Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Transl Med ; 16(1): 164, 2018 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-29898734

RESUMO

BACKGROUND: Hypoxia-mediated chemoresistance has been regarded as an important obstacle in the development of cancer treatment. Knockdown of krüppel-like factor 5 (KLF5) was reported to inhibit hypoxia-induced cell survival and promote cell apoptosis in non-small cell lung cancer (NSCLC) cells via direct regulation of hypoxia inducible factor-1α (HIF-1α) expression. However, the roles of KLF5 in the development of hypoxia-induced cisplatin (DDP) resistance and its underlying mechanism in NSCLC cells remain to be further elucidated. METHODS: Western blot was performed to determine the protein levels of KLF5, P-glycoprotein (P-gp) and HIF-1α in treated NSCLC cells. Cell survival was examined by MTT assay. The effect of KLF5 knockdown on hypoxia-induced glycolysis was assessed by measuring glucose consumption and lactate production. The effect of KLF5 knockdown on the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) pathway was analyzed by western blot. RESULTS: Hypoxia upregulated the expression of KLF5 in NSCLC cells. KLF5 knockdown suppressed hypoxia-induced DDP resistance in NSCLC cells, as demonstrated by the increased cytotoxic effects of DDP and reduced P-gp expression in NSCLC cells in hypoxia. Moreover, KLF5 knockdown inhibited hypoxia-induced HIF-1α expression and glycolysis, and KLF5 knockdown suppressed hypoxia-induced DDP resistance by inhibiting HIF-1α-dependent glycolysis in NSCLC cells. Furthermore, KLF5 knockdown suppressed hypoxia-induced activation of the PI3K/Akt/mTOR pathway in NSCLC cells and KLF5 overexpression promoted hypoxia-induced DDP resistance in NSCLC cells through activation of the PI3K/Akt/mTOR pathway. CONCLUSIONS: KLF5 knockdown could suppress hypoxia-induced DDP resistance, and its mechanism may be due to the inhibition of HIF-1α-dependent glycolysis via inactivation of the PI3K/Akt/mTOR pathway.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Glicólise/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Neoplasias Pulmonares/metabolismo , Transdução de Sinais/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/patologia , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Pulmonares/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo
2.
J Pharmacol Sci ; 136(2): 79-85, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29422382

RESUMO

Human organic anion transporter-3 (hOAT3) is richly expressed in the kidney, where it plays critical roles in the secretion, from the blood to urine, of clinically important drugs, such as anti-viral therapeutics, anti-cancer drugs, antibiotics, antihypertensives, and anti-inflammatories. In the current study, we examined the role of dexamethasone in hOAT3 transport activity in the kidney HEK293 cells. Cis-inhibition study showed that dexamethasone exhibited a concentration-dependent inhibition of hOAT3-mediated uptake of estrone sulfate, a prototypical substrate for the transporter, with IC50 value of 49.91 µM. Dixon plot analysis revealed that inhibition by dexamethasone was competitive with a Ki = 47.08 µM. In contrast to the cis-inhibition effect of dexamethasone, prolonged incubation (6 h) of hOAT3-expressing cells with dexamethasone resulted in an upregulation of hOAT3 expression and transport activity, kinetically revealed as an increase in the maximum transport velocity Vmax without meaningful alteration in substrate-binding affinity Km. Such upregulation was abrogated by GSK650394, a specific inhibitor for serum- and glucocorticoid-inducible kinases (sgk). Dexamethasone also enhanced sgk1 phosphorylation. Our study demonstrated that dexamethasone exhibits dual effects on hOAT3: it is a competitive inhibitor for hOAT3-mediated transport, and interestingly, when entering the cells, it stimulates hOAT3 expression and transport activity through sgk1.


Assuntos
Dexametasona/farmacologia , Estrona/análogos & derivados , Transportadores de Ânions Orgânicos Sódio-Independentes/fisiologia , Benzoatos/farmacologia , Transporte Biológico/efeitos dos fármacos , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Relação Dose-Resposta a Droga , Estrona/metabolismo , Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Proteínas Imediatamente Precoces/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/genética , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Tempo , Regulação para Cima/efeitos dos fármacos
3.
BMC Complement Altern Med ; 17(1): 216, 2017 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-28415997

RESUMO

BACKGROUND: Dusuqing granules (DSQ) have been used in the treatment of bacterial pneumonia clinically, with remarkable benefits. This study was initiated to explore the effects of DSQ on pulmonary inflammation by regulating nuclear factor (NF)-κB/mitogen-activated protein kinase (MAPK) signaling in bacterial pneumonia rats. METHODS: Rat model was duplicated with Klebsiella pneumonia by a one-time intratracheal injection. Rats were randomized into control, model, DSQ and levofloxacin (LVX) groups. After administrated with appropriate medicines for 7 days, lung tissues were harvested and prepared for pathological analysis, and interleukin (IL)-1, IL-6, monocyte chemotactic protein (MCP)-1and macrophage inflammatory protein (MIP)-2 detections. NF-κB mRNA was measured by real-time qPCR, and the phosphorylation and total proteins of P38MAPK, JNK46/54, ERK42/44 were determined by Western blotting. RESULTS: Marked pathological impairments were observed in model rats, whereas were improved in DSQ group. The cytokines levels, NF-κB mRNA expression and the phosphorylation of P38MAPK, JNK46/54 and ERK42/44 proteins were significantly higher in model group, and were significantly depressed in DSQ group. CONCLUSION: The protective effects of DSQ on Klebsiella pneumonia might be attributed to its inactivative effects of NF-κB/ MAPK pathway.


Assuntos
Medicamentos de Ervas Chinesas/farmacologia , Infecções por Klebsiella/metabolismo , Klebsiella pneumoniae , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Pneumonia Bacteriana/metabolismo , Pneumonia/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Quimiocina CCL2/metabolismo , Quimiocina CXCL2/metabolismo , Medicamentos de Ervas Chinesas/uso terapêutico , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Interleucinas/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Infecções por Klebsiella/tratamento farmacológico , Infecções por Klebsiella/microbiologia , Infecções por Klebsiella/patologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Magnoliopsida , Masculino , Fosforilação , Fitoterapia , Pneumonia/tratamento farmacológico , Pneumonia/microbiologia , Pneumonia Bacteriana/tratamento farmacológico , Pneumonia Bacteriana/microbiologia , Pneumonia Bacteriana/patologia , Ratos Sprague-Dawley , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
4.
Biopharm Drug Dispos ; 38(8): 449-457, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28608480

RESUMO

Human organic anion transporter 3 (hOAT3) is localized at the basolateral membrane of renal proximal tubule cells and facilitates the renal secretion of numerous clinical drugs, including anti-HIV therapeutics, anti-tumor drugs, antibiotics, antihypertension drugs and anti-inflammatories. The present study explored the role of serum and glucocorticoid-inducible kinase 1 (sgk1) in the regulation of hOAT3. It was shown that over-expression of sgk1 in hOAT3-expressing cells stimulated hOAT3 transport activity by enhancing the transporter expression at the plasma membrane, kinetically reflected as an increased maximal transport velocity Vmax without substantial change in the substrate-binding affinity Km . In contrast, treatment of cells with the sgk-specific inhibitor GSK650394 resulted in a dose-dependent inhibition of hOAT3 transport activity. Evidence was further provided that sgk1 regulation of hOAT3 activity was mediated by ubiquitin ligase Nedd4-2, an enzyme previously shown to have an inhibitory effect on hOAT3. It was shown that sgk1 phosphorylated Nedd4-2, weakened the association between Nedd4-2 and hOAT3, and decreased hOAT3 ubiquitination. Functionally, the sgk1-stimulated hOAT3 transport activity was attenuated in the presence of a ligase-dead mutant of Nedd4-2. In summary, the investigation established for the first time that sgk1 stimulates hOAT3 transport activity by interfering with the inhibitory effect of Nedd4-2 on the transporter.


Assuntos
Proteínas Imediatamente Precoces/metabolismo , Ubiquitina-Proteína Ligases Nedd4/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Benzoatos/farmacologia , Biotinilação , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Células COS , Chlorocebus aethiops , Estrona/análogos & derivados , Estrona/metabolismo , Humanos , Proteínas Imediatamente Precoces/antagonistas & inibidores , Proteínas Imediatamente Precoces/genética , Ubiquitina-Proteína Ligases Nedd4/genética , Transportadores de Ânions Orgânicos Sódio-Independentes/genética , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais
5.
Med Res Rev ; 36(5): 964-79, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27291023

RESUMO

Organic anion transporters (OATs) encoded by solute carrier 22 family are localized in the epithelia of multiple organs, where they mediate the absorption, distribution, and excretion of a diverse array of negatively charged environmental toxins and clinically important drugs. Alterations in the expression and function of OATs play important roles in intra- and interindividual variability of the therapeutic efficacy and the toxicity of many drugs. As a result, the activity of OATs must be under tight regulation so as to carry out their normal functions. The regulation of OAT transport activity in response to various stimuli can occur at several levels such as transcription, translation, and posttranslational modification. Posttranslational regulation is of particular interest, because it usually happens within a very short period of time (minutes to hours) when the body has to deal with rapidly changing amounts of substances as a consequence of variable intake of drugs, fluids, or meals as well as metabolic activity. This review article highlights the recent advances from our laboratory in uncovering several posttranslational mechanisms underlying OAT regulation. These advances offer the promise of identifying targets for novel strategies that will maximize therapeutic efficacy in drug development.


Assuntos
Transportadores de Ânions Orgânicos/metabolismo , Animais , Humanos , Túbulos Renais Proximais/metabolismo , Farmacocinética , Ubiquitinação
6.
Am J Physiol Renal Physiol ; 310(9): F821-31, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-26823285

RESUMO

Human organic anion transporter 1 (hOAT1) expressed at the membrane of the kidney proximal tubule cells mediates the body disposition of a diverse array of clinically important drugs, including anti-HIV therapeutics, antitumor drugs, antibiotics, antihypertensives, and antiinflammatories. Therefore, understanding the regulation of hOAT1 will provide significant insights into kidney function and dysfunction. We previously established that hOAT1 transport activity is inhibited by activation of protein kinase C (PKC) through accelerating hOAT1 internalization from cell surface into intracellular endosomes and subsequent degradation. We further established that PKC-induced hOAT1 ubiquitination is an important step preceding hOAT1 internalization. In the current study, we identified two closely related E3 ubiquitin ligases, neural precursor cell expressed, developmentally downregulated 4-1 and 4-2 (Nedd4-1 and Nedd4-2), as important regulators for hOAT1: overexpression of Nedd4-1 or Nedd4-2 enhanced hOAT1 ubiquitination, reduced the hOAT1 amount at the cell surface, and suppressed hOAT1 transport activity. In further exploring the relationship among PKC, Nedd4-1, and Nedd4-2, we discovered that PKC-dependent changes in hOAT1 ubiquitination, expression, and transport activity were significantly blocked in cells transfected with the ligase-dead mutant of Nedd4-2 (Nedd4-2/C821A) or with Nedd4-2-specific siRNA to knockdown endogenous Nedd4-2 but not in cells transfected with the ligase-dead mutant of Nedd4-1 (Nedd4-1/C867S) or with Nedd4-1-specific siRNA to knockdown endogenous Nedd4-1. In conclusion, this is the first demonstration that both Nedd4-1 and Nedd4-2 are important regulators for hOAT1 ubiquitination, expression, and function. Yet they play distinct roles, as Nedd4-2 but not Nedd4-1 is a critical mediator for PKC-regulated hOAT1 ubiquitination, expression, and transport activity.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Proteína 1 Transportadora de Ânions Orgânicos/biossíntese , Proteína 1 Transportadora de Ânions Orgânicos/genética , Proteína Quinase C/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Transporte Biológico , Células COS , Chlorocebus aethiops , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Córtex Renal/efeitos dos fármacos , Córtex Renal/metabolismo , Masculino , Mutagênese Sítio-Dirigida , Ubiquitina-Proteína Ligases Nedd4 , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , Transfecção , Ubiquitinação
7.
Am J Physiol Renal Physiol ; 311(2): F320-9, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27226107

RESUMO

Human organic anion transporter 1 (hOAT1), expressed at the basolateral membrane of kidney proximal tubule cells, mediates the active renal secretion of a diverse array of clinically important drugs, including anti-human immunodeficiency virus therapeutics, antitumor drugs, antibiotics, antihypertensives, and anti-inflammatories. We have previously demonstrated that posttranslational modification of hOAT1 by ubiquitination is an important mechanism for the regulation of this transporter. The present study aimed at identifying the ubiquitin ligase for hOAT1 and its mechanism of action. We showed that overexpression of neural precursor cell expressed, developmentally downregulated (Nedd)4-1, an E3 ubiquitin ligase, enhanced hOAT1 ubiquitination, decreased hOAT1 expression at the cell surface, and inhibited hOAT1 transport activity. In contrast, overexpression of the ubiquitin ligase-dead mutant Nedd4-1/C867S was without effects on hOAT1. Furthermore, knockdown of endogenously expressed Nedd4-1 by Nedd4-1-specific small interfering RNA reduced hOAT1 ubiquitination. Immunoprecipitation experiments in cultured cells and rat kidney slices and immunofluorescence experiments in rat kidney slices showed that there was a physical interaction between OAT1 and Nedd4-1. Nedd4-1 contains four protein-protein interacting WW domains. When these WW domains were inactivated by mutating two amino acid residues in each of the four WW domains (Mut-WW1: V210W/H212G, Mut-WW2: V367W/H369G, Mut-WW3: I440W/H442G, and Mut-WW4: I492W/H494G, respectively), only Mut-WW2 and Mut-WW3 significantly lost their ability to bind and to ubiquitinate hOAT1. As a result, Mut-WW2 and Mut-WW3 were unable to suppress hOAT1-mediated transport as effectively as wild-type Nedd4-1. In conclusion, this is the first demonstration that Nedd4-1 regulates hOAT1 ubiquitination, expression, and transport activity through its WW2 and WW3 domains.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Proteína 1 Transportadora de Ânions Orgânicos/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Biotinilação , Células COS , Chlorocebus aethiops , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Células HEK293 , Humanos , Técnicas In Vitro , Rim/metabolismo , Proteínas de Membrana/metabolismo , Mutagênese Sítio-Dirigida , Ubiquitina-Proteína Ligases Nedd4 , Proteína 1 Transportadora de Ânions Orgânicos/genética , Ligação Proteica , Processamento de Proteína Pós-Traducional , Ratos , Ratos Sprague-Dawley , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
8.
Mol Pharm ; 13(2): 621-30, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26651153

RESUMO

Organic anion transporter-3 (OAT3) is a member of the organic anion transporter family that mediates the body disposition of a diverse array of clinically important drugs. We previously demonstrated that activation of protein kinase C (PKC) inhibits OAT3 transport activity by accelerating OAT3 internalization from cell surface into intracellular compartments. In the current study, we established that PKC-induced inhibition of OAT3 transport activity occurred through an enhanced OAT3 ubiquitination, a process catalyzed by an E3 ubiquitin-protein ligase Nedd4-2 (neural precursor cell expressed, developmentally downregulated 4-2). Overexpression of Nedd4-2 enhanced OAT3 ubiquitination, decreased OAT3 expression at the cell surface, and inhibited OAT3 transport activity. In contrast, overexpression of the ubiquitin ligase-dead mutant Nedd4-2/C821A or siRNA knockdown of endogenous Nedd4-2 had opposite effects on OAT3. Furthermore, immunoprecipitation experiments conducted both in culture cells and with rat kidney slices showed that there was a physical interaction between OAT3 and Nedd4-2. In conclusion, our results provided the first evidence that Nedd4-2 is an important regulator for OAT3 ubiquitination, expression, and transport activity.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Rim/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina/metabolismo , Animais , Células COS , Chlorocebus aethiops , Endocitose/fisiologia , Células HEK293 , Humanos , Immunoblotting , Imunoprecipitação , Rim/citologia , Masculino , Ubiquitina-Proteína Ligases Nedd4 , Proteína Quinase C/metabolismo , Transporte Proteico , Ratos , Ratos Sprague-Dawley , Ubiquitinação
9.
Biochem Pharmacol ; 215: 115702, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37487877

RESUMO

Human organic anion transporter 4 (hOAT4), mainly expressed in the kidney and placenta, is essential for the disposition of numerous drugs, toxins, and endogenous substances. Insulin-like growth factor 1 (IGF-1) is a hormone generated in the liver and plays important roles in systemic growth, development, and metabolism. In the current study, we explored the regulatory effects of IGF-1 and downstream signaling on the transport activity, protein expression, and SUMOylation of hOAT4. We showed that IGF-1 significantly increased the transport activity, expression, and maximal transport velocity Vmax of hOAT4 in kidney-derived cells. This stimulatory effect of IGF-1 on hOAT4 activity was also confirmed in cells derived from the human placenta. The increased activity and expression were correlated well with the reduced degradation rate of hOAT4 at the cell surface. Furthermore, IGF-1 significantly increased hOAT4 SUMOylation, and protein kinase B (PKB)-specific inhibitors blocked the IGF-1-induced regulations on hOAT4. In conclusion, our study demonstrates that the hepatic hormone IGF-1 regulates hOAT4 expressed in the kidney and placenta through the PKB signaling pathway. Our results support the remote sensing and signaling theory, where OATs play a central role in the remote communications among distal tissues.


Assuntos
Fator de Crescimento Insulin-Like I , Proteínas Proto-Oncogênicas c-akt , Gravidez , Feminino , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Transdução de Sinais , Hormônios
10.
Biochem Pharmacol ; 208: 115387, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36549459

RESUMO

Organic anion transporter 3 (OAT3), an indispensable basolateral membrane transporter predominantly distributed in the kidney proximal tubules, mediated the systemic clearance of substrates including clinical drugs, nutrients, endogenous and exogenous metabolites, toxins, and critically sustains body homeostasis. Preliminary data in this study showed that classical proteasome inhibitors (e.g., MG132), but not lysosome inhibitors, significantly increased the OAT3 ubiquitination and OAT3-mediated transport of estrone sulfate (ES) in OAT3 stable expressing cells, indicating that proteasome rather than lysosome is involved in the intracellular fate of OAT3. Next, bortezomib and carfilzomib, two FDA-approved and widely applied anticancer agents through selective targeting proteasome, were further used to define the role of inhibiting proteasome in OAT3 regulation and related molecular mechanisms. The results showed that 20S proteasome activity in cell lysates was suppressed with bortezomib and carfilzomib treatment, leading to the increased OAT3 ubiquitination, stimulated transport activity of ES, enhanced OAT3 surface and total expression. The upregulated OAT3 function by proteasome inhibition was attributed to the augment in maximum transport velocity and stability of membrane OAT3. Lastly, in vivo study using Sprague Dawley rats validated that proteasome inhibition using bortezomib induced enhancement of OAT3 ubiquitination and membrane expression in kidney. These data suggest that activity of proteasome but not lysosome could have an impact on the physiological function of OAT3, and proteasome displayed a promising target for OAT3 regulation in vitro and in vivo, and could be used in restoring OAT3 impairment under pathological conditions, avoiding OAT3-associated toxicity and diseases, ensuring drug efficacy and safety.


Assuntos
Antineoplásicos , Complexo de Endopeptidases do Proteassoma , Ratos , Animais , Complexo de Endopeptidases do Proteassoma/metabolismo , Bortezomib/farmacologia , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Ratos Sprague-Dawley , Proteína 1 Transportadora de Ânions Orgânicos
11.
Pharmacol Ther ; 217: 107647, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32758646

RESUMO

The members of the organic anion transporter (OAT) family are mainly expressed in kidney, liver, placenta, intestine, and brain. These transporters play important roles in the disposition of clinical drugs, pesticides, signaling molecules, heavy metal conjugates, components of phytomedicines, and toxins, and therefore critical for maintaining systemic homeostasis. Alterations in the expression and function of OATs contribute to the intra- and inter-individual variability of the therapeutic efficacy and the toxicity of many drugs, and to many pathophysiological conditions. Consequently, the activity of these transporters must be highly regulated to carry out their normal functions. This review will present an update on the recent advance in understanding the cellular and molecular mechanisms underlying the regulation of renal OATs, emphasizing on the post-translational modification (PTM), the crosstalk among these PTMs, and the remote sensing and signaling network of OATs. Such knowledge will provide significant insights into the roles of these transporters in health and disease.


Assuntos
Rim/metabolismo , Transportadores de Ânions Orgânicos/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Animais , Transporte Biológico , Vias de Eliminação de Fármacos , Interações Medicamentosas/fisiologia , Glicosilação , Humanos , Nefropatias/metabolismo , Nefropatias/patologia , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Transportadores de Ânions Orgânicos/genética , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Fosforilação/fisiologia , Polimorfismo Genético , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo
12.
Biochim Biophys Acta Biomembr ; 1861(7): 1293-1301, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31054272

RESUMO

Organic anion transporter 3 (OAT3) plays a vital role in removing a broad array of anionic drugs from kidney, thereby avoiding their possibly toxic side effects in the body. We earlier demonstrated that OAT3 is subjected to a specific type of post-translational modification called SUMOylation. SUMOylation is a dynamic event, where de-SUMOylation is catalyzed by a class of SUMO-specific proteases. In the present investigation, we assessed the role of SUMO-specific protease Senp2 in OAT3 SUMOylation, expression and function. We report here that overexpression of Senp2 in COS-7 cells led to a reduced OAT3 SUMOylation, which correlated well with a decreased OAT3 expression and transport activity. Such phenomenon was not observed in cells overexpressing an inactive mutant of Senp2. Furthermore, transfection of cells with Senp2-specific siRNA to knockdown the endogenous Senp2 resulted in an increased OAT3 SUMOylation, which correlated well with an enhanced OAT3 expression and transport activity. Coimmunoprecipitation experiments showed that Senp2 directly interacted with OAT3 in the kidneys of rats. Together these results provided first demonstration that Senp2 is a significant regulator for OAT3-mediated organic anion/drug transport.


Assuntos
Cisteína Endopeptidases/fisiologia , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Sumoilação/fisiologia , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Cisteína Endopeptidases/genética , Humanos , Rim/citologia , Rim/metabolismo , Masculino , Transportadores de Ânions Orgânicos Sódio-Independentes/fisiologia , RNA Interferente Pequeno , Ratos Sprague-Dawley
13.
Int J Pharm ; 555: 165-174, 2019 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-30453017

RESUMO

Human organic anion transporter 4 (hOAT4) belongs to a class of organic anion transporters that exert critical function in the secretion, absorption, and distribution of numerous drugs in the body, such as anti-viral drugs, anti-cancer therapeutics, antibiotics, antihypertensive medicine, and anti-inflammatory drugs. hOAT4 is richly existent in the kidney and placenta. We previously established that serum- and glucocorticoid-inducible kinases (sgk) stimulate hOAT4 expression and transport activity by abrogating the inhibitory effect of a ubiquitin ligase Nedd4-2. Insulin is one of the upstream signaling molecules for sgk. We therefore investigated the effect of insulin on hOAT4 function. We showed that insulin stimulated hOAT4 expression and transport activity, and the action of insulin was abolished in cells overexpressing Nedd4-2-specific siRNA to knockdown the endogenous Nedd4-2. We further showed that insulin phosphorylated serine 327 on Nedd4-2 and weakened the interaction between hOAT4 and Nedd4-2. Interestingly, in cells overexpressing sgk2, the stimulatory effect of insulin on hOAT4 was diminished. In addition, the stimulatory effect of insulin on hOAT4 was blocked by wortmannin and buparlisib, two PI3K inhibitors. In conclusion, our study demonstrated that insulin stimulates hOAT4 expression and transport activity by abrogating the inhibition effect of Nedd4-2 on the transporter. Moreover, insulin regulates hOAT4 by competing with sgk2 rather than through sgk2.


Assuntos
Insulina/farmacologia , Ubiquitina-Proteína Ligases Nedd4/genética , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Aminopiridinas/farmacologia , Animais , Células COS , Chlorocebus aethiops , Técnicas de Silenciamento de Genes , Humanos , Proteínas Imediatamente Precoces/genética , Morfolinas/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Serina-Treonina Quinases/genética , RNA Interferente Pequeno/administração & dosagem , Wortmanina/farmacologia
14.
AAPS J ; 21(2): 30, 2019 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-30761470

RESUMO

Organic anion transporter 3 (OAT3) plays a vital role in removing a broad variety of anionic drugs from kidney, thus avoiding their possible toxicity in the body. We earlier established that activation of protein kinase C (PKC) enhances OAT3 ubiquitination, which promotes OAT3 internalization from the cell plasma membrane to intracellular endosomes and consequent degradation. As a result, OAT3 expression and transport activity are reduced. In the current study, we discovered that protein kinase A (PKA) had an opposite effect to PKC on the regulation of OAT3. We showed that activation of PKA by Bt2-cAMP stimulated OAT3 transport activity, which was largely caused by an enhanced plasma membrane expression of the transporter, kinetically reflected as an augmented maximal transport velocity Vmax without notable alteration in substrate-binding affinity Km. Additionally, we showed that PKA activation accelerated the rate of OAT3 recycling from intracellular compartments to the plasma membrane and decelerated the rate of OAT3 degradation. We further showed that OAT3 is subjected to post-translational modification by SUMO-2 and SUMO-3 not by SUMO-1. PKA activation enhanced OAT3 SUMOylation, which was accompanied by a reduced OAT3 ubiquitination. Finally, insulin-like growth factor 1 significantly stimulated OAT3 transport activity and SUMOylation through PKA signaling pathway. In conclusion, this is the first demonstration that PKA stimulated OAT3 expression and transport activity by altering the trafficking kinetics of OAT3 possibly through the crosstalk between SUMOylation and ubiquitination. Our studies are consistent with a remote sensing and signaling model for transporters (Wu et al. in Mol Pharmacol. 79(5):795-805, 2011).


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Ubiquitinas/metabolismo , Animais , Células COS , Chlorocebus aethiops , Fator de Crescimento Insulin-Like I/metabolismo , Proteína SUMO-1/metabolismo , Sumoilação , Transfecção , Enzimas de Conjugação de Ubiquitina/metabolismo
15.
Oncol Lett ; 17(6): 4859-4864, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31186693

RESUMO

Correlation of p16 and cyclin D1 expression with the incidence and prognosis of cardiac carcinoma was investigated. Thirty-six patients with cardiac carcinoma treated in The Second Affiliated Hospital of Zhengzhou University were selected. After the radical operation of cardiac carcinoma, carcinoma tissues were taken, and the corresponding para-carcinoma tissues were used as controls. p16 and cyclin D1 messenger ribonucleic acid (mRNA) and protein expression in cardiac carcinoma tissues and para-carcinoma tissues were detected via quantitative polymerase chain reaction (qPCR) and western blot analysis. The survival time and pathological conditions of patients with cardiac carcinoma were recorded in detail, and correlation of p16 and cyclin D1 with incidence and prognosis of cardiac carcinoma was studied. In cardiac carcinoma tissues, the p16 mRNA and protein expression levels were significantly lower than those in para-carcinoma tissues (P<0.01), but the cyclin D1 mRNA and protein expression levels were significantly higher than those in para-carcinoma tissues (P<0.01). The expression of p16 and cyclin D1 protein had correlation with the tumor size, lymph node metastasis and tumor-node-metastasis stage of cardiac carcinoma (P<0.01). There was a negative correlation between expression of p16 and cyclin D1 in cardiac carcinoma (P<0.01). According to Kaplan-Meier survival analysis, the survival rate of patients with high expression of p16 was obviously higher than that of patients with low expression of p16 (P<0.01), while the survival rate of patients with high expression of cyclin D1 was obviously lower than that of patients with low expression of cyclin D1 (P<0.01). Both p16 and cyclin D1 are closely related to the incidence and prognosis of cardiac carcinoma, which may become indexes for the incidence and prognosis of cardiac carcinoma.

16.
Discov Med ; 25(137): 99-112, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29641972

RESUMO

Myeloid-derived suppressor cells (MDSCs) are thought to play a critical immunosuppressive role in tumorigenesis. In this project, we aimed to investigate subset alteration of MDSCs in gastric cancer (GC), and the effects of epirubicin (EPI) and paclitaxel (TAX) on MDSCs. The frequencies of MDSC subsets in peripheral blood were observed by using flow cytometry after treatment with EPI- or TAX- based chemotherapy in GC patients. After treatment with EPI or TAX in vitro, the subsets, apoptosis, cell cycle, and MAPK and NF-κB protein expressions of mouse bone marrow MDSCs were analyzed. The frequency of MDSCs in the peripheral blood of GC patients was higher than that in healthy controls. Granulocyte-type MDSCs (G-MDSCs) were significantly more than monocyte-type MDSCs (M-MDSCs) in GC patients. The frequencies of MDSC subsets in the peripheral blood decreased after EPI- or TAX-based chemotherapy. High levels of MDSC subsets were correlated with low cancer differentiation degree. High level of M-MDSCs was related to lymph node metastasis, and was negatively correlated with the overall survival of GC patients. After treatment with EPI or TAX, levels of mouse bone marrow MDSC subsets decreased significantly in vitro. Arg-1 secretion and expression of total and phosphorylated MAPK and NF-κB by MDSCs decreased. EPI or TAX decreases the levels of MDSCs, inhibits the proliferation and function of MDSCs in vitro, and induces their apoptosis via the MAPK and NF-κB signaling pathways.


Assuntos
Apoptose/efeitos dos fármacos , Epirubicina/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Células Supressoras Mieloides/metabolismo , Paclitaxel/farmacologia , Neoplasias Gástricas/metabolismo , Idoso , Animais , Feminino , Citometria de Fluxo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , Células Supressoras Mieloides/patologia , Proteínas de Neoplasias/metabolismo , Prognóstico , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/patologia
17.
Biochem Pharmacol ; 102: 120-129, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26740304

RESUMO

Human organic anion transporter 4 (hOAT4) belongs to a family of organic anion transporters that play critical roles in the body disposition of clinically important drugs, including anti-viral therapeutics, anti-cancer drugs, antibiotics, antihypertensives, and anti-inflammatories. hOAT4 is abundantly expressed in the kidney and placenta. In the current study, we examined the regulation of hOAT4 by serum- and glucocorticoid-inducible kinase 2 (sgk2) in the kidney COS-7 cells. We showed that sgk2 stimulated hOAT4 transport activity. Such stimulation mainly resulted from an increased cell surface expression of the transporter, kinetically revealed as an increased maximal transport velocity Vmax without significant change in substrate-binding affinity Km. We further showed that regulation of hOAT4 activity by sgk2 was mediated by ubiquitin ligase Nedd4-2. Overexpression of Nedd4-2 enhanced hOAT4 ubiquitination, and inhibited hOAT4 transport activity, whereas overexpression of ubiquitin ligase-dead mutant Nedd4-2/C821A or siRNA knockdown of endogenous Nedd4-2 had opposite effects on hOAT4. Our co-immunoprecipitation experiment revealed that sgk2 weakened the association between hOAT4 and Nedd4-2. In conclusion, our study demonstrated for the first time that sgk2 stimulated hOAT4 transport activity by abrogating the inhibitory effect of Nedd4-2 on the transporter.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Proteínas Imediatamente Precoces/fisiologia , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Células COS , Chlorocebus aethiops , Humanos , Ubiquitina-Proteína Ligases Nedd4 , Transporte Proteico/fisiologia
18.
Int J Biochem Mol Biol ; 7(1): 11-8, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27335682

RESUMO

Human organic anion transporter 4 (hOAT4) belongs to a family of organic anion transporters which play critical roles in the body disposition of clinically important drugs. hOAT4 is expressed in the kidney and placenta. In the current study, we examined the inhibitory effects of 101 anticancer drugs from a clinical drug library on hOAT4 transport activity. The studies were carried out in hOAT4-expressing human kidney HEK-293 cells and human placenta BeWo cells. Among these drugs, only chlorambucil and cabazitaxel demonstrated more than 50% cis-inhibitory effect on hOAT4-mediated uptake of (3)H-labeled estrone sulfate, a prototypical substrate for the transporter. The IC50 values for chlorambucil and cabazitaxel were 44.28 and 3.5 µM respectively. Dixon plot analysis revealed that inhibition by chlorambucil was competitive with a Ki = 55.73 µM whereas inhibition by cabazitaxel was non-competitive with a Ki = 1.78 µM. Our results demonstrated that chlorambucil and cabazitaxel were inhibitors of hOAT4. Furthermore, by comparing our data with clinically relevant exposures of these drugs, we conclude that the propensity for chlorambucil and cabazitaxel to cause drug-drug interaction through inhibition of hOAT4 is low.

19.
Exp Ther Med ; 10(4): 1591-1601, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26622532

RESUMO

The aim of the present study was to validate, and if necessary update, a predictive model previously developed using a classification and regression tree (CART) algorithm for predicting successful extubation (ES) using a new cohort. This prospective cohort study enrolled adults admitted to 10 intensive care units, who had successfully passed a spontaneous breathing trial (SBT) and were considered ready for extubation. After extubation, the patients were followed up for 48 h. The primary outcome measure was ES, defined as the ability to maintain spontaneous unassisted breathing for >48 h after extubation. The 3-factor CART model was applied to patients in this cohort. The predicted probability of ES for each patient in this validation cohort was calculated based on the original CART model using the Laplace correction method. The performance was assessed by discrimination and calibration. A decision curve analysis was used assess the clinical net benefit (NB). Extubation failure (EF) occurred in 90/530 patients (17%). Among the 90 patients, 72 (13.6%) were reintubated, while 18 patients remained on rescue noninvasive ventilation within 48 h after extubation. The original CART model showed high discrimination but only moderate calibration with predicted probabilities that were systematically lower than expected. The original CART model was updated, and the updated model preserved excellent discrimination (area under the receiver operating characteristic curve, 0.91; 95% confidence interval, 0.87 to 0.93), but exhibited near-perfect calibration (calibration slope, 1; intercept, 0). Between threshold probabilities of 50 and 80%, the NB of using this updated model is significantly improved compared with the current strategy. The updated CART model may be used to estimate the predicted probability of ES after a successful SBT for individual patients. Applying this model appears to produce a substantial clinical consequence with regard to potential reduction in unexpected EFs.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA