Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Pharmaceuticals (Basel) ; 16(10)2023 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-37895906

RESUMO

During tumorigenesis, urokinase (uPA) and uPA receptor (uPAR) play essential roles in mediating pathological progression in many cancers. To understand the crosstalk between the uPA/uPAR signaling and cancer, as well as to decipher their cellular pathways, we proposed to use cancer driver genes to map out the uPAR signaling. In the study, an integrated pharmaceutical bioinformatics approach that combined modulator identification, driver gene ontology networking, protein targets prediction and networking, pathway analysis and uPAR modulator screening platform construction was employed to uncover druggable targets in uPAR signaling for developing a novel anti-cancer modality. Through these works, we found that uPAR signaling interacted with 10 of 21 KEGG cancer pathways, indicating the important role of uPAR in mediating intracellular cancerous signaling. Furthermore, we verified that receptor tyrosine kinases (RTKs) and ribosomal S6 kinases (RSKs) could serve as signal hubs to relay uPAR-mediated cellular functions on cancer hallmarks such as angiogenesis, proliferation, migration and metastasis. Moreover, we established an in silico virtual screening platform and a uPAR-driver gene pair rule for identifying potential uPAR modulators to combat cancer. Altogether, our results not only elucidated the complex networking between uPAR modulation and cancer but also provided a paved way for developing new chemical entities and/or re-positioning clinically used drugs against cancer.

2.
Lab Invest ; 103(7): 100146, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37004912

RESUMO

Urokinase plasminogen activator (uPA) is a crucial activator of the fibrinolytic system that modulates tissue remodeling, cancer progression, and inflammation. However, its role in membranous nephropathy (MN) remains unclear. To clarify this issue, an established BALB/c mouse model mimicking human MN induced by cationic bovine serum albumin (cBSA), with a T helper cell type 2-prone genetic background, was used. To induce MN, cBSA was injected into Plau knockout (Plau-/-) and wild-type (WT) mice. The blood and urine samples were collected to measure biochemical parameters, such as serum concentrations of immunoglobulin (Ig)G1 and IgG2a, using enzyme-linked immunoassay. The kidneys were histologically examined for the presence of glomerular polyanions, reactive oxygen species (ROS), and apoptosis, and transmission electron microscopy was used to examine subepithelial deposits. Lymphocyte subsets were determined using flow cytometry. Four weeks post-cBSA administration, Plau-/- mice exhibited a significantly higher urine protein-to-creatine ratio, hypoalbuminemia, and hypercholesterolemia than WT mice. Histologically, compared to WT mice, Plau-/- mice showed more severe glomerular basement thickening, mesangial expansion, IgG granular deposition, intensified podocyte effacement, irregular thickening of glomerular basement membrane and subepithelial deposits, and abolishment of the glycocalyx. Moreover, increased renal ROS levels and apoptosis were observed in Plau-/- mice with MN. B-lymphocyte subsets and the IgG1-to-IgG2a ratio were significantly higher in Plau-/- mice after MN induction. Thus, uPA deficiency induces a T helper cell type 2-dominant immune response, leading to increased subepithelial deposits, ROS levels, and apoptosis in the kidneys, subsequently exacerbating MN progression in mice. This study provides a novel insight into the role of uPA in MN progression.


Assuntos
Glomerulonefrite Membranosa , Humanos , Animais , Camundongos , Glomerulonefrite Membranosa/metabolismo , Glomerulonefrite Membranosa/patologia , Soroalbumina Bovina/efeitos adversos , Ativador de Plasminogênio Tipo Uroquinase/genética , Ativador de Plasminogênio Tipo Uroquinase/efeitos adversos , Espécies Reativas de Oxigênio , Imunoglobulina G/efeitos adversos , Imunidade , Linfócitos T Auxiliares-Indutores/metabolismo , Linfócitos T Auxiliares-Indutores/patologia
3.
J Food Drug Anal ; 30(1): 150-162, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35647726

RESUMO

Drug substances are at risk of contamination with N-nitrosamines (NAs), well-known carcinogenic agents, during synthesis processes and/or long-term storage. Therefore, in this study, we developed an efficient data-based screening approach to systemically assess marketed products and investigated its scalability for benefiting both regulatory agencies and pharmaceutical industries. A substructure-based screening method employing DataWarrior, an open-source software, was established to evaluate the risks of NA impurities in drug substances. Eight NA substructures containing susceptible amino sources for N-nitrosation have been identified as screening targets: dimethylamine (DMA), diethylamine, isopropylethylamine, diisopropylamine, N-methyl-2-pyrrolidone, dibutylamine, methylphenylamine, and tetrazoles. Our method detected 192 drug substances with a theoretical possibility of NA impurity, 141 of which had not been reported previously. In addition, the DMA moiety was significantly dominant among the eight NA substructures. The results were validated using data from the literature, and a high detection sensitivity of 0.944 was demonstrated. Furthermore, our approach has the advantage of scalability, owing to which 31 additional drugs with suspected NA-contaminated substructures were identified using the substructures of 1-methyl-4-piperazine in rifampin and 1-cyclopentyl-4-piperazine in rifapentine. In conclusion, the reported substructure-based approach provides an effective and scalable method for the screening and investigation of NA impurities in various pharmaceuticals and might be used as an ancillary technique in the field of pharmaceutical quality control for risk assessments of potential NA impurities.


Assuntos
Contaminação de Medicamentos , Nitrosaminas , Piperazinas , Controle de Qualidade , Medição de Risco
4.
Nutrition ; 69: 110554, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31536856

RESUMO

OBJECTIVES: Indoxyl sulfate (IS), a uremic toxin, has been shown to promote the epithelial-to-mesenchymal transition (EMT) of human proximal tubular cells and to accelerate the progression of chronic kidney disease (CKD). Despite the well-known protective role of 1,25-dihydroxyvitamin D3 [1,25(OH)2 D3] in EMT, the effect of 1,25(OH)2 D3 on IS-induced EMT in human proximal tubular epithelial cells and the underlying mechanism remain unclear. The aim of this study was to determine whether IS (0-1 mM) dose-dependently inhibited the protein expression of E-cadherin and increased the protein expression of alpha-smooth muscle actin, N-cadherin, and fibronectin. METHODS: This study investigated the molecular mechanism by which 1,25(OH)2 D3 attenuates IS-induced EMT. HK-2 human renal tubular epithelial cells was used as the study model, and the MTT assay, Western Blotting, siRNA knockdown technique were used to explore the effects of 1,25(OH)2 D3 on EMT in the presence of IS. RESULTS: Pretreatment with 1,25(OH)2 D3 inhibited the IS-induced EMT-associated protein expression in HK-2 cells. IS induced the phosphorylation of Akt (S473) and ß-catenin (S552) and subsequently increased the nuclear accumulation of ß-catenin. Pretreatment with 1,25(OH)2 D3 and LY294002 (phosphoinositide 3-kinase [PIK3] inhibitor) significantly inhibited the IS-induced phosphorylation of Akt and ß-catenin, nuclear ß-catenin accumulation, and EMT-associated protein expression. CONCLUSIONS: Results from the present study revealed that the anti-EMT effect of 1,25(OH)2 D3 is likely through inhibition of the PI3K/Akt/ß-catenin pathway, which leads to down-regulation of IS-driven EMT-associated protein expression in HK-2 human renal tubular epithelial cells.


Assuntos
Calcitriol/farmacologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Indicã/administração & dosagem , Túbulos Renais/citologia , Transdução de Sinais/efeitos dos fármacos , Caderinas/metabolismo , Linhagem Celular , Relação Dose-Resposta a Droga , Células Epiteliais , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , beta Catenina/metabolismo
5.
J Biomed Sci ; 23: 24, 2016 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-26846181

RESUMO

BACKGROUND: suPAR biomarker generally considered a pathogenic factor in FSGS. However, studies have been published that dispute this conclusion. The current study was designed to investigate the roles of uPA and suPAR in FSGS in clinical and mouse models. METHODS: Clinical subjects including those with biopsy-proven FSGS and MCD were enrolled. To verify the role of uPA in FSGS, Adriamycin was used to induce FSGS in uPA knockout (uPA(-/-)) and BALB/c (WT) mice. Proteinuria and suPAR, the cleaved/intact forms of the circulating suPAR, and possible proteases involving cleavage of the suPAR were also studied. RESULTS: FSGS clinical cases presented significantly higher serum levels of suPAR and Cr and lower serum levels of uPA. In the mice model, the uPA(-/-) group exhibited faster disease progression and worsening proteinuria than the WT group. In addition, the uPA(-/-) group had higher plasma suPAR levels, glomerular cell apoptosis, and dysregulation of the Th1/Th2 balance. In an analysis of suPAR variants in FSGS, both the intact and cleaved forms of the suPAR were higher in clinical subjects and the mouse model. However, the process of suPAR cleavage was not mediated by enzymatic activities of the uPA, elastase, or cathepsin G. CONCLUSIONS: A deficiency of uPA accelerated the progression of Adriamycin-induced mouse FSGS model. Decrease of serum uPA levels may be an indicator of the progression of FSGS in clinical subjects and animal models.


Assuntos
Glomerulosclerose Segmentar e Focal/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Glomerulosclerose Segmentar e Focal/genética , Glomerulosclerose Segmentar e Focal/patologia , Humanos , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Células Th1/metabolismo , Células Th1/patologia , Células Th2/metabolismo , Células Th2/patologia , Ativador de Plasminogênio Tipo Uroquinase/genética
6.
Clin Biochem ; 48(18): 1324-9, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26162494

RESUMO

OBJECTIVES: Soluble urokinase plasminogen activator receptor (suPAR), derived from membrane bound uPAR, is associated with inflammatory diseases. In the present study, we explored the expression of uPAR/suPAR in common biopsy-proven kidney diseases and the relationship between suPAR and staging of type 2 diabetic nephropathy (DN). DESIGN AND METHODS: Serum samples for suPAR and renal tissues for uPAR staining were investigated in various common kidney diseases. The levels of serum suPAR were measured and adequate cut-off values of different stage of DN were calculated by receiver operating characteristic (ROC) curve. RESULTS: In our results, the expression of uPAR on renal tissues was pronounced in the majority of kidney diseases. Comparing of expression of uPAR among different kidney diseases, it was strongest in minimal change disease (MCD) and weakest in chronic interstitial nephritis. Serum suPAR in most kidney diseases, except of MCD, was significantly elevated and was highest in DN. As for DN and suPAR, we found that suPAR progressively increased with staging of DN. Moreover, suPAR was linearly and negatively related to estimated glomerular filtration rate and positively related to the amount of proteinuria. By ROC curve, the cut-off values of suPAR in DN for assessing development increased with the progression of the disease. CONCLUSIONS: We concluded that uPAR/suPAR is elevated in most kidney diseases and that suPAR is a useful biomarker for assessing stages of DN.


Assuntos
Nefropatias Diabéticas/diagnóstico , Nefrite Intersticial/diagnóstico , Nefrose Lipoide/diagnóstico , Proteinúria/diagnóstico , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Adulto , Idoso , Biomarcadores/sangue , Nefropatias Diabéticas/sangue , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/patologia , Progressão da Doença , Feminino , Expressão Gênica , Taxa de Filtração Glomerular , Humanos , Masculino , Pessoa de Meia-Idade , Nefrectomia , Nefrite Intersticial/sangue , Nefrite Intersticial/genética , Nefrite Intersticial/patologia , Nefrose Lipoide/sangue , Nefrose Lipoide/genética , Nefrose Lipoide/patologia , Proteinúria/genética , Proteinúria/patologia , Curva ROC , Receptores de Ativador de Plasminogênio Tipo Uroquinase/sangue , Índice de Gravidade de Doença
7.
Dis Markers ; 2015: 548101, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25944975

RESUMO

In cell model, we discovered the association between chaperonin-containing t-complex polypeptide 1 subunit ß (TCP-1ß) and early diabetic nephropathy (DN). In this study, we further explored the relationships between TCP-1ß and type 2 diabetic mellitus (DM). To mimic the clinical hyperfiltration state, a type 2 DM mice model was established by feeding a high-fat diet in combination with treatment of streptozotocin and nicotinamide. Blood and urine were collected to determine creatinine clearance (C cr), and kidney tissues were harvested for evaluation of TCP-1ß expression by immunohistochemistry and Western blot. Meanwhile, clinical subjects of healthy controls and type 2 DM were recruited to strengthen the evidence with urine TCP-1ß. Results showed that C cr and the expression of TCP-1ß in kidney were significantly higher one week after hyperglycemia development, suggesting that the hyperfiltration state was successfully established in the mice model. TCP-1ß was expressed predominantly on renal tubules. By using the estimated glomerular filtration rate to index progression in clinical investigation, urine TCP-1ß level was associated with the hyperfiltration phase in type 2 DM patients. Conclusively, we confirmed that TCP-1ß is a possible biomarker for early nephropathy of type 2 DM, but further mechanistic study to elucidate its cause and pathway is needed.


Assuntos
Chaperonina com TCP-1/urina , Nefropatias Diabéticas/urina , Taxa de Filtração Glomerular , Idoso , Animais , Biomarcadores/metabolismo , Biomarcadores/urina , Estudos de Casos e Controles , Chaperonina com TCP-1/genética , Chaperonina com TCP-1/metabolismo , Nefropatias Diabéticas/metabolismo , Feminino , Humanos , Rim/metabolismo , Rim/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade
8.
Biomed Res Int ; 2014: 814869, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24795896

RESUMO

The importance of B-cell activation and immune complex-mediated Fc-receptor activation in the pathogenesis of immunologically mediated glomerulonephritis has long been recognized. The two nonreceptor tyrosine kinases, spleen tyrosine kinase (Syk) and Bruton's tyrosine kinase (Btk), are primarily expressed by hematopoietic cells, and participate in B-cell-receptor- and Fc-receptor-mediated activation. Pharmacological inhibitors of Syk or Btk are undergoing preclinical development and clinical trials for several immune diseases; and Syk inhibitors have been shown to reduce disease activity in rheumatoid arthritis patients. However, the clinical therapeutic efficacies of these inhibitors in glomerulonephritis have not been evaluated. Herein, we review recent studies of Syk and Btk inhibitors in several experimental primary and secondary glomerulonephritis models. These inhibitors suppressed development of glomerular injury, and also ameliorated established kidney disease. Thus, targeting Syk and Btk signaling pathways is a potential therapeutic strategy for glomerulonephritis, and further evaluation is recommended.


Assuntos
Glomerulonefrite , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Inibidores de Proteínas Quinases , Proteínas Tirosina Quinases/antagonistas & inibidores , Tirosina Quinase da Agamaglobulinemia , Animais , Glomerulonefrite/tratamento farmacológico , Glomerulonefrite/imunologia , Humanos , Camundongos , Transdução de Sinais , Quinase Syk
9.
Biochim Biophys Acta ; 1833(8): 1799-810, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23542129

RESUMO

Glucocorticoid receptor-interacting protein 1 (GRIP1), a p160 family nuclear receptor co-activator protein, has three activation domains that recruit at least three secondary co-activators: CBP/p300, co-activator-associated arginine methyltransferase 1, and coiled-coil co-activator, which exhibits histone acetyltransferase and/or arginine methyltransferase activities. The regulatory mechanisms underlying the co-activation functions of GRIP1, which associates with promyelocytic leukemia protein (PML) in PML-nuclear bodies, are not well-understood. This study showed that PML specifically and dramatically enhanced the C-terminal transactivation activity of GRIP1 by directly binding to GRIP1 but only when it was sumoylated. Most of the transactivation activity resided in the N-terminal PML regions that are conserved among isoforms. Three N-terminal sumoylation residues (Lys 65, 160, and 490) exhibited differential roles in the regulation of GRIP1 activity, and the sumoylation of Lys 490 acted as the primary nuclear localization signal of PML. While GRIP1 transactivation was stimulated to a similar degree by PML (K490R), located in the nucleus, and wild-type PML, PML (K490D) and the C-truncated mutant PML1-489 both displayed an epinuclear localization and were mostly inactive in stimulating GRIP. Based on these data, nuclear foci, nuclear localization, and the sumoylation status of Lys 490 were not essential for the enhancement of GRIP1 activity by PML, but the charge status of Lys 490 was important for subcellular localization of PML and cross-talk between its N- and C-terminal regions to modulate transcriptional activation. Taken together, these results provide insight into the regulatory mechanisms of PML that control the functional activities of GRIP1.


Assuntos
Proteínas de Transporte/metabolismo , Lisina/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Células HeLa , Humanos , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/metabolismo , Lisina/genética , Proteínas Nucleares/genética , Proteína da Leucemia Promielocítica , Ligação Proteica/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/genética , Ativação Transcricional , Proteínas Supressoras de Tumor/genética
10.
Cancer Chemother Pharmacol ; 71(2): 379-88, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23143189

RESUMO

PURPOSE: The chemotherapy agent gemcitabine is currently administered intravenously because the drug has poor oral bioavailability. In order to assess the pharmacokinetics and antitumor activity of D07001-F4, a new self-microemulsifying oral drug delivery system preparation of gemcitabine, this study was performed to compare the effect of D07001-F4 with administered gemcitabine in vitro and in vivo. METHODS: D07001-F4 pharmacokinetics was examined by evaluation of in vitro deamination of D07001-F4 and gemcitabine hydrochloride by recombinant human cytidine deaminase (rhCDA) and in vivo evaluation of D07001-F4 pharmacokinetics in mice. Antitumor activity was evaluated by comparing the effect of D07001-F4 and gemcitabine hydrochloride in inhibiting growth in nine cancer cell lines and by examining the effect of D07001-F4 and gemcitabine in two xenograft tumor models in mice. RESULTS: In vitro deamination of D07001-F4 by rhCDA was 3.3-fold slower than deamination of gemcitabine hydrochloride. Growth inhibition by D07001-F4 of 7 of the 8 cancer cell lines was increased compared with that seen with gemcitabine hydrochloride, and D07001-F4 inhibited the growth of pancreatic and colon cancer xenografts. In vivo pharmacokinetics showed the oral bioavailability of D07001-F4 to be 34%. CONCLUSIONS: D07001-F4 was effective against several cancer types, was metabolized more slowly than gemcitabine hydrochloride, and exhibited enhanced oral bioavailability.


Assuntos
Antimetabólitos Antineoplásicos/administração & dosagem , Desoxicitidina/análogos & derivados , Administração Oral , Animais , Linhagem Celular Tumoral , Química Farmacêutica , Desoxicitidina/administração & dosagem , Desoxicitidina/farmacocinética , Desoxicitidina/farmacologia , Feminino , Humanos , Camundongos , Camundongos Nus , Ensaios Antitumorais Modelo de Xenoenxerto , Gencitabina
11.
Exp Diabetes Res ; 2011: 565647, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22144988

RESUMO

The aim of this study is to investigate the role of chaperonin-containing t-complex polypeptide 1 beta (CCT2) in the regulation of mouse mesangial cell (mMC) contraction, proliferation, and migration with filamentous/globular-(F/G-) actin ratio under high glucose induction. A low CCT2 mMC model induced by treatment of small interference RNA was established. Groups with and without low CCT2 induction examined in normal and high (H) glucose conditions revealed the following major results: (1) low CCT2 or H glucose showed the ability to attenuate F/G-actin ratio; (2) groups with low F/G-actin ratio all showed less cell contraction; (3) suppression of CCT2 may reduce the proliferation and migration which were originally induced by H glucose. In conclusion, CCT2 can be used as a specific regulator for mMC contraction, proliferation, and migration affected by glucose, which mechanism may involve the alteration of F-actin, particularly for cell contraction.


Assuntos
Actinas/metabolismo , Chaperonina com TCP-1/metabolismo , Células Mesangiais/fisiologia , Animais , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Chaperonina com TCP-1/antagonistas & inibidores , Chaperonina com TCP-1/genética , Glucose/metabolismo , Glucose/farmacologia , Células Mesangiais/citologia , Células Mesangiais/efeitos dos fármacos , Camundongos , Modelos Biológicos , RNA Interferente Pequeno/genética
12.
Arch Pathol Lab Med ; 134(12): 1804-12, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21128779

RESUMO

CONTEXT: Plasminogen kringle domain (K) 5 is known to inhibit endothelial cell growth, but limited data are available investigating the relationship between K5 and glomerulonephritis (GN). OBJECTIVE: To understand the relationships among K5, GN, and glomerular endothelial cells in GN mice models and human subjects. DESIGN: Two mice models of GN and 2 categories of human GN biopsy samples were collected to gain insight into the disease mechanism from the laboratory to bedside. In the mechanistic animal study, membranous nephropathy (MN) and focal segmental glomerulosclerosis mice models were used. Kringle domain 5 in the diseased kidney was located by immunofluorescence and quantified by Western blotting. In the kinetic animal study, different MN time points were stained with K5, immunoglobulin G, and C3 by immunofluorescence. CD31 and proliferating cell nuclear antigen were evaluated by immunohistochemical double staining for alterations in the glomerular endothelial cells. Biopsy samples from patients diagnosed with antibody (Ab)-mediated and non-Ab-mediated GN were collected for K5 analysis. RESULTS: The expression level of K5 was found to be significant in MN, but not in focal segmental glomerulosclerosis, and was markedly elevated in the diseased glomeruli along the capillary walls. Kringle domain 5 levels increased steadily with the evolution of MN, appearing after the deposition of Abs. In altered glomerular endothelial cells, CD31 decreased with the evolution of MN. In human subjects, K5 occurred only in patients with Ab GN. CONCLUSIONS: Kringle domain 5 might be involved in the progression of Ab-mediated GN and associated with the alteration of MN glomerular endothelial cell growth.


Assuntos
Inibidores da Angiogênese/metabolismo , Endotélio Vascular/metabolismo , Glomerulonefrite Membranosa/metabolismo , Glomerulosclerose Segmentar e Focal/metabolismo , Fragmentos de Peptídeos/metabolismo , Plasminogênio/metabolismo , Animais , Biópsia , Modelos Animais de Doenças , Endotélio Vascular/ultraestrutura , Feminino , Glomerulonefrite Membranosa/patologia , Glomerulosclerose Segmentar e Focal/patologia , Humanos , Glomérulos Renais/irrigação sanguínea , Glomérulos Renais/ultraestrutura , Camundongos , Camundongos Endogâmicos BALB C , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo
13.
Transl Res ; 156(2): 80-90, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20627192

RESUMO

Glomerular hyperfiltration is associated with mesangial cell hypocontractility. How 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors (statins) influence mesangial cell contraction is unclear. We investigated the effect of statins on mesangial cell hypocontractility and identified candidate proteins and filamentous/globular (F/G)-actin involved in this process. A high-glucose-induced mouse mesangial cell hypocontractility model was treated with fungal statins, simvastatin (Sim), lovastatin (Lov), and pravastatin (Pra). The optimum statin dose was determined by an 3-(4,5-Dimethylthiazol-2-Yl)-2,5-Diphenyltetrazolium Bromide (MTT) assay and then applied to a cell model. A 2-dimensional gel/matrix-assisted laser desorption/ionization time-of-flight mass spectrometer analysis was used to evaluate protein expression cells incubated in the presence of a normal level of glucose (N), a high level of glucose (H), and a high level of glucose plus Sim (H + S). Candidate proteins were analyzed. Finally, the ratio of F/G actin in groups N, H, and H+S was evaluated. The MTT assay showed that Sim and Lov exerted dose- and time-related inhibition of proliferation of mesangial cells at N, but Pra had no effect. The optimum doses selected for Sim was 1 microM and for Lov was 3 microM, which were 1 increment before significant proliferation inhibition. Both doses reversed cell hypocontractility significantly, but Sim was chosen for further proteomic and F/G actin analyses. Proteomic analysis of groups N, H, and H + S showed that 18 proteins were involved in hypocontractility. These proteins were grouped and analyzed based on their known functions. Two selected proteins, TCP-1beta and GRP78, that were upregulated and downregulated, respectively, were confirmed by Western blot and immunohistochemistry. In regard to the F/G actin, group H had a significantly lower ratio than that of group N, and group H + S returned to a level similar to that of group N. In conclusion, Sim and Lov both seem to reverse mesangial cell hypocontractility. The process of Sim reversal of mesangial cell hypocontractility may involve F-actin, TCP-1beta, and GRP78.


Assuntos
Actinas/metabolismo , Mesângio Glomerular/fisiologia , Glucose/farmacologia , Sinvastatina/farmacologia , Actinas/efeitos dos fármacos , Acil Coenzima A/metabolismo , Animais , Divisão Celular/efeitos dos fármacos , Chaperona BiP do Retículo Endoplasmático , Mesângio Glomerular/citologia , Mesângio Glomerular/efeitos dos fármacos , Proteínas de Choque Térmico/efeitos dos fármacos , Proteínas de Choque Térmico/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Glomérulos Renais/efeitos dos fármacos , Glomérulos Renais/fisiologia , Lovastatina/farmacologia , Camundongos , Pravastatina/farmacologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Acetato de Tetradecanoilforbol/farmacologia
14.
J Control Release ; 130(3): 252-8, 2008 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-18652856

RESUMO

Macromolecular drugs such as proteins and gene products are presumably the most desirable therapeutic agents due to their unmatched substrate specificity and reaction efficiency. Yet, clinical use of these drugs has met with limited success, primarily due to the impermeable nature of the cell membrane that restricts cellular drug uptake to only small (<600 Da) and hydrophobic molecules. The recent discovery of the protein transduction domain (PTD) membrane-penetrating peptides, such as HIV-TAT, has finally offered the possibility of resolving this cell-membrane barrier for macromolecular drug delivery. Via covalent linkages, these PTD peptides have been shown to ferry the attached macromolecular species across membranes of all cell types, both in vitro and in vivo. Nevertheless, the lack of selectivity for PTD-mediated internalization restricts the application of this cell uptake method in clinical practice, due to concerns of inducing systemic toxicity caused by the carried drugs. Presented herein is a modified version of our previously established "ATTEMPTS" approach in delivery of macromolecular drugs, which integrates the cell-penetrating PTDs into a heparin/protamine-regulated delivery system. In vitro findings using asparaginase (ASNase) as a model macromolecular anti-tumor agent were able to validate the feasibility of this delivery system. The chemically constructed TAT-ASNase conjugates not only were able to translocate into the MOLT-4 cells and elicit the cytotoxic effects, but also this PTD-mediated intracellular drug uptake could be regulated (with on/off control) by the addition of heparin and protamine. This modified ATTEMPTS system therefore presents a new avenue of treatment of various types of cancers and other diseases with macromolecular drugs. In vitro characterization and a preliminary proof-of-concept animal investigation that demonstrates the feasibility of this PTD-mediated ASNase therapeutic system is subsequently described.


Assuntos
Asparaginase/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Linfoma/tratamento farmacológico , Sinais Direcionadores de Proteínas , Sequência de Aminoácidos , Animais , Asparaginase/química , Asparaginase/uso terapêutico , Catálise , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Feminino , Células HeLa , Heparina/química , Humanos , Linfoma/patologia , Camundongos , Camundongos Endogâmicos DBA , Fragmentos de Peptídeos/química , Pró-Fármacos/administração & dosagem , Pró-Fármacos/química , Pró-Fármacos/uso terapêutico , Protaminas/química , Análise de Sobrevida , Produtos do Gene tat do Vírus da Imunodeficiência Humana/química
15.
Mol Cancer Res ; 6(7): 1204-14, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18644983

RESUMO

Zac1 is a novel seven-zinc finger protein which possesses the ability to bind specifically to GC-rich DNA elements. Zac1 not only promotes apoptosis and cell cycle arrest but also acts as a transcriptional cofactor for p53 and a number of nuclear receptors. Our previous study indicated that the enhancement of p53 activity by Zac1 is much more pronounced in HeLa cells compared with other cell lines tested. This phenomenon might be due to the coactivator effect of Zac1 on p53 and the ability of Zac1 to reverse E6 inhibition of p53. In the present study, we showed that Zac1 acted synergistically with either p53 or a histone deacetylase inhibitor, trichostatin A, to enhance p21(WAF1/Cip1) promoter activity. We showed that Zac1 physically interacted with some nuclear receptor corepressors such as histone deacetylase 1 (HDAC1) and mSin3a, and the induction of p21(WAF1/Cip1) gene and protein by Zac1 was suppressed by either overexpressing HDAC1 or its deacetylase-dead mutant. In addition, our data suggest that trichostatin A-induced p21(WAF1/Cip1) protein expression might be mediated through a p53-independent and HDAC deacetylase-independent pathway. Taken together, our data suggest that Zac1 might be involved in regulating the p21(WAF1/Cip1) gene and protein expression through its protein-protein interaction with p53 and HDAC1 in HeLa cells.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/genética , Histona Desacetilases/metabolismo , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Células COS , Proteínas de Ciclo Celular/genética , Chlorocebus aethiops , Regulação para Baixo/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes Reporter , Células HeLa , Histona Desacetilase 1 , Humanos , Ácidos Hidroxâmicos/farmacologia , Modelos Biológicos , Regiões Promotoras Genéticas/genética , Ligação Proteica/efeitos dos fármacos , Fatores de Transcrição/genética , Ativação Transcricional/efeitos dos fármacos , Proteínas Supressoras de Tumor/genética
16.
J Biomed Mater Res A ; 84(1): 238-46, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17607767

RESUMO

Camptothecin (CPT) is a naturally occurring alkaloid that shows promise in antitumor activity in vitro against various tumor cell lines. Its potential clinical uses, however, are hindered by a lack of reaction selectivity and poor water solubility. Presented herein is a novel polyrotaxane (PR)-based delivery system that could potentially lead to a highly effective yet less toxic CPT therapy. The approach involves the synthesis of the PR-CPT conjugates via hydrolyzable linkages. To enhance the therapeutic efficacy of CPT, a cell-penetrating peptide, LMWP, is linked to the conjugate to allow specific, intratumoral delivery of CPT. To avoid nonselective uptake of the conjugates by normal tissues following administration, the cell-penetrating function of LMWP on the conjugates is masked by heparin binding. This system was designed such that after accumulation at the tumor via the enhanced permeability and retention (EPR) effect, protamine can be subsequently administered to unmask heparin inhibition on LMWP, permitting intracellular uptake of the LMWP-PR-CPT conjugates. Once inside the tumor, CPT molecules are detached from the PR chain by hydrolysis, yielding a sustained concentration of CPT within tumor cells. In this paper, we demonstrated the in vitro feasibility of this delivery system. The LMWP-PR-CPT conjugates yielded a sevenfold increase in the overall CPT solubility, as well as a sustained release of CPT over a period of more than 7 days. Intracellular uptake of these conjugates by A2780 human ovarian cancer cells and regulation of such uptake by heparin and protamine were tested by MTT assay and confocal/flow cytometric methods, respectively.


Assuntos
Camptotecina/química , Camptotecina/toxicidade , Ciclodextrinas/química , Sistemas de Liberação de Medicamentos/métodos , Poloxâmero/química , Rotaxanos/química , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Estudos de Viabilidade , Citometria de Fluxo , Heparina/química , Humanos , Espectroscopia de Ressonância Magnética , Estrutura Molecular , Protaminas/química
17.
FASEB J ; 19(11): 1555-7, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16033808

RESUMO

Naturally derived, nontoxic peptides from protamine by the authors, termed low molecular weight protamines (LMWPs), possess high arginine content and carry significant sequence similarity to that of TAT, by far the most potent protein transduction domain peptide. Therefore, it was hypothesized that these LMWPs would also inherit the similar translocation activity across the cell membrane, which enables any impermeable species to be transduced into the cells. LMWPs were prepared by enzymatic digestion of protamine, examined their capability of transducing an impermeable protein toxin into the tumor cells by chemical conjugation, and determined cytotoxicity of transduced protein toxin (e.g., gelonin) against cancer cell lines and a tumor-bearing mouse. In vitro results showed that LMWPs could indeed translocate themselves into several mammalian cell lines as efficiently as TAT, thereby transducing impermeable gelonin into the cells by chemical conjugation. In vivo studies further confirmed that LMWP could carry an impermeable gelonin across the tumor mass and subsequently inhibit the tumor growth. In conclusion, the presence of equivalent cell translocation potency, absence of toxicity of peptide itself, and the suitability for low-cost production by simple enzymatic digestion could expand the range of clinical applications of LMWPs, including medical imaging and gene/protein therapies.


Assuntos
Proteínas de Membrana Transportadoras/farmacologia , Fragmentos de Peptídeos/farmacologia , Protaminas/farmacologia , Animais , Linhagem Celular Tumoral , Feminino , Produtos do Gene tat/farmacologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Peso Molecular , Neoplasias Experimentais/tratamento farmacológico , Proteínas de Plantas/farmacologia , Protaminas/uso terapêutico , Transporte Proteico , Proteínas Inativadoras de Ribossomos Tipo 1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA