Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 218
Filter
1.
Amino Acids ; 55(10): 1213-1222, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37572157

ABSTRACT

Amino acids are essential for the survival of all living organisms and living cells. Amino acid transporters mediate the transport and absorption of amino acids, and the dysfunction of these proteins can induce human diseases. Cationic amino acid transporters (CAT family, SLC7A1-4, and SLC7A14) are considered to be a group of transmembrane transporters, of which SLC7A1-3 are essential for arginine transport in mammals. Numerous studies have shown that CAT family-mediated arginine transport is involved in signal crosstalk between malignant tumor cells and immune cells, especially T cells. The modulation of extracellular arginine concentration has entered a number of clinical trials and achieved certain therapeutic effects. Here, we review the role of CAT family on tumor cells and immune infiltrating cells in malignant tumors and explore the therapeutic strategies to interfere with extracellular arginine concentration, to elaborate its application prospects. CAT family members may be used as biomarkers for certain cancer entities and might be included in new ideas for immunotherapy of malignant tumors.


Subject(s)
Amino Acid Transport Systems, Basic , Neoplasms , Animals , Humans , Amino Acid Transport Systems, Basic/genetics , Amino Acid Transport Systems, Basic/metabolism , Arginine/metabolism , Amino Acids/metabolism , Cationic Amino Acid Transporter 1/metabolism , Biological Transport , Cationic Amino Acid Transporter 2/metabolism , Mammals/metabolism , Tumor Microenvironment
2.
J Biomol Struct Dyn ; 41(23): 13580-13594, 2023.
Article in English | MEDLINE | ID: mdl-36762692

ABSTRACT

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.


Subject(s)
Arginine , Cationic Amino Acid Transporter 1 , Animals , Humans , Arginine/metabolism , Cationic Amino Acid Transporter 1/genetics , Cationic Amino Acid Transporter 1/metabolism , Nitric Oxide Synthase , Molecular Dynamics Simulation , Amino Acid Transport Systems, Basic/metabolism , Water/metabolism , Mammals/metabolism
3.
Arthritis Res Ther ; 24(1): 234, 2022 10 17.
Article in English | MEDLINE | ID: mdl-36253807

ABSTRACT

BACKGROUND: Abnormal proliferation of fibroblast-like synoviocytes (FLSs) in the synovial lining layer is the primary cause of synovial hyperplasia and joint destruction in rheumatoid arthritis (RA). Currently, the relationship between metabolic abnormalities and FLS proliferation is a new focus of investigation. However, little is known regarding the relationship between amino acid metabolism and RA. METHODS: The concentrations of amino acids and cytokines in the synovial fluid of RA (n = 9) and osteoarthritis (OA, n = 9) were detected by LC-MS/MS and CBA assay, respectively. The mRNA and protein expression of cationic amino acid transporter-1 (CAT-1) were determined in FLSs isolated from RA and OA patients by real-time PCR and western blotting. MTT assay, cell cycle, apoptosis, invasion, and cytokine secretion were determined in FLSs knocked down of CAT-1 using siRNA or treated with D-arginine under normoxic and hypoxic culture conditions. A mouse collagen-induced arthritis (CIA) model was applied to test the therapeutic potential of blocking the uptake of L-arginine in vivo. RESULTS: L-rginine was upregulated in the synovial fluid of RA patients and was positively correlated with the elevation of the cytokines IL-1ß, IL-6, and IL-8. Further examination demonstrated that CAT-1 was the primary transporter for L-arginine and was overexpressed on RA FLSs compared to OA FLSs. Moreover, knockdown of CAT-1 using siRNA or inhibition of L-arginine uptake using D-arginine significantly suppressed L-arginine metabolism, cell proliferation, migration, and cytokine secretion in RA FLSs under normoxic and hypoxic culture conditions in vitro but increased cell apoptosis in a dose-dependent manner. Meanwhile, in vivo assays revealed that an L-arginine-free diet or blocking the uptake of L-arginine using D-arginine suppressed arthritis progression in CIA mice. CONCLUSION: CAT-1 is upregulated and promotes FLS proliferation by taking up L-arginine, thereby promoting RA progression.


Subject(s)
Arginine , Arthritis, Experimental , Arthritis, Rheumatoid , Cationic Amino Acid Transporter 1 , Synoviocytes , Animals , Mice , Amino Acids/metabolism , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/drug therapy , Cationic Amino Acid Transporter 1/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Chromatography, Liquid , Cytokines/metabolism , Fibroblasts/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Mice, Inbred CBA , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , RNA, Small Interfering/therapeutic use , Synovial Membrane/metabolism , Synoviocytes/metabolism , Tandem Mass Spectrometry
4.
Parasit Vectors ; 15(1): 383, 2022 Oct 21.
Article in English | MEDLINE | ID: mdl-36271393

ABSTRACT

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.


Subject(s)
Aedes , Animals , Aedes/genetics , Aedes/metabolism , Cationic Amino Acid Transporter 1/genetics , Cationic Amino Acid Transporter 1/metabolism , RNA, Double-Stranded/metabolism , Prokaryotic Initiation Factor-2/genetics , Prokaryotic Initiation Factor-2/metabolism , Saccharomyces cerevisiae/genetics , Amino Acid Transport Systems, Basic/genetics , Amino Acid Transport Systems, Basic/metabolism , Amino Acids/genetics , Fertility
5.
J Interferon Cytokine Res ; 42(9): 501-512, 2022 09.
Article in English | MEDLINE | ID: mdl-35900262

ABSTRACT

Arginine is one of the host semiessential amino acids with diverse biological activities, and arginine depletion is associated with the incidence of many diseases. Arginine depletion induced by diet-derived interferon gamma (IFN-γ) leads to malignant transformation and impaired milk quality in healthy lactating bovine mammary epithelial cells (BMECs). However, the molecular mechanism of IFN-γ-induced arginine depletion is unclear. In this study, the BMEC cell line, mammary alveolar cells-large T antigen cells (MAC-T), was stimulated with IFN-γ (10 ng/mL) for 24 h, and cellular arginine and ornithine quantified by liquid chromatography-tandem mass spectrometry. Carnosine synthase 1 (CARNS1) was identified from RNA-seq data, CARNS1 knockdown was achieved using an shRNA interfering plasmid. The expression levels of CARNS1, argininosuccinate synthetase 1 (ASS1), mitogen-activated protein kinase 11 (p38 MAPK), and phosphorylated (p)-p38, and their cognate genes, were analyzed by Western blotting and real-time quantitative polymerase chain reaction. The results showed that IFN-γ inhibited the biosynthesis of arginine, but enhanced its catalysis via disruption of key enzymes involved in arginine metabolism. IFN-γ also inhibited the expression of CARNS1, ASS1, and cationic amino acid transporter 1, while activating the expression and phosphorylation of p38. However, knockdown of CARNS1 reduced arginine level and ASS1 expression and block of either the IFN-γ receptor IFN-γ receptor 2 or p38 relieved both the expression of Carnosine synthase 1 (CARNS1) and ASS1. In summary, these results indicate that IFN-γ induced arginine depletion through inhibition of CARNS1 signaling via activation of p38 in BMECs. These findings provide a novel insight for IFN-γ-related disease control strategies in dairy cows.


Subject(s)
Carnosine , Interferon-gamma , Animals , Antigens, Viral, Tumor/metabolism , Arginine/metabolism , Arginine/pharmacology , Argininosuccinate Synthase/metabolism , Carnosine/metabolism , Cationic Amino Acid Transporter 1/metabolism , Cattle , Epithelial Cells/metabolism , Female , Lactation , Mitogen-Activated Protein Kinase 11/metabolism , Ornithine/metabolism , RNA, Small Interfering , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Amino Acids ; 54(7): 1101-1108, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35377022

ABSTRACT

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.


Subject(s)
Arginine , Cationic Amino Acid Transporter 1 , Biological Transport , Cationic Amino Acid Transporter 1/genetics , Cationic Amino Acid Transporter 1/metabolism , Homoarginine/metabolism
7.
Drug Deliv ; 29(1): 316-327, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35037525

ABSTRACT

Ultrasound nanodroplets (NDs) have been reported as a promising nanocarrier for siRNA delivery depending on its unique strengths of sonoporation. Presently, common means for NDs-mediated siRNA delivery is through electrostatic interaction, but challenges like cationic toxicity still exist. In this study, we demonstrated a novel strategy to construct negatively charged and ultrasound (US)-responsive O-carboxymethyl chitosan (O-CMS) NDs as a siRNA targeted delivery system through three-way junction of bacteriophage phi29 DNA packaging motor (3WJ-pRNA) nanotechnology. 39nt A10-3.2 aptamer targeting prostate specific membrane antigen (PSMA) and 21nt siRNA against cationic amino acid transporter 1 (siCAT-1) were annealed to 3WJ-pRNA scaffold via complementation with an extended sequence. The cholesterol molecule attached to one branch facilitates the 3WJ-pRNA nanoparticles anchoring onto NDs. The desired O-CMS NDs with siRNA-loading and RNA-aptamer modification (A10-3.2/siCAT-1/3WJ-NDs) were successfully prepared, which were with spherical shapes, core-shell structures and uniform in sizes (198 nm with PDI 0.3). As a main proportion of shell, O-CMC showed a certain anti-tumor effects. In vitro studies demonstrated that A10-3.2/siCAT-1/3WJ-NDs exhibited good contrast-enhanced US imaging, buffering capacity and high bio-safety, were able to deliver siCAT-1 to PSMA-overexpressed prostate cancer cells under US irradiation, thus silence the CAT-1 expression, and consequently suppressing 22RV1 cell proliferation and migration. Taken overall, our findings provide a promising strategy to develop negatively charged and US-responsive NDs for tumor-targeted siRNA delivery.


Subject(s)
Aptamers, Nucleotide/pharmacology , Cationic Amino Acid Transporter 1/pharmacology , Chitosan/analogs & derivatives , Nanoparticle Drug Delivery System/chemistry , RNA, Small Interfering/pharmacology , Ultrasonography, Interventional/methods , Aptamers, Nucleotide/administration & dosage , Bacillus Phages/drug effects , Cationic Amino Acid Transporter 1/administration & dosage , Cell Line, Tumor , Cell Movement/drug effects , Chemistry, Pharmaceutical , Chitosan/chemistry , Drug Carriers/chemistry , Drug Liberation , Humans , Particle Size , Prostate-Specific Antigen/drug effects , RNA, Small Interfering/administration & dosage , Surface Properties
8.
Cancer Sci ; 113(1): 65-78, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34699654

ABSTRACT

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.


Subject(s)
Carcinoma, Renal Cell/pathology , Ferric Compounds/adverse effects , Gene Regulatory Networks , Kidney Neoplasms/pathology , Nitrilotriacetic Acid/analogs & derivatives , Sequence Deletion , Animals , Carcinoma, Renal Cell/chemically induced , Carcinoma, Renal Cell/genetics , Cationic Amino Acid Transporter 1/genetics , Cyclin-Dependent Kinase Inhibitor p15/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Ferritins/genetics , Ferroptosis , Gene Expression Regulation, Neoplastic , Homozygote , Injections, Intraperitoneal , Kidney Neoplasms/chemically induced , Kidney Neoplasms/genetics , Lipid Peroxidation , Lipocalin-2/genetics , Male , Mice , Neoplasms, Experimental , Nitrilotriacetic Acid/adverse effects , Oxidative Stress , Receptors, Transferrin/genetics , Species Specificity , Up-Regulation
9.
Amino Acids ; 53(9): 1441-1454, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34245369

ABSTRACT

L-arginine is a versatile amino acid with a number of bioactive metabolites. Increasing evidence implicates altered arginine metabolism in the aging and neurodegenerative processes. The present study, for the first time, determined the effects of sex and estrous cycle on the brain and blood (plasma) arginine metabolic profile in naïve rats. Female rats displayed significantly lower levels of L-arginine in the frontal cortex and three sub-regions of the hippocampus when compared to male rats. Moreover, female rats had significantly higher levels of L-arginine and γ-aminobutyric acid, but lower levels of L-ornithine, agmatine and putrescine, in plasma relative to male rats. The observed sex difference in brain L-arginine appeared to be independent of the enzymes involved in its metabolism, de novo synthesis and blood-to-brain transport (cationic acid transporter 1 protein expression at least), as well as circulating L-arginine. While the estrous cycle did not affect L-arginine and its metabolites in the brain, there were estrous cycle phase-dependent changes in plasma L-arginine. These findings demonstrate the sex difference in brain L-arginine in the estrous cycle-independent manner. Since peripheral blood has been increasingly used to identify biomarkers of brain pathology, the influences of sex and estrous cycle on blood arginine metabolic profile need attention when experimental research involves female rodents.


Subject(s)
Arginine/metabolism , Brain/metabolism , Cationic Amino Acid Transporter 1/metabolism , Estrous Cycle , Metabolome , Animals , Arginine/blood , Biological Transport , Female , Male , Rats , Rats, Sprague-Dawley , Sex Factors
10.
Biochim Biophys Acta Mol Basis Dis ; 1867(12): 166216, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34314821

ABSTRACT

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.


Subject(s)
Arginase/genetics , Cationic Amino Acid Transporter 1/genetics , Diabetes, Gestational/blood , Dyslipidemias/blood , Adult , Arginase/metabolism , Arginine/metabolism , Cholesterol/blood , Diabetes, Gestational/pathology , Dyslipidemias/genetics , Dyslipidemias/pathology , Endothelium/metabolism , Endothelium/pathology , Female , Fetal Development/genetics , Fetus/metabolism , Humans , Ion Transport/genetics , Nitric Oxide/genetics , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/genetics , Placental Function Tests , Pregnancy , Signal Transduction/genetics , Triglycerides/blood
11.
Clin Sci (Lond) ; 135(1): 105-108, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33404050

ABSTRACT

Although the numbers of patients affected by cardiorenal syndrome keeps increasing, we lack a complete understanding of the molecular pathways involved in its development and progression. Nitric oxide synthase (NOS) may play a role in cardiorenal syndrome, particularly cardiorenal syndrome type 2 (CRS2). However, complexities and paradoxical clinical findings have limited translation. In the current Clinical Science, Giam et al. (Clinical Science (2020) 134, 2755-2769) highlight the role of a key NOS substrate transporter, the cationic amino acid transporter-1, in preserving renal function in CRS2. In this commentary, we introduce the cardiorenal syndrome and the putative role that nitric oxide (NO) may play in the development of this disease and discuss the exciting findings of Giam et al. (Clinical Science (2020) 134, 2755-2769) and their tantalizing translational implications.


Subject(s)
Cardio-Renal Syndrome , Cationic Amino Acid Transporter 1 , Arginine , Humans , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II/metabolism
12.
Cancer Sci ; 112(2): 563-574, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33211385

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Cationic Amino Acid Transporter 1/antagonists & inhibitors , Colorectal Neoplasms/genetics , Animals , Cationic Amino Acid Transporter 1/genetics , Cell Line, Tumor , Gene Amplification , Heterografts , Humans , Mice , Mice, Nude , Rats
13.
Clin Sci (Lond) ; 134(20): 2755-2769, 2020 10 30.
Article in English | MEDLINE | ID: mdl-33034619

ABSTRACT

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.


Subject(s)
Cationic Amino Acid Transporter 1/metabolism , Endothelial Cells/metabolism , Heart Failure/physiopathology , Kidney Function Tests , Kidney/physiopathology , Animals , Blood Pressure , Body Weight , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/physiopathology , Cationic Amino Acid Transporter 1/genetics , Fibrosis , Gene Expression Regulation , Heart Failure/blood , Heart Failure/genetics , Inflammation/genetics , Inflammation/pathology , Kidney/immunology , Kidney/pathology , Male , Mice, Transgenic , Myocardium/pathology , Nitrates/blood , Nitrites/blood , Organ Size , Organ Specificity , RNA, Messenger/genetics , RNA, Messenger/metabolism
14.
Fish Physiol Biochem ; 46(6): 2281-2298, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32980952

ABSTRACT

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.


Subject(s)
Cationic Amino Acid Transporter 1/genetics , Zebrafish Proteins/genetics , Zebrafish/embryology , Zebrafish/genetics , Amino Acid Sequence , Animals , Base Sequence , Brain/metabolism , Branchial Region/metabolism , Cationic Amino Acid Transporter 1/chemistry , Embryo, Nonmammalian , Eye/metabolism , Gene Expression Regulation, Developmental , Nephrons/metabolism , Phylogeny , Sequence Analysis, DNA , Sequence Analysis, Protein , Somites/metabolism , Zebrafish Proteins/chemistry
15.
Virol J ; 17(1): 57, 2020 04 22.
Article in English | MEDLINE | ID: mdl-32321527

ABSTRACT

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.


Subject(s)
Cationic Amino Acid Transporter 1/genetics , Giant Cells/virology , Leukemia Virus, Bovine/immunology , Luminescent Measurements/methods , Receptors, Virus/genetics , Animals , Cattle , Cell Line , Coculture Techniques , Green Fluorescent Proteins/genetics , Leukemia Virus, Bovine/genetics , Leukemia Virus, Bovine/pathogenicity , Sensitivity and Specificity
16.
Science ; 368(6486): 85-89, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32241947

ABSTRACT

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.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cysteine/deficiency , Ferroptosis , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Animals , Cationic Amino Acid Transporter 1/genetics , Cell Line, Tumor , Cystathionine gamma-Lyase/administration & dosage , Cystathionine gamma-Lyase/pharmacology , Cystine/metabolism , Ferroptosis/drug effects , Ferroptosis/genetics , Gene Deletion , Humans , Mice , Mice, Mutant Strains
17.
Nitric Oxide ; 99: 7-16, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32165314

ABSTRACT

Insulin regulates the l-arginine/nitric oxide (NO) pathway in human umbilical vein endothelial cells (HUVECs), increasing the plasma membrane expression of the l-arginine transporter hCAT-1 and inducing vasodilation in umbilical and placental veins. Placental vascular relaxation induced by insulin is dependent of large conductance calcium-activated potassium channels (BKCa), but the role of KCa channels on l-arginine transport and NO synthesis is still unknown. The aim of this study was to determine the contribution of KCa channels in both insulin-induced l-arginine transport and NO synthesis, and its relationship with placental vascular relaxation. HUVECs, human placental vein endothelial cells (HPVECs) and placental veins were freshly isolated from umbilical cords and placenta from normal pregnancies. Cells or tissue were incubated in absence or presence of insulin and/or tetraethylammonium, 1-[(2-chlorophenyl)diphenylmethyl]-1H-pyrazole, iberiotoxin or NG-nitro-l-arginine methyl ester. l-Arginine uptake, plasma membrane polarity, NO levels, hCAT-1 expression and placenta vascular reactivity were analyzed. The inhibition of intermediate-conductance KCa (IKCa) and BKCa increases l-arginine uptake, which was related with protein abundance of hCAT-1 in HUVECs. IKCa and BKCa activities contribute to NO-synthesis induced by insulin but are not directly involved in insulin-stimulated l-arginine uptake. Long term incubation (8 h) with insulin increases the plasma membrane hyperpolarization and hCAT-1 expression in HUVECs and HPVECs. Insulin-induced relaxation in placental vasculature was reversed by KCa inhibition. The results show that the activity of IKCa and BKCa channels are relevant for both physiological regulations of NO synthesis and vascular tone regulation in the human placenta, acting as a part of negative feedback mechanism for autoregulation of l-arginine transport in HUVECs.


Subject(s)
Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Nitric Oxide/metabolism , Umbilical Veins/metabolism , Adult , Arginine/metabolism , Cationic Amino Acid Transporter 1/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Female , Human Umbilical Vein Endothelial Cells , Humans , Insulin/pharmacology , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Large-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Peptides/pharmacology , Placenta/drug effects , Placenta/metabolism , Potassium Channel Blockers/pharmacology , Pregnancy , Pyrazoles/pharmacology , Umbilical Veins/drug effects , Young Adult
19.
Amino Acids ; 52(3): 499-503, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32008093

ABSTRACT

A direct inhibiting effect of NO on the function of CAT-1 and -2A has been postulated to occur via nitrosylation of cysteine residues in the transporters. Neither the NO donor SNAP nor a mixture of SIN-1 and Spermine NONOate, that generates the strong nitrosating agent N2O3, reduced CAT-mediated L-arginine transport. Direct nitros(yl)ation does either not occur in CATs or does not affect their transport function. A regulatory effect of NO or nitrosating agents on CAT-mediated transport under physiological conditions seems, therefore, unlikely.


Subject(s)
Amino Acid Transport Systems, Basic/metabolism , Cationic Amino Acid Transporter 1/metabolism , Cysteine/metabolism , Nitric Oxide/metabolism , Animals , Arginine/metabolism , Humans , Oocytes , Xenopus laevis
20.
Biochim Biophys Acta Mol Basis Dis ; 1866(2): 165370, 2020 02 01.
Article in English | MEDLINE | ID: mdl-30660686

ABSTRACT

Gestational diabetes mellitus (GDM) is a disease of pregnancy associated with maternal and foetal hyperglycaemia and altered foetoplacental vascular function. Human foetoplacental microvascular and macrovascular endothelium from GDM pregnancy show increased maximal l-arginine transport capacity via the human cationic amino acid transporter 1 (hCAT-1) isoform and nitric oxide (NO) synthesis by the endothelial NO synthase (eNOS). These alterations are paralleled by lower maximal transport activity of the endogenous nucleoside adenosine via the human equilibrative nucleoside transporter 1 (hENT1) and activation of adenosine receptors. A causal relationship has been described for adenosine-activation of A2A adenosine receptors, hCAT-1, and eNOS activity (i.e. the Adenosine/l-Arginine/Nitric Oxide, ALANO, signalling pathway). Insulin restores these alterations in GDM via activation of insulin receptor A (IR-A) form in the macrovascular but IR-A and IR-B forms in the microcirculation of the human placenta. Adipokines are secreted from adipocytes influencing the foetoplacental metabolic and vascular function. Various adipokines are dysregulated in GDM, with adiponectin and leptin playing major roles. Abnormal plasma concentration of these adipokines and the activation or their receptors are involved in the pathophysiology of GDM. However, involvement of adipokines, adenosine, and insulin receptors and membrane transporters in the aetiology of this disease of pregnancy is unknown. This review focuses on the pathophysiology of insulin and adenosine receptors and l-arginine and adenosine membranes transporters giving an overview of the key adipokines leptin and adiponectin in the foetoplacental vasculature in GDM. This article is part of a Special Issue entitled: Membrane Transporters and Receptors in Pregnancy Metabolic Complications edited by Luis Sobrevia.


Subject(s)
Adenosine/metabolism , Diabetes, Gestational/metabolism , Endothelium, Vascular/metabolism , Insulin/metabolism , Receptors, Adipokine/metabolism , Adipokines/blood , Antigens, CD/metabolism , Arginine/metabolism , Biological Transport/physiology , Cationic Amino Acid Transporter 1/metabolism , Endothelium/metabolism , Equilibrative Nucleoside Transporter 1/metabolism , Female , GTPase-Activating Proteins , Humans , Nitric Oxide , Nitric Oxide Synthase Type III/metabolism , Placenta/metabolism , Pregnancy , Protein Isoforms , Receptor, Insulin/metabolism , Receptors, Purinergic P1/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...