Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Osteoarthritis Cartilage ; 30(9): 1210-1221, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35513246

RESUMO

OBJECTIVE: This study was performed to characterize selected rhodanine derivatives as potential preclinical disease-modifying drugs for experimental osteoarthritis (OA) in mice. METHODS: Three rhodanine derivatives, designated rhodanine (R)-501, R-502, and R-503, were selected as candidate OA disease-modifying drugs. Their effects were evaluated by intra-articular (IA) injection in OA mouse models induced by DMM (destabilization of the medial meniscus) or adenoviral overexpression in joint tissues of hypoxia-inducible factor (HIF)-2α or zinc importer ZIP8. The regulatory mechanisms impacted by the rhodanine derivatives were examined in primary-culture chondrocytes and fibroblast-like synoviocytes (FLS). RESULTS: All three rhodanine derivatives inhibited OA development caused by DMM or overexpression of HIF-2α or ZIP8. Compared to vehicle-treated group, for example, IA injection of R-501 in DMM-operated mice reduced median OARSI grade from 3.78 (IQR 3.00-5.00) to 1.89 (IQR 0.94-2.00, P = 0.0001). R-502 and R-503 also reduced from 3.67 (IQR 2.11-4.56) to 2.00 (IQR 1.00-2.00, P = 0.0030) and 2.00 (IQR 1.83-2.67, P = 0.0378), respectively. Mechanistically, the rhodanine derivatives inhibited the nuclear localization and transcriptional activity of HIF-2α in chondrocytes and FLS. They did not bind to Zn2+ or modulate Zn2+ homeostasis in chondrocytes or FLS; instead, they inhibited the nuclear localization and transcriptional activity of the Zn2+-dependent transcription factor, MTF1. HIF-2α, ZIP8, and interleukin-1ß could upregulate matrix-degrading enzymes in chondrocytes and FLS, and the rhodanine derivatives inhibited these effects. CONCLUSION: IA administration of rhodanine derivatives significantly reduced OA pathogenesis in various mouse models, demonstrating that these derivatives have disease-modifying therapeutic potential against OA pathogenesis.


Assuntos
Cartilagem Articular , Osteoartrite , Rodanina , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Cartilagem Articular/patologia , Condrócitos/metabolismo , Modelos Animais de Doenças , Camundongos , Osteoartrite/metabolismo , Preparações Farmacêuticas/metabolismo , Rodanina/metabolismo , Rodanina/farmacologia
2.
Osteoarthritis Cartilage ; 29(4): 558-567, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33485930

RESUMO

OBJECTIVE: Osteoarthritic cartilage destruction can be regulated by the balance between proteases and anti-proteases. Here, we sought to identify novel cellular protease inhibitors associated with osteoarthritis (OA) pathogenesis. METHODS: Candidate molecules were screened from microarray data of chondrocytes treated with OA-associated catabolic factors. The functions of candidate molecules in OA pathogenesis were examined in primary-culture mouse articular chondrocytes and mouse models of OA, such as those stimulated by destabilization of the medial meniscus (DMM) or intra-articular (IA) injection of adenovirus expressing the candidate gene. The value of the selected candidate molecule as a biomarker of OA was examined by measuring its circulating levels in human and mouse blood. RESULTS: Bioinformatic analysis identified secretory leukocyte peptidase inhibitor (SLPI) as a highly upregulated cellular protease inhibitor in chondrocytes treated with pathogenic catabolic factors, including interleukin (IL)-1ß, hypoxia-inducible factor (HIF)-2α, and zinc importer ZIP8. The adenovirus-mediated overexpression of SLPI in joint tissues did not cause any OA-like change or modulate DMM- or HIF-2α-induced experimental OA in mice. SLPI also did not markedly modulate the expression of OA-associated catabolic or anabolic factors in chondrocytes. However, SLPI was specifically upregulated in OA cartilage, and the serum SLPI levels were significantly elevated in human OA patients and experimental OA mice, suggesting that SLPI may be a biomarker of OA. CONCLUSION: Although SLPI is upregulated in OA chondrocytes, it does not appear to per se modulate OA development in mice. However, it may be a potential biomarker of OA in humans and animal models.


Assuntos
Artrite Experimental/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Condrócitos/metabolismo , Osteoartrite do Joelho/genética , Inibidor Secretado de Peptidases Leucocitárias/genética , Inibidor Secretado de Peptidases Leucocitárias/metabolismo , Animais , Artrite Experimental/metabolismo , Cartilagem Articular , Humanos , Meniscos Tibiais/cirurgia , Camundongos , Osteoartrite/genética , Osteoartrite/metabolismo , Osteoartrite do Joelho/metabolismo , Cultura Primária de Células , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sinoviócitos
3.
Osteoarthritis Cartilage ; 25(3): 401-405, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27477830

RESUMO

OBJECTIVE: Lipocalin-2 (LCN2) is a recently characterized adipokine that is upregulated in chondrocytes treated with pro-inflammatory mediators and in the synovial fluid of osteoarthritis (OA) patients. Here, we explored the in vivo functions of LCN2 in OA cartilage destruction in mice. METHODS: The expression levels of LCN2 were determined at the mRNA and protein levels in primary cultured mouse chondrocytes and in human and mouse OA cartilage. Experimental OA was induced in wild-type (WT) or Lcn2-knockout (KO) mice by destabilization of the medial meniscus (DMM) or intra-articular (IA) injection of adenoviruses expressing hypoxia-inducible factor (HIF)-2α (Ad-Epas1), ZIP8 (Ad-Zip8), or LCN2 (Ad-Lcn2). The effect of LCN2 overexpression on the cartilage of WT mice was examined by IA injection of Ad-Lcn2. RESULTS: LCN2 mRNA levels in chondrocytes were markedly increased by the pro-inflammatory cytokines, interleukin (IL)-1ß and tumor necrosis factor-α (TNF-α), and by previously identified catabolic regulators of OA, such as HIF-2α and components of the zinc-ZIP8-MTF1 axis. LCN2 protein levels were also markedly increased in human OA cartilage and cartilage from various experimental mouse models of OA. However, overexpression of LCN2 in chondrocytes did not modulate the expression of cartilage matrix molecules or matrix-degrading enzymes. Furthermore, LCN2 overexpression in mouse cartilage via IA injection of Ad-Lcn2 did not cause OA pathogenesis, and Lcn2 KO mice showed no alteration in DMM-induced OA cartilage destruction. CONCLUSIONS: Our observations collectively suggest that upregulation of LCN2 in OA cartilage is not sufficient or necessary for OA cartilage destruction in mice.


Assuntos
Cartilagem/metabolismo , Lipocalina-2/metabolismo , Osteoartrite/metabolismo , Animais , Cartilagem/patologia , Condrócitos/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoartrite/patologia , Regulação para Cima
4.
Osteoarthritis Cartilage ; 25(3): 397-400, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27480933

RESUMO

OBJECTIVE: In a preliminary study, we found that recently identified catabolic regulators of osteoarthritis (OA), including hypoxia-inducible factor (HIF)-2α and members of the zinc-ZIP8-MTF1 axis, upregulate the E3 ubiquitin ligase, Atrogin-1 (encoded by Fbxo32), in chondrocytes. As the ubiquitination/proteasomal degradation pathways are tightly regulated to modulate the expression of catabolic factors in chondrocytes, we examined the in vivo functions of Atrogin-1 in mouse models of OA. METHODS: The mRNA and protein levels of Atrogin-1 and other regulators of OA were determined in primary cultured mouse chondrocytes, OA human cartilage, and OA cartilage from wild-type (WT) and Fbxo32-knockout (KO) mice subjected to destabilization of the medial meniscus or intra-articular (IA) injection of adenoviruses expressing HIF-2α (Ad-Epas1), ZIP8 (Ad-Zip8), or Atrogin-1 (Ad-Fbxo32). The effect of Atrogin-1 overexpression on the cartilage of WT mice was examined by IA injection of Ad-Fbxo32. RESULTS: Atrogin-1 mRNA levels in chondrocytes were markedly increased by treatment with interleukin-1ß, HIF-2α, and members of the zinc-ZIP8-MTF1 axis. Atrogin-1 protein levels were also increased in OA cartilage from humans and various mouse OA models. However, the forced overexpression of Atrogin-1 in chondrocytes did not modulate the expression of cartilage matrix molecules or matrix-degrading enzymes. Moreover, overexpression of Atrogin-1 in the mouse joint tissues failed to cause OA pathogenesis, and Fbxo32 knockout failed to affect post-traumatic OA cartilage destruction in mice. CONCLUSIONS: Although Atrogin-1 is upregulated in OA cartilage, overexpression of Atrogin-1 in the joint tissues or knockout of Fbxo32 does not affect OA cartilage destruction in mice.


Assuntos
Cartilagem/metabolismo , Modelos Animais de Doenças , Proteínas Musculares/metabolismo , Osteoartrite/metabolismo , Proteínas Ligases SKP Culina F-Box/metabolismo , Animais , Cartilagem/patologia , Condrócitos/metabolismo , Condrócitos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoartrite/patologia , Regulação para Cima
5.
Osteoarthritis Cartilage ; 24(1): 134-45, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26241779

RESUMO

OBJECTIVE: Hypoxia-inducible factor (HIF)-2α and the zinc-ZIP8-MTF1 axis in chondrocytes serve as catabolic regulators of osteoarthritic cartilage destruction by regulating the expression of catabolic factor genes. We explored possible crosstalk between these signaling pathways and its biological significance in osteoarthritis (OA). METHODS: Microarray analysis, various mRNA and protein assays were conducted using primary cultured mouse articular chondrocytes and experimental OA cartilage to reveal molecular mechanisms underlying the crosstalk between HIF-2α and the zinc-ZIP8-MTF1 axis. Experimental OA in mice was induced by intra-articular (IA) injection of adenovirus expressing HIF-2α (Ad-Epas1), ZIP8 (Ad-Zip8), or MTF1 (Ad-Mtf1) in wild-type mice or mice with cartilage-specific conditional knockout of HIF-2α (Epas1(fl/fl);Col2a1-Cre), ZIP8 (Zip8(fl/fl);Col2a1-Cre), or MTF1 (Mtf1(fl/fl);Col2a1-Cre). RESULTS: HIF-2α activated the zinc-ZIP8-MTF1 axis in chondrocytes by upregulating the Zn(2+) transporter ZIP8, thereby increasing Zn(2+) influx and activating the downstream transcription factor MTF1. The zinc-ZIP8-MTF1 axis, in turn, acted as a novel transcriptional regulator of HIF-2α. HIF-2α-induced activation of the zinc-ZIP8-MTF1 axis amplified HIF-2α regulation of OA cartilage destruction by synergistically promoting expression of matrix-degrading enzymes. Thus, HIF-2α-induced activation of the zinc-ZIP8-MTF1 axis, together with zinc-ZIP8-MTF1 regulation of HIF-2α, acted collectively to synergistically promote expression of matrix-degrading enzymes and OA cartilage destruction. CONCLUSION: Our findings identify a reciprocal activation mechanism involving HIF-2α and the zinc-ZIP8-MTF1 axis during OA pathogenesis that amplifies catabolic signaling and cartilage destruction.


Assuntos
Artrite Experimental/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Cartilagem Articular/metabolismo , Proteínas de Transporte de Cátions/genética , Condrócitos/metabolismo , Proteínas de Ligação a DNA/genética , Osteoartrite do Joelho/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição/genética , Zinco/metabolismo , Animais , Artrite Experimental/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Cartilagem Articular/citologia , Proteínas de Transporte de Cátions/metabolismo , Células Cultivadas , Imunoprecipitação da Cromatina , Proteínas de Ligação a DNA/metabolismo , Técnicas de Introdução de Genes , Masculino , Camundongos , Camundongos Knockout , Osteoartrite do Joelho/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Fatores de Transcrição/metabolismo , Fator MTF-1 de Transcrição
6.
Osteoarthritis Cartilage ; 23(12): 2288-2296, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26209889

RESUMO

OBJECTIVE: Hypoxia-inducible factor-2α (HIF-2α) transcriptionally upregulates Nampt in articular chondrocytes. NAMPT, which exhibits nicotinamide phosphoribosyltransferase activity, in turn causes osteoarthritis (OA) in mice by stimulating the expression of matrix-degrading enzymes. Here, we sought to elucidate whether HIF-2α activates the NAMPT-NAD(+)-SIRT axis in chondrocytes and thereby contributes to the pathogenesis of OA. METHODS: Assays of NAD levels, SIRT activity, reporter gene activity, mRNA, and protein levels were conducted in primary cultured mouse articular chondrocytes. Experimental OA in mice was induced by intra-articular (IA) injection of adenovirus expressing HIF-2α (Ad-Epas1) or NAMPT (Ad-Nampt). The functions of SIRT in OA were examined by IA co-injection of SIRT inhibitors or adenovirus expressing individual SIRT isoforms or shRNA targeting specific SIRT isoforms. RESULTS: HIF-2α activated the NAMPT-NAD(+)-SIRT axis in chondrocytes by upregulating NAMPT, which stimulated NAD(+) synthesis and thereby activated SIRT family members. The activated NAMPT-SIRT pathway, in turn, promoted HIF-2α protein stability by negatively regulating its hydroxylation and 26S proteasome-mediated degradation, resulting in increased HIF-2α transcriptional activity. Among SIRT family members (SIRT1-7), SIRT2 and SIRT4 were positively associated with HIF-2α stability and transcriptional activity in chondrocytes. This reciprocal regulation was required for the expression of catabolic matrix metalloproteinases (MMP3, MMP12, and MMP13) and OA cartilage destruction caused by IA injection of Ad-Epas1 Ad-Nampt. CONCLUSION: The reciprocal regulation of HIF-2α and the NAMPT-NAD(+)-SIRT axis in articular chondrocytes is involved in OA cartilage destruction caused by HIF-2α or NAMPT.


Assuntos
Artrite Experimental/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Cartilagem Articular/metabolismo , Condrócitos/metabolismo , Citocinas/genética , NAD/metabolismo , Nicotinamida Fosforribosiltransferase/genética , Osteoartrite do Joelho/genética , Sirtuína 1/genética , Sirtuína 2/genética , Animais , Artrite Experimental/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Western Blotting , Citocinas/metabolismo , Imunoprecipitação , Metaloproteinase 12 da Matriz/genética , Metaloproteinase 12 da Matriz/metabolismo , Metaloproteinase 13 da Matriz/genética , Metaloproteinase 13 da Matriz/metabolismo , Metaloproteinase 3 da Matriz/genética , Metaloproteinase 3 da Matriz/metabolismo , Camundongos , Nicotinamida Fosforribosiltransferase/metabolismo , Osteoartrite do Joelho/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sirtuína 1/metabolismo , Sirtuína 2/metabolismo , Sirtuínas/genética , Sirtuínas/metabolismo , Regulação para Cima
7.
Cell Death Differ ; 19(3): 440-50, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21869830

RESUMO

Apoptosis of articular chondrocytes is associated with the pathogenesis of osteoarthritis (OA). Recently, we demonstrated that hypoxia-inducible factor (HIF)-2α, encoded by Epas1, causes OA cartilage destruction by regulating the expression of various matrix-degrading enzymes. Here, we investigated the involvement of HIF-2α in chondrocyte apoptosis and OA cartilage destruction. HIF-2α levels in human and mouse OA chondrocytes were markedly elevated in association with increased apoptosis of articular chondrocytes. Overexpression or knockdown of HIF-2α alone did not cause chondrocyte apoptosis. However, HIF-2α expression markedly increased chondrocyte apoptosis in the presence of an agonistic anti-Fas (CD95) antibody. HIF-2α enhanced Fas expression and potentiated downstream signaling pathways, increasing the activity of initiator and executioner caspases. Overexpression of HIF-2α in mouse cartilage tissue, either by intra-articular injection of Epas1 adenovirus (Ad-Epas1) or in the context of chondrocyte-specific Epas1 transgenic mice, increased chondrocyte apoptosis and cartilage destruction. In contrast, chondrocyte-specific knockout of Epas1 in mice suppressed DMM (destabilization of the medial meniscus)-induced chondrocyte apoptosis and inhibited OA cartilage destruction. Moreover, Fas-deficient mice exhibited diminished chondrocyte apoptosis and OA cartilage destruction in response to Ad-Epas1 injection or DMM surgery. Taken together, our results demonstrate that HIF-2α potentiates Fas-mediated chondrocyte apoptosis, which is associated with OA cartilage destruction.


Assuntos
Apoptose , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Cartilagem/metabolismo , Condrócitos/metabolismo , Osteoartrite/metabolismo , Receptor fas/biossíntese , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Cartilagem/patologia , Células Cultivadas , Condrócitos/patologia , Regulação da Expressão Gênica/genética , Humanos , Camundongos , Camundongos Transgênicos , Osteoartrite/genética , Osteoartrite/patologia , Transdução de Sinais/genética , Receptor fas/genética
8.
Clin Exp Dermatol ; 36(7): 769-74, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21623876

RESUMO

BACKGROUND: Expression of Runt-related transcription factor 3 (RUNX3) is reduced in a large number of cancers. However, a few studies have reported higher expression of RUNX3 in several cancers, including basal cell carcinoma (BCC). In light of this, we explored the expression of RUNX3 in skin cancers generally, to determine whether it acts as an oncogene or a tumour-suppressor gene in skin tumours. AIM: To investigate the expression of RUNX3 in normal skin and malignant skin tumours. METHODS: RUNX3 expression was evaluated by western blotting in 24 specimens, comprising 6 malignant melanoma (MM), 6 squamous cell carcinoma (SCC), 6 BCC and 6 normal skin specimens. Immunohistochemical staining was carried out to analyse RUNX3 expression in 16 MM, 16 SCC and 16 BCC specimens. To identify where the protein was expressed, the cytoplasmic and nuclear protein expression of RUNX3 in skin cancer tissues was determined. A cell-proliferation study was performed on an MM line (G361) by small interfering (si)RNA transfection. RESULTS: The western blotting experiments showed that RUNX3 was not expressed in normal skin tissues, but it was overexpressed in all MM and SCC samples, and in five of the six BCC samples. Using immunochemistry, RUNX3 was found to be overexpressed in all cancer tissues analysed. Subcellular fraction analysis revealed that RUNX3 was expressed in the nuclei but not the cytoplasm of all the skin cancer tissues analysed, and RUNX3 silencing by siRNA in G361 cells resulted in a decrease in proliferation. CONCLUSIONS: Based on these results, we suggest that RUNX3 has an oncogenic potential and does not act as a tumour suppressor in skin cancers.


Assuntos
Carcinoma Basocelular/metabolismo , Carcinoma de Células Escamosas/metabolismo , Subunidade alfa 3 de Fator de Ligação ao Core/metabolismo , Melanoma/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Cutâneas/metabolismo , Idoso , Idoso de 80 Anos ou mais , Western Blotting , Carcinoma Basocelular/genética , Carcinoma de Células Escamosas/genética , Linhagem Celular , Núcleo Celular/metabolismo , Proliferação de Células , Feminino , Regulação da Expressão Gênica , Inativação Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas de Neoplasias/genética , RNA Interferente Pequeno/genética , Pele/metabolismo , Neoplasias Cutâneas/genética
9.
Clin Exp Dermatol ; 34(8): e798-801, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19778310

RESUMO

Dapsone has potent anti-inflammatory effects, and is used in the treatment of leprosy, cutaneous vasculitis, neutrophilic dermatoses, and dermatitis herpetiformis and other blistering disorders. However, it may cause severe adverse reactions such as hypersensitivity syndrome, which is characterized by fever, skin rash, hepatitis and lymphadenopathy. We report a 44-year-old female Korean patient with dapsone hypersensitivity syndrome (DHS) that presented as a bullous skin eruption. The patient had a 1-year history of urticarial vasculitis, treated with antihistamines, prednisolone and dapsone. Although the skin lesions improved, she reported fever, nausea, abdominal pain, jaundice, fatigue and skin rashes. On physical examination, there were generalized erythematous macules and purpura with facial oedema that developed into vesicles on the upper limbs. Histological examination of a skin biopsy of a vesicular lesion found subepidermal oedema with a mixed inflammatory cell infiltrate, including eosinophils in the dermis. Indirect immunofluorescence testing using normal foreskin as substrate revealed IgG deposits in the basement membrane zone. Circulating autoantibodies against antigens of 190 and 230 kDa were found by immunoblotting analysis using epidermal extracts. This case illustrates DHS with the formation of circulating autoantibodies.


Assuntos
Dapsona/efeitos adversos , Fármacos Dermatológicos/efeitos adversos , Toxidermias/imunologia , Hipersensibilidade/imunologia , Vasculite/tratamento farmacológico , Adulto , Autoanticorpos/imunologia , Feminino , Humanos , Síndrome
10.
Br J Dermatol ; 159(1): 61-7, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18489589

RESUMO

BACKGROUND: 5-Aminolaevulinic acid (ALA) and its esters act as precursors to the fluorescent photosensitizer protoporphyrin IX (PpIX) in photodynamic therapy (PDT). There is little information about how ALA and its esters induce PpIX synthesis and photodynamic effects in cell lines derived from the skin. OBJECTIVES: We compared the amount of PpIX synthesis induced by ALA and its esters in skin cell lines, and evaluated the relationship of PpIX synthesis to photodynamic effects by ALA and its esters in vitro. METHODS: Four cell lines, including human epidermal keratinocytes (HEK), human dermal fibroblast (hF), A431, and TXM13 were used. Cell survival was evaluated by the MTT assay. Fluorescence spectroscopy was used to measure the amount of PpIX synthesis induced by ALA and its esters. Flow cytometry measured cell death induced by ALA- and its esters-mediated PDT. RESULTS: ALA and its esters were not toxic at concentrations lower than 2 mmol L(-1) in all cell lines. PpIX synthesis was dose-dependent at low doses (0.01-0.1 mmol L(-1)), and ALA esters were more effective than ALA. Cell death occurred from necrosis rather than apoptosis just after light irradiation illumination on both ALA and its esters-treated cells. Cell death related more to PpIX synthesis than the irradiation light dose. CONCLUSIONS: PpIX production by ALA and its esters was induced on both normal and malignant cell lines derived from the skin, and cell death of PDT responses is closely related to the amount of PpIX synthesis rather than to the irradiation dose.


Assuntos
Ácido Aminolevulínico/farmacologia , Fotoquimioterapia , Fármacos Fotossensibilizantes/farmacologia , Protoporfirinas/biossíntese , Neoplasias Cutâneas/tratamento farmacológico , Pele/metabolismo , Ácido Aminolevulínico/análogos & derivados , Morte Celular/efeitos dos fármacos , Linhagem Celular , Relação Dose-Resposta a Droga , Ésteres/farmacologia , Citometria de Fluxo/métodos , Humanos , Microscopia de Fluorescência/métodos , Pele/efeitos dos fármacos , Células Tumorais Cultivadas
11.
Childs Nerv Syst ; 24(4): 525-8, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18097670

RESUMO

CASE REPORT: We report an infant who presented with clinical manifestations of incontinentia pigmenti (IP). Despite experiencing seizures in the early neonatal period, the patient had normal growth and development until recently. However, follow-up magnetic resonance imaging revealed sequential changes in white matter lesions. DISCUSSION: The pathogenesis of neurological involvement in IP has not been clearly elucidated and appears to be associated with various mechanisms, including developmental, destructive, and vascular processes. We have attempted to explain the pathogenesis of IP through these changes.


Assuntos
Lesões Encefálicas/patologia , Incontinência Pigmentar/complicações , Incontinência Pigmentar/patologia , Neuroglia/patologia , Lesões Encefálicas/complicações , Progressão da Doença , Feminino , Seguimentos , Humanos , Lactente , Imageamento por Ressonância Magnética/métodos
12.
Osteoarthritis Cartilage ; 16(1): 115-24, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17683952

RESUMO

OBJECTIVE: Proinflammatory cytokine-induced expression of matrix metalloproteinases (MMPs) is a major cause of arthritic cartilage destruction. The neuropeptide, alpha-melanocyte-stimulating hormone (alpha-MSH), has been detected in the synovial fluid of arthritis patients, where it is thought to play an anti-inflammatory role. Here, we examined whether alpha-MSH acts via downregulation of MMP expression, and sought to elucidate the intracellular signal pathways underlying this effect. DESIGN: Human chondrosarcoma cell line, HTB-94 (SW1353) was pretreated with or without alpha-MSH and then treated with tumor necrosis factor-alpha (TNF-alpha). The effect of alpha-MSH on TNF-alpha-induced MMP-13 expression and mitogen-activated protein kinases' (MAPKs) activation were determined by reverse transcriptase-polymerase chain reaction and Western blot analysis. Additionally, the intracellular signaling of alpha-MSH was investigated using the inhibitors of MAPK and nuclear factor kappaB (NF-kappaB) and plasmids encoding dominant negative (dn) forms of inhibitor kappaB kinase-alpha (IKKalpha) and inhibitor kappaB kinase-beta (IKKbeta). RESULTS: We found that alpha-MSH pretreatment inhibited TNF-alpha-induced MMP-13 expression and p38 kinase phosphorylation in HTB-94 human chondrosarcoma cells. TNF-alpha-induced MMP-13 expression was not suppressed by extracellular signal-regulated kinase (ERK) inhibitors (PD98059 and U0126) or a c-jun terminal kinase (JNK) inhibitor (SP600125), but was inhibited by inhibitors of p38 kinase (SB203580) and NF-kappaB (SN-50 peptide) and dnIKKalpha and dnIKKbeta. CONCLUSIONS: Our results suggest that alpha-MSH regulates TNF-alpha-induced MMP-13 expression by decreasing p38 kinase phosphorylation and subsequent NF-kappaB activation in human chondrocytes and may be an effective inhibitor of MMP-13-mediated collagen degradation, providing new potential opportunities for the development of anti-arthritis therapeutics.


Assuntos
Condrossarcoma/enzimologia , Hormônios/farmacologia , Metaloproteinase 13 da Matriz/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , alfa-MSH/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Western Blotting , Comunicação Celular , Linhagem Celular Tumoral , Condrossarcoma/imunologia , Feminino , Humanos , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Cell Death Differ ; 12(3): 201-12, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15665819

RESUMO

This study investigated the molecular mechanisms underlying inhibition of protein kinase C (PKC) zeta by p38 kinase during nitric oxide (NO)-induced apoptosis of chondrocytes. Coimmunoprecipitation experiments showed that activation of p38 kinase following addition of an NO donor resulted in a physical association between PKCzeta and p38 kinase. Direct interaction of p38 kinase with PKCzeta was confirmed in vitro using p38 kinase and PKCzeta recombinant proteins. p38 kinase interacts with the regulatory domain of PKCzeta and its association blocked PKCzeta autophosphorylation. Micro LC-MS/MS analysis using recombinant proteins indicated that the interaction of p38 kinase with PKCzeta blocked autophosphorylation of PKCzeta on Thr-560, which is required for PKCzeta activation. Collectively, our results demonstrate a novel mechanism of PKCzeta regulation: following activation by the production of NO, p38 kinase binds directly to the PKCzeta regulatory domain, preventing PKCzeta autophosphorylation on Thr-560, thereby inhibiting PKCzeta activation.


Assuntos
Apoptose , Condrócitos/fisiologia , Óxido Nítrico/fisiologia , Proteína Quinase C/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Animais , Cartilagem Articular/citologia , Células Cultivadas , Condrócitos/efeitos dos fármacos , Condrócitos/enzimologia , Cromatografia Líquida , Ativação Enzimática , Espectrometria de Massas , Doadores de Óxido Nítrico/farmacologia , Fosforilação , Ligação Proteica , Proteína Quinase C/química , Proteína Quinase C/metabolismo , Estrutura Terciária de Proteína , Coelhos , Proteínas Recombinantes/química , Serina/química , Transdução de Sinais , Treonina/química
14.
Biochem Biophys Res Commun ; 289(2): 507-12, 2001 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-11716502

RESUMO

Comparative proteome analysis was performed between human normal (BEAS 2B) and malignant (A549) lung epithelial cells in an attempt to identify novel biomarkers of lung cancer. Approximately 500 protein spots could be separated by mini two-dimensional electrophoresis and visualized with Coomassie blue R-250. Among those relatively abundant proteins, eight spots were changed more than twofold reproducibly and identified by peptide mass fingerprints using mass spectrometry and database search. The increased proteins in A549 were aldehyde dehydrogenase, peroxiredoxin I, fatty acid binding protein, aldoketoreductase, and destrin, whereas the decreased proteins were galectin-1, transgelin, and stathmin. Since human lung is exposed to continuous oxidative stress, antioxidant enzyme peroxiredoxin I was selected for further investigation and its augmented expression was confirmed in cancer tissues compared to normal tissues from lung cancer patients, suggesting peroxiredoxin I as a potential biomarker of lung cancer.


Assuntos
Neoplasias Pulmonares/metabolismo , Pulmão/metabolismo , Peroxidases/biossíntese , Sequência de Aminoácidos , Antioxidantes/farmacologia , Biomarcadores Tumorais/metabolismo , Western Blotting , Linhagem Celular , Eletroforese em Gel Bidimensional , Eletroforese em Gel de Poliacrilamida , Humanos , Immunoblotting , Espectrometria de Massas , Dados de Sequência Molecular , Peroxidases/química , Peroxirredoxinas , Biossíntese de Proteínas , Isoformas de Proteínas
15.
Eur J Pharmacol ; 427(3): 175-85, 2001 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-11567647

RESUMO

Immunosuppressants are now known to modulate bone metabolism, including bone formation and resorption. Because cartilage, formed by differentiated chondrocytes, serves as a template for endochondral bone formation, we examined the effects of the immunosuppressant rapamycin on the chondrogenesis of mesenchymal cells and on the cell signaling that is required for chondrogenesis, such as protein kinase C, extracellular signal-regulated kinase-1 (ERK-1), and p38 mitogen-activated protein (MAP) kinase pathways. Rapamycin inhibited the expression of type II collagen and the accumulation of sulfate glycosaminoglycan, indicating inhibition of the chondrogenesis of mesenchymal cells. Rapamycin treatment did not affect precartilage condensation, but it prevented cartilage nodule formation. Exposure of chondrifying mesenchymal cells to rapamycin blocked activation of the protein kinase C alpha and p38 MAP kinase, but had no discernible effect on ERK-1 signaling. Selective inhibition of PKCalpha or p38 MAP kinase activity, which is dramatically increased during chondrogenesis, with specific inhibitors in the absence of rapamycin blocked the chondrogenic differentiation of mesenchymal cells. Taken together, our data indicate that the immunosuppressant rapamycin inhibits the chondrogenesis of mesenchymal cells at the post-precartilage condensation stage by modulating signaling pathways including those of PKCalpha and p38 MAP kinase.


Assuntos
Condrogênese/efeitos dos fármacos , Imunossupressores/farmacologia , Isoenzimas/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteína Quinase C/antagonistas & inibidores , Sirolimo/farmacologia , Animais , Cartilagem/efeitos dos fármacos , Cartilagem/embriologia , Técnicas de Cultura de Células/métodos , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Embrião de Galinha , Ativação Enzimática/efeitos dos fármacos , Isoenzimas/metabolismo , Mesoderma/citologia , Mesoderma/efeitos dos fármacos , Mesoderma/enzimologia , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Proteína Quinase C/metabolismo , Proteína Quinase C-alfa , Proteínas Quinases S6 Ribossômicas/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno
16.
Biochem Biophys Res Commun ; 285(3): 825-9, 2001 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-11453667

RESUMO

Exposure of mammalian cells to ultraviolet (UV) light elicits a cellular response and also lead to apoptotic cell death. However, the role of Rac, a member of Rho family GTPases, in the UV-induced apoptosis has never been examined. In UV-irradiated Rat-2 fibroblasts, nuclear fragmentation began to be observed within 2 h and the total viability of Rat-2 cells were only about 15% at 6 h following by UV irradiation, whereas the total viability in Rat2-Rac(N17) cells stably expressing RacN17, a dominant negative Rac1 mutant, was almost close to 67%. Pretreatment with SB203580, a specific inhibitor of p38 kinase, likewise attenuated UV-induced cell death, but PD98059, a MEK inhibitor, did not. Thus, Rac1 and p38 kinase appear to be components in the apoptotic signaling pathway induced by UV irradiation in Rat-2 fibroblasts. In addition, our results show that p38 kinase stimulation by UV is dramatically inhibited by RacN17, suggesting that p38 kinase is situated downstream of Rac1 in the UV signaling to apoptosis.


Assuntos
Apoptose , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Raios Ultravioleta , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Bisbenzimidazol , Linhagem Celular , Sobrevivência Celular/efeitos da radiação , Inibidores Enzimáticos/farmacologia , Fibroblastos/citologia , Corantes Fluorescentes , Genes Dominantes , Imidazóis/farmacologia , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Mutação , Fosforilação/efeitos dos fármacos , Piridinas/farmacologia , Ratos , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno , Proteínas rac1 de Ligação ao GTP/genética
17.
J Immunol ; 167(3): 1663-71, 2001 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-11466390

RESUMO

Histamine, through H(2) receptors, triggers a prominent rise in intracellular free Ca(2+) concentration ([Ca(2+)](i)) in addition to an elevation of cAMP level in HL-60 promyelocytes. Here we show that the histamine-induced [Ca(2+)](i) rise was due to influx of Ca(2+) from the extracellular space, probably through nonselective cation channels, as incubation of the cells with SKF 96365 abolished the histamine-induced [Ca(2+)](i) rise, Na(+) influx, and membrane depolarization. The Ca(2+) influx was specifically inhibited by pretreatment of the cells with PMA or extracellular ATP with 50% inhibitory concentrations of 0.12 +/- 0.03 nM and 185 +/- 17 microM, respectively. Western blot analysis of protein kinase C (PKC) isoforms revealed that PMA (< or =1 nM) and ATP (300 microM) caused selective translocation of PKC-delta to the particulate/membrane fraction. Costimulation of the cells with histamine and SKF 96365 partially reduced histamine-induced granulocytic differentiation, which was evaluated by looking at the extent of fMet-Leu-Phe-induced [Ca(2+)](i) rise and superoxide generation. In conclusion, nonselective cation channels are opened by stimulation of the H(2) receptor, and the channels are at least in part involved in the induction of histamine-mediated differentiation processes. Both effects of histamine were selectively inhibited probably by the delta isoform of PKC in HL-60 cells.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/metabolismo , Granulócitos/metabolismo , Antagonistas dos Receptores H2 da Histamina/farmacologia , Proteína Quinase C/fisiologia , Receptores Histamínicos H2/fisiologia , Trifosfato de Adenosina/farmacologia , Trifosfato de Adenosina/fisiologia , Bloqueadores dos Canais de Cálcio/metabolismo , Canais de Cálcio/fisiologia , Cátions Bivalentes/metabolismo , Diferenciação Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/enzimologia , Granulócitos/efeitos dos fármacos , Granulócitos/enzimologia , Inibidores do Crescimento/fisiologia , Células HL-60 , Histamina/farmacologia , Humanos , Imidazóis/farmacologia , Isoenzimas/metabolismo , Isoenzimas/fisiologia , Proteína Quinase C/metabolismo , Proteína Quinase C-delta , Acetato de Tetradecanoilforbol/farmacologia
18.
J Biol Chem ; 276(27): 24645-53, 2001 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-11323430

RESUMO

Activation of Ras signaling by growth factors has been associated with gene regulation and cell proliferation. Here we characterize the contributory role of cytosolic phospholipase A(2) in the oncogenic Ha-Ras(V12) signaling pathway leading to activation of c-fos serum response element (SRE) and transformation in Rat-2 fibroblasts. Using a c-fos SRE-luciferase reporter gene, we showed that the transactivation of SRE by Ha-Ras(V12) is mainly via a Rac-linked cascade, although the Raf-mitogen-activated protein kinase cascade is required for full activation. In addition, Ha-Ras(V12)-induced DNA synthesis was significantly attenuated by microinjection of recombinant Rac(N17), a dominant negative mutant of Rac1. To identify the mediators downstream of Rac in the Ha-Ras(V12) signaling, we investigated the involvement of cytosolic phospholipase A(2). Oncogenic Ha-Ras(V12)-induced SRE activation was significantly inhibited by either pretreatment with mepacrine, a phospholipase A(2) inhibitor, or cotransfection with the antisense oligonucleotide of cytosolic phospholipase A(2). We also found cytosolic phospholipase A(2) to be situated downstream of Ha-Ras(V12) in a signal pathway leading to transformation. Together, these results are indicative of mediatory roles of Rac and cytosolic phospholipase A(2) in the signaling pathway by which Ha-Ras(V12) transactivates c-fos SRE and transformation. Our findings point to cytosolic phospholipase A(2) as a novel potential target for suppressing oncogenic Ha-Ras(V12) signaling in the cell.


Assuntos
Genes ras/fisiologia , Fosfolipases A/metabolismo , Transdução de Sinais , Proteínas ras/fisiologia , Animais , Sequência de Bases , Replicação do DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Genes Reporter , Luciferases/genética , Microinjeções , Dados de Sequência Molecular , Proteínas Nucleares/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosfolipases A2 , Quinacrina/farmacologia , Ratos , Fator de Resposta Sérica , Ativação Transcricional/efeitos dos fármacos , Transfecção , Proteínas rac1 de Ligação ao GTP/metabolismo
19.
J Biol Chem ; 276(16): 12871-8, 2001 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-11278500

RESUMO

In the yeast two-hybrid screening, we have isolated a cDNA clone from a human heart library using Nck Src homology 3 (SH3) domains as bait. The full-length cDNA, which encoded 722 amino acids, was identified as a VIP54-related gene containing an SH3 domain, proline-rich motifs, a serine/threonine-rich region, and a long C-terminal hydrophobic region. We refer to this protein as SPIN90 (SH3 Protein Interacting with Nck, 90 kDa). The amino acid sequence of the SH3 domain has the highest homology with those of Fyn, Yes, and c-Src. SPIN90 was broadly expressed in human tissues; in particular, it was highly expressed in heart, brain, and skeletal muscle, and its expression was developmentally regulated during cardiac myocyte differentiation. SPIN90 is able to bind to the first and third SH3 domains of Nck, in vitro, and is colocalized with Nck at sarcomere Z-discs within cardiac myocytes. Moreover, treatment with antisera raised against SPIN90 disrupted sarcomere structure, suggesting that this protein may play an important role in the maintenance of sarcomere structure and/or in the assembly of myofibrils into sarcomeres.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Bactérias , Regulação da Expressão Gênica no Desenvolvimento , Coração/fisiologia , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Miocárdio/citologia , Proteínas Oncogênicas/metabolismo , Adulto , Envelhecimento , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Sequência de Bases , Diferenciação Celular , Biblioteca Gênica , Humanos , Proteínas de Filamentos Intermediários/química , Dados de Sequência Molecular , Proteínas Musculares/química , Especificidade de Órgãos , RNA Mensageiro/genética , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Sarcômeros/fisiologia , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transcrição Gênica , Domínios de Homologia de src
20.
J Bone Miner Res ; 15(11): 2197-205, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11092400

RESUMO

Chondrogenesis of mesenchymal cells during in vitro micromass culture requires the generation of cyclic adenosine monophosphate (cAMP) and subsequent activation of cAMP-dependent protein kinase A (PKA). In this study, we investigated the regulatory activity of PKA during chondrogenesis of chick limb bud mesenchymal cells. PKA activity was high in 1-day and 2-day cultures, which was followed by a slight decrease in 4-day and 5-day old cultures. Inhibition of PKA blocked chondrogenesis. It did not affect precartilage condensation, but it blocked the progression from the precartilage condensation stage to cartilage nodule formation. The PKA inhibition-induced blockage of chondrogenesis was accompanied by an altered expression of N-cadherin. Although expression of N-cadherin was detected during the early period of chondrogenesis, it became reduced as chondrogenesis proceeded. Still, inhibition of PKA maintained expression of N-cadherin throughout the micromass culture period. The inhibition of PKA did not affect expression of protein kinase C-alpha (PKCalpha), PKCepsilon, PKCdelta, and PKClambda/iota, which are the isoforms expressed in differentiating mesenchymal cells. However, PKA inhibition completely blocked activation of PKCalpha. Because PKC activity regulates N-cadherin expression and chondrogenesis, the PKA-mediated regulation of PKCalpha appears to be responsible for the PKA regulation of N-cadherin expression and chondrogenesis. Taken together, our results suggest that PKA regulates chondrogenesis by activating PKCalpha at the stage of post-precartilage condensation.


Assuntos
Carbazóis , Cartilagem/citologia , Cartilagem/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Isoenzimas/metabolismo , Mesoderma/metabolismo , Proteína Quinase C/metabolismo , Sulfonamidas , Animais , Caderinas/metabolismo , Cartilagem/efeitos dos fármacos , Diferenciação Celular , Embrião de Galinha , Condrogênese/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Indução Embrionária , Inibidores Enzimáticos/farmacologia , Fibronectinas/metabolismo , Flavonoides/farmacologia , Indóis/farmacologia , Isoenzimas/efeitos dos fármacos , Isoquinolinas/farmacologia , Mesoderma/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteína Quinase C-alfa , Pirróis/farmacologia , Receptores de Fibronectina/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA