment of fibrotic diseases.
Asunto(s)
Araquidonato 12-Lipooxigenasa/metabolismo , Araquidonato 15-Lipooxigenasa/metabolismo , Fibroblastos/enzimología , Fibroblastos/patología , Esclerodermia Sistémica/metabolismo , Esclerodermia Sistémica/patología , Animales , Antibióticos Antineoplásicos/farmacología , Araquidonato 12-Lipooxigenasa/genética , Araquidonato 15-Lipooxigenasa/genética , Bleomicina/farmacología , Células Cultivadas , Dermis/enzimología , Dermis/patología , Eicosanoides/metabolismo , Fibroblastos/efectos de los fármacos , Fibrosis/inducido químicamente , Fibrosis/enzimología , Fibrosis/patología , Lipoxinas/metabolismo , Mesodermo/enzimología , Mesodermo/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Proteínas Serina-Treonina Quinasas/genética , Esclerodermia Sistémica/genética , Factor de Crecimiento Transformador beta/farmacologíaRESUMEN
OBJECTIVE: We have previously demonstrated that in response to transforming growth factor ß (TGFß), Fli-1 activity is repressed through a series of sequential posttranslational modifications, consisting of protein kinase Cδ (PKCδ)-induced Thr312 phosphorylation, acetylation by p300/CREB binding protein-associated factor, and detachment from the collagen promoter. The purpose of this study was to further investigate the upstream events that lead to Fli-1 phosphorylation in response to TGFß. METHODS: Dermal fibroblasts were isolated from systemic sclerosis (SSc) patients and healthy control subjects matched for age, sex, and ethnicity. Western blotting was used to analyze protein levels and real-time quantitative reverse transcription-polymerase chain reaction analysis was used to measure messenger RNA expression. Cells were transduced with constitutively active PKCδ adenovirus or were transiently transfected with a Bcr-Abl-overexpressing plasmid. Subcellular localization of PKCδ was examined by immunocytochemistry. RESULTS: Western blot analysis of cell lysates demonstrated that the levels of phospho-Fli-1 (Thr312) were up-regulated in SSc fibroblasts, correlating with increased levels of type I collagen and c-Abl protein. Experiments using a constitutively activated form of c-Abl, small interfering RNA against c-Abl and the specific tyrosine kinase inhibitor imatinib, demonstrated the requirement of c-Abl for the TGFß-induced phosphorylation of Fli-1. Additionally, we showed that c-Abl kinase activity was required for nuclear localization of PKCδ. CONCLUSION: Our results demonstrate that in SSc fibroblasts, c-Abl is an upstream regulator of the profibrotic PKCδ/phospho-Fli-1 pathway, via induction of PKCδ nuclear localization. Additionally, the finding that Fli-1 is phosphorylated at higher levels in SSc fibroblasts supports the notion that the c-Abl/PKCδ/phospho-Fli-1 pathway is constitutively activated in these cells. Thus, blocking the TGFß/c-Abl/PKCδ/phospho-Fli-1 pathway could be an attractive alternative approach to therapy for scleroderma.
Asunto(s)
Dermis/enzimología , Fibroblastos/enzimología , Proteínas de Microfilamentos/metabolismo , Proteína Quinasa C-delta/metabolismo , Proteínas Proto-Oncogénicas c-abl/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Esclerodermia Sistémica/enzimología , Benzamidas , Núcleo Celular/metabolismo , Células Cultivadas , Colágeno Tipo I/biosíntesis , Dermis/efectos de los fármacos , Femenino , Fibroblastos/efectos de los fármacos , Humanos , Mesilato de Imatinib , Masculino , Fosforilación , Piperazinas/farmacología , Proteína Quinasa C-delta/genética , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-abl/biosíntesis , Proteínas Proto-Oncogénicas c-abl/genética , Pirimidinas/farmacología , ARN Interferente Pequeño/farmacología , Esclerodermia Sistémica/genética , Transactivadores , Transducción Genética , Factor de Crecimiento Transformador beta/farmacologíaRESUMEN
Reciprocal communication between hematopoietic cells and their surrounding bone marrow stroma is crucial for normal progression of hematopoiesis. This complex network of cell-to-cell signals in the microenvironment involves both cell contact-mediated and paracrine cues. In hematological malignancies the intricate balance is, however, disrupted to support cancer progression. In order to detect altered microenvironmental reactivity of a hematopoietic cell sample, cellular functional assays can be designed to measure the cells' capacity to modulate stromal stress reactions, such as inflammation.Recently, we showed that human leukemic cell lines of monocytic origin can actively participate in modulation of stromal inflammation. In order to further functionally evaluate the hematopoietic cells' capacity to modulate stromal inflammation, we utilized an in vitro model of nemosis-induced inflammation of fibroblasts in a three-dimensional culture setting. This process of stromal inflammation in fibroblast aggregates is consistent, requires both cell-contact and paracrine signals, and can be produced on a large scale to support dose-dependent analyses. To extend our previous observations, we evaluated the effect of a wide panel of leukemia cell lines on cyclooxygenase- 2 induction in fibroblast aggregates in co-culture. We also assessed the feasibility of the model to support clinical functional testing by utilizing the hematopoietic fraction of leukemia patients' bone marrow aspirates after immunophenotyping. Our results suggest that the stromal inflammation-modulating activity of these samples is differently modulated in cancer and in normal bone marrow. Moreover, differences in the samples' anti-inflammatory activity may reflect disease state.
Asunto(s)
Antiinflamatorios/metabolismo , Médula Ósea/patología , Ciclooxigenasa 2/metabolismo , Fibroblastos/patología , Células Madre Hematopoyéticas/patología , Leucemia/patología , Microambiente Tumoral , Adolescente , Adulto , Médula Ósea/enzimología , Médula Ósea/inmunología , Comunicación Celular , Células Cultivadas , Niño , Preescolar , Técnicas de Cocultivo , Dermis/enzimología , Dermis/inmunología , Dermis/patología , Femenino , Fibroblastos/enzimología , Fibroblastos/inmunología , Citometría de Flujo , Células Madre Hematopoyéticas/enzimología , Células Madre Hematopoyéticas/inmunología , Humanos , Immunoblotting , Inmunofenotipificación , Lactante , Leucemia/enzimología , Leucemia/inmunología , Masculino , Persona de Mediana Edad , Células del Estroma/enzimología , Células del Estroma/inmunología , Células del Estroma/patología , Adulto JovenRESUMEN
We examined the interaction of ECM1 (extracellular matrix protein 1) using yeast two-hybrid screening and identified the type II transmembrane protein, PLSCR1 (phospholipid scramblase 1), as a binding partner. This interaction was then confirmed by in vitro and in vivo co-immunoprecipitation experiments, and additional pull-down experiments with GST-tagged ECM1a fragments localized this interaction to occur within the tandem repeat region of ECM1a. Furthermore, immunohistochemical staining revealed a partial overlap of ECM1 and PLSCR1 in human skin at the basal epidermal cell layer. Moreover, in human skin equivalents, both proteins are expressed at the basal membrane in a dermal fibroblast-dependent manner. Next, immunogold electron microscopy of ultrathin human skin sections showed that ECM1 and PLSCR1 co-localize in the extracellular matrix, and using antibodies against ECM1 or PLSCR1 cross-linked to magnetic immunobeads, we were able to demonstrate PLSCR1-ECM1 interaction in human skin extracts. Furthermore, whereas ECM1 is secreted by the endoplasmic/Golgi-dependent pathway, PLSCR1 release from HaCaT keratinocytes occurs via a lipid raft-dependent mechanism, and is deposited in the extracellular matrix. In summary, we here demonstrate that PLSCR1 interacts with the tandem repeat region of ECM1a in the dermal epidermal junction zone of human skin and provide for the first time experimental evidence that PLSCR1 is secreted by an unconventional secretion pathway. These data suggest that PLSCR1 is a multifunctional protein that can function both inside and outside of the cell and together with ECM1 may play a regulatory role in human skin.
Asunto(s)
Dermis/metabolismo , Epidermis/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Proteínas de Transferencia de Fosfolípidos/metabolismo , Vías Secretoras , Piel/metabolismo , Línea Celular , Células Cultivadas , Dermis/enzimología , Epidermis/enzimología , Matriz Extracelular/enzimología , Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/química , Proteínas de la Matriz Extracelular/genética , Humanos , Uniones Intercelulares/enzimología , Uniones Intercelulares/metabolismo , Queratinocitos/enzimología , Queratinocitos/metabolismo , Metabolismo de los Lípidos , Proteínas de Transferencia de Fosfolípidos/genética , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Piel/enzimologíaRESUMEN
Systemic sclerosis results in tissue fibrosis due to the activation of fibroblasts and the ensuing overproduction of the extracellular matrix. We previously reported that the absence of alpha2-antiplasmin (alpha2AP) attenuated the process of dermal fibrosis; however, the detailed mechanism of how alpha2AP affects the progression of fibrosis remained unclear. The goal of the present study was to examine the role of alpha2AP in fibrotic change. We observed significantly higher levels of alpha2AP expression in the skin of bleomycin-injected systemic sclerosis model mice in comparison with the levels seen in control mice. We also demonstrated that alpha2AP induced myofibroblast differentiation, and the absence of alpha2AP attenuated the induction of myofibroblast differentiation. Moreover, we found that connective tissue growth factor induced the expression of alpha2AP through both the extracellular signal-regulated kinase 1/2 (ERK1/2) and c-Jun N-terminal kinase (JNK) pathways in fibroblasts. Interestingly, alpha2AP also induced transforming growth factor-beta expression through the same pathways, and the inhibition of ERK1/2 and JNK slowed the progression of bleomycin-induced fibrosis. Our findings suggest that alpha2AP is associated with the progression of fibrosis, and regulation of alpha2AP expression by the ERK1/2 and JNK pathways may be an effective antifibrotic therapy for the treatment of systemic sclerosis.
Asunto(s)
Dermis/patología , alfa 2-Antiplasmina/metabolismo , Animales , Bleomicina , Diferenciación Celular/efectos de los fármacos , Factor de Crecimiento del Tejido Conjuntivo/farmacología , Dermis/efectos de los fármacos , Dermis/enzimología , Progresión de la Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Fibroblastos/patología , Fibrosis , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Células 3T3 NIH , Inhibidores de Proteínas Quinasas/farmacología , Factor de Crecimiento Transformador beta/metabolismo , alfa 2-Antiplasmina/farmacologíaRESUMEN
Prostaglandin D2 (PGD2), an arachidonic acid metabolite, has been implicated in allergic responses. A major source of PGD2 in the skin is mast cells that express hematopoietic PGD synthase (H-PGDS). In this study, we show the expression of H-PGDS in human dendritic cells (DCs) and the regulatory mechanisms by which DCs produce PGD2. We detected H-PGDS in epidermal Langerhans cells, dermal DCs, plasmacytoid DCs, and myeloid DCs. Monocyte-derived DCs rapidly secreted PGD2 when stimulated with the calcium ionophore A23187. More importantly, pretreatment of monocyte-derived DCs with PMA (phorbol 12-myrisate 13-acetate) synergistically enhanced the rapid PGD2 secretion induced by A23187, whereas PMA alone did not induce PGD2 secretion. Lipopolysaccharide (LPS) reduced H-PGDS expression, but interferon-gamma followed by LPS induced significant PGD2 production in a delayed time course at 6 hours. This effect was associated with inhibition of LPS-induced H-PGDS reduction. Interestingly, an irritant compound, SDS, also induced a rapid PGD2 release. PGD2 synergistically enhanced CCL22/macrophage-derived chemokine synthesis in interferon-gamma-treated human keratinocytes. In addition, bone marrow-derived DCs from wild-type mice stimulated lymph node cells to produce higher amounts of interleukin-17 than did DCs from mice lacking the H-PGDS gene. Thus, DCs could be an important source of skin PGD2 and may mediate or regulate skin inflammation by releasing PGD2 in response to various stimuli, contributing to the innate and/or acquired immune responses.
Asunto(s)
Células Dendríticas/enzimología , Hematopoyesis , Oxidorreductasas Intramoleculares/metabolismo , Lipocalinas/metabolismo , Prostaglandina D2/biosíntesis , Piel/enzimología , Animales , Células Sanguíneas/efectos de los fármacos , Células Sanguíneas/enzimología , Quimiocina CCL22/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/patología , Dermis/efectos de los fármacos , Dermis/enzimología , Dermis/patología , Hematopoyesis/efectos de los fármacos , Humanos , Interferón gamma/farmacología , Interleucina-17/biosíntesis , Ionóforos/farmacología , Irritantes/farmacología , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Células de Langerhans/enzimología , Células de Langerhans/patología , Lipopolisacáridos/farmacología , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Prostaglandina D2/farmacología , Piel/efectos de los fármacos , Piel/patología , Dodecil Sulfato de Sodio/farmacología , Acetato de Tetradecanoilforbol/farmacologíaRESUMEN
The interaction of cancer cells with surrounding normal tissue cells is of utmost importance for their survival and tumor progression. For these purposes the cancer cells exploit normal tissue responses associated with inflammation and tissue repair. In the immediate tumor microenvironment one of the early stromal markers is cyclooxygenase-2 (COX-2). In this study we evaluated the effect of leukemia cell lines on nemosis-induced COX-2 expression in stromal fibroblasts. We found that THP-1 cells were the most potent leukemic cells (IC50=746) to suppress COX-2 expression. The U-937 cell line exhibited similar suppressive potency (IC50=921), whereas the KG-1 cell line (IC50=3519) was the least potent to affect COX-2 expression in the stromal cells. Our study shows that human leukemic cells can actively participate in modulation of stromal inflammation via inhibition of COX-2 expression. In a co-culture model of leukemia cell lines and stromal fibroblasts, our data suggest that the tumor-stromal interactions are complexly regulated, and the straightforward association of COX-2 expression with tumor progression may require re-evaluation since some tumor cells, e.g. from hematologic malignancies, may differentially modulate inflammation and COX-2 expression.
Asunto(s)
Ciclooxigenasa 2/metabolismo , Fibroblastos/patología , Leucemia/patología , Células del Estroma/patología , Microambiente Tumoral , Células Cultivadas , Técnicas de Cocultivo , Dermis/citología , Dermis/efectos de los fármacos , Dermis/enzimología , Fibroblastos/enzimología , Regulación Neoplásica de la Expresión Génica , Humanos , Immunoblotting , Leucemia/enzimología , Transducción de Señal , Esferoides Celulares , Células del Estroma/enzimologíaRESUMEN
Hepatoma-derived growth factor (HDGF) is a novel mitogenic growth factor that has been implicated in many different carcinomas. Its role in keloid biology has not yet been investigated. The present study is aimed at examining the role of HDGF in keloid pathogenesis. Immunohistochemical staining and Western blot analyses were used to examine in vivo localization and expression of HDGF in keloid and normal skin tissue. This was followed by the detection of HDGF expression in fibroblasts cultured in vitro and fibroblasts exposed to serum. To investigate the effect of epithelial-mesenchymal interactions, a two-chamber system was employed in which keratinocytes on membrane inserts were co-cultured with the fibroblasts. HDGF expression levels in all cell extracts and conditioned media were assayed through Western blot analysis. In another set of experiments, the effect of exogenous recombinant HDGF on keloid fibroblasts (KF) and normal fibroblasts (NF) was examined. Cell proliferation was assessed by the MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay and by quantifying proliferating cell nuclear antigen (PCNA) expression. Downstream targets of HDGF were identified by detecting their expression through Western blot analysis. Our results indicate that there was an increase in HDGF expression in the dermis of keloid compared with normal skin tissue. The application of serum and epithelial-mesenchymal interactions did not seem to have any effect on intracellular HDGF expression levels. However, co-culturing keloid keratinocytes with KFs resulted in increased HDGF secretion when compared with monoculture or normal controls. Furthermore, treatment with exogenous recombinant HDGF was found to increase the proliferation of KFs, activate the extracellular signal-regulated kinase (ERK) pathway and up-regulate the secretion of vascular endothelial growth factor (VEGF).