Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Yakugaku Zasshi ; 144(4): 397-402, 2024.
Artigo em Japonês | MEDLINE | ID: mdl-38556314

RESUMO

Cysts are abnormal fluid-filled sacs found in various human organs, including the liver. Liver cysts can be associated with known causes such as parasite infections and gene mutations, or simply aging. Among these causes, simple liver cysts are often found in elderly people. While they are generally benign, they may occasionally grow but rarely shrink with age, indicating their clear association with aging. However, the mechanism behind the formation of simple liver cysts has not been thoroughly investigated. Recently, we have generated transgenic mice that specifically overexpress fibroblast growth factor (FGF)18 in hepatocytes. These mice exhibit severe liver fibrosis without inflammation and spontaneously develop liver cysts that grow with age. Our findings suggest that simple liver cysts can be induced by fibrosis accompanied by sterile inflammation or injury, whereas fibrosis accompanied by severe inflammation or injury may lead to cirrhosis. We also discuss the detrimental effects of disease- and aging-associated fibrosis in various organs, such as the heart, lungs, and kidneys. Additionally, we provide a brief summary of the two currently approved anti-fibrotic drugs for idiopathic pulmonary fibrosis, nintedanib and pirfenidone, as well as their possibility of future expansion of application toward other fibrotic diseases.


Assuntos
Cistos , Pulmão , Humanos , Camundongos , Animais , Idoso , Pulmão/metabolismo , Fibrose , Inflamação , Envelhecimento/genética , Cistos/metabolismo , Cistos/patologia
2.
Cureus ; 16(1): e52506, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38371142

RESUMO

Nirmatrelvir/ritonavir is a treatment for COVID-19 consisting of nirmatrelvir, which has anti-SARS-CoV-2 activity, and ritonavir, a booster to maintain blood levels. Ritonavir is known to be a potent inhibitor of cytochrome P450 3A (CYP3A), and interactions with CYP3A-metabolized drugs, such as the immunosuppressant tacrolimus, can be problematic. Ritonavir's inhibition of CYP3A is irreversible due to covalent binding, and its inhibitory effects are expected to persist until replaced by new CYP3A. Here, we report a case where the combination of nirmatrelvir/ritonavir and tacrolimus resulted in toxic tacrolimus blood levels. A patient on tacrolimus for systemic lupus erythematosus (SLE) developed COVID-19 and was prescribed nirmatrelvir/ritonavir. After starting the combination of nirmatrelvir/ritonavir and tacrolimus, the patient's tacrolimus blood levels became abnormally high, leading to the discontinuation of these drugs due to symptoms of tacrolimus toxicity. Even after ritonavir blood levels had fallen below the detection limit, the decline in tacrolimus blood levels was delayed. The CYP3A inhibition of ritonavir persists even when its blood concentration decreases, emphasizing the need for careful consideration of concomitant medications before starting nirmatrelvir/ritonavir therapy. Adjustments or discontinuation may be necessary.

3.
Nat Commun ; 14(1): 6304, 2023 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-37813881

RESUMO

Liver fibrosis results from chronic liver injury triggered by factors such as viral infection, excess alcohol intake, and lipid accumulation. However, the mechanisms underlying liver fibrosis are not fully understood. Here, we demonstrate that the expression of fibroblast growth factor 18 (Fgf18) is elevated in mouse livers following the induction of chronic liver fibrosis models. Deletion of Fgf18 in hepatocytes attenuates liver fibrosis; conversely, overexpression of Fgf18 promotes liver fibrosis. Single-cell RNA sequencing reveals that overexpression of Fgf18 in hepatocytes results in an increase in the number of Lrat+ hepatic stellate cells (HSCs), thereby inducing fibrosis. Mechanistically, FGF18 stimulates the proliferation of HSCs by inducing the expression of Ccnd1. Moreover, the expression of FGF18 is correlated with the expression of profibrotic genes, such as COL1A1 and ACTA2, in human liver biopsy samples. Thus, FGF18 promotes liver fibrosis and could serve as a therapeutic target to treat liver fibrosis.


Assuntos
Células Estreladas do Fígado , Cirrose Hepática , Camundongos , Animais , Humanos , Células Estreladas do Fígado/metabolismo , Cirrose Hepática/patologia , Fígado/metabolismo , Fibrose , Proliferação de Células
4.
Biochem Biophys Res Commun ; 675: 71-77, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37453260

RESUMO

Fibroblast growth factor 18 (FGF18) is elevated in several human cancers, such as gastrointestinal and ovarian cancers, and stimulates the proliferation of tumor cells. This suggests that FGF18 may be a promising candidate biomarker in cancer patients. However, the lack of a high-sensitivity enzyme-linked immunosorbent assay (ELISA) does not permit testing of this possibility. In this study, we generated monoclonal antibodies against human FGF18 and developed a high-sensitivity ELISA to measure human FGF18 at concentrations as low as 10 pg/mL. Of the eight tumor cell lines investigated, we detected human FGF18 in culture supernatants from four tumor cell lines, including HeLa, OVCAR-3, BxPC-3, and SW620 cells, albeit the production levels were relatively low in the latter two cell lines. Moreover, the in-house ELISA could detect murine FGF18 in sera from mice overexpressing murine Fgf18 in hepatocytes, although the sensitivity in detecting murine FGF18 was relatively low. This FGF18 ELISA could be a valuable tool to validate FGF18 as a potential biomarker for cancer patients and to test the contribution of FGF18 for various disease models invivo and in vitro.


Assuntos
Apoptose , Neoplasias Ovarianas , Humanos , Camundongos , Animais , Feminino , Linhagem Celular Tumoral , Neoplasias Ovarianas/patologia , Fatores de Crescimento de Fibroblastos/metabolismo , Ensaio de Imunoadsorção Enzimática
5.
Ther Drug Monit ; 45(5): 653-659, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-36645709

RESUMO

BACKGROUND: Perampanel (PER) is an oral antiepileptic drug and its concomitant use with carbamazepine (CBZ) leads to decreased PER concentrations. However, the magnitude of its influence may vary, depending on the dynamics of the enzyme induction properties of CBZ. This study aimed to develop a population pharmacokinetic (PPK) model considering the dynamics of enzyme induction and evaluate the effect of CBZ on PER pharmacokinetics. METHODS: We retrospectively collected data on patient background, laboratory tests, and prescribed drugs from electronic medical records. We developed 2 PPK models incorporating the effect of CBZ-mediated enzyme induction to describe time-concentration profiles of PER using the following different approaches: (1) treating the concomitant use of CBZ as a categorical covariate (empirical PPK model) and (2) incorporating the time-course of changes in the amount of enzyme by CBZ-mediated induction (semimechanistic PPK model). The bias and precision of the predictions were investigated by calculating the mean error, mean absolute error, and root mean squared error. RESULTS: A total of 133 PER concentrations from 64 patients were available for PPK modelling. PPK analyses showed that the co-administration of CBZ increased the clearance of PER. Goodness-of-fit plots indicated a favorable description of the observed data and low bias. The mean error, mean absolute error, and root mean square error values based on the semimechanistic model were smaller than those obtained using the empirical PPK model for predicting PER concentrations in patients with CBZ. CONCLUSIONS: We developed 2 PPK models to describe PER pharmacokinetics based on different approaches, using electronic medical record data. Our PPK models support the use of PER in clinical practice.


Assuntos
Carbamazepina , Epilepsia , Humanos , Estudos Retrospectivos , Indução Enzimática , Carbamazepina/uso terapêutico , Epilepsia/tratamento farmacológico , Benzodiazepinas/uso terapêutico , Interações Medicamentosas
6.
Toxicol Appl Pharmacol ; 449: 116112, 2022 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-35688184

RESUMO

Neurotoxicity is one of the major side effects caused by calcineurin inhibitors such as tacrolimus in clinical practice. The underlying mechanisms remain unclear, and no potential protective agents have been identified yet. Here, we aimed to investigate tacrolimus-induced neurotoxicity and assess the protective effects of ibudilast, a nonselective phosphodiesterase inhibitor with neuroprotective effects, against tacrolimus-induced neurotoxicity. An in vitro assay of human neuroblastoma SH-SY5Y cells showed that ibudilast reduced tacrolimus-induced cell death. Subsequently, using in vivo studies, we assessed the pathological mechanism of neurotoxicity and evaluated the protective effect of ibudilast. Wistar rats were subcutaneously administered tacrolimus (2.5 or 5.0 mg/kg/day) for 14 d, and ibudilast (7.5 mg/kg/day) was intraperitoneally administered once a day beginning 2 d prior to tacrolimus (5 mg/kg/day) administration. We observed that ibudilast significantly reduced the tacrolimus-induced neurotoxic events. From the assessment of excised brains, we found that tacrolimus was penetrated to brain and the brain concentration was correlated with the neurotoxicity-score, although ibudilast had no effect on this pharmacokinetics. Tacrolimus-induced neuronal damage was histopathologically evaluated using Nissl and TUNEL staining, where only the cerebral cortex and CA1 region in hippocampus exhibited neuronal death, but not the CA3 region, dendrite gyrus, and cerebellum. Co-administration of ibudilast significantly attenuated these histopathological changes. In conclusion, these results suggest that tacrolimus translocation into the brain and neuronal damage in the cerebral cortex and CA1 are the underlying mechanisms of tacrolimus-induced neurotoxicity and that ibudilast could be a protective agent against this adverse event.


Assuntos
Neuroblastoma , Fármacos Neuroprotetores , Síndromes Neurotóxicas , Animais , Humanos , Fármacos Neuroprotetores/farmacologia , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/prevenção & controle , Piridinas , Ratos , Ratos Wistar , Tacrolimo/toxicidade
7.
Ther Drug Monit ; 44(4): 592-596, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35149666

RESUMO

BACKGROUND: Gilteritinib, a novel oral tyrosine kinase inhibitor, is used to treat acute myeloid leukemia (AML) with FMS-like tyrosine kinase-3 (FLT3) mutations. Therapeutic drug monitoring (TDM) of gilteritinib is important for improving clinical outcomes and ensuring safety. Therefore, this study aimed to develop a simplified method for quantifying gilteritinib in human plasma using liquid chromatography-tandem mass spectrometry. METHODS: Liquid chromatography was performed by using an Acquity BEH C18 column (50 mm × 2.1 mm, 1.7 µm) and a gradient elution with 0.1% formic acid in water (A) and acetonitrile (B). Detection was performed by using a Shimadzu tandem mass spectrometer through multiple reaction monitoring in the positive-ion mode. RESULTS: The developed method enabled quantification of gilteritinib in 4 minutes and was validated by evaluating selectivity, calibration curve (10-1000 ng/mL, r 2 > 0.99), a lower limit of quantification (LLOQ), accuracy (overall bias -4.2% to 1.9%), precision (intraday CV ≤ 7.9%; interday CV ≤ 13.6%), carryover, recovery, matrix effect, dilution integrity, and stability according to the US Food and Drug Administration (FDA) guidelines. This method was successfully applied to the TDM of gilteritinib trough concentrations in 3 patients with AML. CONCLUSIONS: The developed method fulfilled the FDA guideline criteria and can easily be implemented to facilitate TDM in patients receiving gilteritinib in a clinical setting.


Assuntos
Leucemia Mieloide Aguda , Espectrometria de Massas em Tandem , Compostos de Anilina , Cromatografia Líquida de Alta Pressão/métodos , Cromatografia Líquida , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Limite de Detecção , Mutação , Pirazinas , Reprodutibilidade dos Testes , Espectrometria de Massas em Tandem/métodos , Tirosina Quinase 3 Semelhante a fms/genética , Tirosina Quinase 3 Semelhante a fms/uso terapêutico
8.
Chem Pharm Bull (Tokyo) ; 69(7): 646-651, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34193713

RESUMO

With the aim of studying the pharmacokinetics of letermovir, which is a newly developed antiviral agent for human cytomegalovirus, a rapid and simple ultra-performance liquid chromatography coupled with mass spectrometry (UPLC/MS) method was developed and validated for the quantification of letermovir in human plasma. Separation was performed in reverse phase mode using an ACQUITY UPLC BEH C18 column (130 Å, 1.7 µm, 2.1 × 50 mm) at a flow rate of 0.3 mL/min, 10 mM ammonium acetate-0.1% formic acid solution as mobile phase A, and acetonitrile as mobile phase B with a gradient elution. The method was validated over a linear range of 10-1000 ng/mL with a coefficient of determination (R2) >0.99 using weighted linear regression analysis. The intra- and inter-assay accuracy (nominal%) and precision (relative standard deviation%) were within ±15 and ≤15%, respectively. The specificity, recovery, matrix effect, stability, and dilution integrity of this method were also within acceptable limits. This method could be useful in studying the pharmacokinetics and pharmacodynamics, as well as performing the therapeutic drug monitoring of letermovir.


Assuntos
Acetatos/sangue , Cromatografia Líquida de Alta Pressão/métodos , Quinazolinas/sangue , Espectrometria de Massas em Tandem/métodos , Acetatos/farmacocinética , Antivirais/sangue , Antivirais/farmacocinética , Meia-Vida , Humanos , Limite de Detecção , Quinazolinas/farmacocinética , Reprodutibilidade dos Testes
9.
Commun Biol ; 4(1): 80, 2021 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33469115

RESUMO

Mind bomb 2 (MIB2) is an E3 ligase involved in Notch signalling and attenuates TNF-induced apoptosis through ubiquitylation of receptor-interacting protein kinase 1 (RIPK1) and cylindromatosis. Here we show that MIB2 bound and conjugated K48- and K63-linked polyubiquitin chains to a long-form of cellular FLICE-inhibitory protein (cFLIPL), a catalytically inactive homologue of caspase 8. Deletion of MIB2 did not impair the TNF-induced complex I formation that mediates NF-κB activation but significantly enhanced formation of cytosolic death-inducing signalling complex II. TNF-induced RIPK1 Ser166 phosphorylation, a hallmark of RIPK1 death-inducing activity, was enhanced in MIB2 knockout cells, as was RIPK1 kinase activity-dependent and -independent apoptosis. Moreover, RIPK1 kinase activity-independent apoptosis was induced in cells expressing cFLIPL mutants lacking MIB2-dependent ubiquitylation. Together, these results suggest that MIB2 suppresses both RIPK1 kinase activity-dependent and -independent apoptosis, through suppression of RIPK1 kinase activity and ubiquitylation of cFLIPL, respectively.


Assuntos
Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Apoptose/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Células HCT116 , Células HEK293 , Células HeLa , Humanos , NF-kappa B/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo , Ubiquitina-Proteína Ligases/fisiologia , Ubiquitinação/efeitos dos fármacos
10.
Yakugaku Zasshi ; 140(8): 1035-1040, 2020.
Artigo em Japonês | MEDLINE | ID: mdl-32741861

RESUMO

For many of the novel antiepileptics, immunoassays, used for routine therapeutic drug monitoring (TDM), cannot be used. We could monitor eight novel antiepileptics using an LC/MS method since July 2017. The purpose of this study was to evaluate the significant changes associated with the transition from outsourcing to in-hospital monitoring of novel antiepileptics. The number of measurements of novel antiepileptics was significantly increased during the first (p<0.01) and second (p<0.001) years of in-hospital monitoring as compared to that one year prior to in-hospital monitoring which was outsourced. The proportion of measurements of novel antiepileptics to all antiepileptics was 19.7%, 31.1%, and 38.4% during outsourcing, and first, and second years of in-hospital monitoring, respectively. The measurement cost was significantly reduced during the first (p<0.001) and second (p<0.001) years of in-hospital monitoring as compared to that during outsourcing. In addition, the revenue from TDM of antiepileptic drugs was significantly increased during the first (p<0.05) and second (p<0.01) years of in-hospital monitoring as compared with that during outsourcing. In conclusion, the switch from outsourcing to in-hospital monitoring led to an increase in the number of orders, a reduction in the measurement-related expenses of novel antiepileptics, and an increase in the revenue from TDM of antiepileptic drugs, which could promote the proper use of novel antiepileptics through TDM.


Assuntos
Anticonvulsivantes , Monitoramento de Medicamentos/métodos , Monitoramento de Medicamentos/estatística & dados numéricos , Serviços Terceirizados/estatística & dados numéricos , Serviço de Farmácia Hospitalar/estatística & dados numéricos , Cromatografia Líquida , Monitoramento de Medicamentos/economia , Humanos , Renda/estatística & dados numéricos , Espectrometria de Massas , Fatores de Tempo
11.
Ther Drug Monit ; 42(3): 400-406, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31568181

RESUMO

BACKGROUND: Therapeutic drug monitoring of tacrolimus is necessary for appropriate dose adjustment for a successful immunosuppressive therapy. Several commercial immunoassays are available for tacrolimus measurements. This study aimed at simultaneously evaluating the analytical performances of 4 such immunoassays, using liquid chromatography-tandem mass spectrometry (LC-MS/MS) as a standard. For the first time, cross-reactivity to tacrolimus metabolites was assessed at concentrations frequently observed in clinical settings, as opposed to the higher concentrations tested by assay manufacturers. METHODS: An affinity column-mediated immunoassay (ACMIA), using upgraded flex reagents; released in 2015, a chemiluminescence immunoassay (CLIA), an electrochemiluminescence immunoassay (ECLIA), and a latex agglutination turbidimetric immunoassay (LTIA) were evaluated using frozen whole blood samples collected from transplantation patients. Cross-reactivities to 3 major tacrolimus metabolites (13-O-demethyl-tacrolimus [M-I], 31-O-demethyl-tacrolimus [M-II], and 15-O-demethyl-tacrolimus [M-III]) were evaluated. RESULTS: Each immunoassay correlated well with LC-MS/MS, and the Pearson's correlation coefficients (R) were 0.974, 0.977, 0.978, and 0.902 for ACMIA, CLIA, ECLIA, and LTIA, respectively. Using Bland-Altman difference plots to compare the immunoassays with LC-MS/MS, the calculated average biases were -6.73%, 6.07%, 7.46%, and 12.27% for ACMIA, CLIA, ECLIA, and LTIA, respectively. The cross-reactivities of ACMIA to the tacrolimus metabolites M-II and M-III were 81% and 78%, respectively, when blood was spiked at 2 ng/mL, and 94% and 68%, respectively, when it was spiked at 5 ng/mL. CONCLUSIONS: Each immunoassay was useful, but had its own characteristics. ACMIA cross-reactivities to M-II and M-III were much higher than the respective 18% and 15% reported on its package insert, suggesting that cross-reactivity should be examined at clinically relevant concentrations.


Assuntos
Monitoramento de Medicamentos/métodos , Imunoensaio/métodos , Imunossupressores/sangue , Tacrolimo/sangue , Cromatografia Líquida , Monitoramento de Medicamentos/normas , Humanos , Tacrolimo/análogos & derivados , Espectrometria de Massas em Tandem , Transplante/métodos
12.
Cell Death Dis ; 10(6): 449, 2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31209199

RESUMO

Nonalcoholic steatohepatitis (NASH) is a metabolic liver disease that progresses from simple steatosis to the disease state of inflammation and fibrosis. Previous studies suggest that apoptosis and necroptosis may contribute to the pathogenesis of NASH, based on several murine models. However, the mechanisms underlying the transition of simple steatosis to steatohepatitis remain unclear, because it is difficult to identify when and where such cell deaths begin to occur in the pathophysiological process of NASH. In the present study, our aim is to investigate which type of cell death plays a role as the trigger for initiating inflammation in fatty liver. By establishing a simple method of discriminating between apoptosis and necrosis in the liver, we found that necrosis occurred prior to apoptosis at the onset of steatohepatitis in the choline-deficient, ethionine-supplemented (CDE) diet model. To further investigate what type of necrosis is involved in the initial necrotic cell death, we examined the effect of necroptosis and ferroptosis inhibition by administering inhibitors to wild-type mice in the CDE diet model. In addition, necroptosis was evaluated using mixed lineage kinase domain-like protein (MLKL) knockout mice, which is lacking in a terminal executor of necroptosis. Consequently, necroptosis inhibition failed to block the onset of necrotic cell death, while ferroptosis inhibition protected hepatocytes from necrotic death almost completely, and suppressed the subsequent infiltration of immune cells and inflammatory reaction. Furthermore, the amount of oxidized phosphatidylethanolamine, which is involved in ferroptosis pathway, was increased in the liver sample of the CDE diet-fed mice. These findings suggest that hepatic ferroptosis plays an important role as the trigger for initiating inflammation in steatohepatitis and may be a therapeutic target for preventing the onset of steatohepatitis.


Assuntos
Ferroptose , Fígado/patologia , Hepatopatia Gordurosa não Alcoólica/patologia , Animais , Apoptose/efeitos dos fármacos , Tetracloreto de Carbono/toxicidade , Cromanos/farmacologia , Citocinas/metabolismo , Dieta , Etionina , Ferroptose/efeitos dos fármacos , Hepatite/imunologia , Hepatite/metabolismo , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Quelantes de Ferro/farmacologia , Fígado/citologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Necroptose/efeitos dos fármacos , Necrose , Hepatopatia Gordurosa não Alcoólica/induzido quimicamente , Hepatopatia Gordurosa não Alcoólica/metabolismo
14.
J Cell Biol ; 217(4): 1287-1301, 2018 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-29507125

RESUMO

In mammalian pancreatic ß cells, the IRE1α-XBP1 pathway is constitutively and highly activated under physiological conditions. To elucidate the precise role of this pathway, we constructed ß cell-specific Ire1α conditional knockout (CKO) mice and established insulinoma cell lines in which Ire1α was deleted using the Cre-loxP system. Ire1α CKO mice showed the typical diabetic phenotype including impaired glycemic control and defects in insulin biosynthesis postnatally at 4-20 weeks. Ire1α deletion in pancreatic ß cells in mice and insulinoma cells resulted in decreased insulin secretion, decreased insulin and proinsulin contents in cells, and decreased oxidative folding of proinsulin along with decreased expression of five protein disulfide isomerases (PDIs): PDI, PDIR, P5, ERp44, and ERp46. Reconstitution of the IRE1α-XBP1 pathway restored the proinsulin and insulin contents, insulin secretion, and expression of the five PDIs, indicating that IRE1α functions as a key regulator of the induction of catalysts for the oxidative folding of proinsulin in pancreatic ß cells.


Assuntos
Endorribonucleases/metabolismo , Células Secretoras de Insulina/enzimologia , Insulina/metabolismo , Proinsulina/metabolismo , Dobramento de Proteína , Proteínas Serina-Treonina Quinases/metabolismo , Proteína 1 de Ligação a X-Box/metabolismo , Fator 6 Ativador da Transcrição/metabolismo , Animais , Sítios de Ligação , Glicemia/metabolismo , Linhagem Celular Tumoral , Diabetes Mellitus/sangue , Diabetes Mellitus/enzimologia , Diabetes Mellitus/genética , Endorribonucleases/deficiência , Endorribonucleases/genética , Insulina/genética , Insulinoma/enzimologia , Insulinoma/genética , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Knockout , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Oxirredução , Neoplasias Pancreáticas/enzimologia , Neoplasias Pancreáticas/genética , Fosforilação , Proinsulina/química , Proinsulina/genética , Regiões Promotoras Genéticas , Isomerases de Dissulfetos de Proteínas/genética , Isomerases de Dissulfetos de Proteínas/metabolismo , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais , Tiorredoxinas/genética , Tiorredoxinas/metabolismo , Proteína 1 de Ligação a X-Box/genética , eIF-2 Quinase/metabolismo
15.
Cell Struct Funct ; 42(1): 61-70, 2017 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-28321016

RESUMO

IRE1α plays an important role in the unfolded protein response (UPR), which is activated by the accumulation of unfolded proteins in the endoplasmic reticulum. 4µ8C, a well-known inhibitor of IRE1α RNase activity, is commonly used to analyze IRE1α function during ER stress in cultured mammalian cells. However, the off-target effects of 4µ8C remain elusive. Pancreatic ß-cells synthesize a large amount of insulin in response to high glucose stimulation, and IRE1α plays an important role in insulin secretion from pancreatic ß-cells. Here, to analyze the role of IRE1α in pancreatic ß-cells, we examined insulin secretion after 4µ8C treatment. Although 4µ8C inhibited insulin secretion within 2 hr, neither insulin synthesis nor maturation was inhibited by 4µ8C under the same conditions. This result prompted us to examine the precise effects of 4µ8C on insulin secretion in pancreatic ß-cells. Unexpectedly, with just 5 min of treatment, 4µ8C blocked insulin secretion in cultured pancreatic ß-cells as well as in pancreatic islets. Furthermore, insulin secretion was prevented by 4µ8C, even in pancreatic ß-cells lacking the IRE1α RNase domain, suggesting that 4µ8C blocked the late stage of the insulin secretory process, independent of the IRE1α-XBP1 pathway. Our results indicate that 4µ8C has an off-target effect on insulin secretion in pancreatic ß-cells. These findings inform the researchers in the field that the use of 4µ8C requires the special consideration for the future studies.Key words: 4µ8C, XBP1, insulin, IRE1α, pancreatic ß-cells.


Assuntos
Aldeídos/farmacologia , Endorribonucleases/metabolismo , Himecromona/análogos & derivados , Insulina/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Linhagem Celular , Endocitose/efeitos dos fármacos , Endorribonucleases/química , Himecromona/farmacologia , Insulina/biossíntese , Secreção de Insulina , Masculino , Camundongos , Domínios Proteicos , Proteínas Serina-Treonina Quinases/química , Splicing de RNA/efeitos dos fármacos , RNA Mensageiro/genética , Fatores de Tempo , Proteína 1 de Ligação a X-Box/genética
16.
Int J Hematol ; 105(3): 361-368, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27822915

RESUMO

The aim of this retrospective study was to identify variable factors affecting tacrolimus blood concentration during the switch from continuous intravenous infusion to twice-daily oral administration in allogeneic hematopoietic stem cell transplant recipients (n = 73). The blood concentration/dose ratio of tacrolimus immediately before the change from continuous infusion (C/Div) was compared with that between 3 and 5 days after the change to oral administration (C/Dpo). Median (C/Dpo)/(C/Div) was 0.21 (range 0.04-0.58). Multiple regression analysis showed that concomitant use of oral itraconazole or voriconazole significantly increased the (C/Dpo)/(C/Div) of tacrolimus (p = 0.002), probably owing to the inhibition of enterohepatic cytochrome P450 3A4. In addition, 5 of 18 (28%) patients who had the lowest quartile (C/Dpo)/(C/Div) values developed acute graft-versus-host-disease (GVHD), which was significantly higher than in others [5 of 55 (9%) patients, p = 0.045]. Although the switch from intravenous to oral administration at a ratio of 1:5 appeared to be appropriate, a lower conversion ratio was suitable in patients taking oral itraconazole or voriconazole. In patients whose blood concentration decreases after the switch, the development of GVHD should be monitored and tacrolimus dosage should be readjusted to maintain an appropriate blood concentration.


Assuntos
Transplante de Células-Tronco Hematopoéticas/métodos , Tacrolimo/administração & dosagem , Administração Oral , Adulto , Idoso , Aloenxertos , Quimioterapia Combinada , Feminino , Doença Enxerto-Hospedeiro/diagnóstico , Humanos , Infusões Intravenosas , Itraconazol/uso terapêutico , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Tacrolimo/sangue , Voriconazol/uso terapêutico
17.
Yakugaku Zasshi ; 136(6): 817-25, 2016.
Artigo em Japonês | MEDLINE | ID: mdl-27252061

RESUMO

The endoplasmic reticulum (ER) is an organelle in which newly synthesized secretory and membrane proteins are folded and assembled. Various stresses cause the accumulation of unfolded or misfolded proteins in the ER, resulting in ER dysfunction. This condition is termed ER stress. To cope with ER stress, cells activate a signaling pathway termed the unfolded protein response (UPR). Recently, accumulating evidence suggests that the UPR plays a pivotal role in pancreatic ß cells. Pancreatic ß cells producing a large amount of insulin are highly sensitive when the UPR is impaired. In mammalian cells, three principal ER stress sensors, PERK, IRE1, and ATF6, initiate the UPR. Activated PERK attenuates protein translation through eIF2α phosphorylation to cope with the ER stress. PERK KO mice develop diabetes by 2-4 weeks of age due to progressive ß-cell loss. IRE1α noncanonically splices the XBP1 mRNA, leading to the upregulation of the ERAD components and ER molecular chaperones. This pathway is constitutively activated in pancreatic ß cells. To clarify the physiological role of the IRE1α pathway in ß cells, we generated pancreatic-ß-cell-specific IRE1α-conditional KO (cKO) mice and IRE1α-cKO insulinoma cell lines. Here, we show that IRE1α is required for the upregulation of insulin-folding enzymes in pancreatic ß cells to balance insulin-folding enzymes with insulin.


Assuntos
Diabetes Mellitus/etiologia , Estresse do Retículo Endoplasmático/fisiologia , Endorribonucleases/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Resposta a Proteínas não Dobradas/fisiologia , Fator 6 Ativador da Transcrição/fisiologia , Animais , Fator de Iniciação 2 em Eucariotos/metabolismo , Humanos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Camundongos , Fosforilação , eIF-2 Quinase/fisiologia
18.
Int J Mol Sci ; 16(12): 30321-41, 2015 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-26694384

RESUMO

cFLIP (cellular FLICE-like inhibitory protein) is structurally related to caspase-8 but lacks proteolytic activity due to multiple amino acid substitutions of catalytically important residues. cFLIP protein is evolutionarily conserved and expressed as three functionally different isoforms in humans (cFLIPL, cFLIPS, and cFLIPR). cFLIP controls not only the classical death receptor-mediated extrinsic apoptosis pathway, but also the non-conventional pattern recognition receptor-dependent apoptotic pathway. In addition, cFLIP regulates the formation of the death receptor-independent apoptotic platform named the ripoptosome. Moreover, recent studies have revealed that cFLIP is also involved in a non-apoptotic cell death pathway known as programmed necrosis or necroptosis. These functions of cFLIP are strictly controlled in an isoform-, concentration- and tissue-specific manner, and the ubiquitin-proteasome system plays an important role in regulating the stability of cFLIP. In this review, we summarize the current scientific findings from biochemical analyses, cell biological studies, mathematical modeling, and gene-manipulated mice models to illustrate the critical role of cFLIP as a switch to determine the destiny of cells among survival, apoptosis, and necroptosis.


Assuntos
Apoptose , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Animais , Homeostase , Humanos , Necrose , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo
19.
J Cell Sci ; 128(9): 1762-72, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25770101

RESUMO

Accumulation of unfolded proteins in the endoplasmic reticulum (ER) accompanies ER stress and causes the type-I transmembrane protein Ire1 (also known as ERN1) to trigger the unfolded protein response (UPR). When dimerized, the core stress-sensing region (CSSR) of Ire1 directly captures unfolded proteins and forms a high-order oligomer, leading to clustering and activation of Ire1. The CSSR is N-terminally flanked by an intrinsically disordered subdomain, which we previously named Subregion I, in Saccharomyces cerevisiae Ire1. In this study, we describe tight repression of Ire1 activity by Subregion I under conditions of no or weak stress. Weak hyperactivation of an Ire1 mutant lacking Subregion I slightly retarded growth of yeast cells cultured under unstressed conditions. Fungal Ire1 orthologs and the animal Ire1 family protein PERK (also known as EIF2AK3) carry N-terminal intrinsically disordered subdomains with a similar structure and function to that of Subregion I. Our observations presented here cumulatively indicate that Subregion I is captured by the CSSR as an unfolded protein substrate. This intramolecular subdomain interaction is likely to compromise self-association of the CSSR, explaining why Subregion I can suppress Ire1 activity when ER-accumulated unfolded proteins are not abundant.


Assuntos
Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Resposta a Proteínas não Dobradas , Animais , Mamíferos , Camundongos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Células NIH 3T3 , Peptídeos/metabolismo , Estrutura Terciária de Proteína , Deleção de Sequência , Fatores de Tempo , Técnicas do Sistema de Duplo-Híbrido , eIF-2 Quinase/metabolismo
20.
FEBS J ; 282(7): 1256-70, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25631627

RESUMO

When intracellular damage accumulates in proliferating somatic cells, the cell cycle usually arrests in G1 or G2 in a checkpoint-dependent manner, either to repair the damage or to die by apoptosis. In contrast, early embryonic cells lack checkpoint-mediated cell-cycle arrest, and it is not clear whether apoptosis in early embryonic cells occurs at a specific cell cycle stage or at random points. Here, we examined the functional molecular link between the embryonic cell cycle and apoptosis using Xenopus egg extracts. When apoptosis was induced in egg extracts by addition of exogenous cytochrome c during cell-cycle progression, cyclin B accumulation was inhibited, Cdc2 was not activated, and the cell cycle arrested at interphase. However, addition of recombinant cyclin B failed to activate Cdc2 due to the strong inhibitory phosphorylation of Cdc2 Tyr15 in apoptotic egg extracts. We found that endogenous Cdc25C, which activates the Cdc2-cyclin B complex by dephosphorylating Cdc2 Tyr15, was inactivated by caspase-mediated cleavage at two sites in the N-terminal regulatory domain. When the hyperactive Cdc25A catalytic fragment was added together with recombinant cyclin B to artificially dephosphorylate Cdc2 Tyr15, M-phase induction was restored in apoptotic egg extracts, indicating that the blockage of cyclin B accumulation and the caspase-mediated inactivation of Cdc25C dually inhibited Cdc2 activation. Apoptosis induction in cytostatic factor-arrested metaphase egg extracts resulted in inactivation of Cdc2 without cyclin B degradation. These results suggest that apoptotic inactivation of Cdc25C plays an important role in arresting the embryonic cell cycle at interphase during apoptosis.


Assuntos
Apoptose , Proteínas de Ciclo Celular/metabolismo , Proteínas Quinases/metabolismo , Proteínas de Xenopus/metabolismo , Fosfatases cdc25/metabolismo , Animais , Caspases/fisiologia , Pontos de Checagem do Ciclo Celular , Ciclina B/metabolismo , Ativação Enzimática , Metáfase , Oócitos/enzimologia , Fosforilação , Biossíntese de Proteínas , Processamento de Proteína Pós-Traducional , Proteólise , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...