Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 135
Filtrar
1.
Eur J Cell Biol ; 103(2): 151406, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38547677

RESUMO

Despite extensive research, targeted delivery of substances to the brain still poses a great challenge due to the selectivity of the blood-brain barrier (BBB). Most molecules require either carrier- or receptor-mediated transport systems to reach the central nervous system (CNS). These transport systems form attractive routes for the delivery of therapeutics into the CNS, yet the number of known brain endothelium-enriched receptors allowing the transport of large molecules into the brain is scarce. Therefore, to identify novel BBB targets, we combined transcriptomic analysis of human and murine brain endothelium and performed a complex screening of BBB-enriched genes according to established selection criteria. As a result, we propose the high-affinity cationic amino acid transporter 1 (SLC7A1) as a novel candidate for transport of large molecules across the BBB. Using RNA sequencing and in situ hybridization assays, we demonstrated elevated SLC7A1 gene expression in both human and mouse brain endothelium. Moreover, we confirmed SLC7A1 protein expression in brain vasculature of both young and aged mice. To assess the potential of SLC7A1 as a transporter for larger proteins, we performed internalization and transcytosis studies using a radiolabelled or fluorophore-labelled anti-SLC7A1 antibody. Our results showed that SLC7A1 internalised a SLC7A1-specific antibody in human colorectal carcinoma (HCT116) cells. Moreover, transcytosis studies in both immortalised human brain endothelial (hCMEC/D3) cells and primary mouse brain endothelial cells clearly demonstrated that SLC7A1 effectively transported the SLC7A1-specific antibody from luminal to abluminal side. Therefore, here in this study, we present for the first time the SLC7A1 as a novel candidate for transport of larger molecules across the BBB.


Assuntos
Barreira Hematoencefálica , Transportador 1 de Aminoácidos Catiônicos , Animais , Humanos , Camundongos , Barreira Hematoencefálica/metabolismo , Transportador 1 de Aminoácidos Catiônicos/metabolismo , Transportador 1 de Aminoácidos Catiônicos/genética , Células Endoteliais/metabolismo , Camundongos Endogâmicos C57BL
2.
J Biomol Struct Dyn ; 41(23): 13580-13594, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36762692

RESUMO

Metabolic and signaling mechanisms in mammalian cells are facilitated by the transportation of L-arginine (Arg) across the plasma membrane through cationic amino acid transporter (CAT) proteins. Due to a lack of argininosuccinate synthase (ASS) activity in various tumor cells such as acute myeloid leukemia, acute lymphocytic leukemia, and chronic lymphocytic leukemia, these tumor entities are arginine-auxotrophic and therefore depend on the uptake of the amino acid arginine. Cationic amino acid transporter-1 (CAT-1) is the leading arginine importer expressed in the aforementioned tumor entities. Hence, in the present study, to investigate the transportation mechanism of arginine in CAT-1, we performed molecular dynamics (MD) simulation methods on the modeled human CAT-1. The MM-PBSA approach was conducted to determine the critical residues interacting with arginine within the corresponding binding site of CAT-1. In addition, we found out that the water molecules have the leading role in forming the transportation channel within CAT-1. The conductive structure of CAT-1 was formed only when the water molecules were continuously distributed across the channel. Steered molecular dynamics (SMD) simulation approach showed various energy barriers against arginine transportation through CAT-1, especially while crossing the bottlenecks of the related channel. These findings at the molecular level might shed light on identifying the crucial amino acids in the binding of arginine to eukaryotic CATs and also provide fundamental insights into the arginine transportation mechanisms through CAT-1. Understanding the transportation mechanism of arginine is essential to developing CAT-1 blockers, which can be potential medications for some types of cancers.Communicated by Ramaswamy H. Sarma.


Assuntos
Arginina , Transportador 1 de Aminoácidos Catiônicos , Animais , Humanos , Arginina/metabolismo , Transportador 1 de Aminoácidos Catiônicos/genética , Transportador 1 de Aminoácidos Catiônicos/metabolismo , Óxido Nítrico Sintase , Simulação de Dinâmica Molecular , Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Água/metabolismo , Mamíferos/metabolismo
3.
Parasit Vectors ; 15(1): 383, 2022 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-36271393

RESUMO

BACKGROUND: The amino acid transporter protein cationic amino acid transporter 1 (CAT1) is part of the nutrient sensor in the fat body of mosquitoes. A member of the SLC7 family of cationic amino acid transporters, it is paramount for the detection of elevated amino acid levels in the mosquito hemolymph after a blood meal and the subsequent changes in gene expression in the fat body. METHODS: We performed a re-annotation of Aedes aegypti cationic amino acid transporters (CATs) and selected the C-terminal tail of CAT1 to perform a yeast two-hybrid screen to identify putative interactors of this protein. One interesting interacting protein we identified was general control nonderepressible 1 (GCN1). We determined the expression pattern of GCN1 in several adult organs and structures using qRT-PCR and western blots. Finally, we knocked down GCN1 using double-stranded RNA and identified changes in downstream signaling intermediates and the effects of knockdown on vitellogenesis and fecundity. RESULTS: In a screen for Ae. aegypti CAT1-interacting proteins we identified GCN1 as a putative interactor. GCN1 is highly expressed in the ovaries and fat body of the mosquito. We provide evidence that eukaryotic translation initiation factor 2 subunit alpha (eIF2α) phosphorylation changed during vitellogenesis and that RNA interference knockdown of GCN1 in whole mosquitoes reduced egg clutch sizes of treated mosquitoes relative to controls. CONCLUSIONS: Aedes aegypti CAT1 and GCN1 are likely interacting partners and GCN1 is likely necessary for proper egg development. Our data suggest that GCN1 is part of a nutrient sensor mechanism in various mosquito tissues involved in vitellogenesis.


Assuntos
Aedes , Animais , Aedes/genética , Aedes/metabolismo , Transportador 1 de Aminoácidos Catiônicos/genética , Transportador 1 de Aminoácidos Catiônicos/metabolismo , RNA de Cadeia Dupla/metabolismo , Fator de Iniciação 2 em Procariotos/genética , Fator de Iniciação 2 em Procariotos/metabolismo , Saccharomyces cerevisiae/genética , Sistemas de Transporte de Aminoácidos Básicos/genética , Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Aminoácidos/genética , Fertilidade
4.
Amino Acids ; 54(7): 1101-1108, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35377022

RESUMO

The cationic amino acid transporter 1 (CAT1/SLC7A1) plays a key role in the cellular uptake or export of L-arginine and some of its derivatives. This study investigated the effect of 113 chemically diverse and commonly used drugs (at 20 and 200 µM) on the CAT1-mediated cellular uptake of L-arginine, L-homoarginine, and asymmetric dimethylarginine (ADMA). Twenty-three (20%) of the tested substances showed weak inhibitory or stimulatory effects, but only verapamil showed consistent inhibitory effects on CAT1-mediated transport of all tested substrates.


Assuntos
Arginina , Transportador 1 de Aminoácidos Catiônicos , Transporte Biológico , Transportador 1 de Aminoácidos Catiônicos/genética , Transportador 1 de Aminoácidos Catiônicos/metabolismo , Homoarginina/metabolismo
5.
Cancer Sci ; 113(1): 65-78, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34699654

RESUMO

Cancer susceptibility is a critical factor in the understanding of carcinogenesis. Intraperitoneal (i.p.) injection of an iron chelate, ferric nitrilotriacetate (Fe-NTA), produces hydroxyl radicals via Fenton reaction to induce ferroptosis in renal proximal tubules. Rats or mice subjected to repeated i.p. injections of Fe-NTA develop renal cell carcinoma (RCC). To elucidate the molecular mechanisms that cause susceptibility to renal carcinogenesis, we first established an inter-strain difference in the susceptibility to Fe-NTA-induced renal carcinogenesis in mice. Based on a previous observation of a low incidence of RCC with this model in C57BL/6J strain mice, we investigated A/J strain mice here, which demonstrated significantly higher susceptibility to Fe-NTA-induced renal carcinogenesis. Homozygous deletion of the Cdkn2a/2b tumor suppressor locus was detected for the first time in A/J strain mice. Focusing on ferroptosis and iron metabolism, we explored the mechanisms involved that lead to the difference in RCC development. We compared the protective responses in the kidney of A/J and C57BL/6J strains after Fe-NTA treatment. After 3-week Fe-NTA treatment, A/J mice maintained higher levels of expression of glutathione peroxidase 4 and xCT (SLC7A11), leading to a lower level of lipid peroxidation. Simultaneously, A/J mice had decreased expression of transferrin receptor and increased expression of ferritin to greater degrees than C57BL/6 mice. After a single Fe-NTA injection, higher levels of oxidative cell damage and cytosolic catalytic Fe(II) were observed in C57BL/6J mice, accompanied by a greater increase in lipocalin-2. Lipocalin-2 deficiency significantly decreased oxidative renal damage. Our results suggest that a genetic trait favoring ferroptosis resistance contributes to high susceptibility to Fe-NTA-induced RCC in A/J strain.


Assuntos
Carcinoma de Células Renais/patologia , Compostos Férricos/efeitos adversos , Redes Reguladoras de Genes , Neoplasias Renais/patologia , Ácido Nitrilotriacético/análogos & derivados , Deleção de Sequência , Animais , Carcinoma de Células Renais/induzido quimicamente , Carcinoma de Células Renais/genética , Transportador 1 de Aminoácidos Catiônicos/genética , Inibidor de Quinase Dependente de Ciclina p15/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Ferritinas/genética , Ferroptose , Regulação Neoplásica da Expressão Gênica , Homozigoto , Injeções Intraperitoneais , Neoplasias Renais/induzido quimicamente , Neoplasias Renais/genética , Peroxidação de Lipídeos , Lipocalina-2/genética , Masculino , Camundongos , Neoplasias Experimentais , Ácido Nitrilotriacético/efeitos adversos , Estresse Oxidativo , Receptores da Transferrina/genética , Especificidade da Espécie , Regulação para Cima
6.
Biochim Biophys Acta Mol Basis Dis ; 1867(12): 166216, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34314821

RESUMO

Maternal physiological dyslipidemia (MPD) supports fetal development in human pregnancy. However, some women develop maternal supraphysiological dyslipidemia (MSPD: increased total cholesterol (TC) and triglycerides (TG) levels). MSPH is present in normal and also in gestational diabetes mellitus (GDM) pregnancies. MSPD and GDM associate with fetoplacental endothelial dysfunction, producing alterations in nitric oxide (NO)-L-arginine/arginase metabolism. Nevertheless, the effect of MSPD on GDM, and how this synergy alters fetoplacental endothelial function is unknown. Therefore, the aim of this study was to evaluate in human umbilical vein endothelial cells, the effects of MSPD in GDM and how these pathologies together affect the fetoplacental endothelial function. 123 women at term of pregnancy were classified as MPD (n = 40), MSPD (n = 35), GDM with normal lipids (GDM-MPD, n = 23) and with increased lipids (GDM-MSPD, n = 25). TC ≥291 mg/dL and TG ≥275 mg/dL were considered as MSPD. Endothelial NO synthase (eNOS), human cationic amino acid transporter 1 (hCat1), and arginase II protein abundance and activity, were assayed in umbilical vein endothelial cells. In MSPD and GDM-MSPD, TC and TG increased respect to MPD and GDM-MPD. eNOS activity was reduced in MSPD and GDM-MSPD, but increased in GDM-MPD compared with MPD. However, decreased tetrahydrobiopterin levels were observed in all groups compared with MPD. Increased hCat1 protein and L-arginine transport were observed in both GDM groups compared with MPD. However, the transport was higher in GDM-MSPD compared to GDM-MPD. Higher Arginase II protein and activity were observed in GDM-MSPD compared with MPD. Thus, MSPD in GDM pregnancies alters fetal endothelial function associated with NO metabolism.


Assuntos
Arginase/genética , Transportador 1 de Aminoácidos Catiônicos/genética , Diabetes Gestacional/sangue , Dislipidemias/sangue , Adulto , Arginase/metabolismo , Arginina/metabolismo , Colesterol/sangue , Diabetes Gestacional/patologia , Dislipidemias/genética , Dislipidemias/patologia , Endotélio/metabolismo , Endotélio/patologia , Feminino , Desenvolvimento Fetal/genética , Feto/metabolismo , Humanos , Transporte de Íons/genética , Óxido Nítrico/genética , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Testes de Função Placentária , Gravidez , Transdução de Sinais/genética , Triglicerídeos/sangue
7.
Cancer Sci ; 112(2): 563-574, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33211385

RESUMO

Copy number alterations detected by comparative genomic hybridization (CGH) can lead to the identification of novel cancer-related genes. We analyzed chromosomal aberrations in a set of 100 human primary colorectal cancers (CRCs) using CGH and found a solute carrier (SLC) 7A1 gene, which encodes cationic amino acid transporter 1 (CAT1) with 14 putative transmembrane domains, in a chromosome region (13q12.3) with a high frequency of gene amplifications. SLC7A1/CAT1 is a transporter responsible for the uptake of cationic amino acids (arginine, lysine, and ornithine) essential for cellular growth. Microarray and PCR analyses have revealed that mRNA transcribed from CAT1 is overexpressed in more than 70% of human CRC samples, and RNA interference-mediated knockdown of CAT1 inhibited the cell growth of CRCs. Rats were immunized with rat hepatoma cells expressing CAT1 tagged with green fluorescent protein (GFP), and rat splenocytes were fused with mouse myeloma cells. Five rat monoclonal antibodies (mAbs) (CA1 ~ CA5) reacting with HEK293 cells expressing CAT1-GFP in a GFP expression-dependent manner were selected from established hybridoma clones. Novel anti-CAT1 mAbs selectively reacted with human CRC tumor tissues compared with adjacent normal tissues according to immuno-histochemical staining and bound strongly to numerous human cancer cell lines by flow cytometry. Anti-CAT1 mAbs exhibited internalization activity, antibody-dependent cellular cytotoxicity, and migration inhibition activity against CRC cell lines. Furthermore, CA2 inhibited the in vivo growth of human HT29 and SW-C4 CRC tumors in nude mice. This study suggested CAT1 to be a promising target for mAb therapy against CRCs.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos Imunológicos/farmacologia , Transportador 1 de Aminoácidos Catiônicos/antagonistas & inibidores , Neoplasias Colorretais/genética , Animais , Transportador 1 de Aminoácidos Catiônicos/genética , Linhagem Celular Tumoral , Amplificação de Genes , Xenoenxertos , Humanos , Camundongos , Camundongos Nus , Ratos
8.
Clin Sci (Lond) ; 134(20): 2755-2769, 2020 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-33034619

RESUMO

Heart failure (HF) is associated with impaired L-arginine transport. In the present study, we tested the hypothesis that augmented L-arginine transport prevents the loss of kidney function in HF. Renal function was assessed in wildtype mice (WT), transgenic mice with HF (dilated cardiomyopathy, DCM) and double transgenic mice (double transgenic mice with DCM and CAT-1 overexpression, HFCAT-1) with HF and endothelial-specific overexpression of the predominant L-arginine transporter, cationic amino acid transporter-1 (CAT-1) (n=4-8/group). Cardiac function was assessed via echocardiography and left ventricular catheterisation. Renal function was assessed via quantification of albuminuria and creatinine clearance. Plasma nitrate and nitrite levels together with renal fibrosis and inflammatory markers were also quantified at study end. Albumin/creatinine ratio was two-fold greater in DCM mice than in WT mice (P=0.002), and tubulointerstitial and glomerular fibrosis were approximately eight- and three-fold greater, respectively, in DCM mice than in WT mice (P≤0.02). Critically, urinary albumin/creatinine ratio and tubulointerstitial and glomerular fibrosis were less in HFCAT-1 mice than in DCM mice (P<0.05). Renal CAT-1 expression and plasma nitrate and nitrite levels were less in DCM mice compared with WT (P≤0.03) but was greater in HFCAT-1 mice than in DCM mice (P≤0.009). Renal expression of IL-10 was less in DCM mice compared with WT (P<0.001) but was greater in HFCAT-1 mice compared with DCM mice (P=0.02). Our data provide direct evidence that augmented L-arginine transport prevents renal fibrosis, inflammation and loss of kidney function in HF.


Assuntos
Transportador 1 de Aminoácidos Catiônicos/metabolismo , Células Endoteliais/metabolismo , Insuficiência Cardíaca/fisiopatologia , Testes de Função Renal , Rim/fisiopatologia , Animais , Pressão Sanguínea , Peso Corporal , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/fisiopatologia , Transportador 1 de Aminoácidos Catiônicos/genética , Fibrose , Regulação da Expressão Gênica , Insuficiência Cardíaca/sangue , Insuficiência Cardíaca/genética , Inflamação/genética , Inflamação/patologia , Rim/imunologia , Rim/patologia , Masculino , Camundongos Transgênicos , Miocárdio/patologia , Nitratos/sangue , Nitritos/sangue , Tamanho do Órgão , Especificidade de Órgãos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
9.
Fish Physiol Biochem ; 46(6): 2281-2298, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32980952

RESUMO

Cationic amino acid transporter 1 (Cat-1 alias Slc7a1) is a Na+-independent carrier system involved in transport and absorption of the cationic amino acids lysine, arginine, histidine, and ornithine and has also been shown to be indispensable in a large variety of biological processes. Starting from isolated full-length zebrafish (Danio rerio) cDNA for slc7a1a, we performed comparative and phylogenetic sequence analysis, investigated the conservation of the gene during vertebrate evolution, and defined tissue expression during zebrafish development. Whole mount in situ hybridization first detected slc7a1a transcripts in somites, eyes, and brain at 14 h post-fertilization (hpf) with additional expression in the distal nephron at 24 hpf and in branchial arches at 3 days post-fertilization (dpf), with significant increase by 5 dpf. Taken together, the expression analysis of the zebrafish Cat-1 system gene slc7a1a suggests a functional role(s) during the early development of the central nervous system, muscle, gills, and kidney. Graphical abstract.


Assuntos
Transportador 1 de Aminoácidos Catiônicos/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Encéfalo/metabolismo , Região Branquial/metabolismo , Transportador 1 de Aminoácidos Catiônicos/química , Embrião não Mamífero , Olho/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Néfrons/metabolismo , Filogenia , Análise de Sequência de DNA , Análise de Sequência de Proteína , Somitos/metabolismo , Proteínas de Peixe-Zebra/química
10.
Virol J ; 17(1): 57, 2020 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-32321527

RESUMO

Bovine leukemia virus (BLV) causes enzootic bovine leukosis, the most common neoplastic disease in cattle. We previously reported the development and protocol of the luminescence syncytium induction assay (LuSIA), a method for evaluating BLV infectivity based on CC81-GREMG cells. These cells form syncytia expressing enhanced green fluorescent protein when co-cultured with BLV-infected cells. Recently, we confirmed CAT1/SLC7A1 functions as a receptor of BLV. Here, we focused on CAT1/SLC7A1 to increase the sensitivity of LuSIA. We constructed a bovine CAT1-expressing plasmid and established a new CC81-GREMG-derived reporter cell line highly expressing bovine CAT1 (CC81-GREMG-CAT1). The new LuSIA protocol using CC81-GREMG-CAT1 cells measures cell-to-cell infectivity and cell-free infectivity of BLV faster and with greater sensitivity than the previous protocol using CC81-GREMG. The new LuSIA protocol is quantitative and more sensitive than the previous assay based on CC81-GREMG cells and will facilitate the development of several new BLV assays.


Assuntos
Transportador 1 de Aminoácidos Catiônicos/genética , Células Gigantes/virologia , Vírus da Leucemia Bovina/imunologia , Medições Luminescentes/métodos , Receptores Virais/genética , Animais , Bovinos , Linhagem Celular , Técnicas de Cocultura , Proteínas de Fluorescência Verde/genética , Vírus da Leucemia Bovina/genética , Vírus da Leucemia Bovina/patogenicidade , Sensibilidade e Especificidade
11.
Science ; 368(6486): 85-89, 2020 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-32241947

RESUMO

Ferroptosis is a form of cell death that results from the catastrophic accumulation of lipid reactive oxygen species (ROS). Oncogenic signaling elevates lipid ROS production in many tumor types and is counteracted by metabolites that are derived from the amino acid cysteine. In this work, we show that the import of oxidized cysteine (cystine) via system xC - is a critical dependency of pancreatic ductal adenocarcinoma (PDAC), which is a leading cause of cancer mortality. PDAC cells used cysteine to synthesize glutathione and coenzyme A, which, together, down-regulated ferroptosis. Studying genetically engineered mice, we found that the deletion of a system xC - subunit, Slc7a11, induced tumor-selective ferroptosis and inhibited PDAC growth. This was replicated through the administration of cyst(e)inase, a drug that depletes cysteine and cystine, demonstrating a translatable means to induce ferroptosis in PDAC.


Assuntos
Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Cisteína/deficiência , Ferroptose , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Animais , Transportador 1 de Aminoácidos Catiônicos/genética , Linhagem Celular Tumoral , Cistationina gama-Liase/administração & dosagem , Cistationina gama-Liase/farmacologia , Cistina/metabolismo , Ferroptose/efeitos dos fármacos , Ferroptose/genética , Deleção de Genes , Humanos , Camundongos , Camundongos Mutantes
12.
FASEB J ; 33(12): 14516-14527, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31648581

RESUMO

Bovine leukemia virus (BLV) is the causative agent of enzootic bovine leukosis, the most common neoplastic disease of cattle, which is closely related to human T-cell leukemia viruses. BLV has spread worldwide and causes a serious problem for the cattle industry. The cellular receptor specifically binds with viral envelope glycoprotein (Env), and this attachment mediates cell fusion to lead virus entry. BLV Env reportedly binds to cationic amino acid transporter 1 (CAT1)/solute carrier family 7 member 1 (SLC7A1), but whether the CAT1/SLC7A1 is an actual receptor for BLV remains unknown. Here, we showed that CAT1 functioned as an infection receptor, interacting with BLV particles. Cells expressing undetectable CAT1 levels were resistant to BLV infection but became highly susceptible upon CAT1 overexpression. CAT1 exhibited specific binding to BLV particles on the cell surface and colocalized with the Env in endomembrane compartments and membrane. Knockdown of CAT1 in permissive cells significantly reduced binding to BLV particles and BLV infection. Expression of CAT1 from various species demonstrated no species specificity for BLV infection, implicating CAT1 as a functional BLV receptor responsible for its broad host range. These findings provide insights for BLV infection and for developing new strategies for treating BLV and preventing its spread.-Bai, L., Sato, H., Kubo, Y., Wada, S., Aida, Y. CAT1/SLC7A1 acts as a cellular receptor for bovine leukemia virus infection.


Assuntos
Transportador 1 de Aminoácidos Catiônicos/metabolismo , Leucose Enzoótica Bovina/metabolismo , Vírus da Leucemia Bovina/metabolismo , Animais , Células CHO , Células COS , Transportador 1 de Aminoácidos Catiônicos/genética , Gatos , Bovinos , Chlorocebus aethiops , Cricetinae , Cricetulus , Leucose Enzoótica Bovina/virologia , Células HEK293 , Células HeLa , Humanos , Vírus da Leucemia Bovina/patogenicidade , Ligação Proteica , Ovinos , Suínos , Proteínas do Envelope Viral/metabolismo
13.
Arch Pharm (Weinheim) ; 352(6): e1800354, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31081964

RESUMO

Nineteen antioxidant pseudopeptides were designed and synthesized. They were confirmed as mild antioxidants, in which L1-11 was the most active antioxidant with a cellular antioxidant activity (CAA) value of 5.65 ± 0.64 µmol QE/g, and L1-12 was the second most active one (5.58 ± 0.66 µmol QE/g). The existence of nonnatural amino acids in L1-12 increased its stability. Pretreatment with L1-12 dose-dependently extended the lifespan of Caenorhabditis elegans. L1-12 improved resistance against UVB irradiation, oxidative stress induced by paraquat, and thermal shock. It decreased the reactive oxygen species level and upregulated the superoxide dismutase activity inside C. elegans. This pseudopeptide sensitively enhanced the expressions of the Cat-1 and Nhr-8 genes to reduce oxidative damage, leading to an extension of the lifespan. All the evidence support that L1-12 may probably be a potential antiageing agent.


Assuntos
Aminoácidos/síntese química , Antioxidantes/síntese química , Caenorhabditis elegans/efeitos dos fármacos , Desenho de Fármacos , Longevidade/efeitos dos fármacos , Peptídeos/síntese química , Aminoácidos/química , Aminoácidos/farmacologia , Animais , Antioxidantes/química , Antioxidantes/farmacologia , Caenorhabditis elegans/crescimento & desenvolvimento , Caenorhabditis elegans/efeitos da radiação , Proteínas de Caenorhabditis elegans/genética , Transportador 1 de Aminoácidos Catiônicos/genética , Relação Dose-Resposta a Droga , Estabilidade de Medicamentos , Expressão Gênica/efeitos dos fármacos , Longevidade/genética , Estrutura Molecular , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Peptídeos/química , Peptídeos/farmacologia , Receptores Citoplasmáticos e Nucleares/genética , Relação Estrutura-Atividade
14.
Virology ; 532: 82-87, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31035110

RESUMO

Cytoplasmic tails of envelope (Env) glycoproteins of many retroviruses inhibit their membrane fusion activity. The cytoplasmic 16-amino acid peptide of ecotropic murine leukemia virus (E-MLV) Env protein, called the R-peptide, also inhibits the membrane fusion activity of the Env protein. However, the molecular mechanism of the inhibition has not been elucidated yet. In this study, we found that R-peptide-containing Env protein of E-MLV binds to the cell surface receptor cationic amino acid transporter-1 (CAT-1) with weaker affinity than R-peptide-truncated Env protein. Consistent with this result, R-peptide-containing Env protein had less efficient inhibition of E-MLV vector infection than R-peptide-truncated Env protein. R-peptide truncation has been reported to induce conformational change in the surface subunit of E-MLV Env protein that interacts with the receptor. Taken together, our findings indicate that R-peptide truncation induces conformational change in the receptor-binding domain of the E-MLV Env protein and facilitates the Env-receptor interaction.


Assuntos
Transportador 1 de Aminoácidos Catiônicos/metabolismo , Interações Hospedeiro-Patógeno/genética , Vírus da Leucemia Murina/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores Virais/metabolismo , Proteínas do Envelope Viral/metabolismo , Animais , Sítios de Ligação , Transportador 1 de Aminoácidos Catiônicos/química , Transportador 1 de Aminoácidos Catiônicos/genética , Regulação da Expressão Gênica , Células HEK293 , Humanos , Vírus da Leucemia Murina/genética , Fusão de Membrana , Camundongos , Células NIH 3T3 , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteólise , Receptores de Superfície Celular/química , Receptores de Superfície Celular/genética , Receptores Virais/química , Receptores Virais/genética , Transdução de Sinais , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
15.
Am J Physiol Lung Cell Mol Physiol ; 316(4): L598-L607, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30628488

RESUMO

The pathology of acute lung injury (ALI) involves inducible nitric oxide (NO) synthase (iNOS)-derived NO-induced apoptosis of pulmonary endothelial cells. In vitro, iNOS-derived NO production has been shown to depend on the uptake of l-arginine by the cationic amino acid transporters (CAT). To test the hypothesis that mice deficient in CAT-2 ( slc7a2-/- on a C57BL/6 background) would be protected from hyperoxia-induced ALI, mice ( slc7a2-/- or wild-type) were placed in >95% oxygen (hyperoxia) or 21% oxygen (control) for 60 h. In wild-type mice exposed to hyperoxia, the exhaled nitric oxide (exNO) was twofold greater than in wild-type mice exposed to normoxia ( P < 0.005), whereas in slc7a2-/- mice there was no significant difference between exNO in animals exposed to hyperoxia or normoxia ( P = 0.95). Hyperoxia-exposed wild-type mice had greater ( P < 0.05) lung resistance and a lower ( P < 0.05) lung compliance than did hyperoxia-exposed slc7a2-/- mice. The lung wet-to-dry weight ratio was greater ( P < 0.005) in the hyperoxia-exposed wild-type mice than in hyperoxia-exposed slc7a2-/- mice. Neutrophil infiltration was lower ( P < 0.05) in the hyperoxia-exposed slc7a2-/- mice than in the hyperoxia-exposed wild-type mice as measured by myeloperoxidase activity. The protein concentration in bronchoalveolar lavage fluid was lower ( P < 0.001) in the hyperoxia-exposed slc7a2-/- mice than in similarly exposed wild-type mice. The percent of TUNEL-positive cells in the lung following hyperoxia exposure was significantly lower ( P < 0.001) in the slc7a2-/- mice than in the wild-type mice. These results are consistent with our hypothesis that lack of CAT-2 protects mice from acute lung injury.


Assuntos
Lesão Pulmonar Aguda/prevenção & controle , Sistemas de Transporte de Aminoácidos Básicos/deficiência , Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/metabolismo , Sistemas de Transporte de Aminoácidos Básicos/genética , Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Animais , Transportador 1 de Aminoácidos Catiônicos/genética , Ciclo-Oxigenase 2/genética , Modelos Animais de Doenças , Feminino , Hiperóxia/complicações , Hiperóxia/metabolismo , Interleucina-6/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo III/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
16.
Animal ; 13(2): 326-332, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29929568

RESUMO

Peptide transporter 1 (SLC15A1, PepT1), excitatory amino acid transporter 3 (SLC1A1, EAAT3) and cationic amino acid transporter 1 (SLC7A1, CAT1) were identified as genes responsible for the transport of small peptides and amino acids. The tissue expression pattern of rabbit (SLC15A1, SLC7A1 and SLC1A1) across the digestive tract remains unclear. The present study investigated SLC15A1, SLC7A1 and SLC1A1 gene expression patterns across the digestive tract at different stages of development and in response to dietary protein levels. Real time-PCR results indicated that SLC15A1, SLC7A1 and SLC1A1 genes throughout the rabbits' entire development and were expressed in all tested rabbit digestive sites, including the stomach, duodenum, jejunum, ileum, colon and cecum. Furthermore, SLC7A1 and SLC1A1 mRNA expression occurred in a tissue-specific and time-associated manner, suggesting the distinct transport ability of amino acids in different tissues and at different developmental stages. The most highly expressed levels of all three genes were in the duodenum, ileum and jejunum in all developmental stages. All increased after lactation. With increased dietary protein levels, SLC7A1 mRNA levels in small intestine and SLC1A1 mRNA levels in duodenum and ileum exhibited a significant decreasing trend. Moreover, rabbits fed a normal level of protein had the highest levels of SLC15A1 mRNA in the duodenum and jejunum (P<0.05). In conclusion, gene mRNA differed across sites and with development suggesting time and sites related differences in peptide and amino acid absorption in rabbits. The effects of dietary protein on expression of the three genes were also site specific.


Assuntos
Transportador 1 de Aminoácidos Catiônicos/genética , Proteínas Alimentares/metabolismo , Digestão/genética , Transportador 3 de Aminoácido Excitatório/genética , Expressão Gênica , Transportador 1 de Peptídeos/genética , Coelhos/genética , Animais , Transportador 1 de Aminoácidos Catiônicos/metabolismo , Transportador 3 de Aminoácido Excitatório/metabolismo , Transportador 1 de Peptídeos/metabolismo , Coelhos/crescimento & desenvolvimento , Coelhos/metabolismo , Distribuição Aleatória
17.
BMC Genet ; 19(1): 103, 2018 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-30419805

RESUMO

BACKGROUND: Identification of genes underlying production traits is a key aim of the mink research community. Recent availability of genomic tools have opened the possibility for faster genetic progress in mink breeding. Availability of mink genome assembly allows genome-wide association studies in mink. RESULTS: In this study, we used genotyping-by-sequencing to obtain single nucleotide polymorphism (SNP) genotypes of 2496 mink. After multiple rounds of filtering, we retained 28,336 high quality SNPs and 2352 individuals for a genome-wide association study (GWAS). We performed the first GWAS for body weight, behavior, along with 10 traits related to fur quality in mink. CONCLUSIONS: Combining association results with existing functional information of genes and mammalian phenotype databases, we proposed WWC3, MAP2K4, SLC7A1 and USP22 as candidate genes for body weight and pelt length in mink.


Assuntos
Tamanho Corporal/genética , Vison/genética , Polimorfismo de Nucleotídeo Único , Animais , Transportador 1 de Aminoácidos Catiônicos/genética , Estudo de Associação Genômica Ampla , Genótipo , MAP Quinase Quinase 4/genética , Vison/fisiologia , Fenótipo , Tioléster Hidrolases/genética
18.
Vet Res ; 49(1): 95, 2018 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-30236161

RESUMO

Transmissible gastroenteritis virus (TGEV) is a coronavirus that causes severe diarrhea in suckling piglets. TGEV primarily targets and infects porcine intestinal epithelial cells, which play an important role in nutrient absorption. However, the effects of TGEV infection on nutrient absorption in swine have not yet been investigated. In this study, we evaluated the impact of TGEV infection on arginine uptake using the porcine small intestinal epithelial cell line IPEC-J2 as a model system. High performance liquid chromatography (HPLC) analyses showed that TGEV infection leads to reduced arginine uptake at 48 hours post-infection (hpi). Expression of cationic amino acid transporter 1 (CAT-1) was attenuated as well. TGEV infection induced activation of phospho-protein kinase C α (p-PKC α), phospho-epidermal growth factor receptor (p-EGFR), and enhanced the expression of caveolin-1, all of which appear to be involved in down-regulating arginine uptake and CAT-1 expression. These results illuminate the relationship between TGEV infection and nutrient absorption, and further our understanding of the mechanisms of TGEV infection.


Assuntos
Arginina/metabolismo , Transportador 1 de Aminoácidos Catiônicos/genética , Regulação para Baixo , Gastroenterite Suína Transmissível/genética , Regulação da Expressão Gênica , Vírus da Gastroenterite Transmissível/fisiologia , Animais , Transportador 1 de Aminoácidos Catiônicos/metabolismo , Linhagem Celular , Gastroenterite Suína Transmissível/fisiopatologia , Gastroenterite Suína Transmissível/virologia , Intestino Delgado/metabolismo , Intestino Delgado/virologia , Transdução de Sinais , Suínos
19.
Nitric Oxide ; 80: 24-31, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30056252

RESUMO

Blockade of the mineralocorticoid receptor (MCR) has been shown to improve endothelial function far beyond blood pressure control. In the current studies we have looked at the effect of MCR antagonists on cationic amino acid transporter-1 (CAT-1), a major modulator of endothelial nitric oxide (NO) generation. Using radio-labeled arginine, {[3H] l-arginine} uptake was determined in human umbilical vein endothelial cells (HUVEC) following incubation with either spironolactone or eplerenone with or without silencing of MCR. Western blotting for CAT-1, PKCα and their phosphorylated forms were performed. NO generation was measured by using Griess reaction assay. Both Spironolactone and eplerenone significantly increased endothelial arginine transport, an effect which was further augmented by co-incubation with aldosterone, and blunted by either silencing of MCR or co-administration of amiloride. Following MCR blockade, we identified two bands for CAT-1. The addition of tunicamycin (an inhibitor of protein glycosylation) or MCR silencing resulted in disappearance of the extra band and prevented the increase in arginine transport. Only spironolactone decreased CAT-1 phosphorylation through inhibition of PKCα (CAT-1 inhibitor). Subsequently, incubation with either MCR antagonists significantly augmented NO2/NO3 levels (stable NO metabolites) and this was attenuated by silencing of MCR or tunicamycin. GO 6076 (PKCα inhibitor) intensified the increase of NO metabolites only in eplerenone treated cells. In conclusion spironolactone and eplerenone augment arginine transport and NO generation through modulation of CAT-1 in endothelial cells. Both MCR antagonists activate CAT-1 by inducing its glycosylation while only spironolactone inhibits PKCα.


Assuntos
Arginina/metabolismo , Transportador 1 de Aminoácidos Catiônicos/metabolismo , Antagonistas de Receptores de Mineralocorticoides/farmacologia , Óxido Nítrico/metabolismo , Espironolactona/farmacologia , Transporte Biológico/efeitos dos fármacos , Transportador 1 de Aminoácidos Catiônicos/genética , Eplerenona/farmacologia , Glicosilação/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana , Humanos , Fosforilação/efeitos dos fármacos , Proteína Quinase C-alfa/antagonistas & inibidores , Receptores de Mineralocorticoides/genética , Receptores de Mineralocorticoides/metabolismo , Transdução de Sinais/efeitos dos fármacos
20.
Placenta ; 66: 26-35, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29884299

RESUMO

INTRODUCTION: Gestational diabetes mellitus (GDM) is associated with fetoplacental endothelial dysfunction, which may be induced by hyperglycemia. We hypothesized that endothelial exosomes, which are extracellular nanovesicles affecting endothelial function, play a role in the high glucose (HG)-induced endothelial dysfunction. METHODS: Exosomes were isolated from HUVECs incubated with basal glucose (5.5 mmol/L; HUVEC- BG; exo-BG) and from HUVECs incubated with HG for 24 h (25 mmol/L; HUVEC-HG; exo-HG) in exosome-free medium. Exosomes were isolated and characterized by ultracentrifugation, sucrose gradient, electron microscopy, nanotracking analysis and Western blotting. HUVEC-BG and HUVEC-HG were exposed to exo-BG and exo-HG in two different concentrations: 5 µg and 1 µg exosome protein/mL. The exosomal effect on endothelial cell function was determined by wound healing assay, expression of endothelial nitric oxide synthase (eNOS), human cationic amino acid transporter type 1 (hCAT-1), vascular endothelial growth factor (VEGF) and intracellular adhesion molecule type 1 (ICAM-1) by Western blotting, qPCR or flow cytometry. RESULTS: HG increased the exosomal release from HUVECs, endothelial wound healing and expression of phosphorylated (P∼Ser1177)-eNOS, hCAT-1, VEGF and ICAM-1. Exo-HG also increased endothelial cell wound healing, P∼Ser1177-eNOS, hCAT-1 and ICAM-1 expression in HUVEC-BG. Exo-BG reverted the effect of HG on endothelial cell wound healing and hCAT-1 mRNA expression to normal values. DISCUSSION: Our results show that HG may induce endothelial dysfunction in HUVECs and that exosomes from HUVEC-HG mimicked some of the effects of HG. This study contributes to the unraveling of the mechanism by which hyperglycemia affects the fetoplacental vasculature in GDM.


Assuntos
Diabetes Gestacional/fisiopatologia , Exossomos/fisiologia , Hiperglicemia/fisiopatologia , Placenta/fisiopatologia , Transportador 1 de Aminoácidos Catiônicos/genética , Diabetes Gestacional/genética , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiopatologia , Exossomos/efeitos dos fármacos , Exossomos/ultraestrutura , Feminino , Glucose/metabolismo , Glucose/farmacologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Molécula 1 de Adesão Intercelular/genética , Modelos Biológicos , Óxido Nítrico Sintase Tipo III/genética , Circulação Placentária/fisiologia , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Cicatrização/efeitos dos fármacos , Cicatrização/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...