51
|
Li Y, Ren Q, Zhu L, Li Y, Li J, Zhang Y, Zheng G, Han T, Sun S, Feng F. Involvement of methylation of MicroRNA-122, -125b and -106b in regulation of Cyclin G1, CAT-1 and STAT3 target genes in isoniazid-induced liver injury. BMC Pharmacol Toxicol 2018; 19:11. [PMID: 29554950 PMCID: PMC5859513 DOI: 10.1186/s40360-018-0201-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 03/09/2018] [Indexed: 12/31/2022] Open
Abstract
Background This investigation aimed to evaluate the role of methylation in the regulation of microRNA (miR)-122, miR-125b and miR-106b gene expression and the expression of their target genes during isoniazid (INH)-induced liver injury. Methods Rats were given INH 50 mg kg− 1·d− 1 once per day for 3, 7, 10, 14, 21 and 28 days and were sacrificed. Samples of blood and liver were obtained. Results We analysed the methylation and expression levels of miR-122, miR-125b and miR-106b and their potential gene targets in livers. Liver tissue pathologies, histological scores and alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities changed, indicating the occurrence of liver injury. Relative expression levels of miR-122, miR-125b and miR-106b genes in the liver decreased after INH administration and correlated with the scores of liver pathology and serum AST and ALT activities, suggesting that miR-122, miR-125b and miR-106b are associated with INH-induced liver injury. The amount of methylated miR-122, miR-125b and miR-106b in the liver increased after INH administration and correlated with their expression levels, suggesting the role of methylation in regulating miRNA gene expression. Two miR-122 gene targets, cell cycle protein G1 (Cyclin G1) and cationic amino acid transporter-1 (CAT-1), also increased at the mRNA and protein levels, which suggests that lower levels of miR-122 contribute to the upregulation of Cyclin G1 and CAT-1 and might play a role in INH-induced liver injury. Signal transducer and activator of transcription 3 (STAT3) was a common target gene of miR-125b and miR-106b, and its expression levels of mRNA and protein increased after INH administration. The protein expression of phosphorylated (p)-STAT3 and the mRNA expression of RAR-related orphan receptor gamma (RORγt) regulated by p-STAT3 also increased. Meanwhile, the mRNA and protein expression of interleukin (IL)-17 regulated by RORγt, and the mRNA and protein expression of CXCL1 and MIP-2 regulated by IL-17 increased after INH administration. These results demonstrate that lower levels of hepatic miR-125b and miR-106b contribute to the upregulation of STAT3 in stimulating the secretion of inflammatory factors during INH-induced liver injury. Conclusions Our results suggested that DNA methylation probably regulates the expression of miRNA genes (miR-122, miR-125b, and miR-106b), affecting the expression of their gene targets (Cyclin G1, CAT-1, and STAT3) and participating in the process of INH-induced liver injury. Electronic supplementary material The online version of this article (10.1186/s40360-018-0201-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yuhong Li
- Hebei Province Key Laboratory of Occupational Health and Safety for Coal Industry, School of Public Health, North China University of Science and Technology, No.21 Bohai Road, Tangshan, 063210, People's Republic of China
| | - Qi Ren
- Hebei Province Key Laboratory of Occupational Health and Safety for Coal Industry, School of Public Health, North China University of Science and Technology, No.21 Bohai Road, Tangshan, 063210, People's Republic of China
| | - Lingyan Zhu
- Hebei Province Key Laboratory of Occupational Health and Safety for Coal Industry, School of Public Health, North China University of Science and Technology, No.21 Bohai Road, Tangshan, 063210, People's Republic of China
| | - Yingshu Li
- Hebei Province Key Laboratory of Occupational Health and Safety for Coal Industry, School of Public Health, North China University of Science and Technology, No.21 Bohai Road, Tangshan, 063210, People's Republic of China
| | - Jinfeng Li
- Hebei Province Key Laboratory of Occupational Health and Safety for Coal Industry, School of Public Health, North China University of Science and Technology, No.21 Bohai Road, Tangshan, 063210, People's Republic of China
| | - Yiyang Zhang
- Hebei Province Key Laboratory of Occupational Health and Safety for Coal Industry, School of Public Health, North China University of Science and Technology, No.21 Bohai Road, Tangshan, 063210, People's Republic of China
| | - Guoying Zheng
- Hebei Province Key Laboratory of Occupational Health and Safety for Coal Industry, School of Public Health, North China University of Science and Technology, No.21 Bohai Road, Tangshan, 063210, People's Republic of China
| | - Tiesheng Han
- Hebei Province Key Laboratory of Occupational Health and Safety for Coal Industry, School of Public Health, North China University of Science and Technology, No.21 Bohai Road, Tangshan, 063210, People's Republic of China
| | - Shufeng Sun
- College of Nursing and Rehabilitation, North China University of Science and Technology, Tangshan, 063210, China
| | - Fumin Feng
- Hebei Province Key Laboratory of Occupational Health and Safety for Coal Industry, School of Public Health, North China University of Science and Technology, No.21 Bohai Road, Tangshan, 063210, People's Republic of China.
| |
Collapse
|
52
|
Kim SH, Roszik J, Grimm EA, Ekmekcioglu S. Impact of l-Arginine Metabolism on Immune Response and Anticancer Immunotherapy. Front Oncol 2018; 8:67. [PMID: 29616189 PMCID: PMC5864849 DOI: 10.3389/fonc.2018.00067] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 03/01/2018] [Indexed: 12/19/2022] Open
Abstract
The progression from neoplastic initiation to malignancy happens in part because of the failure of immune surveillance. Cancer cells successfully escape immune recognition and elimination and create an immune-suppressive microenvironment. A suppressive metabolic microenvironment may also contribute to ineffective T-cell function. Tumor progression is characterized by a complex network of interactions among different cell types that cooperatively exploit metabolic reprogramming. As we start to recognize that cancer cells use different metabolism processes than normal cells do, a better understanding of the functional mechanisms of the regulation and reprogramming of the metabolic landscape in cancer cells is crucial to successful immunotherapy strategies. However, the exact role of metabolism in T cells and in the tumor microenvironment is not known. One pathway that plays an important role in the regulation of immune cell reactivity is arginine metabolism, which has complex cellular functions. l-arginine and its downstream metabolites (e.g., ornithine and citrulline) could be essential to T-cell activation and thus modulate innate and adaptive immunity to further promote tumor survival and growth. Identifying metabolic targets that mediate immunosuppression and are fundamental to sustaining tumor growth is key to increasing the efficacy of immunotherapies.
Collapse
Affiliation(s)
- Sun-Hee Kim
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jason Roszik
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Elizabeth A Grimm
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Suhendan Ekmekcioglu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
53
|
Prudner BC, Sun F, Kremer JC, Xu J, Huang C, Sai KKS, Morgan Z, Leeds H, McConathy J, Van Tine BA. Amino Acid Uptake Measured by [ 18F]AFETP Increases in Response to Arginine Starvation in ASS1-Deficient Sarcomas. Am J Cancer Res 2018; 8:2107-2116. [PMID: 29721066 PMCID: PMC5928874 DOI: 10.7150/thno.22083] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 02/13/2018] [Indexed: 01/04/2023] Open
Abstract
Rational: In a subset of cancers, arginine auxotrophy occurs due to the loss of expression of argininosuccinate synthetase 1 (ASS1). This loss of ASS1 expression makes cancers sensitive to arginine starvation that is induced by PEGylated arginine deiminase (ADI-PEG20). Although ADI-PEG20 treatment is effective, it does have important limitations. Arginine starvation is only beneficial in patients with cancers that are ASS1-deficient. Also, these tumors may metabolically reprogram to express ASS1, transforming them from an auxotrophic phenotype to a prototrophic phenotype and thus rendering ADI-PEG20 ineffective. Due to these limitations of ADI-PEG20 treatment and the potential for developing resistance, non-invasive tools to monitor sensitivity to arginine starvation are needed. Methods: Within this study, we assess the utility of a novel positron emission tomography (PET) tracer to determine sarcomas reliant on extracellular arginine for survival by measuring changes in amino acid transport in arginine auxotrophic sarcoma cells treated with ADI-PEG20. The uptake of the 18F-labeled histidine analogue, (S)-2-amino-3-[1-(2-[18F]fluoroethyl)-1H-[1,2,3]triazol-4-yl]propanoic acid (AFETP), was assessed in vitro and in vivo using human-derived sarcoma cell lines. In addition, we examined the expression and localization of cationic amino acid transporters in response to arginine starvation with ADI-PEG20. Results: In vitro studies revealed that in response to ADI-PEG20 treatment, arginine auxotrophs increase the uptake of L-[3H]arginine and [18F]AFETP due to an increase in the expression and localization to the plasma membrane of the cationic amino acid transporter CAT-1. Furthermore, in vivo PET imaging studies in mice with arginine-dependent osteosarcoma xenografts showed increased [18F]AFETP uptake in tumors 4 days after ADI-PEG20 treatment compared to baseline. Conclusion: CAT-1 transporters localizes to the plasma membrane as a result of arginine starvation with ADI-PEG20 in ASS1-deficient tumor cells and provides a mechanism for using cationic amino acid transport substrates such as [18F]AFETP for identifying tumors susceptible to ADI-PEG20 treatment though non-invasive PET imaging techniques. These findings indicate that [18F]AFETP-PET may be suitable for the early detection of tumor response to arginine depletion due to ADI-PEG20 treatment.
Collapse
|
54
|
Muxel SM, Aoki JI, Fernandes JCR, Laranjeira-Silva MF, Zampieri RA, Acuña SM, Müller KE, Vanderlinde RH, Floeter-Winter LM. Arginine and Polyamines Fate in Leishmania Infection. Front Microbiol 2018; 8:2682. [PMID: 29379478 PMCID: PMC5775291 DOI: 10.3389/fmicb.2017.02682] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 12/22/2017] [Indexed: 01/22/2023] Open
Abstract
Leishmania is a protozoan parasite that alternates its life cycle between the sand fly and the mammalian host macrophages, involving several environmental changes. The parasite responds to these changes by promoting a rapid metabolic adaptation through cellular signaling modifications that lead to transcriptional and post-transcriptional gene expression regulation and morphological modifications. Molecular approaches such as gene expression regulation, next-generation sequencing (NGS), microRNA (miRNA) expression profiling, in cell Western blot analyses and enzymatic activity profiling, have been used to characterize the infection of murine BALB/c and C57BL/6 macrophages, as well as the human monocytic cell-lineage THP-1, with Leishmania amazonensis wild type (La-WT) or arginase knockout (La-arg-). These models are being used to elucidate physiological roles of arginine and polyamines pathways and the importance of arginase for the establishment of the infection. In this review, we will describe the main aspects of Leishmania-host interaction, focusing on the arginine and polyamines pathways and pointing to possible targets to be used for prognosis and/or in the control of the infection. The parasite enzymes, arginase and nitric oxide synthase-like, have essential roles in the parasite survival and in the maintenance of infection. On the other hand, in mammalian macrophages, defense mechanisms are activated inducing alterations in the mRNA, miRNA and enzymatic profiles that lead to the control of infection. Furthermore, the genetic background of both parasite and host are also important to define the fate of infection.
Collapse
Affiliation(s)
- Sandra M Muxel
- Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - Juliana I Aoki
- Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - Juliane C R Fernandes
- Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | | | - Ricardo A Zampieri
- Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - Stephanie M Acuña
- Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - Karl E Müller
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Rubia H Vanderlinde
- Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - Lucile M Floeter-Winter
- Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
55
|
Polyamine concentration is increased in thoracic ascending aorta of patients with bicuspid aortic valve. Heart Vessels 2017; 33:327-339. [DOI: 10.1007/s00380-017-1087-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 11/13/2017] [Indexed: 12/26/2022]
|
56
|
Osmanyan AK, Ghazi Harsini S, Mahdavi R, Fisinin VI, Arkhipova AL, Glazko TT, Kovalchuk SN, Kosovsky GY. Intestinal amino acid and peptide transporters in broiler are modulated by dietary amino acids and protein. Amino Acids 2017; 50:353-357. [DOI: 10.1007/s00726-017-2510-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Accepted: 11/04/2017] [Indexed: 01/05/2023]
|
57
|
Hyun H, Lee SE, Son YJ, Shin MY, Park YG, Kim EY, Park SP. Cell Synchronization by Rapamycin Improves the Developmental Competence of Porcine SCNT Embryos. Cell Reprogram 2017; 18:195-205. [PMID: 27253629 DOI: 10.1089/cell.2015.0090] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The cell cycle stage of donor cells influences the success of somatic cell nuclear transfer (SCNT). This study investigated the effects of rapamycin treatment on synchronization of porcine fibroblasts in comparison with control and serum-starved cells, SCNT donor cell viability, and SCNT-derived embryo development. Porcine fibroblasts were treated with 0.1, 1, 10, and 100 μM rapamycin for 1 or 3 days. The proportion of cells in G0/G1 phase was significantly higher among cells treated with 1 μM rapamycin for 3 days (D3-1R) than among control and serum-starved cells (p < 0.05). In comparison with control cells, rapamycin-treated cells exhibited reduced proliferation, similar to serum-starved cells. The viability (as assessed by the MTT assay) of D3-1R-treated cells was good, similar to control cells, showing their quality was maintained. To confirm nutrient regulation by rapamycin treatment, we checked the transcript levels of nutrient transporter genes (SLC2A2, SLC2A4, SLC6A14, and SLC7A1). These levels were significantly lower in D3-1R-treated cells than in control cells (p < 0.01). We performed SCNT with D3-1R-treated cells (SCNT(D3-1R)) to confirm the effect of cell cycle synchronization by rapamycin treatment. Although SCNT(D3-1R) embryos did not have an increased fusion rate, their cleavage and blastocyst formation rates were significantly higher than those of control embryos (p < 0.05). Regarding embryo quality, the numbers of total and apoptotic cells per blastocyst were increased and decreased, respectively, in SCNT(D3-1R) blastocysts. The mRNA levels of developmental (CDX2 and CDH1) and proapoptotic (FAS and CASP3) genes were significantly higher and lower, respectively, in SCNT(D3-1R) blastocysts than in control blastocysts (p < 0.05). These results demonstrate that rapamycin treatment affects the cell cycle synchronization of donor cells and enhances the developmental potential of porcine SCNT embryos.
Collapse
Affiliation(s)
- Hyuk Hyun
- 1 Stem cell Research Center, Jeju National University , Jeju, Jeju Special Self-Governing Province, Korea.,2 Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University , Jeju, Jeju Special Self-Governing Province, Korea
| | - Seung-Eun Lee
- 1 Stem cell Research Center, Jeju National University , Jeju, Jeju Special Self-Governing Province, Korea.,2 Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University , Jeju, Jeju Special Self-Governing Province, Korea
| | - Yeo-Jin Son
- 1 Stem cell Research Center, Jeju National University , Jeju, Jeju Special Self-Governing Province, Korea.,2 Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University , Jeju, Jeju Special Self-Governing Province, Korea
| | - Min-Young Shin
- 1 Stem cell Research Center, Jeju National University , Jeju, Jeju Special Self-Governing Province, Korea.,2 Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University , Jeju, Jeju Special Self-Governing Province, Korea
| | - Yun-Gwi Park
- 1 Stem cell Research Center, Jeju National University , Jeju, Jeju Special Self-Governing Province, Korea.,2 Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University , Jeju, Jeju Special Self-Governing Province, Korea
| | - Eun-Young Kim
- 1 Stem cell Research Center, Jeju National University , Jeju, Jeju Special Self-Governing Province, Korea.,2 Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University , Jeju, Jeju Special Self-Governing Province, Korea.,3 Mirae Cell Bio , Gwangjin-gu, Seoul, Korea
| | - Se-Pill Park
- 1 Stem cell Research Center, Jeju National University , Jeju, Jeju Special Self-Governing Province, Korea.,2 Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University , Jeju, Jeju Special Self-Governing Province, Korea.,3 Mirae Cell Bio , Gwangjin-gu, Seoul, Korea
| |
Collapse
|
58
|
Bhatta A, Yao L, Xu Z, Toque HA, Chen J, Atawia RT, Fouda AY, Bagi Z, Lucas R, Caldwell RB, Caldwell RW. Obesity-induced vascular dysfunction and arterial stiffening requires endothelial cell arginase 1. Cardiovasc Res 2017; 113:1664-1676. [PMID: 29048462 PMCID: PMC6410953 DOI: 10.1093/cvr/cvx164] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 03/16/2017] [Accepted: 08/09/2017] [Indexed: 02/04/2023] Open
Abstract
AIMS Elevation of arginase activity has been linked to vascular dysfunction in diabetes and hypertension by a mechanism involving decreased nitric oxide (NO) bioavailability due to L-arginine depletion. Excessive arginase activity also can drive L-arginine metabolism towards the production of ornithine, polyamines, and proline, promoting proliferation of vascular smooth muscle cells and collagen formation, leading to perivascular fibrosis. We hypothesized that there is a specific involvement of arginase 1 expression within the vascular endothelial cells in this pathology. METHODS AND RESULTS To test this proposition, we used models of type 2 diabetes and metabolic syndrome. Studies were performed using wild type (WT), endothelial-specific arginase 1 knockout (EC-A1-/-) and littermate controls(A1con) mice fed high fat-high sucrose (HFHS) or normal diet (ND) for 6 months and isolated vessels exposed to palmitate-high glucose (PA/HG) media. Some WT mice or isolated vessels were treated with an arginase inhibitor, ABH [2-(S)-amino-6-boronohexanoic acid. In WT mice, the HFHS diet promoted increases in body weight, fasting blood glucose, and post-prandial insulin levels along with arterial stiffening and fibrosis, elevated blood pressure, decreased plasma levels of L-arginine, and elevated L-ornithine. The HFHS diet or PA/HG treatment also induced increases in vascular arginase activity along with oxidative stress, reduced vascular NO levels, and impaired endothelial-dependent vasorelaxation. All of these effects except obesity and hypercholesterolemia were prevented or significantly reduced by endothelial-specific deletion of arginase 1 or ABH treatment. CONCLUSION Vascular dysfunctions in diet-induced obesity are prevented by deletion of arginase 1 in vascular endothelial cells or arginase inhibition. These findings indicate that upregulation of arginase 1 expression/activity in vascular endothelial cells has an integral role in diet-induced cardiovascular dysfunction and metabolic syndrome.
Collapse
MESH Headings
- Animals
- Arginase/antagonists & inhibitors
- Arginase/genetics
- Arginase/metabolism
- Arginine/blood
- Blood Glucose/metabolism
- Blood Pressure
- Diabetes Mellitus, Experimental/enzymology
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Experimental/prevention & control
- Diabetes Mellitus, Type 2/enzymology
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/physiopathology
- Diabetes Mellitus, Type 2/prevention & control
- Diet, High-Fat
- Dietary Sucrose
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/pathology
- Endothelium, Vascular/physiopathology
- Enzyme Inhibitors/pharmacology
- Fibrosis
- Genetic Predisposition to Disease
- Insulin/blood
- Male
- Metabolic Syndrome/enzymology
- Metabolic Syndrome/genetics
- Metabolic Syndrome/physiopathology
- Metabolic Syndrome/prevention & control
- Mice, Inbred C57BL
- Mice, Knockout
- Nitric Oxide/metabolism
- Obesity/drug therapy
- Obesity/enzymology
- Obesity/genetics
- Obesity/physiopathology
- Ornithine/blood
- Oxidative Stress
- Phenotype
- Signal Transduction
- Vascular Diseases/enzymology
- Vascular Diseases/genetics
- Vascular Diseases/physiopathology
- Vascular Diseases/prevention & control
- Vascular Stiffness/drug effects
- Vasodilation
Collapse
Affiliation(s)
- Anil Bhatta
- Department of Pharmacology and Toxicology, Medical College of Georgia,
Augusta University, Augusta, GA 30912, USA
| | - Lin Yao
- Department of Pharmacology and Toxicology, Medical College of Georgia,
Augusta University, Augusta, GA 30912, USA
- School of Pharmaceutical Sciences, South China Research Centre for
Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR
China
| | - Zhimin Xu
- Vascular Biology Centre, Medical College of Georgia, Augusta University,
Augusta, GA 30912, USA
| | - Haroldo A. Toque
- Department of Pharmacology and Toxicology, Medical College of Georgia,
Augusta University, Augusta, GA 30912, USA
| | - Jijun Chen
- Department of Pharmacology and Toxicology, Medical College of Georgia,
Augusta University, Augusta, GA 30912, USA
| | - Reem T. Atawia
- Department of Pharmacology and Toxicology, Medical College of Georgia,
Augusta University, Augusta, GA 30912, USA
| | - Abdelrahman Y. Fouda
- Department of Pharmacology and Toxicology, Medical College of Georgia,
Augusta University, Augusta, GA 30912, USA
- Vascular Biology Centre, Medical College of Georgia, Augusta University,
Augusta, GA 30912, USA
| | - Zsolt Bagi
- Vascular Biology Centre, Medical College of Georgia, Augusta University,
Augusta, GA 30912, USA
- Department of Medicine, Medical College of Georgia, Augusta University,
Augusta, GA 30912, USA
| | - Rudolf Lucas
- Department of Pharmacology and Toxicology, Medical College of Georgia,
Augusta University, Augusta, GA 30912, USA
- Vascular Biology Centre, Medical College of Georgia, Augusta University,
Augusta, GA 30912, USA
| | - Ruth B. Caldwell
- Vascular Biology Centre, Medical College of Georgia, Augusta University,
Augusta, GA 30912, USA
- Department of Cell Biology and Anatomy, Medical College of Georgia, Augusta
University, Augusta, GA 30912, USA
- Veterans Administration Medical Centre, Augusta, GA 30912, USA
| | - Robert W. Caldwell
- Department of Pharmacology and Toxicology, Medical College of Georgia,
Augusta University, Augusta, GA 30912, USA
- Vascular Biology Centre, Medical College of Georgia, Augusta University,
Augusta, GA 30912, USA
| |
Collapse
|
59
|
Yi-lin L, Ke Z, Dan W, Xi-hong Z, Zheng R, Xin W, Yu-long Y. Dynamic feeding low and high methionine diets affect the diurnal rhythm of amino acid transporters and clock related genes in jejunum of laying hens. BIOL RHYTHM RES 2017. [DOI: 10.1080/09291016.2017.1395531] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Liu Yi-lin
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, China
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, China
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
- State Laboratory of Food Science and Technology, School of Food Science and Technology, Nanchang University, Nanchang, China
| | - Zhang Ke
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, China
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, China
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| | - Wan Dan
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, China
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, China
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| | - Zhou Xi-hong
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, China
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, China
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| | - Ruan Zheng
- State Laboratory of Food Science and Technology, School of Food Science and Technology, Nanchang University, Nanchang, China
| | - Wu Xin
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, China
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, China
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
- State Laboratory of Food Science and Technology, School of Food Science and Technology, Nanchang University, Nanchang, China
| | - Yin Yu-long
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, China
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, China
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
- State Laboratory of Food Science and Technology, School of Food Science and Technology, Nanchang University, Nanchang, China
| |
Collapse
|
60
|
Amino acid homeostasis and signalling in mammalian cells and organisms. Biochem J 2017; 474:1935-1963. [PMID: 28546457 PMCID: PMC5444488 DOI: 10.1042/bcj20160822] [Citation(s) in RCA: 319] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/08/2017] [Accepted: 03/10/2017] [Indexed: 12/19/2022]
Abstract
Cells have a constant turnover of proteins that recycle most amino acids over time. Net loss is mainly due to amino acid oxidation. Homeostasis is achieved through exchange of essential amino acids with non-essential amino acids and the transfer of amino groups from oxidised amino acids to amino acid biosynthesis. This homeostatic condition is maintained through an active mTORC1 complex. Under amino acid depletion, mTORC1 is inactivated. This increases the breakdown of cellular proteins through autophagy and reduces protein biosynthesis. The general control non-derepressable 2/ATF4 pathway may be activated in addition, resulting in transcription of genes involved in amino acid transport and biosynthesis of non-essential amino acids. Metabolism is autoregulated to minimise oxidation of amino acids. Systemic amino acid levels are also tightly regulated. Food intake briefly increases plasma amino acid levels, which stimulates insulin release and mTOR-dependent protein synthesis in muscle. Excess amino acids are oxidised, resulting in increased urea production. Short-term fasting does not result in depletion of plasma amino acids due to reduced protein synthesis and the onset of autophagy. Owing to the fact that half of all amino acids are essential, reduction in protein synthesis and amino acid oxidation are the only two measures to reduce amino acid demand. Long-term malnutrition causes depletion of plasma amino acids. The CNS appears to generate a protein-specific response upon amino acid depletion, resulting in avoidance of an inadequate diet. High protein levels, in contrast, contribute together with other nutrients to a reduction in food intake.
Collapse
|
61
|
Hellsten SV, Lekholm E, Ahmad T, Fredriksson R. The gene expression of numerous SLC transporters is altered in the immortalized hypothalamic cell line N25/2 following amino acid starvation. FEBS Open Bio 2017; 7:249-264. [PMID: 28174690 PMCID: PMC5292668 DOI: 10.1002/2211-5463.12181] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/09/2016] [Accepted: 12/10/2016] [Indexed: 12/20/2022] Open
Abstract
Amino acids are known to play a key role in gene expression regulation, and in mammalian cells, amino acid signaling is mainly mediated via two pathways, the mammalian target of rapamycin complex 1 (mTORC1) pathway and the amino acid responsive (AAR) pathway. It is vital for cells to have a system to sense amino acid levels, in order to control protein and amino acid synthesis and catabolism. Amino acid transporters are crucial in these pathways, due to both their sensing and transport functions. In this large-scale study, an immortalized mouse hypothalamic cell line (N25/2) was used to study the gene expression changes following 1, 2, 3, 5 or 16 h of amino acid starvation. We focused on genes encoding solute carriers (SLCs) and putative SLCs, more specifically on amino acid transporters. The microarray contained 28 270 genes and 86.2% of the genes were expressed in the cell line. At 5 h of starvation, 1001 genes were upregulated and 848 genes were downregulated, and among these, 47 genes from the SLC superfamily or atypical SLCs were found. Of these, 15 were genes encoding amino acid transporters and 32 were genes encoding other SLCs or atypical SLCs. Increased expression was detected for genes encoding amino acid transporters from system A, ASC, L, N, T, xc-, and y+. Using GO annotations, genes involved in amino acid transport and amino acid transmembrane transporter activity were found to be most upregulated at 3 h and 5 h of starvation.
Collapse
Affiliation(s)
- Sofie V Hellsten
- Department of Pharmaceutical Bioscience, Molecular Neuropharmacology Uppsala University Sweden; Department of Neuroscience, Functional Pharmacology Uppsala University Sweden
| | - Emilia Lekholm
- Department of Pharmaceutical Bioscience, Molecular Neuropharmacology Uppsala University Sweden
| | - Tauseef Ahmad
- Department of Neuroscience, Functional Pharmacology Uppsala University Sweden
| | - Robert Fredriksson
- Department of Pharmaceutical Bioscience, Molecular Neuropharmacology Uppsala University Sweden
| |
Collapse
|
62
|
O'Connor MA, Rastad JL, Green WR. The Role of Myeloid-Derived Suppressor Cells in Viral Infection. Viral Immunol 2017; 30:82-97. [PMID: 28051364 DOI: 10.1089/vim.2016.0125] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are heterogeneous immature myeloid cells that are well described as potent immune regulatory cells during human cancer and murine tumor models. Reports of MDSCs during viral infections remain limited, and their association with immunomodulation of viral diseases is still being defined. Here, we provide an overview of MDSCs or MDSC-like cells identified during viral infections, including murine viral models and human viral diseases. Understanding the similarities and/or differences of virally induced versus tumor-derived MDSCs will be important for designing future immunotherapeutic approaches.
Collapse
Affiliation(s)
- Megan A O'Connor
- 1 Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth , Lebanon , New Hampshire
| | - Jessica L Rastad
- 1 Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth , Lebanon , New Hampshire
| | - William R Green
- 1 Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth , Lebanon , New Hampshire.,2 Norris Cotton Cancer Center , Geisel School of Medicine at Dartmouth, Lebanon , New Hampshire
| |
Collapse
|
63
|
Mathieu C, Macêdo JP, Hürlimann D, Wirdnam C, Haindrich AC, Suter Grotemeyer M, González-Salgado A, Schmidt RS, Inbar E, Mäser P, Bütikofer P, Zilberstein D, Rentsch D. Arginine and Lysine Transporters Are Essential for Trypanosoma brucei. PLoS One 2017; 12:e0168775. [PMID: 28045943 PMCID: PMC5207785 DOI: 10.1371/journal.pone.0168775] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 12/06/2016] [Indexed: 12/14/2022] Open
Abstract
For Trypanosoma brucei arginine and lysine are essential amino acids and therefore have to be imported from the host. Heterologous expression in Saccharomyces cerevisiae mutants identified cationic amino acid transporters among members of the T. brucei AAAP (amino acid/auxin permease) family. TbAAT5-3 showed high affinity arginine uptake (Km 3.6 ± 0.4 μM) and high selectivity for L-arginine. L-arginine transport was reduced by a 10-times excess of L-arginine, homo-arginine, canavanine or arginine-β-naphthylamide, while lysine was inhibitory only at 100-times excess, and histidine or ornithine did not reduce arginine uptake rates significantly. TbAAT16-1 is a high affinity (Km 4.3 ± 0.5 μM) and highly selective L-lysine transporter and of the compounds tested, only L-lysine and thialysine were competing for L-lysine uptake. TbAAT5-3 and TbAAT16-1 are expressed in both procyclic and bloodstream form T. brucei and cMyc-tagged proteins indicate localization at the plasma membrane. RNAi-mediated down-regulation of TbAAT5 and TbAAT16 in bloodstream form trypanosomes resulted in growth arrest, demonstrating that TbAAT5-mediated arginine and TbAAT16-mediated lysine transport are essential for T. brucei. Growth of induced RNAi lines could partially be rescued by supplementing a surplus of arginine or lysine, respectively, while addition of both amino acids was less efficient. Single and double RNAi lines indicate that additional low affinity uptake systems for arginine and lysine are present in T. brucei.
Collapse
Affiliation(s)
| | - Juan P. Macêdo
- Institute of Plant Sciences, University of Bern, Bern, Switzerland
| | - Daniel Hürlimann
- Institute of Plant Sciences, University of Bern, Bern, Switzerland
| | - Corina Wirdnam
- Institute of Plant Sciences, University of Bern, Bern, Switzerland
| | | | | | | | - Remo S. Schmidt
- Swiss Tropical and Public Health Institute and University of Basel, Basel, Switzerland
| | - Ehud Inbar
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Pascal Mäser
- Swiss Tropical and Public Health Institute and University of Basel, Basel, Switzerland
| | - Peter Bütikofer
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Dan Zilberstein
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Doris Rentsch
- Institute of Plant Sciences, University of Bern, Bern, Switzerland
- * E-mail:
| |
Collapse
|
64
|
Morris SM. Arginine Metabolism Revisited. J Nutr 2016; 146:2579S-2586S. [PMID: 27934648 DOI: 10.3945/jn.115.226621] [Citation(s) in RCA: 226] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 01/22/2016] [Accepted: 02/05/2016] [Indexed: 01/20/2023] Open
Abstract
Mammalian arginine metabolism is complex due to the expression of multiple enzymes that utilize arginine as substrate and to interactions or competition between specific enzymes involved in arginine metabolism. Moreover, cells may contain multiple intracellular arginine pools that are not equally accessible to all arginine metabolic enzymes, thus presenting additional challenges to more fully understanding arginine metabolism. At the whole-body level, arginine metabolism ultimately results in the production of a biochemically diverse range of products, including nitric oxide, urea, creatine, polyamines, proline, glutamate, agmatine, and homoarginine. Included in this group of compounds are the methylated arginines (e.g., asymmetric dimethylarginine), which are released upon degradation of proteins containing methylated arginine residues. Changes in arginine concentration also can regulate cellular metabolism and function via a variety of arginine sensors. Although much is known about arginine metabolism, elucidation of the physiologic or pathophysiologic roles for all of the pathways and their metabolites remains an active area of investigation, as exemplified by current findings highlighted in this review.
Collapse
Affiliation(s)
- Sidney M Morris
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
65
|
Huber L, de Lange CFM, Ernst CW, Krogh U, Trottier NL. Impact of improving dietary amino acid balance for lactating sows on efficiency of dietary amino acid utilization and transcript abundance of genes encoding lysine transporters in mammary tissue. J Anim Sci 2016; 94:4654-4665. [PMID: 27898953 DOI: 10.2527/jas.2016-0697] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Lactating multiparous Yorkshire sows ( = 64) were used in 2 experiments to test the hypothesis that reducing dietary CP intake and improving AA balance through crystalline AA (CAA) supplementation improves apparent dietary AA utilization efficiency for milk production and increases transcript abundance of genes encoding Lys transporter proteins in mammary tissue. In Exp. 1, 40 sows were assigned to 1 of 4 diets: 1) high CP (HCP; 16.0% CP, as-fed basis; analyzed concentration), 2) medium-high CP (MHCP; 15.7% CP), 3) medium-low CP (MLCP; 14.3% CP), and 4) low CP (LCP; 13.2% CP). The HCP diet was formulated using soybean meal and corn as the only Lys sources. The reduced-CP diets contained CAA to meet estimated requirements for essential AA that became progressively limiting with reduction in CP concentration, that is, Lys, Ile, Met + Cys, Thr, Trp, and Val. Dietary standardized ileal digestible (SID) Lys concentration was 80% of the estimated requirement. In Exp. 2, 24 sows were assigned to the HCP or LCP diets. In Exp. 1, blood samples were postprandially collected 15 h on d 3, 7, 14, and 18 of lactation and utilization efficiency of dietary AA for milk production was calculated during early (d 3 to 7) and peak (d 14 to 18) lactation. Efficiency values were estimated from daily SID AA intakes and milk AA yield, with corrections for maternal AA requirement for maintenance and AA contribution from body protein losses. In Exp. 2, mammary tissue was biopsied on d 4 and 14 of lactation to determine the mRNA abundance of genes encoding Lys transporter proteins. In peak lactation, Lys, Thr, Trp, and Val utilization efficiency increased with decreasing dietary CP (linear for Trp and Val, < 0.05; in sows fed the MHCP diet vs. sows fed the HCP diet for Lys and Thr, < 0.05). Total essential and nonessential 15-h postprandial serum AA concentrations increased with decreasing dietary CP (linear, = 0.09 and < 0.05, respectively), suggesting increased maternal body protein mobilization. Transcript abundance of several genes involved in Lys transport in mammary tissue did not differ between sows fed the LCP and HCP diets. Feeding lactating sows low-CP diets supplemented with CAA increases the efficiency of utilizing dietary Lys, Thr, Trp, and Val for milk protein production but is unrelated to abundance in mRNA of genes encoding Lys transport proteins in the mammary gland. Dietary Lys utilization for milk protein production in lactating sows appears to be optimized when crystalline Lys is included at a minimum of 0.10% in a diet containing 15.70% CP.
Collapse
|
66
|
Singh K, Al-Greene NT, Verriere TG, Coburn LA, Asim M, Barry DP, Allaman MM, Hardbower DM, Delgado AG, Piazuelo MB, Vallance BA, Gobert AP, Wilson KT. The L-Arginine Transporter Solute Carrier Family 7 Member 2 Mediates the Immunopathogenesis of Attaching and Effacing Bacteria. PLoS Pathog 2016; 12:e1005984. [PMID: 27783672 PMCID: PMC5081186 DOI: 10.1371/journal.ppat.1005984] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 10/06/2016] [Indexed: 12/17/2022] Open
Abstract
Solute carrier family 7 member 2 (SLC7A2) is an inducible transporter of the semi-essential amino acid L-arginine (L-Arg), which has been implicated in immune responses to pathogens. We assessed the role of SLC7A2 in murine infection with Citrobacter rodentium, an attaching and effacing enteric pathogen that causes colitis. Induction of SLC7A2 was upregulated in colitis tissues, and localized predominantly to colonic epithelial cells. Compared to wild-type mice, Slc7a2–/–mice infected with C. rodentium had improved survival and decreased weight loss, colon weight, and histologic injury; this was associated with decreased colonic macrophages, dendritic cells, granulocytes, and Th1 and Th17 cells. In infected Slc7a2–/–mice, there were decreased levels of the proinflammatory cytokines G-CSF, TNF-α, IL-1α, IL-1β, and the chemokines CXCL1, CCL2, CCL3, CCL4, CXCL2, and CCL5. In bone marrow chimeras, the recipient genotype drove the colitis phenotype, indicative of the importance of epithelial, rather than myeloid SLC7A2. Mice lacking Slc7a2 exhibited reduced adherence of C. rodentium to the colonic epithelium and decreased expression of Talin-1, a focal adhesion protein involved in the attachment of the bacterium. The importance of SLC7A2 and Talin-1 in the intimate attachment of C. rodentium and induction of inflammatory response was confirmed in vitro, using conditionally-immortalized young adult mouse colon (YAMC) cells with shRNA knockdown of Slc7a2 or Tln1. Inhibition of L-Arg uptake with the competitive inhibitor, L-lysine (L-Lys), also prevented attachment of C. rodentium and chemokine expression. L-Lys and siRNA knockdown confirmed the role of L-Arg and SLC7A2 in human Caco-2 cells co-cultured with enteropathogenic Escherichia coli. Overexpression of SLC7A2 in human embryonic kidney cells increased bacterial adherence and chemokine expression. Taken together, our data indicate that C. rodentium enhances its own pathogenicity by inducing the expression of SLC7A2 to favor its attachment to the epithelium and thus create its ecological niche. Intestinal infections by attaching and effacing (A/E) bacteria widely impact human health, with major social and economic repercussions. Mucosal immunity plays a critical role in determining the outcome of these infections. The amino acid L-arginine regulates inflammatory responses to bacterial pathogens. We studied the role of the L-arginine transporter solute carrier family 7 member 2 (SLC7A2) during infection with the A/E pathogen Citrobacter rodentium. SLC7A2 is induced in colonic epithelial cells during the infection and facilitates the intimate attachment of the bacteria, thus initiating the inflammatory response of the infected mucosa. These data were confirmed in vitro using C. rodentium-infected mouse cells and human colonic epithelial cells infected with enteropathogenic Escherichia coli. Our work describes a mechanism by which A/E bacteria manipulate host response to favor their colonization, thereby positioning SLC7A2 as an unrecognized therapeutic target to limit infection with enterobacteria.
Collapse
Affiliation(s)
- Kshipra Singh
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Nicole T. Al-Greene
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Thomas G. Verriere
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Lori A. Coburn
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee, United States of America
| | - Mohammad Asim
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Daniel P. Barry
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Margaret M. Allaman
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Dana M. Hardbower
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Alberto G. Delgado
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - M. Blanca Piazuelo
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Bruce A. Vallance
- Division of Gastroenterology, Department of Pediatrics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Alain P. Gobert
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Keith T. Wilson
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee, United States of America
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
67
|
Protein Restriction with Amino Acid-Balanced Diets Shrinks Circulating Pool Size of Amino Acid by Decreasing Expression of Specific Transporters in the Small Intestine. PLoS One 2016; 11:e0162475. [PMID: 27611307 PMCID: PMC5017764 DOI: 10.1371/journal.pone.0162475] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 08/23/2016] [Indexed: 01/10/2023] Open
Abstract
Dietary protein restriction is not only beneficial to health and longevity in humans, but also protects against air pollution and minimizes feeding cost in livestock production. However, its impact on amino acid (AA) absorption and metabolism is not quite understood. Therefore, the study aimed to explore the effect of protein restriction on nitrogen balance, circulating AA pool size, and AA absorption using a pig model. In Exp.1, 72 gilts weighting 29.9 ± 1.5 kg were allocated to 1 of the 3 diets containing 14, 16, or 18% CP for a 28-d trial. Growth (n = 24), nitrogen balance (n = 6), and the expression of small intestinal AA and peptide transporters (n = 6) were evaluated. In Exp.2, 12 barrows weighting 22.7 ± 1.3 kg were surgically fitted with catheters in the portal and jejunal veins as well as the carotid artery and assigned to a diet containing 14 or 18% CP. A series of blood samples were collected before and after feeding for determining the pool size of circulating AA and AA absorption in the portal vein, respectively. Protein restriction did not sacrifice body weight gain and protein retention, since nitrogen digestibility was increased as dietary protein content reduced. However, the pool size of circulating AA except for lysine and threonine, and most AA flux through the portal vein were reduced in pigs fed the low protein diet. Meanwhile, the expression of peptide transporter 1 (PepT-1) was stimulated, but the expression of the neutral and cationic AA transporter systems was depressed. These results evidenced that protein restriction with essential AA-balanced diets, decreased AA absorption and reduced circulating AA pool size. Increased expression of small intestinal peptide transporter PepT-1 could not compensate for the depressed expression of jejunal AA transporters for AA absorption.
Collapse
|
68
|
Bose M, Bhattacharyya SN. Target-dependent biogenesis of cognate microRNAs in human cells. Nat Commun 2016; 7:12200. [PMID: 27448149 PMCID: PMC4961841 DOI: 10.1038/ncomms12200] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 06/10/2016] [Indexed: 12/22/2022] Open
Abstract
Extensive research has established how miRNAs regulate target mRNAs by translation repression and/or endonucleolytic degradation in metazoans. However, information related to the effect of target mRNA on biogenesis and stability of corresponding miRNAs in animals is limited. Here we report regulated biogenesis of cognate miRNAs by their target mRNAs. Enhanced pre-miRNA processing by AGO-associated DICER1 contributes to this increased miRNP formation. The processed miRNAs are loaded onto AGO2 to form functionally competent miRISCs both in vivo and also in a cell-free in vitro system. Thus, we identify an additional layer of posttranscriptional regulation that helps the cell to maintain requisite levels of mature forms of respective miRNAs by modulating their processing in a target-dependent manner, a process happening for miR-122 during stress reversal in human hepatic cells. MicroRNAs are a widespread regulatory mechanism and are themselves extensively regulated. Here the authors show regulated miRNA biogenesis by the target mRNA, a layer of regulation that modulates miRNA levels dependent on target availability.
Collapse
Affiliation(s)
- Mainak Bose
- RNA Biology Research Laboratory, Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja SC Mullick Road, Kolkata 700032, India
| | - Suvendra N Bhattacharyya
- RNA Biology Research Laboratory, Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja SC Mullick Road, Kolkata 700032, India
| |
Collapse
|
69
|
Cervantes M, Cota M, Arce N, Castillo G, Avelar E, Espinoza S, Morales A. Effect of heat stress on performance and expression of selected amino acid and glucose transporters, HSP90, leptin and ghrelin in growing pigs. J Therm Biol 2016; 59:69-76. [DOI: 10.1016/j.jtherbio.2016.04.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 12/05/2015] [Accepted: 04/17/2016] [Indexed: 10/21/2022]
|
70
|
Kishikawa T, Otsuka M, Tan PS, Ohno M, Sun X, Yoshikawa T, Shibata C, Takata A, Kojima K, Takehana K, Ohishi M, Ota S, Noyama T, Kondo Y, Sato M, Soga T, Hoshida Y, Koike K. Decreased miR122 in hepatocellular carcinoma leads to chemoresistance with increased arginine. Oncotarget 2016; 6:8339-52. [PMID: 25826076 PMCID: PMC4480756 DOI: 10.18632/oncotarget.3234] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 01/28/2015] [Indexed: 12/12/2022] Open
Abstract
Reduced expression of microRNA122 (miR122), a liver-specific microRNA, is
frequent in hepatocellular carcinoma (HCC). However, its biological
significances remain poorly understood. Because deregulated amino acid levels in
cancers can affect their biological behavior, we determined the amino acid
levels in miR122-silenced mouse liver tissues, in which intracellular arginine
levels were significantly increased. The increased intracellular arginine levels
were through upregulation of the solute carrier family 7 (SLC7A1), a transporter
of arginine and a direct target of miR122. Arginine is the substrate for nitric
oxide (NO) synthetase, and intracellular NO levels were increased in
miR122-silenced HCC cells, with increased resistance to sorafenib, a multikinase
inhibitor. Conversely, maintenance of the miR122-silenced HCC cells in
arginine-depleted culture media, as well as overexpression of miR122 in
miR122-low-expressing HCC cells, reversed these effects and rendered the cells
more sensitive to sorafenib. Using a reporter knock-in construct, chemical
compounds were screened, and Wee1 kinase inhibitor was identified as
upregulators of miR122 transcription, which increased the sensitivity of the
cells to sorafenib. These results provide an insight into sorafenib resistance
in miR122-low HCC, and suggest that arginine depletion or a combination of
sorafenib with the identified compound may provide promising approaches to
managing this HCC subset.
Collapse
Affiliation(s)
- Takahiro Kishikawa
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Motoyuki Otsuka
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan.,Japan Science and Technology Agency, PRESTO, Kawaguchi, Saitama 332-0012, Japan
| | - Poh Seng Tan
- Liver Cancer Program, Tisch Cancer Institute, Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, NY 10029, USA.,Division of Gastroenterology and Hepatology, University Medicine Cluster, National University Health System, 119228, Singapore
| | - Motoko Ohno
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Xiaochen Sun
- Liver Cancer Program, Tisch Cancer Institute, Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, NY 10029, USA
| | - Takeshi Yoshikawa
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Chikako Shibata
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Akemi Takata
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Kentaro Kojima
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Kenji Takehana
- Pharmacology Research Laboratory, Research Institute, Ajinomoto Pharmaceutical Co., Ltd., Kawasaki, Kanagawa 210-8681, Japan
| | - Maki Ohishi
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0052, Japan
| | - Sana Ota
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0052, Japan
| | - Tomoyuki Noyama
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Yuji Kondo
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Masaya Sato
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Tomoyoshi Soga
- Pharmacology Research Laboratory, Research Institute, Ajinomoto Pharmaceutical Co., Ltd., Kawasaki, Kanagawa 210-8681, Japan
| | - Yujin Hoshida
- Liver Cancer Program, Tisch Cancer Institute, Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, NY 10029, USA
| | - Kazuhiko Koike
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| |
Collapse
|
71
|
Cremades A, Del Rio-Garcia J, Lambertos A, López-Garcia C, Peñafiel R. Tissue-specific regulation of potassium homeostasis by high doses of cationic amino acids. SPRINGERPLUS 2016; 5:616. [PMID: 27330882 PMCID: PMC4870509 DOI: 10.1186/s40064-016-2224-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 04/22/2016] [Indexed: 01/19/2023]
Abstract
The administration of l-arginine hydrochloride has been used for testing pituitary secretion in humans, and as an experimental model for induction of acute pancreatitis in rats and mice. Whereas in the first case, the administration of the amino acid is associated with hiperkalemia, in the model of acute pancreatitis no data are available on possible changes in potassium homeostasis. The present study shows that the acute administration to mice of l-arginine hydrochloride or other cationic amino acids almost duplicate plasma potassium levels. This effect was associated to a marked decrease of tissue potassium in both pancreas and liver. No changes were found in other tissues. These changes cannot be ascribed to the large load of chloride ions, since similar effects were produced when l-ornithine aspartate was administered. The changes in potassium levels were dependent on the dose. The displacement of intracellular potassium from the liver and pancreas to the extracellular compartment appears to be dependent on the entry of the cationic amino acid, since the administration of an equivalent dose of alfa-difluoromethyl ornithine HCl (DFMO), a non physiological analog of l-ornithine, which is poorly taken by the tissues in comparison with the physiological cationic amino acids, did not produce any change in potassium levels in pancreas and liver. The analyses of the expression of cationic amino acid transporters (CAT) suggest that the CAT-2 transporter may be implicated in the potassium/cationic amino acid interchange in liver and pancreas. The possible physiological or pathological relevance of these findings is discussed.
Collapse
Affiliation(s)
- Asunción Cremades
- Department of Pharmacology, Faculty of Medicine, University of Murcia, Murcia, Spain ; Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Jesús Del Rio-Garcia
- Department of Pharmacology, Faculty of Medicine, University of Murcia, Murcia, Spain
| | - Ana Lambertos
- Department of Biochemistry, Molecular Biology B and Immunology, School of Medicine, University of Murcia, 30100 Murcia, Spain
| | - Carlos López-Garcia
- Department of Pharmacology, Faculty of Medicine, University of Murcia, Murcia, Spain
| | - Rafael Peñafiel
- Department of Biochemistry, Molecular Biology B and Immunology, School of Medicine, University of Murcia, 30100 Murcia, Spain ; Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| |
Collapse
|
72
|
Catabolism and safety of supplemental L-arginine in animals. Amino Acids 2016; 48:1541-52. [PMID: 27156062 DOI: 10.1007/s00726-016-2245-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 04/25/2016] [Indexed: 12/14/2022]
Abstract
L-arginine (Arg) is utilized via multiple pathways to synthesize protein and low-molecular-weight bioactive substances (e.g., nitric oxide, creatine, and polyamines) with enormous physiological importance. Furthermore, Arg regulates cell signaling pathways and gene expression to improve cardiovascular function, augment insulin sensitivity, enhance lean tissue mass, and reduce obesity in humans. Despite its versatile roles, the use of Arg as a dietary supplement is limited due to the lack of data to address concerns over its safety in humans. Data from animal studies are reviewed to assess arginine catabolism and the safety of long-term Arg supplementation. The arginase pathway was responsible for catabolism of 76-85 and 81-96 % Arg in extraintestinal tissues of pigs and rats, respectively. Dietary supplementation with Arg-HCl or the Arg base [315- and 630-mg Arg/(kg BW d) for 91 d] had no adverse effects on male or female pigs. Similarly, no safety issues were observed for male or female rats receiving supplementation with 1.8- and 3.6-g Arg/(kg BW d) for at least 91 d. Intravenous administration of Arg-HCl to gestating sheep at 81 and 180 mg Arg/(kg BW d) is safe for at least 82 and 40 d, respectively. Animals fed conventional diets can well tolerate large amounts of supplemental Arg [up to 630-mg Arg/(kg BW d) in pigs or 3.6-g Arg/(kg BW d) in rats] for 91 d, which are equivalent to 573-mg Arg/(kg BW d) for humans. Collectively, these results can help guide studies to determine the safety of long-term oral administration of Arg in humans.
Collapse
|
73
|
Li YH, Li FN, Wu L, Liu YY, Wei HK, Li TJ, Tan BE, Kong XF, Wu F, Duan YH, Oladele OA, Yin YL. Reduced dietary protein level influences the free amino acid and gene expression profiles of selected amino acid transceptors in skeletal muscle of growing pigs. J Anim Physiol Anim Nutr (Berl) 2016; 101:96-104. [PMID: 27045856 DOI: 10.1111/jpn.12514] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 03/12/2016] [Indexed: 12/22/2022]
Abstract
This study was conducted to evaluate the effect of reduced dietary protein level on growth performance, muscle mass weight, free amino acids (FAA) and gene expression profile of selected amino acid transceptors in different fibre type of skeletal muscle tissues (longissimus dorsi, psoas major, biceps femoris) of growing pigs. A total of 18 cross-bred growing pigs (Large White × Landrace × Duroc) with initial body weight (9.57 ± 0.67 kg) were assigned into three dietary treatments: 20% crude protein (CP) diet (normal recommended, NP), 17% CP diet (low protein, LP) and 14% CP diet (very low protein, VLP). The results indicated improved feed-to-gain ratio was obtained for pigs fed LP and NP diets (p < 0.01), while the pigs fed VLP diet showed the worst growth performance (p < 0.01). There was no significant difference in the weights of longissimus dorsi and psoas major muscle between LP and NP groups (p > 0.05). Majority of the determined FAA concentration of LP group were greater than or equal to those of NP group in both longissimus dorsi and psoas major muscle (p < 0.01). Further, the mRNA expression levels of sodium-coupled neutral amino acid transceptor 2, L-type amino acid transceptor 1 and proton-assisted amino acid transceptors 2 were higher in skeletal muscle tissue in LP group compared to those of the pigs fed NP or VLP diet. These results suggested that reduced dietary protein level (3 points of percentage less than recommended level) would upregulate the mRNA expression of amino acid transceptors to enhance the absorption of FAA in skeletal muscle of growing pigs. There seems to be a relationship between response of AA transceptors to the dietary protein level in skeletal muscle tissue of different fibre type. To illustrate the underlying mechanisms will be beneficial to animal nutrition.
Collapse
Affiliation(s)
- Y H Li
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - F N Li
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China.,Hunan Co-Innovation Center of Animal Production Safety (CICAPS), Changsha, China
| | - L Wu
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - Y Y Liu
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - H K Wei
- College of Animal Sciences, Huazhong Agricultural University, Wuhan, Hubei, China
| | - T J Li
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - B E Tan
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - X F Kong
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - F Wu
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - Y H Duan
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - O A Oladele
- Animal Nutrition Department, College of Animal Science and Livestock Production, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| | - Y L Yin
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center of Healthy Livestock, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| |
Collapse
|
74
|
Li H, Jiang JD, Peng ZG. MicroRNA-mediated interactions between host and hepatitis C virus. World J Gastroenterol 2016; 22:1487-1496. [PMID: 26819516 PMCID: PMC4721982 DOI: 10.3748/wjg.v22.i4.1487] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 09/25/2015] [Accepted: 10/13/2015] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs. More than 2500 mature miRNAs are detected in plants, animals and several types of viruses. Hepatitis C virus (HCV), which is a positive-sense, single-stranded RNA virus, does not encode viral miRNA. However, HCV infection alters the expression of host miRNAs, either in cell culture or in patients with liver disease progression, such as liver fibrosis, cirrhosis, and hepatocellular carcinoma. In turn, host miRNAs regulate HCV life cycle through directly binding to HCV RNAs or indirectly targeting cellular mRNAs. Increasing evidence demonstrates that miRNAs are one of the centered factors in the interaction network between virus and host. The competitive viral and host RNA hypothesis proposes a latent cross-regulation pattern between host mRNAs and HCV RNAs. High loads of HCV RNA sequester and de-repress host miRNAs from their normal host targets and thus disturb host gene expression, indicating a means of adaptation for HCV to establish a persistent infection. Some special miRNAs are closely correlated with liver-specific disease progression and the changed levels of miRNAs are even higher sensitivity and specificity than those of traditional proteins. Therefore, some of them can serve as novel diagnostic/prognostic biomarkers in HCV-infected patients with liver diseases. They are also attractive therapeutic targets for development of new anti-HCV agents.
Collapse
|
75
|
Zhang H, Gilbert ER, Zhang K, Ding X, Luo Y, Wang J, Zeng Q, Bai S. Uptake of manganese from manganese-lysine complex in the primary rat intestinal epithelial cells. J Anim Physiol Anim Nutr (Berl) 2015; 101:147-158. [DOI: 10.1111/jpn.12430] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 09/30/2015] [Indexed: 11/30/2022]
Affiliation(s)
- H. Zhang
- Animal Nutrition Institute; Sichuan Agricultural University Ya'an; Sichuan China
| | - E. R. Gilbert
- Department of Animal and Poultry Sciences; Virginia Tech University; Blacksburg VA USA
| | - K. Zhang
- Animal Nutrition Institute; Sichuan Agricultural University Ya'an; Sichuan China
| | - X. Ding
- Animal Nutrition Institute; Sichuan Agricultural University Ya'an; Sichuan China
| | - Y. Luo
- Animal Nutrition Institute; Sichuan Agricultural University Ya'an; Sichuan China
| | - J. Wang
- Animal Nutrition Institute; Sichuan Agricultural University Ya'an; Sichuan China
| | - Q. Zeng
- Animal Nutrition Institute; Sichuan Agricultural University Ya'an; Sichuan China
| | - S. Bai
- Animal Nutrition Institute; Sichuan Agricultural University Ya'an; Sichuan China
| |
Collapse
|
76
|
Werner A, Amann E, Schnitzius V, Habermeier A, Luckner-Minden C, Leuchtner N, Rupp J, Closs EI, Munder M. Induced arginine transport via cationic amino acid transporter-1 is necessary for human T-cell proliferation. Eur J Immunol 2015; 46:92-103. [PMID: 26449889 DOI: 10.1002/eji.201546047] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 09/03/2015] [Accepted: 10/05/2015] [Indexed: 11/10/2022]
Abstract
Availability of the semiessential amino acid arginine is fundamental for the efficient function of human T lymphocytes. Tumor-associated arginine deprivation, mainly induced by myeloid-derived suppressor cells, is a central mechanism of tumor immune escape from T-cell-mediated antitumor immune responses. We thus assumed that transmembranous transport of arginine must be crucial for T-cell function and studied which transporters are responsible for arginine influx into primary human T lymphocytes. Here, we show that activation via CD3 and CD28 induces arginine transport into primary human T cells. Both naïve and memory CD4(+) T cells as well as CD8(+) T cells specifically upregulated the human cationic amino acid transporter-1 (hCAT-1), with an enhanced and persistent expression under arginine starvation. When hCAT-1 induction was suppressed via siRNA transfection, arginine uptake, and cellular proliferation were impaired. In summary, our results demonstrate that hCAT-1 is a key component of efficient T-cell activation and a novel potential target structure to modulate adaptive immune responses in tumor immunity or inflammation.
Collapse
Affiliation(s)
- Anke Werner
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Eva Amann
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Vanessa Schnitzius
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Alice Habermeier
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Claudia Luckner-Minden
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Nadine Leuchtner
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Johanna Rupp
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Ellen I Closs
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Markus Munder
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.,Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
77
|
Chertow JH, Alkaitis MS, Nardone G, Ikeda AK, Cunnington AJ, Okebe J, Ebonyi AO, Njie M, Correa S, Jayasooriya S, Casals-Pascual C, Billker O, Conway DJ, Walther M, Ackerman H. Plasmodium Infection Is Associated with Impaired Hepatic Dimethylarginine Dimethylaminohydrolase Activity and Disruption of Nitric Oxide Synthase Inhibitor/Substrate Homeostasis. PLoS Pathog 2015; 11:e1005119. [PMID: 26407009 PMCID: PMC4583463 DOI: 10.1371/journal.ppat.1005119] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 07/29/2015] [Indexed: 12/04/2022] Open
Abstract
Inhibition of nitric oxide (NO) signaling may contribute to pathological activation of the vascular endothelium during severe malaria infection. Dimethylarginine dimethylaminohydrolase (DDAH) regulates endothelial NO synthesis by maintaining homeostasis between asymmetric dimethylarginine (ADMA), an endogenous NO synthase (NOS) inhibitor, and arginine, the NOS substrate. We carried out a community-based case-control study of Gambian children to determine whether ADMA and arginine homeostasis is disrupted during severe or uncomplicated malaria infections. Circulating plasma levels of ADMA and arginine were determined at initial presentation and 28 days later. Plasma ADMA/arginine ratios were elevated in children with acute severe malaria compared to 28-day follow-up values and compared to children with uncomplicated malaria or healthy children (p<0.0001 for each comparison). To test the hypothesis that DDAH1 is inactivated during Plasmodium infection, we examined DDAH1 in a mouse model of severe malaria. Plasmodium berghei ANKA infection inactivated hepatic DDAH1 via a post-transcriptional mechanism as evidenced by stable mRNA transcript number, decreased DDAH1 protein concentration, decreased enzyme activity, elevated tissue ADMA, elevated ADMA/arginine ratio in plasma, and decreased whole blood nitrite concentration. Loss of hepatic DDAH1 activity and disruption of ADMA/arginine homeostasis may contribute to severe malaria pathogenesis by inhibiting NO synthesis. During a malaria infection, the vascular endothelium becomes more adhesive, permeable, and prone to trigger blood clotting. These changes help the parasite adhere to blood vessels, but endanger the host by obstructing blood flow through small vessels. Endothelial nitric oxide (NO) would normally counteract these pathological changes, but NO signalling is diminished malaria. NO synthesis is inhibited by asymmetric dimethylarginine (ADMA), a methylated derivative of arginine that is released during normal protein turnover. We found the ratio of ADMA to arginine to be elevated in Gambian children with severe malaria, a metabolic disturbance known to inhibit NO synthesis. ADMA was associated with markers of endothelial activation and impaired tissue perfusion. In parallel experiments using mice, the enzyme responsible for metabolizing ADMA, dimethylarginine dimethylaminohydrolase (DDAH), was inactivated after infection with a rodent malaria. Based on these studies, we propose that decreased metabolism of ADMA by DDAH might contribute to the elevated ADMA/arginine ratio observed during an acute episode of malaria. Strategies to preserve or increase DDAH activity might improve NO synthesis and help to prevent the vascular manifestations of severe malaria.
Collapse
Affiliation(s)
- Jessica H. Chertow
- Laboratory of Malaria and Vector Research, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Matthew S. Alkaitis
- Laboratory of Malaria and Vector Research, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
- Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Headington Oxford, United Kingdom
| | - Glenn Nardone
- Research Technology Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Allison K. Ikeda
- Laboratory of Malaria and Vector Research, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | | | | | | | | | | | | | | | - Oliver Billker
- Wellcome Trust Sanger Institute, Hinxton Cambridge, United Kingdom
| | - David J. Conway
- MRC Unit, Fajara, The Gambia
- London School of Hygiene and Tropical Medicine, Bloomsbury, London, United Kingdom
| | | | - Hans Ackerman
- Laboratory of Malaria and Vector Research, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
- * E-mail:
| |
Collapse
|
78
|
Henriques C, Miller MP, Catanho M, de Carvalho TMU, Krieger MA, Probst CM, de Souza W, Degrave W, Amara SG. Identification and functional characterization of a novel arginine/ornithine transporter, a member of a cationic amino acid transporter subfamily in the Trypanosoma cruzi genome. Parasit Vectors 2015; 8:346. [PMID: 26109388 PMCID: PMC4486710 DOI: 10.1186/s13071-015-0950-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 06/13/2015] [Indexed: 01/03/2023] Open
Abstract
Background Trypanosoma cruzi, the etiological agent of Chagas disease, is auxotrophic for arginine. It obtains this amino acid from the host through transporters expressed on the plasma membrane and on the membranes of intracellular compartments. A few cationic amino acid transporters have been characterized at the molecular level, such as the novel intracellular arginine/ornithine transporter, TcCAT1.1, a member of the TcCAT subfamily that is composed of four almost identical open reading frames in the T. cruzi genome. Methods The functional characterization of the TcCAT1.1 isoform was performed in two heterologous expression systems. TcCAT subfamily expression was evaluated by real-time PCR in polysomal RNA fractions, and the cellular localization of TcCAT1.1 fused to EGFP was performed by confocal and immunoelectron microscopy. Results In the S. cerevisiae expression system, TcCAT1.1 showed high affinity for arginine (Km = 0.085 ± 0.04 mM) and low affinity for ornithine (Km = 1.7 ± 0.2 mM). Xenopus laevis oocytes expressing TcCAT1.1 showed a 7-fold increase in arginine uptake when they were pre-loaded with arginine, indicating that transport is enhanced by substrates on the trans side of the membrane (trans-stimulation). Oocytes that were pre-loaded with [3H]-arginine displayed a 16-fold higher efflux of [3H]-arginine compared with that of the control. Analysis of polysomal RNA fractions demonstrated that the expression of members of the arginine transporter TcCAT subfamily is upregulated under nutritional stress and that this upregulation precedes metacyclogenesis. To investigate the cellular localization of the transporter, EGFP was fused to TcCAT1.1, and fluorescence microscopy and immunocytochemistry revealed the intracellular labeling of vesicles in the anterior region, in a network of tubules and vesicles. Conclusions TcCAT1.1 is a novel arginine/ornithine transporter, an exchanger expressed in intracellular compartments that is physiologically involved in arginine homeostasis throughout the T. cruzi life cycle. The properties and estimated kinetic parameters of TcCAT1.1 can be extended to other members of the TcCAT subfamily.
Collapse
Affiliation(s)
- Cristina Henriques
- Fundação Oswaldo Cruz, Fiocruz-Mato Grosso do Sul, Rua Gabriel Abrão 92-Jardim das Nações, Campo Grande, MS, 89081-746, Brazil. .,Instituto de Biofísica Carlos Chagas Filho-UFRJ, CCS-Bloco G-Laboratório de Ultraestrutura Celular Hertha Meyer, Rio de Janeiro, RJ, 21949-900, Brazil. .,Nucleo de Biologia Estrutural e Biomagens, Universidade Federal do Rio de Janeiro-CENABIO, Rio de Janeiro, RJ, Brazil.
| | - Megan P Miller
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15260, USA.
| | - Marcos Catanho
- Fiocruz, Instituto Oswaldo Cruz, Laboratório de Genômica Funcional e Bioinformática, Av. Brasil 4365, Manguinhos, 21040-900, Rio de Janeiro, RJ, Brazil.
| | - Técia Maria Ulisses de Carvalho
- Instituto de Biofísica Carlos Chagas Filho-UFRJ, CCS-Bloco G-Laboratório de Ultraestrutura Celular Hertha Meyer, Rio de Janeiro, RJ, 21949-900, Brazil.
| | | | | | - Wanderley de Souza
- Instituto de Biofísica Carlos Chagas Filho-UFRJ, CCS-Bloco G-Laboratório de Ultraestrutura Celular Hertha Meyer, Rio de Janeiro, RJ, 21949-900, Brazil. .,Nucleo de Biologia Estrutural e Biomagens, Universidade Federal do Rio de Janeiro-CENABIO, Rio de Janeiro, RJ, Brazil. .,Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Biomagens-INBEB, Rio de Janeiro, Brazil.
| | - Wim Degrave
- Fiocruz, Instituto Oswaldo Cruz, Laboratório de Genômica Funcional e Bioinformática, Av. Brasil 4365, Manguinhos, 21040-900, Rio de Janeiro, RJ, Brazil.
| | - Susan Gaye Amara
- National Institute of Mental Health, NIH Building 10 Center Driver, Room 4N222, MSC 1381, Bethesda, MD, 20892-1381, USA. .,Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15260, USA.
| |
Collapse
|
79
|
Morales A, Buenabad L, Castillo G, Arce N, Araiza BA, Htoo JK, Cervantes M. Low-protein amino acid-supplemented diets for growing pigs: effect on expression of amino acid transporters, serum concentration, performance, and carcass composition. J Anim Sci 2015; 93:2154-64. [PMID: 26020311 DOI: 10.2527/jas.2014-8834] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023] Open
Abstract
Pigs fed protein-bound AA appear to have a higher abundance of AA transporters for their absorption in the jejunum compared with the duodenum. However, there is limited data about the effect of dietary free AA, readily available in the duodenum, on the duodenal abundance of AA transporters and its impact on pig performance. Forty-eight pigs (24.3 kg initial BW) distributed in 4 treatments were used to evaluate the effect of the CP level and form (free vs. protein bound) in which AA are added to diets on the expression of AA transporters in the 3 small intestine segments, serum concentration of AA, and performance. Dietary treatments based on wheat and soybean meal (SBM) were 1) low-CP (14%) diet supplemented with L-Lys, L-Thr, DL-Met, L-Leu, L-Ile, L-Val, L-His, L-Trp, and L-Phe (LPAA); 2) as in the LPAA but with added L-Gly as a N source (LPAA+N); 3) intermediate CP content (16%) supplemented with L-Lys HCl, L-Thr, and DL-Met (MPAA); and 4) high-CP (22%) diet (HP) without free AA. At the end of the experiment, 8 pigs from LPAA and HP were sacrificed to collect intestinal mucosa and blood samples and to dissect the carcasses. There were no differences in ADG, ADFI, G:F, and weights of carcass components and some visceral organs between treatments. Weights of the large intestine and kidney were higher in HP pigs (P < 0.01). Expression of b(0,+) in the duodenum was higher in pigs fed the LPAA compared with the HP diet (P= 0.036) but there was no difference in the jejunum and ileum. In the ileum, y+ L expression tended to be higher in pigs fed the LPAA diet (P = 0.098). Expression of b(0,+) in LPAA pigs did not differ between the duodenum and the jejunum, but in HP pigs, the expression of all AA transporters was higher in the jejunum than in the duodenum or ileum (P < 0.05). The serum concentration of Arg, His, Ile, Leu, Phe, and Val was higher but serum Lys and Met were lower in pigs fed the HP diet (P < 0.05). These results indicate that LPAA can substitute up to 8 percentage units of protein in HP wheat-SBM diets without affecting pig performance; nonessential N does not seem to be limiting in very low-protein wheat-SBM diets for growing pigs. Also, the inclusion of free AA in the diet appears to affect their serum concentration and the expression of the AA transporter b0,+ in the duodenum of pigs.
Collapse
|
80
|
Glushko AA, Voronkov AV, Chernikov MV. [Molecular targets for searching of endothelial-protective substances]. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2015; 40:515-27. [PMID: 25895347 DOI: 10.1134/s1068162014050069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Endothelial dysfunction underlies the development of many cardiovascular diseases. Thus endothelium becomes an independent therapeutic target, and the search of new substances with endothelial-protective action to date is one of the promising tasks for pharmacotherapy and medicinal chemistry. Molecular modeling is an effective tool for solving this problem. Computer chemistry methods use is only possible in combination with detailed information on three dimensional structure and functions of molecular targets: receptors and enzymes, involved in signal transduction inside and outside of endothelial cells. Information on structure and function of various macromolecules involved in vascular tone regulation is collected in the review. The structure of endothelial NO-synthase (EC 1.14.13.39) (eNOS)--enzyme, responsible for the nitric oxide synthesis and involved in vascular tone regulation process is reviewed. The importance of eNOS substrate--L-arginine is underlined in the review in terms of this enzyme activity, regulation, the information on structure and functions of L-arginine transport system is provided. Also different ways of eNOS activity regulation are reviewed, among which are enzyme activation and concurrent inhibition by substances interaction with active center of enzyme, inhibition by caveoline binding with oxigenase domain, and also regulation by phosphorylation of certain amino acids of eNOS by proteinkinase and dephoshphorylation of them by phosphatases. The importance of membrane receptors of endothelial cells as targets for endothelial-protective substances is underlined. Among them are receptors of endothelin, platelet activation factor, prostanoids, bradykinin, histamine, serotonin and protease activated receptors. The important role of potassium and calcium ion channels of vascular cells in endothelial-protective activity is underlined. Macromolecules presented in the review finally are considered as targets for searching for medicinal substances with endothelial-protective activity using proposed ways and methods of molecular modeling.
Collapse
|
81
|
González M, Rojas S, Avila P, Cabrera L, Villalobos R, Palma C, Aguayo C, Peña E, Gallardo V, Guzmán-Gutiérrez E, Sáez T, Salsoso R, Sanhueza C, Pardo F, Leiva A, Sobrevia L. Insulin reverses D-glucose-increased nitric oxide and reactive oxygen species generation in human umbilical vein endothelial cells. PLoS One 2015; 10:e0122398. [PMID: 25875935 PMCID: PMC4397070 DOI: 10.1371/journal.pone.0122398] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 02/23/2015] [Indexed: 01/14/2023] Open
Abstract
Vascular tone is controlled by the L-arginine/nitric oxide (NO) pathway, and NO bioavailability is strongly affected by hyperglycaemia-induced oxidative stress. Insulin leads to high expression and activity of human cationic amino acid transporter 1 (hCAT-1), NO synthesis and vasodilation; thus, a protective role of insulin on high D-glucose-alterations in endothelial function is likely. Vascular reactivity to U46619 (thromboxane A2 mimetic) and calcitonin gene related peptide (CGRP) was measured in KCl preconstricted human umbilical vein rings (wire myography) incubated in normal (5 mmol/L) or high (25 mmol/L) D-glucose. hCAT-1, endothelial NO synthase (eNOS), 42 and 44 kDa mitogen-activated protein kinases (p42/44mapk), protein kinase B/Akt (Akt) expression and activity were determined by western blotting and qRT-PCR, tetrahydrobiopterin (BH4) level was determined by HPLC, and L-arginine transport (0-1000 μmol/L) was measured in response to 5-25 mmol/L D-glucose (0-36 hours) in passage 2 human umbilical vein endothelial cells (HUVECs). Assays were in the absence or presence of insulin and/or apocynin (nicotinamide adenine dinucleotide phosphate-oxidase [NADPH oxidase] inhibitor), tempol or Mn(III)TMPyP (SOD mimetics). High D-glucose increased hCAT-1 expression and activity, which was biphasic (peaks: 6 and 24 hours of incubation). High D-glucose-increased maximal transport velocity was blocked by insulin and correlated with lower hCAT-1 expression and SLC7A1 gene promoter activity. High D-glucose-increased transport parallels higher reactive oxygen species (ROS) and superoxide anion (O2•-) generation, and increased U46619-contraction and reduced CGRP-dilation of vein rings. Insulin and apocynin attenuate ROS and O2•- generation, and restored vascular reactivity to U46619 and CGRP. Insulin, but not apocynin or tempol reversed high D-glucose-increased NO synthesis; however, tempol and Mn(III)TMPyP reversed the high D-glucose-reduced BH4 level. Insulin and tempol blocked the high D-glucose-increased p42/44mapk phosphorylation. Vascular dysfunction caused by high D-glucose is likely attenuated by insulin through the L-arginine/NO and O2•-/NADPH oxidase pathways. These findings are of interest for better understanding vascular dysfunction in states of foetal insulin resistance and hyperglycaemia.
Collapse
Affiliation(s)
- Marcelo González
- Vascular Physiology Laboratory, Department of Physiology, Faculty of Biological Sciences, Universidad de Concepción, P.O. Box 160-C, Concepción 4070386, Chile
- Group of Research and Innovation in Vascular Health (GRIVAS-Health), PO-Box 114-D, Chillán 3800708, Chile
| | - Susana Rojas
- Vascular Physiology Laboratory, Department of Physiology, Faculty of Biological Sciences, Universidad de Concepción, P.O. Box 160-C, Concepción 4070386, Chile
| | - Pía Avila
- Vascular Physiology Laboratory, Department of Physiology, Faculty of Biological Sciences, Universidad de Concepción, P.O. Box 160-C, Concepción 4070386, Chile
| | - Lissette Cabrera
- Vascular Physiology Laboratory, Department of Physiology, Faculty of Biological Sciences, Universidad de Concepción, P.O. Box 160-C, Concepción 4070386, Chile
- Department of Morphophysiology, Faculty of Medicine, Universidad Diego Portales, Santiago 8370076, Chile
| | - Roberto Villalobos
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, P.O. Box 114-D, Santiago 8330024, Chile
| | - Carlos Palma
- Vascular Physiology Laboratory, Department of Physiology, Faculty of Biological Sciences, Universidad de Concepción, P.O. Box 160-C, Concepción 4070386, Chile
| | - Claudio Aguayo
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, Universidad de Concepción, P.O. Box 160-C, Concepción 4070386, Chile
- Group of Research and Innovation in Vascular Health (GRIVAS-Health), PO-Box 114-D, Chillán 3800708, Chile
| | - Eduardo Peña
- Department of Physiopathology, Faculty of Biological Sciences, Universidad de Concepción, P.O. Box 160-C, Concepción 4070386, Chile
| | - Victoria Gallardo
- Department of Physiopathology, Faculty of Biological Sciences, Universidad de Concepción, P.O. Box 160-C, Concepción 4070386, Chile
| | - Enrique Guzmán-Gutiérrez
- Group of Research and Innovation in Vascular Health (GRIVAS-Health), PO-Box 114-D, Chillán 3800708, Chile
- Faculty of Health Sciences, Universidad San Sebastián, Concepción 4080871, Chile
| | - Tamara Sáez
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, P.O. Box 114-D, Santiago 8330024, Chile
| | - Rocío Salsoso
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, P.O. Box 114-D, Santiago 8330024, Chile
| | - Carlos Sanhueza
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, P.O. Box 114-D, Santiago 8330024, Chile
| | - Fabián Pardo
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, P.O. Box 114-D, Santiago 8330024, Chile
| | - Andrea Leiva
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, P.O. Box 114-D, Santiago 8330024, Chile
| | - Luis Sobrevia
- University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, QLD 4029, Queensland, Australia
- Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville E-41012, Spain
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, P.O. Box 114-D, Santiago 8330024, Chile
| |
Collapse
|
82
|
Morales A, García H, Arce N, Cota M, Zijlstra RT, Araiza BA, Cervantes M. Effect of L-lysine on expression of selected genes, serum concentration of amino acids, muscle growth and performance of growing pigs. J Anim Physiol Anim Nutr (Berl) 2014; 99:701-9. [PMID: 25354230 DOI: 10.1111/jpn.12267] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 09/22/2014] [Indexed: 12/22/2022]
Abstract
Lysine (Lys) is the first limiting amino acid (AA) in most feed formulations for pigs and most abundant, along with leucine, in muscle proteins. An experiment was conducted with 17 pigs (17.7 ± 0.05 kg initial BW) to identify a role of dietary Lys in the control of protein synthesis in pigs. Fourteen pigs were randomly assigned to one of the two wheat-based dietary treatments: Lys-deficient, 3.0 g/kg (DEF) and Lys-adequate, 10.8 g/kg (ADE). Samples from jejunum mucosa, liver, Longissumus and Semitendinosus muscles, and blood were collected. The other three pigs were sacrificed at the beginning of the trial to measure basal carcass composition. Weight gain, gain:feed ratio, Lys intake and loin eye area were greater in ADE than in DEF pigs (p < 0.01). Muscle-related carcass characteristics were better, and myosin heavy chain IIb expression (MyHC IIb) in Semitendinosus was higher in ADE than in DEF pigs. Expression of AA transporters CAT-1 was lower (p < 0.05), serum Lys was higher and serum Val was lower in pigs fed the ADE diet. The higher muscularity, MyHC IIb expression in Semitendinosus muscle and Lys serum of pigs fed the ADE diet suggest that Lys increases growth rate not only by functioning as protein construction unit but also as potential control of the protein synthesis process.
Collapse
Affiliation(s)
- A Morales
- Instituto de Ciencias Agrícolas, Universidad Autónoma de Baja California, Mexicali, BC, México
| | - H García
- Instituto de Ciencias Agrícolas, Universidad Autónoma de Baja California, Mexicali, BC, México
| | - N Arce
- Instituto de Ciencias Agrícolas, Universidad Autónoma de Baja California, Mexicali, BC, México
| | - M Cota
- Instituto de Ciencias Agrícolas, Universidad Autónoma de Baja California, Mexicali, BC, México
| | - R T Zijlstra
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - B A Araiza
- Instituto de Ciencias Agrícolas, Universidad Autónoma de Baja California, Mexicali, BC, México
| | - M Cervantes
- Instituto de Ciencias Agrícolas, Universidad Autónoma de Baja California, Mexicali, BC, México
| |
Collapse
|
83
|
Rath M, Müller I, Kropf P, Closs EI, Munder M. Metabolism via Arginase or Nitric Oxide Synthase: Two Competing Arginine Pathways in Macrophages. Front Immunol 2014; 5:532. [PMID: 25386178 PMCID: PMC4209874 DOI: 10.3389/fimmu.2014.00532] [Citation(s) in RCA: 784] [Impact Index Per Article: 78.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 10/09/2014] [Indexed: 12/23/2022] Open
Abstract
Macrophages play a major role in the immune system, both as antimicrobial effector cells and as immunoregulatory cells, which induce, suppress or modulate adaptive immune responses. These key aspects of macrophage biology are fundamentally driven by the phenotype of macrophage arginine metabolism that is prevalent in an evolving or ongoing immune response. M1 macrophages express the enzyme nitric oxide synthase, which metabolizes arginine to nitric oxide (NO) and citrulline. NO can be metabolized to further downstream reactive nitrogen species, while citrulline might be reused for efficient NO synthesis via the citrulline–NO cycle. M2 macrophages are characterized by expression of the enzyme arginase, which hydrolyzes arginine to ornithine and urea. The arginase pathway limits arginine availability for NO synthesis and ornithine itself can further feed into the important downstream pathways of polyamine and proline syntheses, which are important for cellular proliferation and tissue repair. M1 versus M2 polarization leads to opposing outcomes of inflammatory reactions, but depending on the context, M1 and M2 macrophages can be both pro- and anti-inflammatory. Notably, M1/M2 macrophage polarization can be driven by microbial infection or innate danger signals without any influence of adaptive immune cells, secondarily driving the T helper (Th)1/Th2 polarization of the evolving adaptive immune response. Since both arginine metabolic pathways cross-inhibit each other on the level of the respective arginine break-down products and Th1 and Th2 lymphocytes can drive or amplify macrophage M1/M2 dichotomy via cytokine activation, this forms the basis of a self-sustaining M1/M2 polarization of the whole immune response. Understanding the arginine metabolism of M1/M2 macrophage phenotypes is therefore central to find new possibilities to manipulate immune responses in infection, autoimmune diseases, chronic inflammatory conditions, and cancer.
Collapse
Affiliation(s)
- Meera Rath
- Department of Pharmacology, Institute of Medical Sciences, Faculty of Medical Sciences, Siksha 'O' Anusandhan University , Bhubaneshwar , India
| | - Ingrid Müller
- Section of Immunology, Department of Medicine, Imperial College London , London , UK
| | - Pascale Kropf
- Section of Immunology, Department of Medicine, Imperial College London , London , UK
| | - Ellen I Closs
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University , Mainz , Germany
| | - Markus Munder
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center, Johannes Gutenberg University , Mainz , Germany ; Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg University , Mainz , Germany
| |
Collapse
|
84
|
L-arginine stimulates CAT-1-mediated arginine uptake and regulation of inducible nitric oxide synthase for the growth of chick intestinal epithelial cells. Mol Cell Biochem 2014; 399:229-36. [PMID: 25336270 DOI: 10.1007/s11010-014-2249-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 10/09/2014] [Indexed: 10/24/2022]
Abstract
L-arginine (L-Arg) uptake is mediated by members of cationic amino acid transporter (CAT) family and may coincide with the induction of nitric oxide synthases (NOS). The present study was conducted to investigate the extracellular concentrations of L-Arg regulating the CAT-1, CAT-4 and inducible NOS (iNOS) in chick intestinal epithelial cells. The cells were cultured for 4 days in Arg-free Dulbecco's modified Eagle's medium containing 10, 100, 200, 400, or 600 μM L-Arg. Cell viability, nitric oxide (NO) concentrations, uptake and metabolism of L-[3H]-Arg as well as expression of CAT-1, CAT-4, and iNOS were determined. Our results showed that L-Arg enhances cell growth with a maximal response at 10-400 μM. Addition of 100, 200, or 400 μM L-Arg increased the L-[3H]-Arg uptake, which was associated with greater conversion of L-[3H]-citrulline and NO production in comparison with 10 μM L-Arg group. Increasing extracellular concentrations of L-Arg from 10 to 400 μM dose dependently increased the levels of CAT-1 mRNA and protein, while no effect on CAT-4 mRNA abundance was found. Furthermore, supplementation of 100, 200, or 400 μM L-Arg upregulated the expression of iNOS mRNA, and the relative protein levels for iNOS in 200 and 400 μM L-Arg groups were higher than those in 10 and 100 μM L-Arg groups. Collectively, we conclude that the CAT-1 isoform plays a role in L-Arg uptake, and L-Arg-mediated elevation of NO via iNOS promotes the growth of chick intestinal epithelial cells.
Collapse
|
85
|
Kuruppu S, Rajapakse NW, Dunstan RA, Smith AI. Nitric oxide inhibits the production of soluble endothelin converting enzyme-1. Mol Cell Biochem 2014; 396:49-54. [DOI: 10.1007/s11010-014-2141-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 07/11/2014] [Indexed: 12/27/2022]
|
86
|
Laufenberg LJ, Pruznak AM, Navaratnarajah M, Lang CH. Sepsis-induced changes in amino acid transporters and leucine signaling via mTOR in skeletal muscle. Amino Acids 2014; 46:2787-98. [PMID: 25218136 DOI: 10.1007/s00726-014-1836-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 09/03/2014] [Indexed: 12/13/2022]
Abstract
The present study tested the hypothesis that sepsis-induced leucine (Leu) resistance in skeletal muscle is associated with a down-regulation of amino acid transporters important in regulating Leu flux or an impairment in the formation of the Leu-sensitive mTOR-Ragulator complex. Sepsis in adult male rats decreased basal protein synthesis in gastrocnemius, associated with a reduction in mTOR activation as indicated by decreased 4E-BP1 and S6K1 phosphorylation. The ability of oral Leu to increase protein synthesis and mTOR kinase after 1 h was largely prevented in sepsis. Sepsis increased CAT1, LAT2 and SNAT2 mRNA content two- to fourfold, but only the protein content for CAT1 (20 % decrease) differed significantly. Conversely, sepsis decreased the proton-assisted amino acid transporter (PAT)-2 mRNA by 60 %, but without a coordinate change in PAT2 protein. There was no sepsis or Leu effect on the protein content for RagA-D, LAMTOR-1 and -2, raptor, Rheb or mTOR in muscle. The binding of mTOR, PRAS40 and RagC to raptor did not differ for control and septic muscle in the basal condition; however, the Leu-induced decrease in PRAS40·raptor and increase in RagC·raptor seen in control muscle was absent in sepsis. The intracellular Leu concentration was increased in septic muscle, compared to basal control conditions, and oral Leu further increased the intracellular Leu concentration similarly in both control and septic rats. Hence, while alterations in select amino acid transporters are not associated with development of sepsis-induced Leu resistance, the Leu-stimulated binding of raptor with RagC and the recruitment of mTOR/raptor to the endosome-lysosomal compartment may partially explain the inability of Leu to fully activate mTOR and muscle protein synthesis.
Collapse
Affiliation(s)
- Lacee J Laufenberg
- Departments of Cellular and Molecular Physiology (H166), and Surgery, Penn State College of Medicine, Hershey, PA, 17033, USA
| | | | | | | |
Collapse
|
87
|
Mechanisms of miRNA-Mediated Gene Regulation from Common Downregulation to mRNA-Specific Upregulation. Int J Genomics 2014; 2014:970607. [PMID: 25180174 PMCID: PMC4142390 DOI: 10.1155/2014/970607] [Citation(s) in RCA: 358] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 07/09/2014] [Accepted: 07/17/2014] [Indexed: 12/12/2022] Open
Abstract
Discovered in 1993, micoRNAs (miRNAs) are now recognized as one of the major regulatory gene families in eukaryotes. To date, 24521 microRNAs have been discovered and there are certainly more to come. It was primarily acknowledged that miRNAs result in gene expression repression at both the level of mRNA stability by conducting mRNA degradation and the level of translation (at initiation and after initiation) by inhibiting protein translation or degrading the polypeptides through binding complementarily to 3′UTR of the target mRNAs. Nevertheless, some studies revealed that miRNAs have the capability of activating gene expression directly or indirectly in respond to different cell types and conditions and in the presence of distinct cofactors. This reversibility in their posttranslational gene regulatory natures enables the bearing cells to rapidly response to different cell conditions and consequently block unnecessary energy wastage or maintain the cell state. This paper provides an overview of the current understandings of the miRNA characteristics including their genes and biogenesis, as well as their mediated downregulation. We also review up-to-date knowledge of miRNA-mediated gene upregulation through highlighting some notable examples and discuss the emerging concepts of their associations with other posttranscriptional gene regulation processes.
Collapse
|
88
|
Li C, Beck BH, Peatman E. Nutritional impacts on gene expression in the surface mucosa of blue catfish (Ictalurus furcatus). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2014; 44:226-234. [PMID: 24378224 DOI: 10.1016/j.dci.2013.12.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 12/20/2013] [Accepted: 12/21/2013] [Indexed: 06/03/2023]
Abstract
Short-term feed deprivation is a common occurrence in both wild and farmed fish species, due to reproductive processes, seasonal variations in temperature, or in response to a disease outbreak. Fasting can have dramatic physiological and biological consequences for fish, including impacts on mucosal immunity which can, in turn, change host susceptibility to pathogens. Culture and selection of blue catfish (Ictalurus furcatus) has gained importance as the production of a channel catfish×blue catfish (Ictalurus punctatus×I. furcatus) hybrid has increased in the Southeast US. Following a recent examination of fasting-induced impacts on mucosal immunity in channel catfish, here we utilized Illumina-based RNA-seq expression profiling to compare changes in blue catfish gill and skin after a brief (7 day) period of fasting. Transcriptome sequencing and de novo assembly of over 194 million 100 base-pair transcript reads was followed by differential expression analysis. Fasting altered a total of 530 genes in the surface mucosa, including genes regulating the immune response, energy metabolism, mucus production, cellular cytoskeletal structure, cell proliferation, and antioxidant responses. In particular, fasting perturbed arginine synthesis and metabolism pathways in a manner likely altering macrophage activation states and immune readiness. Our findings highlight key mediators of the critical interaction between nutrition and immunity at points of pathogen adherence and entry.
Collapse
Affiliation(s)
- Chao Li
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Benjamin H Beck
- United States Department of Agriculture, Agricultural Research Service, Stuttgart National Aquaculture Research Center, Stuttgart, AR 72160, USA
| | - Eric Peatman
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA.
| |
Collapse
|
89
|
Anderson DJ, Politch JA. Remarks on the article of Hadas et al.: Transmission of chimeric HIV by mating in conventional mice: prevention by pre-exposure antiretroviral therapy and reduced susceptibility during estrus. Dis Model Mech 2014; 7:177-8. [PMID: 24713273 PMCID: PMC3917237 DOI: 10.1242/dmm.014043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Deborah J Anderson
- Department of Obstetrics and Gynecology, Boston University School of Medicine, Boston, MA 02215, USA
| | | |
Collapse
|
90
|
Morales A, Grageola F, García H, Arce N, Araiza B, Yáñez J, Cervantes M. Performance, serum amino acid concentrations and expression of selected genes in pair-fed growing pigs exposed to high ambient temperatures. J Anim Physiol Anim Nutr (Berl) 2014; 98:928-35. [DOI: 10.1111/jpn.12161] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 12/06/2013] [Indexed: 12/25/2022]
Affiliation(s)
- A. Morales
- ICA; Universidad Autónoma de Baja California; Mexicali México
| | - F. Grageola
- ICA; Universidad Autónoma de Baja California; Mexicali México
| | - H. García
- ICA; Universidad Autónoma de Baja California; Mexicali México
| | - N. Arce
- ICA; Universidad Autónoma de Baja California; Mexicali México
| | - B. Araiza
- ICA; Universidad Autónoma de Baja California; Mexicali México
| | - J. Yáñez
- FMVZ; Universidad Autónoma de Tlaxcala; Tlaxcala México
| | - M. Cervantes
- ICA; Universidad Autónoma de Baja California; Mexicali México
| |
Collapse
|
91
|
The SLC3 and SLC7 families of amino acid transporters. Mol Aspects Med 2013; 34:139-58. [PMID: 23506863 DOI: 10.1016/j.mam.2012.10.007] [Citation(s) in RCA: 475] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 08/15/2012] [Indexed: 01/18/2023]
Abstract
Amino acids are necessary for all living cells and organisms. Specialized transporters mediate the transfer of amino acids across plasma membranes. Malfunction of these proteins can affect whole-body homoeostasis giving raise to diverse human diseases. Here, we review the main features of the SLC3 and SLC7 families of amino acid transporters. The SLC7 family is divided into two subfamilies, the cationic amino acid transporters (CATs), and the L-type amino acid transporters (LATs). The latter are the light or catalytic subunits of the heteromeric amino acid transporters (HATs), which are associated by a disulfide bridge with the heavy subunits 4F2hc or rBAT. These two subunits are glycoproteins and form the SLC3 family. Most CAT subfamily members were functionally characterized and shown to function as facilitated diffusers mediating the entry and efflux of cationic amino acids. In certain cells, CATs play an important role in the delivery of L-arginine for the synthesis of nitric oxide. HATs are mostly exchangers with a broad spectrum of substrates and are crucial in renal and intestinal re-absorption and cell redox balance. Furthermore, the role of the HAT 4F2hc/LAT1 in tumor growth and the application of LAT1 inhibitors and PET tracers for reduction of tumor progression and imaging of tumors are discussed. Finally, we describe the link between specific mutations in HATs and the primary inherited aminoacidurias, cystinuria and lysinuric protein intolerance.
Collapse
|
92
|
Määttä K, Kunnas T, Nikkari ST. Contribution of SLC7A1 genetic variant to hypertension, the TAMRISK study. BMC MEDICAL GENETICS 2013; 14:69. [PMID: 23841815 PMCID: PMC3710090 DOI: 10.1186/1471-2350-14-69] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Accepted: 07/01/2013] [Indexed: 01/01/2023]
Abstract
BACKGROUND The rs41318021 polymorphism in the SLC7A1 gene affects endothelial NO production through changes in L-arginine transport. This variation could thus hypothetically cause dysfunction of endothelium and lead to hypertension. The association of rs41318021 with hypertension was therefore studied in a Finnish cohort. METHODS A total of 412 hypertensive cases and 771 non-hypertensive controls from a Finnish 50-year-old cohort were included in this study. The data was collected from the Tampere adult population cardiovascular risk study (TAMRISK). DNA was extracted from buccal swabs and amplified using PCR. A subpopulation of men and women who had available follow-up data of blood pressure measurements at the age of 35-, 40-, 45- and 50 years was also analyzed. RESULTS There was no difference between the variant frequencies of the hypertension group and normotensive group at the age of 50 years (p = 0.209). However, repeated measures analysis from the 15-year follow-up showed that subjects having gene variants CT or TT had slightly higher diastolic blood pressure than subjects having genotype CC (p = 0.047). By post-hoc analysis, this was most pronounced at the age of 35 years (p = 0.044). CONCLUSION The rs41318021 polymorphism in the SLC7A1 gene was not associated with essential hypertension in 50-year-old subjects. However, a borderline effect of this variation upon diastolic blood pressure was seen in these same subjects in a 15-year follow-up from a 35-year-old cohort to 50 years of age.
Collapse
|
93
|
Sai KKS, Huang C, Yuan L, Zhou D, Piwnica-Worms D, Garbow JR, Engelbach JA, Mach RH, Rich KM, McConathy J. 18F-AFETP, 18F-FET, and 18F-FDG imaging of mouse DBT gliomas. J Nucl Med 2013; 54:1120-6. [PMID: 23650628 DOI: 10.2967/jnumed.112.113217] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED The goal of this study was to evaluate the (18)F-labeled nonnatural amino acid (S)-2-amino-3-[1-(2-(18)F-fluoroethyl)-1H-[1,2,3]triazol-4-yl]propanoic acid ((18)F-AFETP) as a PET imaging agent for brain tumors and to compare its effectiveness with the more-established tracers O-(2-(18)F-fluoroethyl)-l-tyrosine ((18)F-FET) and (18)F-FDG in a murine model of glioblastoma. The tracer (18)F-AFETP is a structural analog of histidine and is a lead compound for imaging cationic amino acid transport, a relatively unexplored target for oncologic imaging. METHODS (18)F-AFETP was prepared using the click reaction. BALB/c mice with intracranially implanted delayed brain tumor (DBT) gliomas (n = 4) underwent biodistribution and dynamic small-animal PET imaging for 60 min after intravenous injection of (18)F-AFETP. Tumor and brain uptake of (18)F-AFETP were compared with those of (18)F-FDG and (18)F-FET through small-animal PET analyses. RESULTS (18)F-AFETP demonstrated focally increased uptake in tumors with good visualization. Peak tumor uptake occurred within 10 min of injection, with stable or gradual decrease over time. All 3 tracers demonstrated relatively high uptake in the DBTs throughout the study. At late time points (47.5-57.5 min after injection), the average standardized uptake value with (18)F-FDG (1.9 ± 0.1) was significantly greater than with (18)F-FET (1.1 ± 0.1) and (18)F-AFETP (0.7 ± 0.2). The uptake also differed substantially in normal brain, with significant differences in the standardized uptake values at late times among (18)F-FDG (1.5 ± 0.2), (18)F-FET (0.5 ± 0.05), and (18)F-AFETP (0.1 ± 0.04). The resulting average tumor-to-brain ratio at the late time points was significantly higher for (18)F-AFETP (7.5 ± 0.1) than for (18)F-FDG (1.3 ± 0.1) and (18)F-FET (2.0 ± 0.3). CONCLUSION (18)F-AFETP is a promising brain tumor imaging agent, providing rapid and persistent tumor visualization, with good tumor-to-normal-brain ratios in the DBT glioma model. High tumor-to-brain, tumor-to-muscle, and tumor-to-blood ratios were observed at 30 and 60 min after injection, with higher tumor-to-brain ratios than obtained with (18)F-FET or (18)F-FDG. These results support further development and evaluation of (18)F-AFETP and its derivatives for tumor imaging.
Collapse
|
94
|
Krokowski D, Han J, Saikia M, Majumder M, Yuan CL, Guan BJ, Bevilacqua E, Bussolati O, Bröer S, Arvan P, Tchórzewski M, Snider MD, Puchowicz M, Croniger CM, Kimball SR, Pan T, Koromilas AE, Kaufman RJ, Hatzoglou M. A self-defeating anabolic program leads to β-cell apoptosis in endoplasmic reticulum stress-induced diabetes via regulation of amino acid flux. J Biol Chem 2013; 288:17202-13. [PMID: 23645676 DOI: 10.1074/jbc.m113.466920] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Endoplasmic reticulum (ER) stress-induced responses are associated with the loss of insulin-producing β-cells in type 2 diabetes mellitus. β-Cell survival during ER stress is believed to depend on decreased protein synthesis rates that are mediated via phosphorylation of the translation initiation factor eIF2α. It is reported here that chronic ER stress correlated with increased islet protein synthesis and apoptosis in β-cells in vivo. Paradoxically, chronic ER stress in β-cells induced an anabolic transcription program to overcome translational repression by eIF2α phosphorylation. This program included expression of amino acid transporter and aminoacyl-tRNA synthetase genes downstream of the stress-induced ATF4-mediated transcription program. The anabolic response was associated with increased amino acid flux and charging of tRNAs for branched chain and aromatic amino acids (e.g. leucine and tryptophan), the levels of which are early serum indicators of diabetes. We conclude that regulation of amino acid transport in β-cells during ER stress involves responses leading to increased protein synthesis, which can be protective during acute stress but can lead to apoptosis during chronic stress. These studies suggest that the increased expression of amino acid transporters in islets can serve as early diagnostic biomarkers for the development of diabetes.
Collapse
Affiliation(s)
- Dawid Krokowski
- Department of Nutrition, Case Western Reserve University, Cleveland, Ohio 44106, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
McCracken AN, Edinger AL. Nutrient transporters: the Achilles' heel of anabolism. Trends Endocrinol Metab 2013; 24:200-8. [PMID: 23402769 PMCID: PMC3617053 DOI: 10.1016/j.tem.2013.01.002] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 01/03/2013] [Accepted: 01/04/2013] [Indexed: 01/08/2023]
Abstract
Highly proliferative cells, including cancer cells, require a constant supply of molecular building blocks to support their growth. To acquire substrates such as glucose and amino acids from the extracellular space, dividing cells rely on transporter proteins in the plasma membrane. Numerous studies link transcriptional and post-translational control of nutrient transporter expression with proliferation, highlighting the importance of nutrient transporters in both physiologic and pathologic growth. Here we review recent work that spotlights the crucial role of nutrient transporters in cell growth and proliferation, discuss post-translational mechanisms for coordinating expression of different transporters, and consider the therapeutic potential of targeting these proteins in cancer and other diseases characterized by inappropriate cell division.
Collapse
Affiliation(s)
| | - Aimee L. Edinger
- Corresponding Author: Aimee L. Edinger 2128 Natural Sciences 1 University of California, Irvine Irvine, CA 92697-2300 Tel: 949-824-1921 FAX: 949-824-4709
| |
Collapse
|
96
|
A versatile proline/alanine transporter in the unicellular pathogen Leishmania donovani regulates amino acid homoeostasis and osmotic stress responses. Biochem J 2013; 449:555-66. [PMID: 22994895 DOI: 10.1042/bj20121262] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Unlike all other organisms, parasitic protozoa of the family Trypanosomatidae maintain a large cellular pool of proline that, together with the alanine pool, serve as alternative carbon sources as well as reservoirs of organic osmolytes. These reflect adaptation to their insect vectors whose haemolymphs are exceptionally rich in the two amino acids. In the present study we identify and characterize a new neutral amino acid transporter, LdAAP24, that translocates proline and alanine across the Leishmania donovani plasma membrane. This transporter fulfils multiple functions: it is the sole supplier for the intracellular pool of proline and contributes to the alanine pool; it is essential for cell volume regulation after osmotic stress; and it regulates the transport and homoeostasis of glutamate and arginine, none of which are its substrates. Notably, we provide evidence that proline and alanine exhibit different roles in the parasitic response to hypotonic shock; alanine affects swelling, whereas proline influences the rate of volume recovery. On the basis of our data we suggest that LdAAP24 plays a key role in parasite adaptation to its varying environments in host and vector, a phenomenon essential for successful parasitism.
Collapse
|
97
|
Cervantes-Ramírez M, Mendez-Trujillo V, Araiza-Piña B, Barrera-Silva M, González-Mendoza D, Morales-Trejo A. Supplemental leucine and isoleucine affect expression of cationic amino acid transporters and myosin, serum concentration of amino acids, and growth performance of pigs. GENETICS AND MOLECULAR RESEARCH 2013; 12:115-26. [DOI: 10.4238/2013.january.24.3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
98
|
Lamas B, Vergnaud-Gauduchon J, Goncalves-Mendes N, Perche O, Rossary A, Vasson MP, Farges MC. Altered functions of natural killer cells in response to L-Arginine availability. Cell Immunol 2012; 280:182-90. [PMID: 23399839 DOI: 10.1016/j.cellimm.2012.11.018] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 10/03/2012] [Accepted: 11/29/2012] [Indexed: 01/09/2023]
Abstract
L-Arginine (L-Arg) availability is crucial in the regulation of immune response. Indeed, L-Arg deficiency induces T-cell dysfunction and could modulate the properties of natural killer (NK) cells involved in the early host defense against infections and tumors. We explored the impact of L-Arg depletion on NK cell functions using two models - an NK-92 cell line and isolated human blood NK cells. Below 5mg/L of L-Arg, NK-92 cell proliferation was decreased and a total L-Arg depletion reduced NK-92 cell viability. NK cell cytotoxicity was significantly inhibited in presence of low L-Arg concentration (2.5 mg/L). L-Arg depletion reduced the expression of NK-92 activating receptors, NKp46 and NKp30, the expression of NK ζ chain and the NK-92 intracellular production of IFN-γ. Whatever the L-Arg concentrations tested, no significant variation in the gene expression of transporters and enzymes involved in L-Arg metabolism was found. Thus, L-Arg availability modulates the phenotypic and functional properties of NK cells.
Collapse
Affiliation(s)
- Bruno Lamas
- Clermont Université, Université d'Auvergne, EA 4233, Nutrition Cancerogenèse et Thérapie anti-tumorale, CLARA, CRNH Auvergne, Clermont-Ferrand, France.
| | | | | | | | | | | | | |
Collapse
|
99
|
García-Villalobos H, Morales-Trejo A, Araiza-Piña BA, Htoo JK, Cervantes-Ramírez M. Effects of dietary protein and amino acid levels on the expression of selected cationic amino acid transporters and serum amino acid concentration in growing pigs. Arch Anim Nutr 2012; 66:257-70. [DOI: 10.1080/1745039x.2012.697351] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
100
|
Dey S, Savant S, Teske BF, Hatzoglou M, Calkhoven CF, Wek RC. Transcriptional repression of ATF4 gene by CCAAT/enhancer-binding protein β (C/EBPβ) differentially regulates integrated stress response. J Biol Chem 2012; 287:21936-49. [PMID: 22556424 DOI: 10.1074/jbc.m112.351783] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Different environmental stresses induce the phosphorylation of eIF2 (eIF2∼P), repressing global protein synthesis coincident with preferential translation of ATF4. ATF4 is a transcriptional activator of genes involved in metabolism and nutrient uptake, antioxidation, and regulation of apoptosis. Because ATF4 is a common downstream target that integrates signaling from different eIF2 kinases and their respective stress signals, the eIF2∼P/ATF4 pathway is collectively referred to as the integrated stress response. Although eIF2∼P elicits translational control in response to many different stresses, there are selected stresses, such as exposure to UV irradiation, that do not increase ATF4 expression despite robust eIF2∼P. The rationale for this discordant induction of ATF4 expression and eIF2∼P in response to UV irradiation is that transcription of ATF4 is repressed, and therefore ATF4 mRNA is not available for preferential translation. In this study, we show that C/EBPβ is a transcriptional repressor of ATF4 during UV stress. C/EBPβ binds to critical elements in the ATF4 promoter, resulting in its transcriptional repression. Expression of C/EBPβ increases in response to UV stress, and the liver-enriched inhibitory protein (LIP) isoform of C/EBPβ, but not the liver-enriched activating protein (LAP) version, represses ATF4 transcription. Loss of the liver-enriched inhibitory protein isoform results in increased ATF4 mRNA levels in response to UV irradiation and subsequent recovery of ATF4 translation, leading to enhanced expression of its target genes. Together these results illustrate how eIF2∼P and translational control combined with transcription factors regulated by alternative signaling pathways can direct programs of gene expression that are specifically tailored to each environmental stress.
Collapse
Affiliation(s)
- Souvik Dey
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | | | | | | | |
Collapse
|