Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Biol Macromol ; 259(Pt 2): 129149, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38176486

RESUMO

Lysine crotonylation (Kcr), a newly discovered post-translational modification, played a crucial role in physiology and disease progression. However, the roles of crotonylation in oocyte meiotic resumption remain elusive. As abnormal cumulus cell development will cause oocyte maturation arrest and female infertility, we report that cumulus cells surrounding human meiotic arrested oocytes showed significantly lower crotonylation, which was associated with decreased EP300 expression and blocked cumulus cell expansion. In cultured human cumulus cells, exogenous crotonylation or EP300 activator promoted cell proliferation and reduced cell apoptosis, whereas EP300 knockdown induced the opposite effect. Transcriptome profiling analysis in human cumulus cells indicated that functions of crotonylation were associated with activation of epidermal growth factor receptor (EGFR) pathway. Importantly, we characterized the Kcr proteomics landscape in cumulus cells by LC-MS/MS analysis, and identified that annexin A2 (ANXA2) was crotonylated in cumulus cells in an EP300-dependent manner. Crotonylation of ANXA2 enhanced the ANXA2-EGFR binding, and then activated the EGFR pathway to affect cumulus cell proliferation and apoptosis. Using mouse oocytes IVM model and EP300 knockout mice, we further confirmed that crotonylation alteration in cumulus cells affected the oocyte maturation. Together, our results indicated that EP300-mediated crotonylation is important for cumulus cells functions and oocyte maturation.


Assuntos
Anexina A2 , Células do Cúmulo , Animais , Camundongos , Feminino , Humanos , Células do Cúmulo/metabolismo , Anexina A2/metabolismo , Anexina A2/farmacologia , Cromatografia Líquida , Espectrometria de Massas em Tandem , Oócitos , Receptores ErbB/metabolismo , Proteína p300 Associada a E1A/metabolismo
2.
Acta Biochim Biophys Sin (Shanghai) ; 55(3): 356-366, 2023 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-36916296

RESUMO

Neuroblastoma (NB) is a pediatric cancer of the peripheral sympathetic nervous system and represents the most frequent solid malignancy in infants. Nectin2 belongs to the immunoglobulin superfamily and has been shown to play a role in tumorigenesis. In the current study, we demonstrate that serum Nectin2 level is increased in NB patients compared with that in healthy controls and Nectin2 level is correlated with neuroblastoma international neuroblastoma staging system (INSS) classification. There is a positive correlation between Nectin2 level and shorter overall survival in NB patients. Knockdown of Nectin2 reduces the migration of SH-SY5Y and SK-N-BE2 cells and induces their apoptosis and cell cycle arrest. RNA-seq analysis demonstrates that Nectin2 knockdown affects the expressions of 258 genes, including 240 that are upregulated and 18 that are downregulated compared with negative controls. qRT-PCR and western blot analysis confirm that ANXA2 expression is decreased in Nectin2-knockdown SH-SY5Y cells, consistent with the RNA-seq results. ANXA2 overexpression rescues the percentage of apoptotic NB cells induced by Nectin2 knockdown and compensates for the impact of Nectin2 knockdown on cleaved caspase3 and bax expressions. In addition, western blot analysis results show that ANXA2 overexpression rescues the effect of Nectin2 knockdown on MMP2 and MMP9 expressions. The current data highlight the importance of Nectin2 in NB progression and the potential of Nectin2 as a novel candidate target for gene therapy.


Assuntos
Anexina A2 , Neuroblastoma , Criança , Humanos , Lactente , Anexina A2/genética , Anexina A2/metabolismo , Anexina A2/farmacologia , Apoptose/genética , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patologia
3.
Curr Eye Res ; 47(4): 579-589, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34894941

RESUMO

PURPOSE: Retinal Neovascularization (RNV) is a pathological characteristic of ocular diseases. Annexin A2 (ANXA2) plays important roles in RNV while the mechanism remains unclear. The study aimed to explore relationship between ANXA2 and PI3K/AKT signaling pathway in RNV. METHODS: We used human retinal vascular endothelial cells (HRECs) and oxygen-induced retinopathy (OIR) mice model to show ANXA2 can promote the development of RNV through PI3K/AKT signaling pathway. We divided HRECs into six groups by infecting lentivirus containing appropriate plasmid and adding corresponding solution. Assays showing ability of HRECs were performed in vitro. Mice were randomly divided into three groups and treated accordingly. RESULTS: Expression of ANXA2 and activity of PI3K/AKT signaling pathway in HRECs were detected. RNV and expression of ANXA2 in mice retinas were detected. Results showed that ANXA2 expression is positively related with RNV-forming ability of HRECs in vitro and development of RNV in vivo while low activity of PI3K/AKT signaling pathway could attenuate the role of ANXA2. CONCLUSIONS: We can make ANXA2 and PI3K/ AKT signaling pathway as a promising target for the regulation of pathological neovascularization of the retina, which also provides a novel idea for effective prevention and treatment of diseases related to RNV in future.


Assuntos
Anexina A2 , Neovascularização Retiniana , Animais , Anexina A2/metabolismo , Anexina A2/farmacologia , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Camundongos , Oxigênio/metabolismo , Oxigênio/toxicidade , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Neovascularização Retiniana/metabolismo , Transdução de Sinais
4.
PLoS One ; 8(3): e60281, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23555942

RESUMO

Annexin A2 (AnxA2) is a widely expressed multifunctional protein found in different cellular compartments. In spite of lacking a hydrophobic signal peptide, AnxA2 is found at the cell surface of endothelial cells, indicative of a role in angiogenesis. Increased extracellular levels of AnxA2 in tumours correlate with neoangiogenesis, metastasis and poor prognosis. We hypothesised that extracellular AnxA2 may contribute to angiogenesis by affecting endothelial cell-cell interactions and motility. To address this question, we studied the effect of heterotetrameric and monomeric forms of AnxA2, as well as its two soluble domains on the formation and maintenance of capillary-like structures by using an in vitro co-culture system consisting of endothelial and smooth muscle cells. In particular, addition of purified domains I and IV of AnxA2 potently inhibited the vascular endothelial growth factor (VEGF)-dependent formation of the capillary-like networks in a dose-dependent manner. In addition, these AnxA2 domains disrupted endothelial cell-cell contacts in preformed capillary-like networks, resulting in the internalisation of vascular endothelial (VE)-cadherin and the formation of VE-cadherin-containing filopodia-like structures between the endothelial cells, suggesting increased cell motility. Addition of monoclonal AnxA2 antibodies, in particular against Tyr23 phosphorylated AnxA2, also strongly inhibited network formation in the co-culture system. These results suggest that extracellular AnxA2, most likely in its Tyr phosphorylated form, plays a pivotal role in angiogenesis. The exogenously added AnxA2 domains most likely mediate their effects by competing with endogenous AnxA2 for extracellular factors necessary for the initiation and maintenance of angiogenesis, such as those involved in the formation/integrity of cell-cell contacts.


Assuntos
Anexina A2/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Animais , Anexina A2/farmacologia , Bovinos , Células Cultivadas , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Microscopia Confocal , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Proteínas S100/metabolismo
5.
Biol Chem ; 393(10): 1151-63, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23091278

RESUMO

Annexin A2 (AnxA2), a 38-kDa member of the Ca2+-binding annexin family, has been implicated in numerous cancer pathways. Withaferin A (WithfA), a natural plant compound, has been reported previously to bind covalently to Cys133 of the AnxA2 core domain leading to a reduction of the invasive capabilities of cancer cells by altering their cytoskeleton. We show here that AnxA2 has an inhibitory effect on actin polymerization, and a modification with WithfA significantly increases this inhibitory role of AnxA2. Using mass spectrometry and single-site mutants, we localized the WithfA-AnxA2 interaction to the N-terminal domain of AnxA2 where WithfA binds covalently to Cys9. Whereas binding to F-actin filaments has been mapped to the C terminus of AnxA2, our results suggest that the N-terminal domain modified by WithfA may also play a role in the AnxA2-actin interaction. The binding of WithfA may regulate the AnxA2-mediated actin dynamics in two distinct ways: (i) the increase of F-actin bundling activity by the Anx2/p11 heterotetramer and (ii) the decrease of actin polymerization as a result of the increased affinity of AnxA2 to the barbed end of actin microfilaments. We demonstrate the susceptibility of Cys9 of AnxA2 to chemical modifications and exclude Cys133 as a binding site for WithfA.


Assuntos
Anexina A2/química , Anexina A2/metabolismo , Vitanolídeos/metabolismo , Actinas/química , Animais , Anexina A2/farmacologia , Sítios de Ligação , Bovinos , Cisteína/metabolismo , Humanos , Modelos Moleculares , Ligação Proteica , Multimerização Proteica/efeitos dos fármacos , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Solventes/química
6.
Pancreas ; 41(8): 1247-54, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22750966

RESUMO

OBJECTIVES: Extracellular microenvironment plays crucial roles in the development of cancers and chemoresistance. Pancreatic carcinoma is resistant to almost all chemotherapeutic agents. In this study, we identified annexin II in the medium from pancreatic cancer cells as a protein released into the extracellular environment. METHODS: Medium from 5-hour cultures of various cancer cells was collected. Proteins in the medium were detected by molecular mass analysis and immunoblotting. Anticancer drug sensitivity of cells preincubated with or without recombinant annexin II (rANX II) was measured using crystal violet assay and colony survival assay. Apoptosis-related molecules were analyzed by immunoblotting. RESULTS: Recombinant ANX II supplementation in the medium confers resistance to anticancer drugs, including cisplatin, 5-fluorouracil, and gemcitabine, in MiaPaCa-2 and AsPC-1 cells. In MiaPaCa-2 cells, rANX II supplementation resulted in suppression of caspase-3 activation associated with increased Bcl-2/Bax ratios. Suppression of cisplatin-induced cell death by rANX II supplementation was canceled by inhibitors of phosphatidylinositol 3-kinase and mitogen-activated protein kinase kinase signal pathways. CONCLUSIONS: The current study is the first report to demonstrate that supplementation of rANX II in the medium increased resistance to anticancer drugs in pancreatic cancer cells. Recombinant ANX II exerts cell death-suppressive function by antagonizing cisplatin-induced apoptosis.


Assuntos
Anexina A2/farmacologia , Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias Pancreáticas/tratamento farmacológico , Anexina A2/metabolismo , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Linhagem Celular Tumoral , Inibidores Enzimáticos/farmacologia , Humanos , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Neoplasias Pancreáticas/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Recombinantes/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Proteína X Associada a bcl-2/metabolismo , Neoplasias Pancreáticas
7.
Blood ; 120(1): 207-13, 2012 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-22517898

RESUMO

Increased fibrinolysis is an important component of acute promyelocytic leukemia (APL) bleeding diathesis. APL blasts overexpress annexin II (ANXII), a receptor for tissue plasminogen activator (tPA), and plasminogen, thereby increasing plasmin generation. Previous studies suggested that ANXII plays a pivotal role in APL coagulopathy. ANXII binding to tPA can be inhibited by homocysteine and hyperhomocysteinemia can be induced by L-methionine supplementation. In the present study, we used an APL mouse model to study ANXII function and the effects of hyperhomocysteinemia in vivo. Leukemic cells expressed higher ANXII and tPA plasma levels (11.95 ng/mL in leukemic vs 10.74 ng/mL in wild-type; P = .004). In leukemic mice, administration of L-methionine significantly increased homocysteine levels (49.0 µmol/mL and < 6.0 µmol/mL in the treated and nontreated groups, respectively) and reduced tPA levels to baseline concentrations. The latter were also decreased after infusion of the LCKLSL peptide, a competitor for the ANXII tPA-binding site (11.07 ng/mL; P = .001). We also expressed and purified the p36 component of ANXII in Pichia methanolica. The infusion of p36 in wild-type mice increased tPA and thrombin-antithrombin levels, and the latter was reversed by L-methionine administration. The results of the present study demonstrate the relevance of ANXII in vivo and suggest that methionine-induced hyperhomocysteinemia may reverse hyperfibrinolysis in APL.


Assuntos
Anexina A2/metabolismo , Fibrinólise/fisiologia , Hiper-Homocisteinemia/induzido quimicamente , Leucemia Promielocítica Aguda , Metionina/farmacologia , Animais , Anexina A2/farmacologia , Coagulação Sanguínea/fisiologia , Transplante de Medula Óssea , Modelos Animais de Doenças , Fibrinolisina/metabolismo , Homocisteína/sangue , Leucemia Promielocítica Aguda/complicações , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Ativador de Plasminogênio Tecidual/sangue
8.
Blood ; 119(8): 1888-96, 2012 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-22223826

RESUMO

Multiple myeloma (MM) is an incurable B-cell malignancy in which the marrow microenvironment plays a critical role in our inability to cure MM. Marrow stromal cells in the microenvironment support homing, lodging, and growth of MM cells through activation of multiple signaling pathways in both MM and stromal cells. Recently, we identified annexin II (AXII) as a previously unknown factor produced by stromal cells and osteoclasts (OCL) that is involved in OCL formation, HSC and prostate cancer (PCa) homing to the BM as well as mobilization of HSC and PCa cells. AXII expressed on stromal cells supports PCa cell lodgment via the AXII receptor (AXIIR) on PCa cells, but the role of AXII and AXIIR in MM is unknown. In this study, we show that MM cells express AXIIR, that stromal/osteoblast-derived AXII facilitates adhesion of MM cells to stromal cells via AXIIR, and OCL-derived AXII enhances MM cell growth. Finally, we demonstrate that AXII activates the ERK1/2 and AKT pathways in MM cells to enhance MM cell growth. These results demonstrate that AXII and AXIIR play important roles in MM and that targeting the AXII/AXIIR axis may be a novel therapeutic approach for MM.


Assuntos
Anexina A2/metabolismo , Medula Óssea/metabolismo , Proliferação de Células , Mieloma Múltiplo/metabolismo , Receptores de Peptídeos/metabolismo , Animais , Anexina A2/genética , Anexina A2/farmacologia , Western Blotting , Células da Medula Óssea/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Células Cultivadas , Microambiente Celular , Técnicas de Cocultura , Humanos , Camundongos , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Osteoclastos/metabolismo , Fosforilação/efeitos dos fármacos , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Receptores de Peptídeos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
9.
Radiat Res ; 176(6): 732-42, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22141411

RESUMO

In this study, we found that refractoriness to ultraviolet (UVC) light-induced cell death was increased in UVC-radiation-sensitive cells derived from Cockayne syndrome patients when the cells were precultured in medium supplemented with recombinant annexin II (rANX II). In CS3BES cells, an immortal cell line derived from Cockayne syndrome patients, the rANX II supplementation-induced UVC-radiation resistance was suppressed by treatment with an anti-annexin II antibody and EGTA. The amount of biotinylated annexin II on the cell surface increased in the rANX II-supplemented cells but did not increase in the cells that were cotreated with rANX II and EGTA. The capacity to remove UVC-radiation-damaged DNA, (6-4) photoproducts and cyclobutane pyrimidine dimers, was the same in cells that were precultured with rANX II and in control cells that did not receive rANX II supplementation. The rANX II supplementation-induced UVC-radiation resistance was also observed in nucleotide excision repair-deficient cells and xeroderma pigmentosum group A-downregulated cells. The Bcl-xL to Bax protein ratios, an index of survival activity in cells exposed to lethal stresses, were increased in the cells that had been precultured in rANX II for 24 h prior to UVC irradiation. Treatment with a phosphatidylinositol 3-kinase inhibitor suppressed the increased UVC-radiation resistance and Bcl-xL to Bax ratios in the cells with rANX II supplementation. Furthermore, downregulation of Bcl-xL by siRNA transfection also suppressed the UVC-radiation resistance that was induced by rANX II supplementation. These results suggest that the increase in the Bcl-xL to Bax ratios may be associated with enhanced resistance to UVC-radiation-induced cell death.


Assuntos
Anexina A2/farmacologia , Espaço Extracelular , Tolerância a Radiação/efeitos dos fármacos , Proteínas Recombinantes/farmacologia , Raios Ultravioleta , Proteína X Associada a bcl-2/metabolismo , Proteína bcl-X/metabolismo , Técnicas de Cultura de Células , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Síndrome de Cockayne/patologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/efeitos da radiação , Inibidores Enzimáticos/farmacologia , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/efeitos da radiação , Células HeLa , Humanos , Inibidores de Fosfoinositídeo-3 Quinase , Tolerância a Radiação/efeitos da radiação
10.
Cancer Lett ; 296(2): 160-7, 2010 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-20435405

RESUMO

beta(2)-Glycoprotein-I (beta(2)gpI), an abundant plasma glycoprotein, functions as a regulator of thrombosis. Previously, we demonstrated that plasmin-clipped beta(2)gpI (cbeta(2)gpI) exerts an anti-angiogenic effect on human umbilical vein endothelial cells (HUVEC). The present study was focused on the molecular background responsible for this phenomenon. cbeta(2)gpI strongly reduced HUVEC growth and proliferation as evidenced by the MTT and BrdU assay and delayed cell cycle progression arresting HUVEC in the S-and G2/M-phase. Western blot analysis indicated that cbeta(2)gpI inhibited cyclin A, B and D1, and enhanced p21 and p27 expression. Activity of p38 was down-regulated independently from the cbeta(2)gpI incubation time. Phosphorylation of ERK1/2 was not changed early (30 and 60 min) but became enhanced later (90 min, 4h). JNK activity was reduced rapidly after cbeta(2)gpI treatment but compared to controls, increased thereafter. Annexin II blockade prevented growth inhibition and cell cycle delay evoked by cbeta(2)gpI. We assume that cbeta(2)gpI's effects on HUVEC growth is mediated via cyclin A, B and D1 suppression, up-regulation of p21 and p27 and coupled to modifications of the mitogen-activated protein (MAP) kinase signalling pathway. cbeta(2)gpI may represent a potential endogenous angiogenesis-targeted compound, opening the possibility of a novel tool to treat cancer.


Assuntos
Ciclina A/genética , Ciclina B/genética , Ciclina D1/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Endotélio Vascular/fisiologia , Antígeno Nuclear de Célula em Proliferação/genética , beta 2-Glicoproteína I/metabolismo , Anexina A2/farmacologia , Ciclo Celular , Divisão Celular/fisiologia , Regulação para Baixo , Fibrinolisina/fisiologia , Humanos , Neovascularização Fisiológica/fisiologia , Veias Umbilicais/citologia , Veias Umbilicais/fisiologia , Regulação para Cima , beta 2-Glicoproteína I/isolamento & purificação , beta 2-Glicoproteína I/fisiologia
11.
J Leukoc Biol ; 82(5): 1174-84, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17715360

RESUMO

On the surface of the macrophage, annexin A2 tetramer (A2t) serves as a docking protein or recognition element for bacterial and viral pathogens. Plasma levels of free A2t have been reported to increase following infection, although the mechanistic significance of this observation is unclear. Although annexin A2 had generally been thought to play an anti-inflammatory role, soluble A2t stimulates MAP kinase activity in bone marrow stromal cells downstream of a recently cloned receptor. This raises the question of whether A2t activates human macrophages via MAP kinases and whether it might be capable of acting as an inflammatory mediator. To this end, human monocyte-derived macrophages were treated with soluble A2t and MAP kinase phosphorylation, p65 NF-kappaB activation, and inflammatory mRNA and protein levels were measured. It was found that A2t caused rapid phosphorylation of several MAP kinases, as well as translocation of p65 NF-kappaB to the nucleus. A2t stimulated the production of TNF-alpha, IL-1beta, and IL-6, as well as several members of the chemokine family within 24 h, which are capable of recruitment and/or activation of a broad range of leukocyte classes. Furthermore, A2t-activated macrophages demonstrated enhanced phagocytic ability for the ingestion of GFP-expressing Escherichia coli. These data are the first to suggest the participation of an annexin in microbial clearance, as well as the establishment of inflammation and the immune response, including the recruitment and activation of immune cells to the site of infection.


Assuntos
Anexina A2/farmacologia , Ativação de Macrófagos/imunologia , Macrófagos/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Monócitos/metabolismo , Western Blotting , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Macrófagos/citologia , Macrófagos/imunologia , Monócitos/citologia , Monócitos/imunologia , NF-kappa B/genética , NF-kappa B/metabolismo , Fagocitose , Fosforilação , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de IgG/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
12.
J Bone Miner Res ; 20(7): 1161-7, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15940368

RESUMO

UNLABELLED: We report that AX-II, in addition to inducing GM-CSF expression, also increases membrane-bound RANKL synthesis by marrow stromal cells and does so through a previously unreported MAPK-dependent pathway. Thus, both GM-CSF and RANKL are required for AX-II stimulation of OCL formation. INTRODUCTION: Annexin II (AX-II) is an autocrine/paracrine factor secreted by osteoclasts (OCLs) that stimulates human OCL formation and bone resorption in vitro by inducing bone marrow stromal cells and activated CD4+ T cells to produce granulocyte-macrophage colony-stimulating factor (GM-CSF). GM-CSF in turn increases OCL precursor proliferation and further enhances OCL formation. However, the induction of GM-CSF by AX-II cannot fully explain its effects on OCL formation. In this study, we tested the capacity of AX-II to induce the expression of RANKL and the corresponding signaling pathways AX-II employs in human marrow stromal cells to induce RANKL. We also showed that both GM-CSF and RANKL are required for OCL formation induced by AX-II. MATERIALS AND METHODS: Real-time RT-PCR and Western blot analysis were used to detect RANKL and osteoprotegerin (OPG) mRNA and protein expression in unfractionated human bone marrow mononuclear cells stimulated with AX-II. Soluble RANKL in the conditioned medium was analyzed by ELISA. Activation of the MAPK pathway by AX-II was tested by Western blot. The effects of OPG and anti-GM-CSF on AX-II-induced OCL formation were also examined. RESULTS AND CONCLUSION: In addition to upregulating GM-CSF mRNA, AX-II increased RANKL mRNA expression dose-dependently in unfractionated human bone marrow mononuclear cells and modestly increased soluble RANKL in unfractionated human bone marrow mononuclear cell conditioned medium. However, AX-II markedly increased membrane-bound RANKL on human bone marrow stromal cells. Treatment of marrow stromal cells with AX-II activated MAP-kinase (ERKs) and PD 98059 abolished the effect but did not block the increase in GM-CSF. Interestingly, OPG, a natural decoy receptor for RANKL, or anti-GM-CSF partially inhibited OCL formation by AX-II in human bone marrow cells, and the combination of OPG and anti-GM-CSF completely blocked AX-II-induced OCL formation. These data show that AX-II stimulates both the proliferation and differentiation of OCL precursors through production of GM-CSF and RANKL respectively.


Assuntos
Anexina A2/fisiologia , Células da Medula Óssea/metabolismo , Proteínas de Transporte/biossíntese , Glicoproteínas de Membrana/biossíntese , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Osteoclastos/fisiologia , Anexina A2/farmacologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/enzimologia , Proteínas de Transporte/genética , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Flavonoides/farmacologia , Glicoproteínas/farmacologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Glicoproteínas de Membrana/genética , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Osteoprotegerina , Inibidores de Proteínas Quinases/farmacologia , Ligante RANK , Receptor Ativador de Fator Nuclear kappa-B , Receptores Citoplasmáticos e Nucleares , Receptores do Fator de Necrose Tumoral , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Regulação para Cima
13.
J Biol Chem ; 280(30): 28103-9, 2005 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-15919660

RESUMO

Caveolin-1 (CAV1) is the structural protein of the filamentous coat that decorates the cytoplasmic surface of each caveola. Cell culture studies have implicated CAV1 in playing an important role in intracellular cholesterol trafficking. In addition, it has been reported that CAV1 forms a detergent-resistant protein complex with Annexin-2 in enterocytes that can be disrupted by the cholesterol absorption inhibitor ezetimibe, suggesting a possible role for CAV1 in cholesterol absorption. In this report, we have evaluated cholesterol homeostasis in Cav1 knock-out mice. Deletion of CAV1 does not result in either a compensatory increase of CAV2 or CAV3 in intestine. In addition, Cav1 knock-out mice display normal mRNA and protein levels of Annexin-2 or the putative cholesterol transport protein Niemann-Pick C1-like 1 (NPC1L1) in proximal intestinal mucosa. Fractional cholesterol absorption and fecal neutral sterol excretion are statistically similar in Cav1 knock-out mice and their wild-type littermates. Moreover, oral administration of ezetimibe is equally effective in decreasing cholesterol absorption in Cav1 null mice and wild-type controls. The mRNA expression levels of genes sensitive to intracellular cholesterol concentration (ATP-binding cassette transporters ABCA1 and ABCG5, hydroxymethylglutaryl-CoA synthase and the LDL receptor) are similarly altered in the proximal intestinal mucosa of Cav1 null and wild-type mice following ezetimibe treatment. These results demonstrate that CAV1 is not required for cholesterol absorption or ezetimibe sensitivity in the mouse.


Assuntos
Caveolinas/fisiologia , Colesterol/metabolismo , Transportador 1 de Cassete de Ligação de ATP , Membro 5 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Trifosfato de Adenosina/química , Animais , Anexina A2/farmacologia , Anticolesterolemiantes/farmacologia , Azetidinas/farmacologia , Transporte Biológico , Northern Blotting , Western Blotting , Caveolina 1 , Caveolina 2 , Caveolina 3 , Caveolinas/metabolismo , Detergentes/farmacologia , Duodeno/metabolismo , Enterócitos/metabolismo , Ezetimiba , Deleção de Genes , Genótipo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Metabolismo dos Lipídeos , Lipoproteínas/metabolismo , Fígado/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA/metabolismo , RNA Mensageiro/metabolismo , Esteróis/metabolismo
14.
Scand J Immunol ; 48(5): 522-6, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9822262

RESUMO

Annexin II (AII) belongs to a family of glycoproteins that bind negatively charged phospholipids in the presence of calcium. The annexins exert various biological functions. We have previously shown that soluble AII suppresses mitogen-induced lymphoproliferation in vitro. In this study we address the question of whether soluble AII may also affect immunoglobulin secretion. Mononuclear cells from peripheral blood were stimulated with pokeweed mitogen in vitro and immunoglobulin-secreting cells were quantified using an ELISPOT assay. Retroplacental serum and soluble AII significantly inhibited secretion of IgG and IgM when added at concentrations that did not affect lymphoproliferation or cell viability. The inhibitory effect was dose- and time dependent. Significant suppression was observed when soluble AII was added at concentrations of 0.1 and 0.01 microg/ml. The strongest inhibition was observed when soluble AII or retroplacental serum was added initially. The data demonstrate that soluble AII can suppress immunoglobulin secretion in vitro. AII seems to be a potent immunosuppressive substance. The presence of high levels of soluble AII in retroplacental serum may indicate a possible immunomodulatory role in normal pregnancy.


Assuntos
Anexina A2/farmacologia , Imunoglobulina G/metabolismo , Imunoglobulina M/metabolismo , Linfócitos/efeitos dos fármacos , Animais , Divisão Celular , Feminino , Humanos , Linfócitos/citologia , Linfócitos/metabolismo , Gravidez , Solubilidade
15.
APMIS ; 105(9): 699-704, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9350213

RESUMO

Annexin II (AII) is a member of a family of glycoproteins which bind negatively charged phospholipids in a calcium-dependent manner. Annexins are membrane-associated proteins, expressed both in normal and malignant cells, but have also been detected as soluble molecules in serum and other body fluids. Because of their adhesive properties, it has been suggested that annexins play a role in the metastatic process. An ELISA was established for quantification of soluble AII. Within-run variation was 5.2-10.4% and run-to-run variation 12.4-15.6%. Soluble AII was detected in all sera studies. A strongly positive serum was arbitrarily given the value 100 AII units and used as reference serum. The mean level in sera from 20 normal blood donors was 49 (SE 5.6) AII units. Sera from peripheral blood of five patients with renal cell carcinoma and sera from blood obtained from the renal vein of the same patients contained 47 (SE 20) and 83 (SE 28) AII units, respectively. In two patients, AII levels were increased in renal vein serum as compared with peripheral blood serum. Interestingly, in both cases, and in none of the three remaining cases, phytohaemagglutinin-stimulated lymphoproliferation was suppressed by renal vein serum as compared with peripheral blood serum. Affinity absorption of AII from the renal vein sera with increased AII levels strongly reduced their immunosuppressive activity. Addition of affinity-purified AII to cell cultures suppressed lymphoproliferation. These data show that the level of AII is markedly increased in renal vein sera from some patients with renal cell carcinoma, suggesting that AII may be locally released in vivo. The study also demonstrates an immunosuppressive effect of soluble AII in vitro. We speculate that soluble AII released by the tumour has immunosuppressive properties. This study identifies soluble AII as a novel immunosuppressive factor in sera from patients with renal cell carcinoma. A further study including a larger number of patients is currently in progress, in order to investigate the pathological significance of this finding.


Assuntos
Anexina A2/farmacologia , Carcinoma de Células Renais/imunologia , Imunossupressores/sangue , Neoplasias Renais/imunologia , Ativação Linfocitária/efeitos dos fármacos , Fito-Hemaglutininas/farmacologia , Anexina A2/sangue , Anexina A2/química , Humanos , Tolerância Imunológica
16.
Am J Reprod Immunol ; 38(5): 313-9, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9352022

RESUMO

PROBLEM: Syncytiotrophoblast microvillous plasma membranes (StMPM) are potent suppressors of lymphoproliferation in vitro. We have previously shown that soluble annexin II (AII) is present at higher levels in retroplacental serum (RPS) than in peripheral serum, and that soluble AII has an immunosuppressive effect. The aims of this study were to determine whether AII can be released from StMPM and whether soluble AII from StMPM exerts any immunosuppressive effect. METHOD OF STUDY: Isolated StMPM were incubated in growth medium for 18 hr and supernatants were prepared by ultracentrifugation. Soluble AII was detected by immunoblotting. StMPM, StMPM supernatant, and affinity-purified AII were analysed in a lymphoproliferation assay for immunomodulating activity. RESULTS: AII heavy chain and its p11 light chain were detected both in StMPM supernatant and in RPS after removal of StMPM particles by ultracentrifugation. StMPM, StMPM supernatant, and purified AII suppressed lymphoproliferation in a dose-dependent manner. Absorption of AII from StMPM supernatant reduced the suppressive activity. The suppressive effect of StMPM supernatant and purified AII was completely reversed by heating at 100 degrees C for 30 min or by adding recombinant interleukin-2 at 100 units/ml. Although StMPM and affinity-purified AII suppressed the proliferation of lymphocytes from all donors tested, StMPM supernatant suppressed the proliferation of lymphocytes from 12 of 23 donors. Six of eight female non-suppressed donors were multiparae, whereas five of five female suppressed donors were nulliparae. CONCLUSIONS: Annexin II is released by isolated placental membranes in vitro and is present in RPS, indicating in vivo release of AII at the fetomaternal interface, probably as AII heterotetramer. AII has immunosuppressive activity and may be important in fetal allograft survival.


Assuntos
Anexina A2/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Placenta/imunologia , Anexina A2/isolamento & purificação , Anexina A2/metabolismo , Feminino , Humanos , Tolerância Imunológica , Técnicas In Vitro , Masculino , Troca Materno-Fetal/imunologia , Microvilosidades/imunologia , Microvilosidades/metabolismo , Placenta/metabolismo , Gravidez , Solubilidade , Fatores Supressores Imunológicos/metabolismo , Fatores Supressores Imunológicos/farmacologia
17.
Pflugers Arch ; 434(3): 261-6, 1997 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9178624

RESUMO

The effect of annexins II, III and V, purified from different species, on the calcium-activated chloride current across the stage-V to stage-VI Xenopus laevis oocyte membrane was tested either directly, using calcium entry mediated by depolarization, by A23187 permeabilization of oocytes or indirectly by quisqualate stimulation of a metabotropic glutamate receptor in the membrane expressed by the oocyte after injection of mRNA. The annexins isolated from the Ehrlich ascites cell, which is a mouse tumor cell, were found to be potent inhibitors of the chloride current, showing half-maximal inhibition at 50 nM, whereas no block was found using bovine or porcine annexins isolated from lung tissue. Of the annexins tested, we found annexin III to be naturally occurring in the oocyte, while only trace amounts of annexins II and V could be demonstrated. The inhibition pattern varied somewhat according to the stimulus method, the inhibition being more complete when an indirect stimulus via the metabotropic receptor was applied compared to a direct calcium stimulus.


Assuntos
Anexinas/farmacologia , Canais de Cloreto/antagonistas & inibidores , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Animais , Anexina A2/isolamento & purificação , Anexina A2/farmacologia , Anexina A3/isolamento & purificação , Anexina A3/farmacologia , Anexina A5/isolamento & purificação , Anexina A5/farmacologia , Anexinas/isolamento & purificação , Anexinas/metabolismo , Calcimicina/farmacologia , Cálcio/metabolismo , Cálcio/farmacologia , Carcinoma de Ehrlich/química , Bovinos , Feminino , Técnicas In Vitro , Ionóforos/farmacologia , Pulmão/química , Potenciais da Membrana , Camundongos , Especificidade da Espécie , Suínos , Xenopus laevis
18.
Arch Biochem Biophys ; 342(2): 322-8, 1997 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-9186494

RESUMO

We investigated the effects of phenothiazines on lung surfactant secretion from rat alveolar epithelial type II cells and on annexin II tetramer (Anx IIt)-mediated membrane fusion. Trifluoperazine and promethazine inhibited ATP-stimulated phosphatidylcholine (PC) secretion from type II cells in a dose-dependent manner. Concentrations that cause 50% inhibition (IC50) were approximately 3 and 25 microM for trifluoperazine and promethazine, respectively. Promethazine also inhibited PC secretion of type II cells stimulated by other secretagogues, including calcium ionophore A23187, phorbol 12-myristate 13-acetate, and terbutaline that are known to stimulate PC secretion via different signal transduction pathways. Since we have recently determined that Anx IIt is involved in PC secretion of type II cells, we examined whether phenothiazines influence Anx IIt's activity. Trifluoperazine and promethazine inhibited Anx IIt's ability to aggregate phosphatidylserine (PS) liposomes, to fuse PS/phosphatidylethanolamine (PE) liposomes, and to fuse PS/PE liposomes with lamellar bodies. These results suggest a relationship between lung surfactant secretion and Anx IIt-mediated membrane fusion.


Assuntos
Anexina A2/farmacologia , Fusão de Membrana/efeitos dos fármacos , Fenotiazinas/farmacologia , Fosfatidilcolinas/metabolismo , Alvéolos Pulmonares/efeitos dos fármacos , Surfactantes Pulmonares/metabolismo , Trifosfato de Adenosina/farmacologia , Animais , Anexina A2/antagonistas & inibidores , Anexina A2/metabolismo , Broncodilatadores/farmacologia , Calcimicina/farmacologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Células Cultivadas , Lipossomos/metabolismo , Masculino , Fosfolipídeos/metabolismo , Prometazina/farmacologia , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/metabolismo , Ratos , Ratos Sprague-Dawley , Terbutalina/farmacologia , Acetato de Tetradecanoilforbol/farmacologia , Trifluoperazina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA