Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Genes Cells ; 28(5): 374-382, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36811310

RESUMEN

Human epidermal growth factor receptor (HER) family proteins are currently major targets of therapeutic monoclonal antibodies against various epithelial cancers. However, the resistance of cancer cells to HER family-targeted therapies, which may be caused by cancer heterogeneity and persistent HER phosphorylation, often reduces overall therapeutic effects. We herein showed that a newly discovered molecular complex between CD98 and HER2 affected HER function and cancer cell growth. The immunoprecipitation of the HER2 or HER3 protein from lysates of SKBR3 breast cancer (BrCa) cells revealed the HER2-CD98 or HER3-CD98 complex. The knockdown of CD98 by small interfering RNAs inhibited the phosphorylation of HER2 in SKBR3 cells. A bispecific antibody (BsAb) that recognized the HER2 and CD98 proteins was constructed from a humanized anti-HER2 (SER4) IgG and an anti-CD98 (HBJ127) single chain variable fragment, and this BsAb significantly inhibited the cell growth of SKBR3 cells. Prior to the inhibition of AKT phosphorylation, BsAb inhibited the phosphorylation of HER2, however, significant inhibition of HER2 phosphorylation was not observed in anti-HER2 pertuzumab, trastuzumab, SER4 or anti-CD98 HBJ127 in SKBR3 cells. The dual targeting of HER2 and CD98 has potential as a new therapeutic strategy for BrCa.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Neoplasias de la Mama/patología , Receptor ErbB-2/metabolismo , Trastuzumab/farmacología , Trastuzumab/metabolismo , Trastuzumab/uso terapéutico , Anticuerpos Monoclonales/metabolismo , Fosforilación , Línea Celular Tumoral
2.
Cell Immunol ; 383: 104652, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36516653

RESUMEN

In this study, we sought to elucidate the roles of the interleukin (IL)-32ß and IL-32γ in mesothelioma cell growth, and vascular endothelial growth factor (VEGF)-A and C-X-C motif chemokine ligand 8 (CXCL8) expression. IL-32 elicited a growth-promoting effect against one of the six mesotheliomas lines and exerted diverse regulatory functions in VEGF-A and CXCL8 secretion from mesotheliomas stimulated with or without IL-17A. Retroviral-mediated transduction of mesothelioma lines with IL-32γ resulted in enhanced IL-32ß expression, which facilitated or suppressed the in vitro growth, and VEGF-A and CXCL8 expression. Overexpressed IL-32ß-augmented growth and VEGF-A and CXCL8 production were mainly mediated through the phosphatidylinositol-3 kinase (PI3K) signaling pathway. On the other hand, overexpressed IL-32ß-deceased growth was mediated through mitogen-activated protein kinase (MAPK) pathway. NCI-H2373IL-32γ tumors grew faster than NCI-H2373Neo tumors in a xenograft model, which was associated with increased vascularity. These findings indicate that IL-32 are involved in the regulation of growth and angiogenic factor production in mesotheliomas.


Asunto(s)
Interleucina-8 , Interleucinas , Mesotelioma Maligno , Factor A de Crecimiento Endotelial Vascular , Humanos , Interleucinas/genética , Interleucinas/metabolismo , Mesotelioma Maligno/metabolismo , Mesotelioma Maligno/patología , Isoformas de Proteínas/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Interleucina-8/metabolismo
3.
Int J Mol Sci ; 23(21)2022 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-36361884

RESUMEN

Under stress conditions, transfer RNAs (tRNAs) are cleaved by stress-responsive RNases such as angiogenin, generating tRNA-derived RNAs called tiRNAs. As tiRNAs contribute to cytoprotection through inhibition of translation and prevention of apoptosis, the regulation of tiRNA production is critical for cellular stress response. Here, we show that RTCB ligase complex (RTCB-LC), an RNA ligase complex involved in endoplasmic reticulum (ER) stress response and precursor tRNA splicing, negatively regulates stress-induced tiRNA production. Knockdown of RTCB significantly increased stress-induced tiRNA production, suggesting that RTCB-LC negatively regulates tiRNA production. Gel-purified tiRNAs were repaired to full-length tRNAs by RtcB in vitro, suggesting that RTCB-LC can generate full length tRNAs from tiRNAs. As RTCB-LC is inhibited under oxidative stress, we further investigated whether tiRNA production is promoted through the inhibition of RTCB-LC under oxidative stress. Although hydrogen peroxide (H2O2) itself did not induce tiRNA production, it rapidly boosted tiRNA production under the condition where stress-responsive RNases are activated. We propose a model of stress-induced tiRNA production consisting of two factors, a trigger and booster. This RTCB-LC-mediated boosting mechanism may contribute to the effective stress response in the cell.


Asunto(s)
Peróxido de Hidrógeno , ARN de Transferencia , Peróxido de Hidrógeno/farmacología , ARN de Transferencia/metabolismo , Estrés Oxidativo , Empalme del ARN , Ligasas/genética
4.
Biochem Biophys Res Commun ; 576: 59-65, 2021 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-34482024

RESUMEN

HER1-and HER2-targeted drugs are effective in cancer therapy, especially against lung, breast and colon malignancies; however, resistance of cancer cells to HER1-and HER2-targeted therapies is becoming a serious problem. The avidity/affinity constant (KA) and growth inhibitory effect of anti-HER3 rat monoclonal antibodies (mAb, Ab1∼Ab6) in the presence of therapeutic mAb or low-molecular-weight inhibitors against HER family proteins were analyzed by flow cytometry-based Scatchard plots (Splot) and cell proliferation assay. The KA of Ab3 and Ab6, but not Ab1 or Ab4, split into dual (high and low) modes of KA, and Ab6 exhibited greater anti-proliferative effects against LS-174T colon cancer cells in the presence of Pertuzumab (anti-HER2 mAb). A high KA by Ab6 and Ab6-mediated increased growth inhibition were observed against NCI-H1838 lung or BT474 breast cancer cells, respectively, in the presence of Panitumumab (anti-HER1 mAb) or Perutuzumab. A high KA by Ab6 and Ab6-mediated increased anti-proliferative effects against NCI-H1838 or BT474 were also respectively observed in the presence of Erlotinib (HER1 inhibitor) or Lapatinib (HER1/HER2 inhibitor). In HER1-knockout (KO) NCI-H1838, the reactivity and KA of Ab4 increased compared with in parent NCI-H1838. In HER1-KO or HER3-KO SW1116 colon cancer cells, dual modes of KA with Pertuzumab were noted, and the combination Ab6 and Pertuzumab promoted growth inhibition of HER1-KO, but not of parent SW1116.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Neoplasias/tratamiento farmacológico , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-3/antagonistas & inhibidores , Animales , Afinidad de Anticuerpos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Línea Celular Tumoral , Proliferación Celular , Resistencia a Antineoplásicos , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/inmunología , Receptores ErbB/metabolismo , Humanos , Neoplasias/inmunología , Neoplasias/metabolismo , Ratas , Receptor ErbB-2/inmunología , Receptor ErbB-2/metabolismo , Receptor ErbB-3/inmunología , Receptor ErbB-3/metabolismo , Transducción de Señal
5.
RNA Biol ; 18(12): 2546-2555, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34085908

RESUMEN

Under adverse conditions, tRNAs are processed into fragments called tRNA-derived stress-induced RNAs (tiRNAs) by stress-responsive ribonucleases (RNases) such as angiogenin (ANG). Recent studies have reported several biological functions of synthetic tiRNAs lacking post-transcriptional modifications found on endogenous tiRNAs. Here we describe a simple and reproducible method to efficiently isolate ANG-cleaved tiRNAs from endogenous tRNAs. Using this in vitro method, more than 50% of mature tRNAs are cleaved into tiRNAs which can be enriched using complementary oligonucleotides. Using this method, the yield of isolated endogenous 5'-tiRNAGly-GCC was increased about fivefold compared to when tiRNAs were obtained by cellular treatment of ANG. Although the non-specific ribonuclease activity of ANG is much lower than that of RNase A, we show that ANG cleaves physiologically folded tRNAs as efficiently as bovine RNase A. These results suggest that ANG is highly specialized to cleave physiologically folded tRNAs. Our method will greatly facilitate the analysis of endogenous tiRNAs to elucidate the physiological functions of ANG.


Asunto(s)
Inductores de la Angiogénesis/metabolismo , Procesamiento Postranscripcional del ARN , ARN de Transferencia/metabolismo , Ribonucleasa Pancreática/metabolismo , Humanos , ARN de Transferencia/genética , Ribonucleasa Pancreática/genética , Células Tumorales Cultivadas
6.
Eur J Immunol ; 49(4): 546-563, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30671932

RESUMEN

In sepsis, the pathology involves a shift from a proinflammatory state toward an immunosuppressive phase. We previously showed that an agonistic anti-TLR4 antibody induced long-term endotoxin tolerance and suppressed antigen-specific secondary IgG production when primed prior to immunization with antigen. These findings led us to speculate that TLR4-induced innate tolerance due to primary infection causes an immunosuppressive pathology in sepsis. Therefore, the mechanism underlying impaired antigen-specific humoral immunity by the TLR4 antibody was investigated. We showed, in a mouse model, that primary antigen-specific IgG responses were impaired in TLR4 antibody-induced tolerized mice, which was the result of reduced numbers of antigen-specific GC B cells and plasma cells. Ovalbumin-specific CD4 and CD8 T-cell responses were impaired in TLR4 antibody-injected OT-I and -II transgenic mice ex vivo. Adoptive transfer studies demonstrated suppression of OVA-specific CD4 and CD8 T-cell responses by the TLR4 antibody in vivo. The TLR4 antibody induced Gr1+ CD11b+ myeloid-derived suppressor cell (MDSC) expansion with suppression of T-cell activation. Monocytic MDSCs were more suppressive and exhibited higher expression of PD-L1 and inducible nitric oxidase compared with granulocytic MDSCs. In conclusion, immune tolerance conferred by TLR4 activation induces the expansion of monocytic MDSCs, which impairs antigen-specific T-cell priming and IgG production.


Asunto(s)
Epítopos de Linfocito T/inmunología , Activación de Linfocitos/inmunología , Linfocitos/inmunología , Linfocitos/metabolismo , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Formación de Anticuerpos/inmunología , Biomarcadores , Epítopos de Linfocito B/inmunología , Tolerancia Inmunológica , Inmunización , Inmunofenotipificación , Ratones
7.
Nephrol Dial Transplant ; 35(2): 250-264, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31411705

RESUMEN

BACKGROUND: Cardiorenal syndrome is a major cause of mortality in patients with chronic kidney disease (CKD). However, the involvement of detrimental humoral mediators in the pathogenesis of cardiorenal syndrome is still controversial. Trimethylamine-N-oxide (TMAO), a hepatic metabolic product of trimethylamine generated from dietary phosphatidylcholine or carnitine derived by the gut microbiota, has been linked directly with progression of cardiovascular disease and renal dysfunction. Thus, targeting TMAO may be a novel strategy for the prevention of cardiovascular disease and chronic kidney disease. METHODS: Linaclotide, a guanylate cyclase C agonist, was administered to adenine-induced renal failure (RF) mice and changes in renal function and levels of gut-derived uremic toxins, as well as the gut microbiota community, were analyzed using metabolomic and metagenomic methods to reveal its cardiorenal effect. RESULTS: Linaclotide decreased the plasma levels of TMAO at a clinically used low dose of 10 µg/kg in the adenine-induced RF mouse model. At a high concentration of 100 µg/kg, linaclotide clearly improved renal function and reduced the levels of various uremic toxins. A reduction in TMAO levels following linaclotide treatment was also observed in a choline-fed pro-atherosclerotic model. Linaclotide ameliorated renal inflammation and fibrosis and cardiac fibrosis, as well as decreased the expression of collagen I, transforming growth factor-ß, galectin-3 (Gal-3) and ST2 genes. Plasma levels of Gal-3 and ST2 were also reduced. Because exposure of cardiomyocytes to TMAO increased fibronectin expression, these data suggest that linaclotide reduced the levels of TMAO and various uremic toxins and may result in not only renal, but also cardiac, fibrosis. F4/80-positive macrophages were abundant in small intestinal crypts in RF mice, and this increased expression was decreased by linaclotide. Reduced colonic claudin-1 levels were also restored by linaclotide, suggesting that linaclotide ameliorated the 'leaky gut' in RF mice. Metagenomic analysis revealed that the microbial order Clostridiales could be responsible for the change in TMAO levels. CONCLUSION: Linaclotide reduced TMAO and uremic toxin levels and could be a powerful tool for the prevention and control of the cardiorenal syndrome by modification of the gut-cardio-renal axis.


Asunto(s)
Adenina/toxicidad , Síndrome Cardiorrenal/tratamiento farmacológico , Microbioma Gastrointestinal/efectos de los fármacos , Guanilato Ciclasa/química , Agonistas de la Guanilato Ciclasa C/farmacología , Péptidos/farmacología , Insuficiencia Renal Crónica/tratamiento farmacológico , Animales , Síndrome Cardiorrenal/inducido químicamente , Síndrome Cardiorrenal/metabolismo , Síndrome Cardiorrenal/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Fibrosis/inducido químicamente , Fibrosis/tratamiento farmacológico , Fibrosis/metabolismo , Fibrosis/patología , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Insuficiencia Renal Crónica/inducido químicamente , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología
8.
Rapid Commun Mass Spectrom ; 34 Suppl 1: e8551, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31412144

RESUMEN

RATIONALE: Hepatocellular carcinoma (HCC) is a highly malignant disease for which the development of prospective or prognostic biomarkers is urgently required. Although metabolomics is widely used for biomarker discovery, there are some bottlenecks regarding the comprehensiveness of detected features, reproducibility of methods, and identification of metabolites. In addition, information on localization of metabolites in tumor tissue is needed for functional analysis. Here, we developed a wide-polarity global metabolomics (G-Met) method, identified HCC biomarkers in human liver samples by high-definition mass spectrometry (HDMS), and demonstrated localization in cryosections using desorption electrospray ionization MS imaging (DESI-MSI) analysis. METHODS: Metabolic profiling of tumor (n = 38) and nontumor (n = 72) regions in human livers of HCC was performed by an ultrahigh-performance liquid chromatography quadrupole time-of-flight MS (UHPLC/QTOFMS) instrument equipped with a mixed-mode column. The HCC biomarker candidates were extracted by multivariate analyses and identified by matching values of the collision cross section and their fragment ions on the mass spectra obtained by HDMS. Cryosections of HCC livers, which included both tumor and nontumor regions, were analyzed by DESI-MSI. RESULTS: From the multivariate analysis, m/z 904.83 and m/z 874.79 were significantly high and low, respectively, in tumor samples and were identified as triglyceride (TG) 16:0/18:1(9Z)/20:1(11Z) and TG 16:0/18:1(9Z)/18:2(9Z,12Z) using the synthetic compounds. The TGs were clearly localized in the tumor or nontumor areas of the cryosection. CONCLUSIONS: Novel biomarkers for HCC were identified by a comprehensive and reproducible G-Met method with HDMS using a mixed-mode column. The combination analysis of UHPLC/QTOFMS and DESI-MSI revealed that the different molecular species of TGs were associated with tumor distribution and were useful for characterizing the progression of tumor cells and discovering prospective biomarkers.


Asunto(s)
Carcinoma Hepatocelular/diagnóstico , Neoplasias Hepáticas/diagnóstico , Hígado/patología , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/metabolismo , Cromatografía Líquida de Alta Presión , Humanos , Hígado/metabolismo , Neoplasias Hepáticas/metabolismo , Metaboloma , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
9.
J Biol Chem ; 293(26): 10186-10201, 2018 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-29760187

RESUMEN

Toll-like receptor 4 (TLR4) is an indispensable immune receptor for lipopolysaccharide (LPS), a major component of the Gram-negative bacterial cell wall. Following LPS stimulation, TLR4 transmits the signal from the cell surface and becomes internalized in an endosome. However, the spatial regulation of TLR4 signaling is not fully understood. Here, we investigated the mechanisms of LPS-induced TLR4 internalization and clarified the roles of the extracellular LPS-binding molecules, LPS-binding protein (LBP), and glycerophosphatidylinositol-anchored protein (CD14). LPS stimulation of CD14-expressing cells induced TLR4 internalization in the presence of serum, and an inhibitory anti-LBP mAb blocked its internalization. Addition of LBP to serum-free cultures restored LPS-induced TLR4 internalization to comparable levels of serum. The secretory form of the CD14 (sCD14) induced internalization but required a much higher concentration than LBP. An inhibitory anti-sCD14 mAb was ineffective for serum-mediated internalization. LBP lacking the domain for LPS transfer to CD14 and a CD14 mutant with reduced LPS binding both attenuated TLR4 internalization. Accordingly, LBP is an essential serum molecule for TLR4 internalization, and its LPS transfer to membrane-anchored CD14 (mCD14) is a prerequisite. LBP induced the LPS-stimulated phosphorylation of TBK1, IKKϵ, and IRF3, leading to IFN-ß expression. However, LPS-stimulated late activation of NF-κB or necroptosis were not affected. Collectively, our results indicate that LBP controls LPS-induced TLR4 internalization, which induces TLR adaptor molecule 1 (TRIF)-dependent activation of the TBK1-IKKϵ-IRF3-IFN-ß pathway. In summary, we showed that LBP-mediated LPS transfer to mCD14 is required for serum-dependent TLR4 internalization and activation of the TRIF pathway.


Asunto(s)
Proteínas de Fase Aguda/metabolismo , Proteínas Portadoras/metabolismo , Quinasa I-kappa B/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Receptores de Lipopolisacáridos/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Toll-Like 4/metabolismo , Humanos , Lipopolisacáridos/farmacología , Antígeno 96 de los Linfocitos/metabolismo , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos
10.
Immunology ; 158(2): 136-149, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31515801

RESUMEN

Immune-checkpoint blockade antibodies have been approved for the treatment of cancer. However, poorly immunogenic tumours are less responsive to such therapies. Agonistic anti-Toll-like receptor 4 (TLR4) monoclonal antibodies (mAbs) activate only cell-surface TLR4; in contrast, lipopolysaccharide (LPS) activates both TLR4 and intracellular inflammatory caspases. In this study, we investigated the adjuvant activity of an anti-TLR4 mAb in T-cell-mediated antitumour immunity. The anti-TLR4 mAb induced the activation of antigen-specific T-cells in adoptive transfer studies. The growth of ovalbumin (OVA)-expressing tumours was significantly suppressed by administration of OVA and the anti-TLR4 mAb in combination, but not individually. The antitumour effect of anti-PD-1 mAb was enhanced in mice administered with OVA plus the anti-TLR4 mAb. The OVA-specific IFN-γ-producing CD8 T-cells were induced by administration of OVA and the anti-TLR4 mAb. The suppression of tumour growth was diminished by depletion of CD8, but not CD4, T-cells. The inflammatory response to the anti-TLR4 mAb was of significantly lesser magnitude than that to LPS, as assessed by NF-κB activation and production of TNF-α, IL-6 and IL-1ß. Administration of LPS (at a dose that elicited levels of proinflammatory cytokines comparable to those by the anti-TLR4 mAb) plus OVA induced no or less-marked activation of OVA-specific T-cells and failed to suppress tumour growth in mice. In conclusion, the agonistic anti-TLR4 mAb induces potent CD8 T-cell-dependent antitumour immunity and an inflammatory response of lesser magnitude than does LPS. The agonistic anti-TLR4 mAb has potential as an adjuvant for use in vaccines against cancer.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Anticuerpos Monoclonales/farmacología , Antineoplásicos Inmunológicos/farmacología , Melanoma Experimental/terapia , Neoplasias Cutáneas/terapia , Receptor Toll-Like 4/antagonistas & inhibidores , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Inmunización , Inmunoterapia/métodos , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Lipopolisacáridos/farmacología , Activación de Linfocitos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , FN-kappa B/genética , FN-kappa B/inmunología , Ovalbúmina/administración & dosificación , Cultivo Primario de Células , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células TH1/patología , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
11.
Int J Mol Sci ; 20(20)2019 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-31658747

RESUMEN

Niemann-Pick disease type C (NPC) is an autosomal recessive disorder caused by the mutation of cholesterol-transporting proteins. In addition, early treatment is important for good prognosis of this disease because of the progressive neurodegeneration. However, the diagnosis of this disease is difficult due to a variety of clinical spectrum. Lysosphingomyelin-509, which is one of the most useful biomarkers for NPC, was applied for the rapid and easy detection of NPC. The fact that its chemical structure was unknown until recently implicates the unrevealed pathophysiology and molecular mechanisms of NPC. In this study, we aimed to elucidate the structure of lysosphingomyelin-509 by various mass spectrometric techniques. As our identification strategy, we adopted analytical and organic chemistry approaches to the serum of patients with NPC. Chemical derivatization and hydrogen abstraction dissociation-tandem mass spectrometry were used for the determination of function groups and partial structure, respectively. As a result, we revealed the exact structure of lysosphingomyelin-509 as N-acylated and O-phosphocholine adducted serine. Additionally, we found that a group of metabolites with N-acyl groups were increased considerably in the serum/plasma of patients with NPC as compared to that of other groups using targeted lipidomics analysis. Our techniques were useful for the identification of lysosphingomyelin-509.


Asunto(s)
Lípidos/química , Lípidos/aislamiento & purificación , Enfermedad de Niemann-Pick Tipo C/diagnóstico , Fosforilcolina/química , Fosforilcolina/aislamiento & purificación , Serina/química , Biomarcadores/sangre , Femenino , Humanos , Masculino , Enfermedad de Niemann-Pick Tipo C/metabolismo , Fosforilcolina/metabolismo , Serina/metabolismo , Espectrometría de Masas en Tándem/métodos
12.
Am J Physiol Renal Physiol ; 315(4): F824-F833, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29167170

RESUMEN

Accumulation of uremic toxins, which exert deleterious effects in chronic kidney disease, is influenced by the intestinal environment; the microbiota contributes to the production of representative uremic toxins, including p-cresyl sulfate and indoxyl sulfate. Canagliflozin is a sodium-glucose cotransporter (SGLT) 2 inhibitor, and it also exerts a modest inhibitory effect on SGLT1. The inhibition of intestinal SGLT1 can influence the gastrointestinal environment. We examined the effect of canagliflozin on the accumulation of uremic toxins in chronic kidney disease using adenine-induced renal failure mice. Two-week canagliflozin (10 mg/kg po) treatment did not influence the impaired renal function; however, it significantly reduced the plasma levels of p-cresyl sulfate and indoxyl sulfate in renal failure mice (a 75% and 26% reduction, respectively, compared with the vehicle group). Additionally, canagliflozin significantly increased cecal short-chain fatty acids in the mice, suggesting the promotion of bacterial carbohydrate fermentation in the intestine. Analysis of the cecal microbiota showed that canagliflozin significantly altered microbiota composition in the renal failure mice. These results indicate that canagliflozin exerts intestinal effects that reduce the accumulation of uremic toxins including p-cresyl sulfate. Reduction of accumulated uremic toxins by canagliflozin could provide a potential therapeutic option in chronic kidney disease.


Asunto(s)
Canagliflozina/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Insuficiencia Renal Crónica/tratamiento farmacológico , Toxinas Biológicas/sangre , Animales , Modelos Animales de Enfermedad , Tracto Gastrointestinal/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Insuficiencia Renal Crónica/sangre , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Toxinas Biológicas/farmacología , Uremia/sangre , Uremia/tratamiento farmacológico
13.
Biol Pharm Bull ; 41(8): 1170-1177, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30068866

RESUMEN

Patients with chronic kidney disease (CKD) have increased blood levels of phenyl sulfate (PS), a circulating uremic toxin. In this study, we produced anti-PS monoclonal antibodies (mAbs) and characterized their cross-reactivity to structural PS analogs. To induce PS-specific mAbs, we synthesized 4-mercaptophenyl sulfate with a sulfhydryl group at the para-position of PS and conjugated it to carrier proteins via bifunctional linkers. Using these PS conjugates as immunogens and as antigens for enzyme-linked immunosorbent assay (ELISA) screening, we produced by a hybridoma method two novel mAbs (YK33.1 and YKS19.2) that react with PS conjugates independent of carrier and linker structures. Although all of the PS analogs tested, with the exception of indoxyl sulfate, were cross-reactive to both mAbs in phosphate buffered saline (PBS), PS specificity for YKS19.2 was enhanced in human plasma and serum. YKS19.2 mAb was cross-reactive only with o-cresyl sulfate, which is absent in human blood. PS sensitivity for YKS19.2 mAb increased to an IC50 of 10.4 µg/mL when 0.1% Tween 20 was added in a primary competitive reaction. To explore potential clinical applications, we determined concentrations of PS in serum samples from 19 CKD patients by inhibition ELISA using YKS19.2 mAb and compared them to those found using an LC-MS/MS method. A good correlation was observed between each value (R2=0.825). Therefore, the unique antigen specificity of YKS19.2 mAb could be useful for prescreening of patients with accumulated PS or for comprehensive analysis of uremic toxins that have a PS-like structure.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Insuficiencia Renal Crónica/sangre , Ésteres del Ácido Sulfúrico/sangre , Ésteres del Ácido Sulfúrico/inmunología , Animales , Antígenos/química , Antígenos/inmunología , Línea Celular Tumoral , Cromatografía Liquida , Ensayo de Inmunoadsorción Enzimática , Femenino , Hemocianinas/química , Hemocianinas/inmunología , Humanos , Inmunoconjugados/química , Inmunoconjugados/inmunología , Ratones Endogámicos BALB C , Ovalbúmina/química , Ovalbúmina/inmunología , Albúmina Sérica Bovina/química , Albúmina Sérica Bovina/inmunología , Ésteres del Ácido Sulfúrico/química , Espectrometría de Masas en Tándem
14.
Kidney Int ; 92(3): 634-645, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28396122

RESUMEN

Gut microbiota is involved in the metabolism of uremic solutes. However, the precise influence of microbiota to the retention of uremic solutes in CKD is obscure. To clarify this, we compared adenine-induced renal failure and control mice under germ-free or specific pathogen-free (SPF) conditions, examining the metabolite profiles of plasma, feces, and urine using a capillary electrophoresis time-of-flight mass spectrometry-based approach. Mice with renal failure under germ-free conditions demonstrated significant changes in plasma metabolites. Among 183 detected solutes, plasma levels of 11 solutes, including major uremic toxins, were significantly lower in germ-free mice than in SPF mice with renal failure. These 11 solutes were considered microbiota-derived uremic solutes and included indoxyl sulfate, p-cresyl sulfate, phenyl sulfate, cholate, hippurate, dimethylglycine, γ-guanidinobutyrate, glutarate, 2-hydroxypentanoate, trimethylamine N-oxide, and phenaceturate. Metabolome profiling showed that these solutes were classified into three groups depending on their origins: completely derived from microbiota (indoxyl sulfate, p-cresyl sulfate), derived from both host and microbiota (dimethylglycine), and derived from both microbiota and dietary components (trimethylamine N-oxide). Additionally, germ-free renal failure conditions resulted in the disappearance of colonic short-chain fatty acids, decreased utilization of intestinal amino acids, and more severe renal damage compared with SPF mice with renal failure. Microbiota-derived short-chain fatty acids and efficient amino acid utilization may have a renoprotective effect, and loss of these factors may exacerbate renal damage in germ-free mice with renal failure. Thus, microbiota contributes substantially to the production of harmful uremic solutes, but conversely, growth without microbiota has harmful effects on CKD progression.


Asunto(s)
Lesión Renal Aguda/metabolismo , Microbioma Gastrointestinal/fisiología , Metaboloma , Insuficiencia Renal Crónica/metabolismo , Toxinas Biológicas/sangre , Uremia/metabolismo , Lesión Renal Aguda/sangre , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/orina , Adenina/toxicidad , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Electroforesis Capilar , Ácidos Grasos Volátiles/análisis , Ácidos Grasos Volátiles/metabolismo , Humanos , Riñón/patología , Espectrometría de Masas , Metabolómica/métodos , Ratones , Insuficiencia Renal Crónica/sangre , Insuficiencia Renal Crónica/patología , Insuficiencia Renal Crónica/orina , Organismos Libres de Patógenos Específicos , Toxinas Biológicas/orina , Uremia/sangre , Uremia/orina
15.
J Am Soc Nephrol ; 27(7): 1925-32, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26609120

RESUMEN

Mitochondrial dysfunction causes increased oxidative stress and depletion of ATP, which are involved in the etiology of a variety of renal diseases, such as CKD, AKI, and steroid-resistant nephrotic syndrome. Antioxidant therapies are being investigated, but clinical outcomes have yet to be determined. Recently, we reported that a newly synthesized indole derivative, mitochonic acid 5 (MA-5), increases cellular ATP level and survival of fibroblasts from patients with mitochondrial disease. MA-5 modulates mitochondrial ATP synthesis independently of oxidative phosphorylation and the electron transport chain. Here, we further investigated the mechanism of action for MA-5. Administration of MA-5 to an ischemia-reperfusion injury model and a cisplatin-induced nephropathy model improved renal function. In in vitro bioenergetic studies, MA-5 facilitated ATP production and reduced the level of mitochondrial reactive oxygen species (ROS) without affecting activity of mitochondrial complexes I-IV. Additional assays revealed that MA-5 targets the mitochondrial protein mitofilin at the crista junction of the inner membrane. In Hep3B cells, overexpression of mitofilin increased the basal ATP level, and treatment with MA-5 amplified this effect. In a unique mitochondrial disease model (Mitomice with mitochondrial DNA deletion that mimics typical human mitochondrial disease phenotype), MA-5 improved the reduced cardiac and renal mitochondrial respiration and seemed to prolong survival, although statistical analysis of survival times could not be conducted. These results suggest that MA-5 functions in a manner differing from that of antioxidant therapy and could be a novel therapeutic drug for the treatment of cardiac and renal diseases associated with mitochondrial dysfunction.


Asunto(s)
Ácidos Indolacéticos/farmacología , Túbulos Renales/citología , Mitocondrias/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Fenilbutiratos/farmacología , Animales , Masculino , Ratones , Ratones Endogámicos C57BL
16.
Neuro Endocrinol Lett ; 38(1): 43-49, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28456147

RESUMEN

OBJECTIVES: Alcohol intake induces brief periods of euphoria; however, its continuous consumption can lead the development of alcohol tolerance. The euphoria, an intense feeling of wellbeing, is deeply associated with dopamine. Dopamine biosynthesis is strictly regulated by tyrosine hydroxylase (TH), a rate-limiting enzyme of dopamine. The aim of this study was to examine the transient or chronic effects of ethanol treatment on TH protein level in vitro. METHODS: Cultured primary mesencephalic neurons were prepared and exposed to 100 mM ethanol for 48 hours or 168 hours. TH and cAMP-responsive element (CRE)-mediated transcriptional activity was measured by reporter gene assay using pTH9.0kb-Luc and pCRE-Luc reporter plasmid. TH protein expression and TH phosphorylation was analyzed by Western blot analysis. Dopamine content was measured by high-performance liquid chromatography (HPLC). RESULTS: Ethanol treatment for 48 hours facilitates TH transcriptional activity and TH protein expression in a cAMP-dependent protein kinase A (PKA) and MAPK/Erk kinase (MEK)-dependent manner in cultured mesencephalic neurons. Ethanol also facilitated TH phosphorylation, which resulted in the elevation of dopamine content. On the other hand, treatment with ethanol for 168 hours did not show significant elevation of TH gene expression and dopamine biosynthesis. Intriguingly, simultaneous treatment with MG-132, a 26S proteasomal inhibitor, recovered the ethanol-induced increase of TH protein expression and dopamine biosynthesis. CONCLUSION: Transient ethanol-treatment facilitates TH gene expression and its phosphorylation in a PKA- and MEK-dependent manner to elevate dopamine biosynthesis, whereas continuous exposure to ethanol abolishes its potent effects on the dopaminergic function to reduce dopamine content. This reduction seems to originate from the decrease of TH protein level by degradation of the protein. Our current data may contribute to the better understanding of alcohol tolerance associated with degradation of TH protein to reduce total-TH level and dopamine biosynthesis.


Asunto(s)
Depresores del Sistema Nervioso Central/farmacología , Dopamina/biosíntesis , Etanol/farmacología , Neuronas/efectos de los fármacos , Tirosina 3-Monooxigenasa/efectos de los fármacos , Animales , Western Blotting , Células Cultivadas , Cromatografía Líquida de Alta Presión , Proteínas Quinasas Dependientes de AMP Cíclico , Inhibidores de Cisteína Proteinasa/farmacología , Expresión Génica/efectos de los fármacos , Leupeptinas/farmacología , Sistema de Señalización de MAP Quinasas , Mesencéfalo , Neuronas/metabolismo , Fosforilación , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Ratas , Ratas Wistar , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo
17.
Tohoku J Exp Med ; 240(1): 47-56, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27594509

RESUMEN

The interleukin (IL)-17 family, consisting of six homodimeric cytokines IL-17A, IL-17B, IL-17C, IL-17D, IL-17E/IL-25, and IL-17F, mediates a variety of biological activities including regulation of chemokine secretion and angiogenesis. Among the IL-17 family members, IL-17A and IL-17E/IL-25 are angiogenesis stimulators, while IL-17B and IL-17F are angiogenesis inhibitors. Recently, IL-17A/F heterodimer, comprised of the IL-17A and IL-17F subunits, was found as another member of the IL-17 cytokine family. However, to date, it has been unknown whether IL-17A/F has biological actions to affect the angiogenesis-related vascular endothelial functions. Therefore, in this study, we investigated the biological effects of IL-17A/F on the growth, migration and capillary-like tube formation of vascular endothelial cells. Recombinant IL-17A/F protein had no direct effects on the growth of human dermal microvascular endothelial cells (HMVECs), whereas, after 4-hour incubation in a modified Boyden Chemotaxicell chamber, IL-17A/F significantly induced migration of HMVECs over a wide range of doses via the phosphatidylinositol-3 kinase (PI3K) signaling pathway. We further investigated the biological effect of IL-17A/F on capillary-like tube formation using a co-culture system of human umbilical vein endothelial cells (HUVECs) and human dermal fibroblasts (HDFs), which mimicked the in vivo microenvironment. In this co-culture system, IL-17A/F significantly promoted capillary-like endothelial tube formation in a dose-dependent fashion via the PI3K and extracellular signal-regulated kinase (ERK) signaling pathways. Additionally, IL-17A/F up-regulated secretion of angiogenic growth factors such as IL-8 and growth-related oncogene (GRO)-α by HDFs. These findings identify a novel biological function for IL-17A/F as an indirect angiogenic agent.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Células Endoteliales/citología , Interleucina-17/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Multimerización de Proteína , Proliferación Celular/efectos de los fármacos , Quimiocinas/metabolismo , Dermis/irrigación sanguínea , Células Endoteliales/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Microvasos/citología , Regulación hacia Arriba/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo
18.
J Am Soc Nephrol ; 26(8): 1787-94, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25525179

RESUMEN

The accumulation of uremic toxins is involved in the progression of CKD. Various uremic toxins are derived from gut microbiota, and an imbalance of gut microbiota or dysbiosis is related to renal failure. However, the pathophysiologic mechanisms underlying the relationship between the gut microbiota and renal failure are still obscure. Using an adenine-induced renal failure mouse model, we evaluated the effects of the ClC-2 chloride channel activator lubiprostone (commonly used for the treatment of constipation) on CKD. Oral administration of lubiprostone (500 µg/kg per day) changed the fecal and intestinal properties in mice with renal failure. Additionally, lubiprostone treatment reduced the elevated BUN and protected against tubulointerstitial damage, renal fibrosis, and inflammation. Gut microbiome analysis of 16S rRNA genes in the renal failure mice showed that lubiprostone treatment altered their microbial composition, especially the recovery of the levels of the Lactobacillaceae family and Prevotella genus, which were significantly reduced in the renal failure mice. Furthermore, capillary electrophoresis-mass spectrometry-based metabolome analysis showed that lubiprostone treatment decreased the plasma level of uremic toxins, such as indoxyl sulfate and hippurate, which are derived from gut microbiota, and a more recently discovered uremic toxin, trans-aconitate. These results suggest that lubiprostone ameliorates the progression of CKD and the accumulation of uremic toxins by improving the gut microbiota and intestinal environment.


Asunto(s)
Alprostadil/análogos & derivados , Agonistas de los Canales de Cloruro/uso terapéutico , Tracto Gastrointestinal/efectos de los fármacos , Fallo Renal Crónico/prevención & control , Microbiota/efectos de los fármacos , Adenina , Alprostadil/farmacología , Alprostadil/uso terapéutico , Animales , Agonistas de los Canales de Cloruro/farmacología , Progresión de la Enfermedad , Evaluación Preclínica de Medicamentos , Tracto Gastrointestinal/microbiología , Fallo Renal Crónico/inducido químicamente , Lubiprostona , Masculino , Ratones Endogámicos C57BL , Distribución Aleatoria , Uremia/prevención & control
19.
Arerugi ; 65(3): 200-5, 2016 May.
Artículo en Japonés | MEDLINE | ID: mdl-27193929

RESUMEN

The patient was a 6-year-old female with milk allergy and persistent asthma. She experienced anaphylactic reactions just after the inhalation of Inavir (Laninamivir Octanoate Hydrate) to treat flu infection. A skin-prick test showed positive reactions for Inavir inhaler powder and lactose used as an excipient but negative for Laninamivir. Same results were obtained in a drug-stimulated basophil activation test. The lactose excipient in Inavir inhaler powder was supposed to contain milk proteins, which caused anaphylactic reactions. To test this possibility, we examined the contamination of allergic milk proteins in the lactose excipient and found the smear band by silver staining, which was identified as ß-lactoglobulin (ß-LG) by Western blotting using specific monoclonal antibody and patient's sera. The ß-LG in Inavir was supposed to be glycosylated with lactose because the molecular weight was slightly higher than ß-LG standard reference as seen in mobility. In fact, the incubation with lactose in vitro tended to increase molecular weight. Following these results, we herein report that the trace amounts of ß-LG contaminated in the lactose excipient of Inavir could cause immediate allergic reactions. The risk that the lactose-containing dry powder inhalers cause allergic reactions for patients with cow's milk allergy need to be reminded. In particular, the use for flu patients should be paid careful attention because of increased airway hypersensitivity in those patients.


Asunto(s)
Anafilaxia/etiología , Lactoglobulinas/efectos adversos , Lactosa/efectos adversos , Hipersensibilidad a la Leche/inmunología , Niño , Femenino , Humanos , Lactoglobulinas/inmunología , Lactosa/inmunología , Nebulizadores y Vaporizadores
20.
Biochem Biophys Res Commun ; 465(1): 53-8, 2015 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-26225746

RESUMEN

The protein amount of tyrosine hydroxylase (TH), that is the rate-limiting enzyme for the biosynthesis of dopamine (DA), should be tightly regulated, whereas its degradation pathway is largely unknown. In this study, we analyzed how the TH protein is chemically modified and subsequently degraded under deficiencies of DA and tetrahydrobiopterin (BH4), a cofactor for TH, by using pharmacological agents in PC12D cells and cultured mesencephalic neurons. When inhibition of DA- or BH4-synthesizing enzymes greatly reduced the DA contents in PC12D cells, a marked and persistent increase in phosphorylated TH at (40)Ser (p40-TH) was concomitantly observed. This phosphorylation was mediated by D2 dopamine auto-receptor and cAMP-dependent protein kinase (PKA). Our immunoprecipitation experiments showed that the increase in the p40-TH level was accompanied with its poly-ubiquitination. Treatment of PC12D cells with cycloheximide showed that total-TH protein level was reduced by the DA- or BH4-depletion. Notably, this reduction in the total-TH protein level was sensitive not only to a 26S proteasomal inhibitor, MG-132, but also to a PKA inhibitor, H-89. These data demonstrated that DA deficiency should induce compensatory activation of TH via phosphorylation at (40)Ser through D2-autoreceptor and PKA-mediated pathways, which in turn give a rise to its degradation through an ubiquitin-proteasome pathway, resulting in a negative spiral of DA production when DA deficiency persists.


Asunto(s)
Biopterinas/análogos & derivados , Dopamina/deficiencia , Neuronas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Ubiquitina/metabolismo , Animales , Biopterinas/deficiencia , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Cicloheximida/farmacología , Inhibidores de Cisteína Proteinasa/farmacología , Regulación de la Expresión Génica , Isoquinolinas/farmacología , Leupeptinas/farmacología , Mesencéfalo/citología , Mesencéfalo/efectos de los fármacos , Mesencéfalo/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Células PC12 , Fosforilación/efectos de los fármacos , Cultivo Primario de Células , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Proteolisis/efectos de los fármacos , Ratas , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Serina/metabolismo , Transducción de Señal , Sulfonamidas/farmacología , Tirosina 3-Monooxigenasa/genética , Ubiquitina/genética , Ubiquitinación/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA