Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
FEBS J ; 291(16): 3628-3652, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38767406

RESUMEN

Cancer cells exhibit a unique metabolic preference for the glycolytic pathway over oxidative phosphorylation for maintaining the tumor microenvironment. Lactate dehydrogenase A (LDHA) is a key enzyme that facilitates glycolysis by converting pyruvate to lactate and has been shown to be upregulated in multiple cancers due to the hypoxic tumor microenvironment. Diclofenac (DCF), a nonsteroidal anti-inflammatory drug, has been shown to exhibit anticancer effects by interfering with the glucose metabolism pathway. However, the specific targets of this drug remain unknown. Using in silico, biochemical, and biophysical studies, we show that DCF binds to LDHA adjacent to the substrate binding site and inhibits its activity in a dose-dependent and allosteric manner in HeLa cells. Thus, DCF inhibits the hypoxic microenvironment and induces apoptosis-mediated cell death. DCF failed to induce cytotoxicity in HeLa cells when LDHA was knocked down, confirming that DCF exerts its antimitotic effects via LDHA inhibition. DCF-induced LDHA inhibition alters pyruvate, lactate, NAD+, and ATP production in cells, and this could be a possible mechanism through which DCF inhibits glucose uptake in cancer cells. DCF-induced ATP deprivation leads to mitochondria-mediated oxidative stress, which results in DNA damage, lipid peroxidation, and apoptosis-mediated cell death. Reduction in intracellular ATP levels additionally activates the sensor kinase, adenosine monophosphate-activated protein kinase (AMPK), which further downregulates phosphorylated ribosomal S6 kinase (p-S6K), leading to apoptosis-mediated cell death. We find that in LDHA knocked down cells, intracellular ATP levels were depleted, resulting in the inhibition of p-S6K, suggesting the involvement of DCF-induced LDHA inhibition in the activation of the AMPK/S6K signaling pathway.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Apoptosis , Diclofenaco , Humanos , Células HeLa , Diclofenaco/farmacología , Apoptosis/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Quinasas Activadas por AMP/genética , L-Lactato Deshidrogenasa/metabolismo , L-Lactato Deshidrogenasa/antagonistas & inhibidores , L-Lactato Deshidrogenasa/genética , Estrés Oxidativo/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Antiinflamatorios no Esteroideos/farmacología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo
3.
Cell Biochem Biophys ; 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38196050

RESUMEN

To survive in the tumour microenvironment, cancer cells undergo rapid metabolic reprograming and adaptability. One of the key characteristics of cancer is increased glycolytic selectivity and decreased oxidative phosphorylation (OXPHOS). Apart from ATP synthesis, glycolysis is also responsible for NADH regeneration and macromolecular biosynthesis, such as amino acid biosynthesis and nucleotide biosynthesis. This allows cancer cells to survive and proliferate even in low-nutrient and oxygen conditions, making glycolytic enzymes a promising target for various anti-cancer agents. Oncogenic activation is also caused by the uncontrolled production and activity of glycolytic enzymes. Nevertheless, in addition to conventional glycolytic processes, some glycolytic enzymes are involved in non-canonical functions such as transcriptional regulation, autophagy, epigenetic changes, inflammation, various signaling cascades, redox regulation, oxidative stress, obesity and fatty acid metabolism, diabetes and neurodegenerative disorders, and hypoxia. The mechanisms underlying the non-canonical glycolytic enzyme activities are still not comprehensive. This review summarizes the current findings on the mechanisms fundamental to the non-glycolytic actions of glycolytic enzymes and their intermediates in maintaining the tumor microenvironment.

4.
Cell Biochem Biophys ; 81(2): 269-283, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37233844

RESUMEN

The tubulin-microtubule system is a major target for a variety of small molecules which can interfere in cell cycle progression. Therefore, it serves as a prospective to control the incessant division of cancer cells. To identify novel inhibitors of the tubulin-microtubule system, a group of estrogen derivatives has been tested with tubulin as a target since literature surveys portray coveted behaviour from the same. Out of them, ß-Estradiol-6-one 6- (O-carboxy methyl Oxime) abbreviated as Oxime, disrupts the cytoskeleton network and induces apoptosis with nuclei fragmentation. It has been revealed from the work that Oxime targets the colchicine binding site and binds tubulin in an entropy-driven manner. This suggests that structural variation might play a key role in modulating the anti-mitotic role of estrogen derivatives. Our work reveals that Oxime might serve as a lead molecule to nurture anti-cancer research, having the potential for recovery of the vast cancer population.


Asunto(s)
Antimitóticos , Antineoplásicos , Tubulina (Proteína)/química , Antimitóticos/metabolismo , Moduladores de Tubulina/farmacología , Moduladores de Tubulina/química , Moduladores de Tubulina/metabolismo , Mitosis , Estudios Prospectivos , Microtúbulos/metabolismo , Estradiol/farmacología , Estradiol/metabolismo , Sitios de Unión , Estrógenos/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/metabolismo
5.
Exp Dermatol ; 28(11): 1328-1335, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31535738

RESUMEN

Tumor necrosis factor-α (TNF-α)-induced keratinocyte inflammation plays a key role in the pathogenesis of multiple inflammatory skin diseases. Here we investigated the anti-inflammatory effect of S-allyl cysteine (SAC) on TNF-α-induced HaCaT keratinocyte cells and the mechanism behind its anti-inflammatory potential. SAC was found to inhibit TNF-α-stimulated cytokine expression. Further, SAC was found to inhibit TNF-α-induced activation of p38, JNK and NF-κB pathways. Interestingly, SAC was found to differentially regulate ERK MAP kinase in cells. TNF-α-induced transient ERK activation and SAC treatment resulted in sustained ERK activation both in the presence and absence of TNF-α. Additionally, SAC failed to inhibit the TNF-α-induced expression of the pro-inflammatory cytokines TNF-α and IL-1ß when cells were treated with the MEK inhibitor PD98059, suggesting that the anti-inflammatory effect of SAC is via sustained activation of the ERK pathway. Since ERK activation has been reported to negatively regulate NF-κB-driven gene expression and we find that SAC activates ERK and negatively regulates NF-κB, we investigated whether there existed any crosstalk between the ERK and the NF-κB pathways. NF-κB-dependent reporter assay, visualization of the nuclear translocation of NF-κB-p65 subunit and determination of the cellular levels of I-κB, the inhibitor of NF-κB, revealed that SAC inhibited TNF-α-induced NF-κB activation, and PD98059 treatment reversed this effect. These results collectively suggest that SAC inhibits TNF-α-induced inflammation in HaCaT cells via a combined effect entailing the inhibition of the p38 and the JNK pathways and NF-κB pathway via the sustained activation of ERK.


Asunto(s)
Cisteína/análogos & derivados , Queratinocitos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Línea Celular Transformada , Cisteína/metabolismo , Humanos , Interleucina-1beta/metabolismo , Sistema de Señalización de MAP Quinasas , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
6.
Biomed Res Int ; 2018: 3169431, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30515391

RESUMEN

Hydrogen peroxide (H2O2) mediated oxidative stress leading to hepatocyte apoptosis plays a pivotal role in the pathophysiology of several chronic liver diseases. This study demonstrates that S-allyl cysteine (SAC) renders cytoprotective effects on H2O2 induced oxidative damage and apoptosis in HepG2 cells. Cell viability assay showed that SAC protected HepG2 cells from H2O2 induced cytotoxicity. Further, SAC treatment dose dependently inhibited H2O2 induced apoptosis via decreasing the Bax/Bcl-2 ratio, restoring mitochondrial membrane potential (∆Ψm), inhibiting mitochondrial cytochrome c release, and inhibiting proteolytic cleavage of caspase-3. SAC protected cells from H2O2 induced oxidative damage by inhibiting reactive oxygen species accumulation and lipid peroxidation. The mechanism underlying the antiapoptotic and antioxidative role of SAC is the induction of the heme oxygenase-1 (HO-1) gene in an NF-E2-related factor-2 (Nrf-2) and Akt dependent manner. Specifically SAC was found to induce the phosphorylation of Akt and enhance the nuclear localization of Nrf-2 in cells. Our results were further confirmed by specific HO-1 gene knockdown studies which clearly demonstrated that HO-1 induction indeed played a key role in SAC mediated inhibition of apoptosis and ROS production in HepG2 cells, thus suggesting a hepatoprotective role of SAC in combating oxidative stress mediated liver diseases.


Asunto(s)
Hemo-Oxigenasa 1/genética , Factor 2 Relacionado con NF-E2/genética , Estrés Oxidativo/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/genética , Apoptosis/efectos de los fármacos , Cisteína/análogos & derivados , Cisteína/farmacología , Citoprotección/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Células Hep G2 , Humanos , Peróxido de Hidrógeno/toxicidad , Especies Reactivas de Oxígeno , Transducción de Señal , Activación Transcripcional/efectos de los fármacos
7.
Sci Rep ; 7(1): 11645, 2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28928443

RESUMEN

Excessive manganese exposure is toxic, but a comprehensive biochemical picture of this assault is poorly understood. Whether oxidative stress or reduced energy metabolism under manganese exposure causes toxicity is still a debate. To address this, we chose Δmnt P Escherichia coli, a highly manganese-sensitive strain, in this study. Combining microarray, proteomics, and biochemical analyses, we show that the chronic manganese exposure rewires diverse regulatory and metabolic pathways. Manganese stress affects protein and other macromolecular stability, and envelope biogenesis. Most importantly, manganese exposure disrupts both iron-sulfur cluster and heme-enzyme biogenesis by depleting cellular iron level. Therefore, the compromised function of the iron-dependent enzymes in the tricarboxylic acid cycle, and electron transport chain impede ATP synthesis, leading to severe energy deficiency. Manganese stress also evokes reactive oxygen species, inducing oxidative stress. However, suppressing oxidative stress does not improve oxidative phosphorylation and cell growth. On the contrary, iron supplementation resumed cell growth stimulating oxidative phosphorylation. Therefore, we hypothesize that affected energy metabolism is the primal cause of manganese toxicity.


Asunto(s)
Metabolismo Energético/efectos de los fármacos , Proteínas de Escherichia coli/biosíntesis , Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Manganeso/farmacología , Estrés Oxidativo/efectos de los fármacos , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Especies Reactivas de Oxígeno/metabolismo
8.
J Biol Chem ; 291(46): 24029-24035, 2016 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-27679485

RESUMEN

δ, a small protein found in most Gram-positive bacteria was, for a long time, thought to be a subunit of RNA polymerase (RNAP) and was shown to be involved in recycling of RNAP at the end of each round of transcription. However, how δ participates in both up-regulation and down-regulation of genes in vivo remains unclear. We have recently shown, in addition to the recycling of RNAP, δ functions as a transcriptional activator by binding to an A-rich sequence located immediately upstream of the -35 element, consequently facilitating the open complex formation. The result had explained the mechanism of up-regulation of the genes by δ. Here, we show that Bacillus subtilis δ could also function as a transcriptional repressor. Our results demonstrate that δ binds to an A-rich sequence located near the -35 element of the spo0B promoter, the gene involved in the regulatory cascade of bacterial sporulation and inhibits the open complex formation due to steric clash with σ region 4.2. We observed a significant increase in the mRNA level of the spo0B gene in a δ-knock-out strain of B. subtilis compared with the wild-type. Thus, the results report a novel function of δ, and suggest the mechanism of down-regulation of genes in vivo by the protein.


Asunto(s)
Bacillus subtilis/fisiología , Proteínas Bacterianas/metabolismo , Proteínas Represoras/metabolismo , Elementos de Respuesta/fisiología , Esporas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Proteínas Represoras/genética , Esporas Bacterianas/genética
9.
J Invest Dermatol ; 130(10): 2448-56, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20505747

RESUMEN

Prostaglandin D(2) (PGD(2)) is known to have antipruritic activity by suppressing histamine release. However, agents that can topically induce PGD(2) for itch relief are not well established. The antimycotic sertaconazole nitrate (STZ) has been shown to exhibit anti-itch properties; however, the mechanism for this activity has not been elucidated. STZ mitigated degranulation of RBL-2H3 (rat basophilic leukemia) mast cells induced by compound 48/80, a pruritogenic agent known to promote the release of histamine, and augmented PGD(2) production in mast cells and macrophages. Addition of exogenous PGD(2) abrogated compound 48/80-induced degranulation by acting through the prostanoid D receptor 1 (DP1). STZ induced p38 mitogen-activated protein kinase (MAPK) phosphorylation in mast cells and a pharmacological inhibitor of p38 MAPK, SB203580, resulted in the attenuation of PGD(2) levels. Finally, in a murine model of pruritus, the scratching behavior induced by compound 48/80 was mitigated by topical application of STZ. This effect was reversed by the addition of the cyclooxygenase inhibitor, ibuprofen, or a DP1 receptor antagonist (MK0524). Collectively, these results suggest that STZ mediates its anti-itch effects by boosting the antipruritic agent, PGD(2), by the activation of the p38-MAPK pathway. This is the first report to demonstrate a promising approach to topically induce PGD(2) for improving pruritus.


Asunto(s)
Imidazoles/farmacología , Macrófagos/efectos de los fármacos , Mastocitos/efectos de los fármacos , Prostaglandina D2/metabolismo , Prurito/tratamiento farmacológico , Tiofenos/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Administración Tópica , Animales , Antifúngicos/farmacología , Degranulación de la Célula/efectos de los fármacos , Línea Celular , Interacciones Farmacológicas , Histamina/metabolismo , Indoles/farmacología , Macrófagos/citología , Macrófagos/metabolismo , Mastocitos/citología , Mastocitos/metabolismo , Ratones , Ratones Endogámicos ICR , Prostaglandina D2/farmacología , Prurito/inducido químicamente , Prurito/metabolismo , Ratas , Receptores de Prostaglandina/metabolismo , p-Metoxi-N-metilfenetilamina/farmacología
10.
Biochem Biophys Res Commun ; 390(3): 786-90, 2009 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-19835845

RESUMEN

We have previously shown that Janus kinase 3, a member of the family of non-receptor protein tyrosine kinases, plays a critical role in the regulation of FcepsilonRI-mediated mast cell responses. In the current study, we investigated the role of another JAK family member, JAK2, in these responses. Our results show that the treatment of IgE-sensitized mouse mast cells with an inhibitor of JAK2 (AG490) blocked the release of leukotriene C(4) in a dose-dependent fashion after antigen challenge. However, prostaglandin PG D(2) production and degranulation were not affected under identical experimental conditions. Transfection of RBL-2H3 mast cells with JAK-2 specific small interfering RNA resulted in a 50% reduction of LTC(4) release in response to FcepsilonRI crosslinking, but did not inhibit mast cell degranulation or calcium ionophore-induced LTC(4) release, indicating involvement of JAK2 in IgE receptor-mediated leukotriene release. Taken together, these data suggest that JAK2 is a critical regulator of IgE/antigen-induced production of LTC(4) in mast cells.


Asunto(s)
Janus Quinasa 2/metabolismo , Leucotrieno C4/biosíntesis , Mastocitos/enzimología , Receptores de IgE/metabolismo , Animales , Células Cultivadas , Janus Quinasa 2/genética , Ratones , Inhibidores de Proteínas Quinasas/farmacología , ARN Interferente Pequeño/genética , Tirfostinos/farmacología
11.
Exp Dermatol ; 18(12): 1070-2, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19624731

RESUMEN

The etiology of acne is a complex process, and acne is one of the most common skin disorders affecting millions of people. The pathogenesis of acne is closely associated with the bacterium, Propionibacterium acnes which was previously known as Corynebacterium parvum. Both viable and non-viable P. acnes/C. parvum have been shown to induce an immunostimulatory effect in vivo, suggesting that even dead bacteria continue to activate an inflammatory response. Acne treatments with lasers or devices, induce a bactericidal effect through heat generation which may not address the immunogenic activity of P. acnes and the resulting acne inflammation. Therefore, we sought to determine whether killed P. acnes is capable of inducing an inflammatory response and therefore could be a contributing factor in acne. Direct heat treatment of P. acnes cultures with temperatures ranging from 50 degrees C to 80 degrees C reduced P. acnes viability. Both viable and heat-killed P. acnes activated the p38 MAP kinase and its downstream substrate Hsp27. Stimulating keratinocytes with normal and heat-inactivated P. acnes resulted in an induction of proinflammatory nitric oxide and IL-8 production. Thus killed P. acnes is capable of inducing inflammation in skin suggesting that therapies that have both bactericidal and anti-inflammatory effects may result in a more effective treatment of patients with acne than treatments that are bactericidal alone.


Asunto(s)
Acné Vulgar/microbiología , Queratinocitos/metabolismo , Propionibacterium acnes , Piel/microbiología , Acné Vulgar/metabolismo , Células Cultivadas , Proteínas de Choque Térmico HSP27/metabolismo , Proteínas de Choque Térmico , Calor , Inflamación/metabolismo , Inflamación/microbiología , Interleucina-8/metabolismo , Queratinocitos/microbiología , Viabilidad Microbiana , Chaperonas Moleculares , Óxido Nítrico/metabolismo , Piel/citología , Enfermedades Cutáneas Bacterianas/metabolismo , Enfermedades Cutáneas Bacterianas/microbiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
Arch Dermatol Res ; 300(10): 569-74, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18461339

RESUMEN

Oatmeal has been used for centuries as a soothing agent to relieve itch and irritation associated with various xerotic dermatoses; however few studies have sought to identify the active phytochemical(s) in oat that mediate this anti-inflammatory activity. Avenanthramides are phenolic compounds present in oats at approximately 300 parts per million (ppm) and have been reported to exhibit anti-oxidant activity in various cell-types. In the current study we investigated whether these compounds exert anti-inflammatory activity in the skin. We found that avenanthramides at concentrations as low as 1 parts per billion inhibited the degradation of inhibitor of nuclear factor kappa B-alpha (IkappaB-alpha) in keratinocytes which correlated with decreased phosphorylation of p65 subunit of nuclear factor kappa B (NF-kappaB). Furthermore, cells treated with avenanthramides showed a significant inhibition of tumor necrosis factor-alpha (TNF-alpha) induced NF-kappaB luciferase activity and subsequent reduction of interleukin-8 (IL-8) release. Additionally, topical application of 1-3 ppm avenanthramides mitigated inflammation in murine models of contact hypersensitivity and neurogenic inflammation and reduced pruritogen-induced scratching in a murine itch model. Taken together these results demonstrate that avenanthramides are potent anti-inflammatory agents that appear to mediate the anti-irritant effects of oats.


Asunto(s)
Avena , Flavonoides/uso terapéutico , Inflamación/tratamiento farmacológico , Fenoles/uso terapéutico , Fitoterapia , Prurito/tratamiento farmacológico , ortoaminobenzoatos/uso terapéutico , Animales , Células Cultivadas , Dermatitis por Contacto/tratamiento farmacológico , Dermatitis por Contacto/etiología , Dermatitis por Contacto/metabolismo , Modelos Animales de Enfermedad , Diterpenos/efectos adversos , Flavonoides/farmacología , Humanos , Inflamación/etiología , Inflamación/metabolismo , Interleucina-8/metabolismo , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Ratones , Ratones Endogámicos ICR , FN-kappa B/metabolismo , Oxazolona/efectos adversos , Fenoles/farmacología , Polifenoles , Prurito/etiología , Prurito/metabolismo , Transducción de Señal/fisiología , ortoaminobenzoatos/farmacología
13.
J Invest Dermatol ; 128(5): 1116-22, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18007587

RESUMEN

Heat-shock protein 27 (Hsp27) is a member of the small Hsp family that functions as molecular chaperones and protects cells against environmental stress. Hsp27 is expressed in the upper epidermal layers of normal human skin and has been reported to play a role in keratinocyte differentiation and apoptosis. In this investigation, we show an additional role of Hsp27 in the regulation of inflammatory pathways in keratinocytes. Downregulation of Hsp27 using Hsp27-specific small interfering RNA increased prostaglandin E(2) (PGE(2)) production in both unstimulated and tumor necrosis factor-alpha (TNF-alpha)-stimulated keratinocytes. Moreover, downregulation of Hsp27 increased the release of the pro-inflammatory cytokine IL-8 from TNF-alpha-stimulated and UV-irradiated keratinocytes, and this increase was inhibited by pretreatment with the NF-kappaB inhibitor BAY11-7082. Further studies showed that downregulation of Hsp27 resulted in induction of NF-kappaB reporter activity in keratinocytes. This correlated with enhanced degradation of IkappaB-alpha protein and accumulation of phosphorylated IkappaB-alpha in Hsp27 knockdown cells. Moreover, Hsp27 associated with the IkappaB kinase (IKK) complex. As synthesis of the pro-inflammatory cytokine IL-8 and the prostanoid PGE(2) are regulated by NF-kappaB, this could be a probable mechanism by which Hsp27 modulates the production of these inflammatory cytokines. Thus, Hsp27 plays a protective role in regulating inflammatory responses in skin.


Asunto(s)
Proteínas de Choque Térmico/metabolismo , Mediadores de Inflamación/farmacología , Queratinocitos/inmunología , FN-kappa B/metabolismo , Proteínas de Neoplasias/metabolismo , Transducción de Señal/inmunología , Línea Celular , Dinoprostona/metabolismo , Proteínas de Choque Térmico HSP27 , Proteínas de Choque Térmico/genética , Humanos , Queratinocitos/citología , Queratinocitos/metabolismo , Chaperonas Moleculares , Proteínas de Neoplasias/genética , Fosforilación , ARN Interferente Pequeño , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología
14.
J Invest Dermatol ; 128(2): 336-44, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17637821

RESUMEN

Sertaconazole nitrate is an antifungal agent that exhibits anti-inflammatory activity; however, the mechanism for this action was unknown. We investigated the cellular mechanisms by which sertaconazole exerts its anti-inflammatory activity in keratinocytes and human peripheral blood mononuclear cells (PBMCs). Paradoxically, sertaconazole was found to activate the proinflammatory p38 mitogen-activated protein kinase. Treatment with sertaconazole also resulted in the induction of cyclooxygenase-2 (COX-2) and the subsequent release of prostaglandin E2 (PGE2). Knocking down p38 in keratinocytes using small interfering RNA resulted in an inhibition of sertaconazole-induced PGE2 release confirming that activation of p38 was required for PGE2 production. Additionally, in stimulated keratinocytes and human PBMCs, sertaconazole was found to suppress the release of cytokines. Treatment with anti-PGE2 antiserum or the COX-2 inhibitor NS398 reversed the inhibitory effects of sertaconazole on the release of proinflammatory cytokines, linking endogenous PGE2 with the anti-inflammatory effects. Finally, in an in vivo mouse model of tetradecanoyl phorbol acetate (TPA)-induced dermatitis, the sertaconazole-mediated inhibition of TPA-induced ear edema was reversed by NS398. Biochemical analysis of tissue biopsies revealed increase in PGE2 levels in sertaconazole-treated mice. Thus, activation of the p38-COX-2-PGE2 pathway by agents such as sertaconazole provides anti-inflammatory therapeutic benefits.


Asunto(s)
Antiinflamatorios/farmacología , Antifúngicos/farmacología , Dermatitis por Contacto/tratamiento farmacológico , Imidazoles/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Tiofenos/farmacología , Acné Vulgar/tratamiento farmacológico , Acné Vulgar/inmunología , Acné Vulgar/microbiología , Animales , Biopsia , Carcinógenos/farmacología , Línea Celular , Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Dermatitis por Contacto/inmunología , Dermatitis por Contacto/patología , Dinoprostona/metabolismo , Activación Enzimática/efectos de los fármacos , Activación Enzimática/inmunología , Infecciones por Bacterias Grampositivas/tratamiento farmacológico , Infecciones por Bacterias Grampositivas/inmunología , Humanos , Irritantes , Queratinocitos/citología , Queratinocitos/efectos de los fármacos , Queratinocitos/inmunología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Ratones , Propionibacterium acnes , ARN Interferente Pequeño , Acetato de Tetradecanoilforbol/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
15.
Arch Dermatol Res ; 300(2): 69-80, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18071724

RESUMEN

The skin is under continual assault from a variety of damaging environmental factors such as ultraviolet irradiation and atmospheric pollutants, and as organisms age the cumulative damage exceeds the capacity of endogenous antioxidant defenses resulting in chronic inflammation and premature aging. Botanical extracts such as Feverfew containing naturally occurring antioxidants could replenish the depleted cutaneous stores and perhaps forestall these degenerative changes. A parthenolide-depleted extract of Feverfew (PD-Feverfew), which was free of sensitization potential, was found to possess free radical scavenging activity against a wide range of reactive oxygen species and with greater activity than Vitamin C. In vitro, PD-Feverfew restored cigarette smoke-mediated depletion of cellular thiols, attenuated the formation of UV-induced hydrogen peroxide and reduced pro-inflammatory cytokine release. In vivo, topical PD-Feverfew reduced UV-induced epidermal hyperplasia, DNA damage and apoptosis. In a clinical study PD-Feverfew treatment significantly reduced erythema versus placebo 24 h post-UV exposure. Through the ability to scavenge free radicals, preserve endogenous antioxidant levels, reduce DNA damage and induce DNA repair enzymes, which can help repair damaged DNA, parthenolide-depleted extract of Feverfew may protect skin from the numerous external aggressions encountered daily by the skin and reduce the damage to oxidatively challenged skin.


Asunto(s)
Exposición a Riesgos Ambientales/efectos adversos , Eritema/prevención & control , Hiperplasia/prevención & control , Extractos Vegetales/uso terapéutico , Piel/patología , Piel/efectos de la radiación , Tanacetum parthenium , Rayos Ultravioleta/efectos adversos , Administración Tópica , Animales , Estudios de Casos y Controles , Células Cultivadas , Reparación del ADN , Eritema/etiología , Femenino , Depuradores de Radicales Libres/uso terapéutico , Humanos , Peróxido de Hidrógeno/antagonistas & inhibidores , Peróxido de Hidrógeno/efectos de la radiación , Hiperplasia/etiología , Inflamación , Masculino , Ratones , Ratones Pelados , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Sesquiterpenos/aislamiento & purificación , Sesquiterpenos/metabolismo , Piel/inmunología , Envejecimiento de la Piel/efectos de la radiación , Fumar/efectos adversos , Porcinos
16.
Int Immunopharmacol ; 7(5): 555-67, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17386403

RESUMEN

Mast cells have long been known to play a detrimental role in the pathogenesis of IgE-associated allergic disorders by their ability to release a wide variety of pro-inflammatory mediators. A number of studies, however, have demonstrated that mast cells play a beneficial role in innate host defense against bacterial infections. Since mast cells clearly play both physiological and pathophysiological functions in the body, it is important to learn about the components of mast cells that drive these responses. The functional roles of mast cell in vivo have been principally characterized by comparing the biological responses in mast cell-deficient mice (WBB6F(1)-W/W(v)), their normal wild-type littermates (WBB6F(1)-+/+) and mast cell deficient mice reconstituted locally or systemically with mast cells cultured from the bone marrow cells of WBB6F(1)-+/+ mice (WBB6F(1)-W/W(v)+MC). Recently investigators have demonstrated that mast cell-deficient mice (WBB6F(1)-W/W(v)) can be reconstituted with mast cells derived in vitro from the bone marrow cells of certain gene knock-out mice or genetically-manipulated embryonic stem cells. This novel approach of analyzing the biological consequences of gene mutations in mast cells will help us to better understand the role of individual gene products in mast cell responses. In this review, we discuss these new approaches to investigate the functions of mast cells in vivo.


Asunto(s)
Inmunoglobulina E/fisiología , Mastocitos/fisiología , Animales , Células Cultivadas , Humanos , Inmunidad Innata/fisiología , Ratones , Ratones Noqueados
17.
Eur J Pharmacol ; 539(3): 195-204, 2006 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-16712837

RESUMEN

Cytosolic phospholipase A(2) (cPLA(2)) plays a pivotal role in inflammation by catalyzing the release of arachidonic acid, a substrate for lipoxygenase and cyclooxygenase enzymes, from membrane phospholipids. In the present study we examined the role of cPLA(2) in inflammatory responses through the use of a specific inhibitor of the enzyme, cPLA(2), arachidonyl trifluoromethyl ketone (AACOCF3). Interestingly, we observed that AACOCF3 is an inhibitor of chronic but not acute inflammatory responses. Specifically, AACOCF3 inhibited phorbol 12-myristate 13-acetate (PMA)-induced chronic ear edema in mice. Additionally, oral treatment of ovalbumin-sensitized/ovalbumin-challenged BALB/c mice with 20 mg/kg AACOCF3 prevented the development of airway hyper-responsiveness in a model of asthma. Furthermore, AACOCF3 decreased cellular recruitment in the airway lumen and airway inflammation after the ovalbumin challenge. Taken together, these results suggest that a potent and specific chemical inhibitor of cPLA(2) may be useful for the treatment of chronic inflammatory diseases including rheumatoid arthritis, inflammatory bowel disease, psoriasis, and asthma.


Asunto(s)
Ácidos Araquidónicos/administración & dosificación , Citosol/enzimología , Sistemas de Liberación de Medicamentos/métodos , Edema/enzimología , Edema/prevención & control , Fosfolipasas A/antagonistas & inhibidores , Fosfolipasas A/metabolismo , Alérgenos/efectos adversos , Animales , Asma/tratamiento farmacológico , Asma/enzimología , Asma/patología , Movimiento Celular/efectos de los fármacos , Enfermedad Crónica , Citosol/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/administración & dosificación , Femenino , Fosfolipasas A2 Grupo IV , Inflamación/tratamiento farmacológico , Inflamación/enzimología , Inflamación/prevención & control , Leucocitos/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Fosfolipasas A2
18.
Biochem J ; 387(Pt 2): 315-24, 2005 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15537391

RESUMEN

The ERK (extracellular-signal regulated-kinase)/MAPK (mitogen-activated protein kinase) pathway can regulate transcription, proliferation, migration and apoptosis. The small DED (death-effector domain) protein PEA-15 (phosphoprotein enriched in astrocytes-15) binds ERK and targets it to the cytoplasm. Other DED-containing proteins including cFLIP and DEDD can also regulate signal transduction events and transcription in addition to apoptosis. In the present study, we report the identification of a novel DED-containing protein called Vanishin. The amino acid sequence of Vanishin is closest in similarly to PEA-15 (61% identical). Vanishin mRNA is expressed in several mouse tissues and in both mouse and human cell lines. Interestingly, Vanishin is regulated by ubiquitinylation and subsequent degradation by the 26 S proteasome. The ubiquitinylation is complex and occurs at both the internal lysine residues and the N-terminus. We further show that Vanishin binds ERK/MAPK but not the DED proteins Fas-associated death domain, caspase 8 or PEA-15. Vanishin is present in both the nucleus and Golgi on overexpression and forces increased ERK accumulation in the nucleus in the absence of ERK stimulation. Moreover, Vanishin expression inhibits ERK activation and ERK-dependent transcription in cells, but does not alter MAPK/ERK activity. Therefore Vanishin is a novel regulator of ERK that is controlled by ubiquitinylation.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Secuencia de Aminoácidos , Animales , Apoptosis/fisiología , Proteínas Reguladoras de la Apoptosis , Línea Celular , Proliferación Celular , Supervivencia Celular/fisiología , Regulación hacia Abajo , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/química , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Humanos , Péptidos y Proteínas de Señalización Intracelular , Datos de Secuencia Molecular , Fosfoproteínas , Unión Proteica , Homología de Secuencia de Aminoácido , Transducción de Señal/fisiología , Ubiquitina/metabolismo
19.
Biochem Pharmacol ; 64(10): 1469-81, 2002 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-12417260

RESUMEN

Nitric oxide is an important mediator of excessive cell growth and inflammation associated with many epidermal proliferative disorders. It is a highly reactive oxidant generated in keratinocytes and macrophages via the inducible form of the enzyme nitric oxide synthase (NOS2). In the present studies, we examined the effects of ultraviolet light (UVB, 2.5-25mJ/cm(2)) on interferon-gamma (IFN-gamma)-induced expression of NOS2 in these cells. Transient transfection assays using wild-type and mutant NOS2 promoter/luciferase reporter constructs showed that DNA binding of the transcription factors Stat1 and NF-kappaB was essential for optimal expression of the NOS2 gene. Whereas NF-kappaB was constitutively expressed in both cell types, Stat1 phosphorylation and nuclear binding activity were dependent upon IFN-gamma. UVB light, which is used therapeutically to treat inflammatory dermatosis, was found to suppress IFN-gamma-induced expression of NOS2 mRNA and protein, and nitric oxide production in both keratinocytes and macrophages. In macrophages, this was associated with complete inhibition of NF-kappaB nuclear binding activity and partial (approximately 20-25%) reduction of Stat1 activity. In keratinocytes, both responses were partially reduced at the highest doses of UVB light (15-25mJ/cm(2)). Whereas in macrophages UVB light suppressed NOS2 wild-type promoter-luciferase reporter activity, this activity was stimulated in keratinocytes. These data suggest that UVB light functions to suppress NOS2 gene expression in macrophages by inhibiting the activity of key regulatory transcription factors. In contrast, in keratinocytes, inhibition occurs downstream of NOS2 promoter activity.


Asunto(s)
Queratinocitos/efectos de la radiación , Macrófagos/efectos de la radiación , Óxido Nítrico/biosíntesis , Animales , Proteínas de Unión al ADN/metabolismo , Expresión Génica/efectos de la radiación , Genes Reporteros , Queratinocitos/metabolismo , Macrófagos/metabolismo , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa de Tipo II , Factor de Transcripción STAT1 , Transducción de Señal/fisiología , Transactivadores/metabolismo , Rayos Ultravioleta
20.
Am J Physiol Cell Physiol ; 283(4): C1267-77, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12225989

RESUMEN

The 60-kDa heat shock protein (HSP60), an endogenous ligand for the toll-like 4 receptor, is generated in response to inflammation, tissue injury, and/or stress and stimulates macrophages to produce cytotoxic and proinflammatory mediators including nitric oxide, tumor necrosis factor (TNF)-alpha, interleukin (IL)-6, and IL-12. In the present studies we report that HSP60 is an effective inducer of cyclooxygenase-2 (COX-2) in macrophages, as well as endothelial cells. In both cell types, the synthesis of COX-2 was coordinate with induction of nitric oxide synthase (NOS)-2 and with nitric oxide production. With the use of promoter constructs in transient transfection assays, optimal expression of COX-2 in macrophages was found to require nuclear factor (NF)-kappaB, the cAMP-response element (CRE), and NF-IL-6, but not the E-box. Mobility shift assays revealed that HSP60 induced NF-kappaB and CRE binding activity, while CCAAT/enhancer binding protein (C/EBP), which binds to NF-IL-6, was constitutively active in the cells. Both c-Jun and CRE binding protein (CREB) bound to the CRE, while C/EBP-beta bound to NF-IL-6. These data indicate that NF-kappaB, C/EBP-beta, c-Jun, and CREB are important in HSP60-induced expression of COX-2. The c-Jun-NH(2)-terminal kinase (JNK), p44/42 mitogen-activated protein (MAP) kinase [extracellular signal-regulated kinase 1/2 (ERK1/2)], and p38 MAP kinase were rapidly activated by HSP60 in the macrophages. PD-98059, an inhibitor of phosphorylation of ERK1/2, caused a marked inhibition of HSP60-induced COX-2 and NOS-2 expression. Unexpectedly, SB-203580, a p38 kinase antagonist, was found to block HSP60-induced expression of COX-2, but not NOS-2. These data indicate that both ERK1/2 kinase and p38 kinase play a role in regulating HSP60-induced expression of COX-2.


Asunto(s)
Chaperonina 60/farmacología , Endotelio/enzimología , Inducción Enzimática/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Isoenzimas/metabolismo , Macrófagos/enzimología , Prostaglandina-Endoperóxido Sintasas/metabolismo , Animales , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Células Cultivadas , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Ciclooxigenasa 2 , Endotelio/citología , Endotelio/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Genes Reporteros , Interferón gamma/farmacología , Isoenzimas/antagonistas & inhibidores , Isoenzimas/genética , Lipopolisacáridos/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mutagénesis Sitio-Dirigida , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Regiones Promotoras Genéticas , Prostaglandina-Endoperóxido Sintasas/genética , ARN Mensajero/biosíntesis , Ratas , Secuencias Reguladoras de Ácidos Nucleicos/fisiología , Factores de Transcripción/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA