1
|
Barroso E, Díaz M, Reguera AC, Peyman M, Balsinde J, Jurado-Aguilar J, Zhang M, Rostami A, Palomer X, Ibáñez L, Vázquez-Carrera M. CHOP upregulation and dysregulation of the mature form of the SNAT2 amino acid transporter in the placentas from small for gestational age newborns. Cell Commun Signal 2023; 21:326. [PMID: 37957724 PMCID: PMC10644500 DOI: 10.1186/s12964-023-01352-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/10/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND The placentas from newborns that are small for gestational age (SGA; birth weight < -2 SD for gestational age) may display multiple pathological characteristics. A key determinant of fetal growth and, therefore, birth weight is placental amino acid transport, which is under the control of the serine/threonine kinase mechanistic target of rapamycin (mTOR). The effects of endoplasmic reticulum (ER) stress on the mTOR pathway and the levels of amino acid transporters are not well established. METHODS Placentas from SGA and appropriate for gestational age (AGA) newborns and the human placental BeWo cell line exposed to the ER stressor tunicamycin were used. RESULTS We detected a significant increase in the levels of C/EBP homologous protein (CHOP) in the placentas from SGA newborns compared with those from AGA newborns, while the levels of other ER stress markers were barely affected. In addition, placental mTOR Complex 1 (mTORC1) activity and the levels of the mature form of the amino acid transporter sodium-coupled neutral amino acid transporter 2 (SNAT2) were also reduced in the SGA group. Interestingly, CHOP has been reported to upregulate growth arrest and DNA damage-inducible protein 34 (GADD34), which in turn suppresses mTORC1 activity. The GADD34 inhibitor guanabenz attenuated the increase in CHOP protein levels and the reduction in mTORC1 activity caused by the ER stressor tunicamycin in the human placental cell line BeWo, but it did not recover mature SNAT2 protein levels, which might be reduced as a result of defective glycosylation. CONCLUSIONS Collectively, these data reveal that GADD34A activity and glycosylation are key factors controlling mTORC1 signaling and mature SNAT2 levels in trophoblasts, respectively, and might contribute to the SGA condition. Video Abstract.
Collapse
Affiliation(s)
- Emma Barroso
- Unitat de Farmacologia, Facultat de Farmàcia I Ciències de L'Alimentació, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Av. Joan XXIII 27-31, 08028, Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Marta Díaz
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, Spain
- Endocrinology, Pediatric Research Institute, Sant Joan de Déu Children's Hospital, Barcelona, Esplugues, Spain
| | - Ana Cristina Reguera
- Unitat de Farmacologia, Facultat de Farmàcia I Ciències de L'Alimentació, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Av. Joan XXIII 27-31, 08028, Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Mona Peyman
- Unitat de Farmacologia, Facultat de Farmàcia I Ciències de L'Alimentació, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Av. Joan XXIII 27-31, 08028, Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Jesús Balsinde
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas, Valladolid, Spain
| | - Javier Jurado-Aguilar
- Unitat de Farmacologia, Facultat de Farmàcia I Ciències de L'Alimentació, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Av. Joan XXIII 27-31, 08028, Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Meijian Zhang
- Unitat de Farmacologia, Facultat de Farmàcia I Ciències de L'Alimentació, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Av. Joan XXIII 27-31, 08028, Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Adel Rostami
- Unitat de Farmacologia, Facultat de Farmàcia I Ciències de L'Alimentació, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Av. Joan XXIII 27-31, 08028, Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Xavier Palomer
- Unitat de Farmacologia, Facultat de Farmàcia I Ciències de L'Alimentació, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Av. Joan XXIII 27-31, 08028, Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Lourdes Ibáñez
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, Spain
- Endocrinology, Pediatric Research Institute, Sant Joan de Déu Children's Hospital, Barcelona, Esplugues, Spain
| | - Manuel Vázquez-Carrera
- Unitat de Farmacologia, Facultat de Farmàcia I Ciències de L'Alimentació, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Av. Joan XXIII 27-31, 08028, Barcelona, Spain.
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain.
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, Spain.
| |
Collapse
|
2
|
Wan Y, Chen M, Li X, Han X, Zhong L, Xiao F, Liu J, Xiang J, Jiang J, Chen X, Liu J, Li H, Li B, Huang H, Hou J. Single-cell RNA sequencing reveals XBP1-SLC38A2 axis as a metabolic regulator in cytotoxic T lymphocytes in multiple myeloma. Cancer Lett 2023; 562:216171. [PMID: 37054944 DOI: 10.1016/j.canlet.2023.216171] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/28/2023] [Accepted: 04/05/2023] [Indexed: 04/15/2023]
Abstract
The mechanisms underlying the functional impairment and metabolic reprogramming of T lymphocytes in multiple myeloma (MM) have not been fully elucidated. In this study, single-cell RNA sequencing was used to compare gene expression profiles in T cells in bone marrow and peripheral blood of 10 newly diagnosed MM patients versus 3 healthy donors. Unbiased bioinformatics analysis revealed 9 cytotoxic T cell clusters. All 9 clusters in MM had higher expression of senescence markers (e.g., KLRG1 and CTSW) than the healthy control; some had higher expression of exhaustion-related markers (e.g., LAG3 and TNFRSF14). Pathway enrichment analyses showed downregulated amino acid metabolism and upregulated unfolded protein response (UPR) pathways, along with absent expression of glutamine transporter SLC38A2 and increased expression of UPR hallmark XBP1 in cytotoxic T cells in MM. In vitro studies revealed that XBP1 inhibited SLC38A2 by directly binding to its promoter, and silencing SLC38A2 resulted in decreased glutamine uptake and immune dysfunction of T cells. This study provided a landscape description of the immunosuppressive and metabolic features in T lymphocytes in MM, and suggested an important role of XBP1-SLC38A2 axis in T cell function.
Collapse
Affiliation(s)
- Yike Wan
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Mengping Chen
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Xin Li
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Xiaofeng Han
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Lu Zhong
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Fei Xiao
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Jia Liu
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Jing Xiang
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Jinxing Jiang
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Xiaotong Chen
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Junling Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hua Li
- Bio-ID Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Bin Li
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Honghui Huang
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Jian Hou
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
3
|
Transcriptomic Responses to Polymyxin B and Analogues in Human Kidney Tubular Cells. Antibiotics (Basel) 2023; 12:antibiotics12020415. [PMID: 36830325 PMCID: PMC9952791 DOI: 10.3390/antibiotics12020415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Polymyxins are last-line antibiotics for the treatment of Gram-negative 'superbugs'. However, nephrotoxicity can occur in up to 60% of patients administered intravenous polymyxins. The mechanisms underpinning nephrotoxicity remain unclear. To understand polymyxin-induced nephrotoxicity, human renal proximal tubule cells were treated for 24 h with 0.1 mM polymyxin B or two new analogues, FADDI-251 or FADDI-287. Transcriptomic analysis was performed, and differentially expressed genes (DEGs) were identified using ANOVA (FDR < 0.2). Cell viability following treatment with polymyxin B, FADDI-251 or FADDI-287 was 66.0 ± 5.33%, 89.3 ± 3.96% and 90.4 ± 1.18%, respectively. Transcriptomics identified 430, 193 and 150 DEGs with polymyxin B, FADDI-251 and FADDI-287, respectively. Genes involved with metallothioneins and Toll-like receptor pathways were significantly perturbed by all polymyxins. Only polymyxin B induced perturbations in signal transduction, including FGFR2 and MAPK signaling. SIGNOR network analysis showed all treatments affected essential regulators in the immune system, autophagy, cell cycle, oxidative stress and apoptosis. All polymyxins caused significant perturbations of metal homeostasis and TLR signaling, while polymyxin B caused the most dramatic perturbations of the transcriptome. This study reveals the impact of polymyxin structure modifications on transcriptomic responses in human renal tubular cells and provides important information for designing safer new-generation polymyxins.
Collapse
|
4
|
Huttunen J, Kronenberger T, Montaser AB, Králová A, Terasaki T, Poso A, Huttunen KM. Sodium-Dependent Neutral Amino Acid Transporter 2 Can Serve as a Tertiary Carrier for l-Type Amino Acid Transporter 1-Utilizing Prodrugs. Mol Pharm 2023; 20:1331-1346. [PMID: 36688491 PMCID: PMC9906736 DOI: 10.1021/acs.molpharmaceut.2c00948] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Membrane transporters are the key determinants of the homeostasis of endogenous compounds in the cells and their exposure to drugs. However, the substrate specificities of distinct transporters can overlap. In the present study, the interactions of l-type amino acid transporter 1 (LAT1)-utilizing prodrugs with sodium-coupled neutral amino acid transporter 2 (SNAT2) were explored. The results showed that the cellular uptake of LAT1-utilizing prodrugs into a human breast cancer cell line, MCF-7 cells, was mediated via SNATs as the uptake was increased at higher pH (8.5), decreased in the absence of sodium, and inhibited in the presence of unselective SNAT-inhibitor, (α-(methylamino)isobutyric acid, MeAIB). Moreover, docking the compounds to a SNAT2 homology model (inward-open conformation) and further molecular dynamics simulations and the subsequent trajectory and principal component analyses confirmed the chemical features supporting the interactions of the studied compounds with SNAT2, which was found to be the main SNAT expressed in MCF-7 cells.
Collapse
Affiliation(s)
- Johanna Huttunen
- School
of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O.
Box 1627, FI-70211 Kuopio, Finland
| | - Thales Kronenberger
- School
of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O.
Box 1627, FI-70211 Kuopio, Finland,Department
of Internal Medicine VIII, University Hospital
Tübingen, Otfried-Müller-Strasse
14, DE 72076 Tübingen, Germany,Department
of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical
Sciences, Eberhard-Karls-Universität,
Tübingen, Auf
der Morgenstelle 8, 72076 Tübingen, Germany,Cluster
of Excellence iFIT (EXC 2180) “Image-Guided and Functionally
Instructed Tumor Therapies”, University
of Tübingen, 72076 Tübingen, Germany,Tübingen
Center for Academic Drug Discovery & Development (TüCAD2), 72076 Tübingen, Germany
| | - Ahmed B. Montaser
- School
of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O.
Box 1627, FI-70211 Kuopio, Finland
| | - Adéla Králová
- School
of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O.
Box 1627, FI-70211 Kuopio, Finland
| | - Tetsuya Terasaki
- School
of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O.
Box 1627, FI-70211 Kuopio, Finland
| | - Antti Poso
- School
of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O.
Box 1627, FI-70211 Kuopio, Finland,Department
of Internal Medicine VIII, University Hospital
Tübingen, Otfried-Müller-Strasse
14, DE 72076 Tübingen, Germany,Department
of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical
Sciences, Eberhard-Karls-Universität,
Tübingen, Auf
der Morgenstelle 8, 72076 Tübingen, Germany,Cluster
of Excellence iFIT (EXC 2180) “Image-Guided and Functionally
Instructed Tumor Therapies”, University
of Tübingen, 72076 Tübingen, Germany,Tübingen
Center for Academic Drug Discovery & Development (TüCAD2), 72076 Tübingen, Germany
| | - Kristiina M. Huttunen
- School
of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O.
Box 1627, FI-70211 Kuopio, Finland,
| |
Collapse
|
5
|
Nielsen CU, Krog NF, Sjekirica I, Nielsen SS, Pedersen ML. SNAT2 is responsible for hyperosmotic induced sarcosine and glycine uptake in human prostate PC-3 cells. Pflugers Arch 2022; 474:1249-1262. [PMID: 36175560 DOI: 10.1007/s00424-022-02752-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/02/2022] [Accepted: 09/19/2022] [Indexed: 10/14/2022]
Abstract
Solute carriers (SLC) are important membrane transport proteins in normal and pathophysiological cells. The aim was to identify amino acid SLC(s) responsible for uptake of sarcosine and glycine in prostate cancer cells and investigate the impact hereon of hyperosmotic stress. Uptake of 14C-sarcosine and 3H-glycine was measured in human prostate cancer (PC-3) cells cultured under isosmotic (300 mOsm/kg) and hyperosmotic (500 mOsm/kg) conditions for 24 h. Hyperosmotic culture medium was obtained by supplementing the medium with 200 mM of the trisaccharide raffinose. Amino acid SLC expression was studied using RT-PCR, real-time PCR, and western blotting. siRNA knockdown of SNAT2 was performed. Experiments were conducted in at least 3 independent cell passages. The uptake of Sar and Gly was increased approximately 8-ninefold in PC-3 cells after 24 h hyperosmotic culture. PAT1 mRNA and protein could not be detected, while SNAT2 was upregulated at the mRNA and protein level. Transfection with SNAT2-specific siRNA reduced Vmax of Sar uptake from 2653 ± 38 to 513 ± 38 nmol mg protein-1 min-1, without altering the Km value (3.19 ± 0.13 vs. 3.42 ± 0.71 mM), indicating that SNAT2 is responsible for at least 80% of Sar uptake in hyperosmotic cultured PC-3 cells. SNAT2 is upregulated in hyperosmotic stressed prostate cancer cells and SNAT2 is responsible for cellular sarcosine and glycine uptake in hyperosmotic cultured PC-3 cells. Sar is identified as a substrate for SNAT2, and this has physiological implications for understanding cellular solute transport in prostate cancer cells.
Collapse
Affiliation(s)
- Carsten Uhd Nielsen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark.
| | - Nanna Friberg Krog
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
| | - Ilham Sjekirica
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
| | - Sidsel Strandgaard Nielsen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
| | - Maria L Pedersen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
| |
Collapse
|
6
|
York A, Everhart A, Vitek MP, Gottschalk KW, Colton CA. Metabolism-Based Gene Differences in Neurons Expressing Hyperphosphorylated AT8- Positive (AT8+) Tau in Alzheimer's Disease. ASN Neuro 2021; 13:17590914211019443. [PMID: 34121475 PMCID: PMC8207264 DOI: 10.1177/17590914211019443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Metabolic adaptations in the brain are critical to the establishment and maintenance of normal cellular functions and to the pathological responses to disease processes. Here, we have focused on specific metabolic pathways that are involved in immune-mediated neuronal processes in brain using isolated neurons derived from human autopsy brain sections of normal individuals and individuals diagnosed as Alzheimer's disease (AD). Laser capture microscopy was used to select specific cell types in immune-stained thin brain sections followed by NanoString technology to identify and quantify differences in mRNA levels between age-matched control and AD neuronal samples. Comparisons were also made between neurons isolated from AD brain sections expressing pathogenic hyperphosphorylated AT8- positive (AT8+) tau and non-AT8+ AD neurons using double labeling techniques. The mRNA expression data showed unique patterns of metabolic pathway expression between the subtypes of captured neurons that involved membrane based solute transporters, redox factors, and arginine and methionine metabolic pathways. We also identified the expression levels of a novel metabolic gene, Radical-S-Adenosyl Domain1 (RSAD1) and its corresponding protein, Rsad1, that impact methionine usage and radical based reactions. Immunohistochemistry was used to identify specific protein expression levels and their cellular location in NeuN+ and AT8+ neurons. APOE4 vs APOE3 genotype-specific and sex-specific gene expression differences in these metabolic pathways were also observed when comparing neurons from individuals with AD to age-matched individuals.
Collapse
Affiliation(s)
- Audra York
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, North Carolina, United States
| | - Angela Everhart
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, North Carolina, United States
| | - Michael P Vitek
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, North Carolina, United States
| | - Kirby W Gottschalk
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, North Carolina, United States
| | - Carol A Colton
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, North Carolina, United States
| |
Collapse
|
7
|
Sedlmeier EM, Meyer DM, Stecher L, Sailer M, Daniel H, Hauner H, Bader BL. Fetal sex modulates placental microRNA expression, potential microRNA-mRNA interactions, and levels of amino acid transporter expression and substrates: INFAT study subpopulation analysis of n-3 LCPUFA intervention during pregnancy and associations with offspring body composition. BMC Mol Cell Biol 2021; 22:15. [PMID: 33657992 PMCID: PMC7931339 DOI: 10.1186/s12860-021-00345-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 12/08/2020] [Indexed: 12/13/2022] Open
Abstract
Background Previously, we revealed sexually dimorphic mRNA expression and responsiveness to maternal dietary supplementation with n-3 long-chain polyunsaturated fatty acids (LCPUFA) in placentas from a defined INFAT study subpopulation. Here, we extended these analyses and explored the respective placental microRNA expression, putative microRNA-mRNA interactions, and downstream target processes as well as their associations with INFAT offspring body composition. Results We performed explorative placental microRNA profiling, predicted microRNA-mRNA interactions by bioinformatics, validated placental target microRNAs and their putative targets by RT-qPCR and western blotting, and measured amino acid levels in maternal and offspring cord blood plasma and placenta. microRNA, mRNA, protein, and amino acid levels were associated with each other and with offspring body composition from birth to 5 years of age. Forty-six differentially regulated microRNAs were found. Validations identified differential expression for microRNA-99a (miR-99a) and its predicted target genes mTOR, SLC7A5, encoding L-type amino acid transporter 1 (LAT1), and SLC6A6, encoding taurine transporter (TauT), and their prevailing significant sexually dimorphic regulation. Target mRNA levels were mostly higher in placentas from control male than from female offspring, whereas respective n-3 LCPUFA responsive target upregulation was predominantly found in female placentas, explaining the rather balanced expression levels between the sexes present only in the intervention group. LAT1 and TauT substrates tryptophan and taurine, respectively, were significantly altered in both maternal plasma at 32 weeks’ gestation and cord plasma following intervention, but not in the placenta. Several significant associations were observed for miR-99a, mTOR mRNA, SLC7A5 mRNA, and taurine and tryptophan in maternal and cord plasma with offspring body composition at birth, 1 year, 3 and 5 years of age. Conclusions Our data suggest that the analyzed targets may be part of a sexually dimorphic molecular regulatory network in the placenta, possibly modulating gene expression per se and/or counteracting n-3 LCPUFA responsive changes, and thereby stabilizing respective placental and fetal amino acid levels. Our data propose placental miR-99, SLC7A5 mRNA, and taurine and tryptophan levels in maternal and fetal plasma as potentially predictive biomarkers for offspring body composition. Supplementary Information The online version contains supplementary material available at 10.1186/s12860-021-00345-x.
Collapse
Affiliation(s)
- Eva-Maria Sedlmeier
- ZIEL-PhD Graduate School 'Epigenetics, Imprinting and Nutrition', ZIEL-Institute for Food and Health, School of Life Sciences Weihenstephan, Technical University of Munich, Gregor-Mendel-Straße 2, 85354, Freising, Germany.,Else Kröner-Fresenius-Center for Nutritional Medicine, School of Life Sciences Weihenstephan, Technical University of Munich, Gregor-Mendel-Straße 2, 85354, Freising, Germany
| | - Dorothy M Meyer
- Institute of Nutritional Medicine, School of Medicine, Technical University of Munich, Georg-Brauchle-Ring 62, 80992, Munich, Germany
| | - Lynne Stecher
- Institute of Nutritional Medicine, School of Medicine, Technical University of Munich, Georg-Brauchle-Ring 62, 80992, Munich, Germany
| | - Manuela Sailer
- Molecular Nutrition Unit, ZIEL-Institute for Food and Health, School of Life Sciences Weihenstephan, Technical University of Munich, Gregor-Mendel-Straße 2, 85354, Freising, Germany
| | - Hannelore Daniel
- Molecular Nutrition Unit, ZIEL-Institute for Food and Health, School of Life Sciences Weihenstephan, Technical University of Munich, Gregor-Mendel-Straße 2, 85354, Freising, Germany
| | - Hans Hauner
- Else Kröner-Fresenius-Center for Nutritional Medicine, School of Life Sciences Weihenstephan, Technical University of Munich, Gregor-Mendel-Straße 2, 85354, Freising, Germany.,Institute of Nutritional Medicine, School of Medicine, Technical University of Munich, Georg-Brauchle-Ring 62, 80992, Munich, Germany.,Clinical Nutritional Medicine Unit, ZIEL-Institute for Food and Health, School of Life Sciences Weihenstephan, Technical University of Munich, Gregor-Mendel-Straße 2, 85354, Freising, Germany
| | - Bernhard L Bader
- ZIEL-PhD Graduate School 'Epigenetics, Imprinting and Nutrition', ZIEL-Institute for Food and Health, School of Life Sciences Weihenstephan, Technical University of Munich, Gregor-Mendel-Straße 2, 85354, Freising, Germany. .,Else Kröner-Fresenius-Center for Nutritional Medicine, School of Life Sciences Weihenstephan, Technical University of Munich, Gregor-Mendel-Straße 2, 85354, Freising, Germany. .,Clinical Nutritional Medicine Unit, ZIEL-Institute for Food and Health, School of Life Sciences Weihenstephan, Technical University of Munich, Gregor-Mendel-Straße 2, 85354, Freising, Germany.
| |
Collapse
|
8
|
Blbas S, Watson E, Butler H, Brown J, Herbert TP, Stover CM, Bevington A, Abbasian N. Dexamethasone acutely suppresses the anabolic SNAT2/SLC38A2 amino acid transporter protein in L6-G8C5 rat skeletal muscle cells. FASEB Bioadv 2021; 3:36-48. [PMID: 33490882 PMCID: PMC7805547 DOI: 10.1096/fba.2020-00076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/23/2020] [Accepted: 09/30/2020] [Indexed: 12/17/2022] Open
Abstract
Chronic metabolic acidosis plays a role in cachexia by enhancing total proteolysis in skeletal muscle. Glucocorticoid also triggers proteolysis and plays a permissive role in the effect of acidosis. The System A amino acid transporter SNAT2/SLC38A2 is ubiquitously expressed in mammalian cells including muscle, performing Na+‐dependent active import of neutral amino acids, and is strongly inhibited by low pH. Exposure of rat skeletal muscle cell line L6‐G8C5 to low pH rapidly inhibits SNAT2 transport activity and enhances total proteolysis rate. Pharmacological inhibition or silencing of SNAT2 also enhances proteolysis. This study tests the hypothesis that the glucocorticoid dexamethasone (DEX), like low pH, inhibits SNAT2 activity in L6‐G8C5 myotubes, thus contributing to total proteolysis. Incubation with 500 nM DEX for 4 h reduced the System A amino acid transport rate to half the rate in control cultures. This inhibition depended on glucocorticoid receptor‐mediated gene transcription, but SNAT2 mRNA levels were unaffected by DEX. In contrast, the SNAT2 protein assessed by immunoblotting was significantly depleted. The co‐inhibitory effects of DEX and low pH on System A transport activity were additive in stimulating total proteolysis. In keeping with this mechanism, DEX’s inhibitory effect on SNAT2 transport activity was significantly blunted by the proteasome inhibitor MG132. Proof of principle was achieved in similar experiments using recombinant expression of a GFP‐tagged SNAT2 fusion protein in HEK293A cells. It is concluded that DEX acutely depletes the SNAT2 transporter protein, at least partly through proteasome‐dependent degradation of this functionally important transporter.
Collapse
Affiliation(s)
- Safia Blbas
- Department of Respiratory Sciences University of Leicester Leicester UK
| | - Emma Watson
- Department of Cardiovascular Sciences University of Leicester Leicester UK
| | - Heather Butler
- John Walls Renal Unit University Hospitals of Leicester Leicester UK
| | - Jeremy Brown
- Department of Cardiovascular Sciences University of Leicester Leicester UK
| | | | - Cordula M Stover
- Department of Respiratory Sciences University of Leicester Leicester UK
| | - Alan Bevington
- Department of Respiratory Sciences University of Leicester Leicester UK
| | - Nima Abbasian
- Department of Respiratory Sciences University of Leicester Leicester UK
| |
Collapse
|
9
|
Weidenfeld S, Chupin C, Langner DI, Zetoun T, Rozowsky S, Kuebler WM. Sodium-coupled neutral amino acid transporter SNAT2 counteracts cardiogenic pulmonary edema by driving alveolar fluid clearance. Am J Physiol Lung Cell Mol Physiol 2021; 320:L486-L497. [PMID: 33439101 DOI: 10.1152/ajplung.00461.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The constant transport of ions across the alveolar epithelial barrier regulates alveolar fluid homeostasis. Dysregulation or inhibition of Na+ transport causes fluid accumulation in the distal airspaces resulting in impaired gas exchange and respiratory failure. Previous studies have primarily focused on the critical role of amiloride-sensitive epithelial sodium channel (ENaC) in alveolar fluid clearance (AFC), yet activation of ENaC failed to attenuate pulmonary edema in clinical trials. Since 40% of AFC is amiloride-insensitive, Na+ channels/transporters other than ENaC such as Na+-coupled neutral amino acid transporters (SNATs) may provide novel therapeutic targets. Here, we identified a key role for SNAT2 (SLC38A2) in AFC and pulmonary edema resolution. In isolated perfused mouse and rat lungs, pharmacological inhibition of SNATs by HgCl2 and α-methylaminoisobutyric acid (MeAIB) impaired AFC. Quantitative RT-PCR identified SNAT2 as the highest expressed System A transporter in pulmonary epithelial cells. Pharmacological inhibition or siRNA-mediated knockdown of SNAT2 reduced transport of l-alanine across pulmonary epithelial cells. Homozygous Slc38a2-/- mice were subviable and died shortly after birth with severe cyanosis. Isolated lungs of Slc38a2+/- mice developed higher wet-to-dry weight ratios (W/D) as compared to wild type (WT) in response to hydrostatic stress. Similarly, W/D ratios were increased in Slc38a2+/- mice as compared to controls in an acid-induced lung injury model. Our results identify SNAT2 as a functional transporter for Na+ and neutral amino acids in pulmonary epithelial cells with a relevant role in AFC and the resolution of lung edema. Activation of SNAT2 may provide a new therapeutic strategy to counteract and/or reverse pulmonary edema.
Collapse
Affiliation(s)
- Sarah Weidenfeld
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada.,Institute of Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Cécile Chupin
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | | | - Tamador Zetoun
- Institute of Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Simon Rozowsky
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Wolfgang M Kuebler
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada.,Department of Surgery, University of Toronto, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada.,Institute of Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
10
|
Morotti M, Zois CE, El-Ansari R, Craze ML, Rakha EA, Fan SJ, Valli A, Haider S, Goberdhan DCI, Green AR, Harris AL. Increased expression of glutamine transporter SNAT2/SLC38A2 promotes glutamine dependence and oxidative stress resistance, and is associated with worse prognosis in triple-negative breast cancer. Br J Cancer 2021; 124:494-505. [PMID: 33028955 PMCID: PMC7852531 DOI: 10.1038/s41416-020-01113-y] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 09/10/2020] [Accepted: 09/17/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Glutamine (Gln) is an abundant nutrient used by cancer cells. Breast cancers cells and particularly triple-receptor negative breast cancer (TNBC) are reported to be dependent on Gln to produce the energy required for survival and proliferation. Despite intense research on the role of the intracellular Gln pathway, few reports have focussed on Gln transporters in breast cancer and TNBC. METHODS The role and localisation of the Gln transporter SLC38A2/SNAT2 in response to Gln deprivation or pharmacological stresses was examined in a panel of breast cancer cell lines. Subsequently, the effect of SLC38A2 knockdown in Gln-sensitive cell lines was analysed. The prognostic value of SLC38A2 in a cohort of breast cancer was determined by immunohistochemistry. RESULTS SLC38A2 was identified as a strongly expressed amino acid transporter in six breast cancer cell lines. We confirmed an autophagic route of degradation for SLC38A2. SLC38A2 knockdown decreased Gln consumption, inhibited cell growth, induced autophagy and led to ROS production in a subgroup of Gln-sensitive cell lines. High expression of SLC38A2 protein was associated with poor breast cancer specific survival in a large cohort of patients (p = 0.004), particularly in TNBC (p = 0.02). CONCLUSIONS These results position SLC38A2 as a selective target for inhibiting growth of Gln-dependent breast cancer cell lines.
Collapse
Affiliation(s)
- Matteo Morotti
- Hypoxia and Angiogenesis Group, Weatherall Institute of Molecular Medicine, Department of Oncology, University of Oxford, Oxford, OX3 9DS, UK.
| | - Christos E Zois
- Hypoxia and Angiogenesis Group, Weatherall Institute of Molecular Medicine, Department of Oncology, University of Oxford, Oxford, OX3 9DS, UK
| | - Rokaya El-Ansari
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, NG7 2RD, UK
| | - Madeleine L Craze
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, NG7 2RD, UK
| | - Emad A Rakha
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, NG7 2RD, UK
| | - Shih-Jung Fan
- Department of Physiology, Anatomy and Genetics, Le Gros Clark Building, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK
| | - Alessandro Valli
- Hypoxia and Angiogenesis Group, Weatherall Institute of Molecular Medicine, Department of Oncology, University of Oxford, Oxford, OX3 9DS, UK
| | - Syed Haider
- Hypoxia and Angiogenesis Group, Weatherall Institute of Molecular Medicine, Department of Oncology, University of Oxford, Oxford, OX3 9DS, UK
- The Breast Cancer Now Toby Robins Research Centre, Division of Breast Cancer Research, The Institute of Cancer Research, London, UK
| | - Deborah C I Goberdhan
- Department of Physiology, Anatomy and Genetics, Le Gros Clark Building, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK
| | - Andrew R Green
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, NG7 2RD, UK
| | - Adrian L Harris
- Hypoxia and Angiogenesis Group, Weatherall Institute of Molecular Medicine, Department of Oncology, University of Oxford, Oxford, OX3 9DS, UK.
| |
Collapse
|
11
|
Zakoji N, Tajima K, Yoneyama D, Akanuma SI, Kubo Y, Hosoya KI. Involvement of sodium-coupled neutral amino acid transporters (system A) in l-proline transport in the rat retinal pericytes. Drug Metab Pharmacokinet 2020; 35:410-416. [PMID: 32771261 DOI: 10.1016/j.dmpk.2020.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 06/02/2020] [Accepted: 06/09/2020] [Indexed: 11/26/2022]
Abstract
The retinal pericytes contribute to the supply of collagen to the basement membrane, and thus, form the structural support of the blood-retinal barrier. Since l-proline (L-Pro) is a major component of collagen, the uptake of L-Pro is an important process for the synthesis of collagen. This study was aimed to elucidate L-Pro transport mechanism(s) in the retinal pericytes. The transport of [3H]L-Pro was evaluated in the conditionally immortalized rat retinal pericyte cell line, TR-rPCT1 cells. The expression of the candidate transporter was examined by qualitative/quantitative reverse transcription-polymerase chain reaction, immunoblot analysis, and immunostaining. The [3H]L-Pro uptake by TR-rPCT1 cells showed Na+-dependence, Cl--independence, and concentration-dependence with a Km of 810 μM. The substrates for system A, such as 2-(methylamino)isobutyric acid (MeAIB), significantly inhibited the L-Pro uptake, suggesting the involvement of system A in the uptake of L-Pro. Among the subtypes of system A, the mRNA expression levels of ATA2 were the highest in TR-rPCT1 cells. Immunostaining analysis of the isolated rat retinal capillaries containing pericytes indicated the protein expression of ATA2 in retinal pericytes. In conclusion, it is suggested that ATA2, at least in part, is involved in the transport of L-Pro in the retinal pericytes.
Collapse
Affiliation(s)
- Nobuyuki Zakoji
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, Japan
| | - Kosuke Tajima
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, Japan
| | - Daisuke Yoneyama
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, Japan
| | - Shin-Ichi Akanuma
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, Japan
| | - Yoshiyuki Kubo
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, Japan
| | - Ken-Ichi Hosoya
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, Japan.
| |
Collapse
|
12
|
Salger SA, Reza J, Deck CA, Wahab MA, Baltzegar DA, Murr AT, Borski RJ. Enhanced biodiversity of gut flora and feed efficiency in pond cultured tilapia under reduced frequency feeding strategies. PLoS One 2020; 15:e0236100. [PMID: 32702020 PMCID: PMC7377384 DOI: 10.1371/journal.pone.0236100] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 06/30/2020] [Indexed: 02/01/2023] Open
Abstract
Feed constitutes 50-70% of total production costs of tilapia, one of the most widely cultured finfishes in the world. We evaluated reduced-feeding strategies for improving production efficiency of Nile tilapia (Oreochromis niloticus). In a 12-week pond trial, fish were fed daily, every other day, every third day, or not at all. Ponds were fertilized to enhance natural foods. In a fifth group fish were fed daily without pond fertilization. Fish fed daily with or without pond fertilization and fish fed every other day had higher specific growth rates, survivability, and net production than the other two treatments. Fish feed efficiency and benefit to cost ratio was highest for treatments fed in a pulsatile manner (i.e. fed every other day or every third day) with fish fed on alternate days providing the best net return among all groups. Fish fed on alternate days had more moderate gene expression levels of intestinal nutrient transporters which may allow for a more balanced and efficient nutrient uptake. Fecal microbe analyses identified 145 families of prokaryotic and 132 genera of eukaryotic organisms in tilapia. The highest diversity of prokaryotes was found in fish fed either every other day or daily in fertilized ponds and the highest diversity of eukaryotes was found in fish fed every other day. These studies indicate feeding Nile tilapia on alternate days along with weekly pond fertilization has no deleterious effects on growth, survivability, or production versus daily feeding regimes, but enhances feed efficiency by 76% and provides the greatest net return on investments. Our studies also suggest for the first time that combining alternate-day feeding with pond fertilization produces the greatest microbial biodiversity in the intestine that could contribute to enhanced feed efficiency and overall health of tilapia.
Collapse
Affiliation(s)
- Scott A. Salger
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Jimi Reza
- Department of Fisheries Management, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Courtney A. Deck
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Md. Abdul Wahab
- Department of Fisheries Management, Bangladesh Agricultural University, Mymensingh, Bangladesh
- WorldFish, Dhaka, Bangladesh
| | - David A. Baltzegar
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, United States of America
- Genomic Sciences Laboratory, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Alexander T. Murr
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Russell J. Borski
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, United States of America
| |
Collapse
|
13
|
Zhang L, Duan Y, Guo Q, Wang W, Li F. A selectively suppressing amino acid transporter: Sodium-coupled neutral amino acid transporter 2 inhibits cell growth and mammalian target of rapamycin complex 1 pathway in skeletal muscle cells. ACTA ACUST UNITED AC 2020; 6:513-520. [PMID: 33364468 PMCID: PMC7750797 DOI: 10.1016/j.aninu.2020.03.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/11/2020] [Accepted: 03/13/2020] [Indexed: 12/18/2022]
Abstract
Sodium-coupled neutral amino acid transporter 2 (SNAT2), also known as solute carrier family 38 member 2 (SLC38A2), is expressed in the skeletal muscle. Our research previously indicated that SNAT2 mRNA expression level in the skeletal muscle was modulated by genotype and dietary protein. The aim of this study was to investigate the key role of the amino acid transporter SNAT2 in muscle cell growth, differentiation, and related signaling pathways via SNAT2 suppression using the inhibitor α-methylaminoisobutyric acid (MeAIB). The results showed that SNAT2 suppression down-regulated both the mRNA and protein expression levels of SNAT2 in C2C12 cells, inhibited cell viability and differentiation of the cell, and regulated the cell distribution in G0/G1 and S phases (P < 0.05). Meanwhile, most of the intercellular amino acid content of the cells after MeAIB co-culturing was significantly lower (P < 0.05). Furthermore, the mRNA expression levels of system L amino acid transporter 1 (LAT1), silent information regulator 1, and peroxisome proliferator-activated receptor-gamma co-activator 1 alpha, as well as the protein expression levels of amino acid transporters LAT1 and vacuolar protein sorting 34, were all down-regulated. The phosphorylated protein expression levels of mammalian target of rapamycin (mTOR), regulatory-associated protein of mTOR, 4E binding protein 1, and ribosomal protein S6 kinase 1 after MeAIB treatment were also significantly down-regulated (P < 0.05), which could contribute to the importance of SNAT2 in amino acid transportation and skeletal muscle cell sensing. In conclusion, SNAT2 suppression inhibited C2C12 cell growth and differentiation, as well as the availability of free amino acids. Although the mTOR complex 1 signaling pathway was found to be involved, its response to different nutrients requires further study.
Collapse
Affiliation(s)
- Lingyu Zhang
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China.,Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, Hunan 410125, China.,University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Yehui Duan
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China.,Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, Hunan 410125, China
| | - Qiuping Guo
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China.,Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, Hunan 410125, China.,University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Wenlong Wang
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China.,Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, Hunan 410125, China.,Laboratory of Animal Nutrition and Human Health, School of Biology, Hunan Normal University, Changsha, 410018, China
| | - Fengna Li
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China.,Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, Hunan 410125, China
| |
Collapse
|
14
|
Bensimon A, Pizzagalli MD, Kartnig F, Dvorak V, Essletzbichler P, Winter GE, Superti-Furga G. Targeted Degradation of SLC Transporters Reveals Amenability of Multi-Pass Transmembrane Proteins to Ligand-Induced Proteolysis. Cell Chem Biol 2020; 27:728-739.e9. [PMID: 32386596 PMCID: PMC7303955 DOI: 10.1016/j.chembiol.2020.04.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 02/21/2020] [Accepted: 04/03/2020] [Indexed: 01/05/2023]
Abstract
With more than 450 members, the solute carrier (SLC) group of proteins represents the largest class of transporters encoded in the human genome. Their several-pass transmembrane domain structure and hydrophobicity contribute to the orphan status of many SLCs, devoid of known cargos or chemical inhibitors. We report that SLC proteins belonging to different families and subcellular compartments are amenable to induced degradation by heterobifunctional ligands. Engineering endogenous alleles via the degradation tag (dTAG) technology enabled chemical control of abundance of the transporter protein, SLC38A2. Moreover, we report the design of d9A-2, a chimeric compound engaging several members of the SLC9 family and leading to their degradation. d9A-2 impairs cellular pH homeostasis and promotes cell death in a range of cancer cell lines. These findings open the era of SLC-targeting chimeric degraders and demonstrate potential access of multi-pass transmembrane proteins of different subcellular localizations to the chemically exploitable degradation machinery.
Collapse
Affiliation(s)
- Ariel Bensimon
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Mattia D Pizzagalli
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Felix Kartnig
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Vojtech Dvorak
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Patrick Essletzbichler
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Georg E Winter
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Giulio Superti-Furga
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria; Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria.
| |
Collapse
|
15
|
Parker SJ, Amendola CR, Hollinshead KER, Yu Q, Yamamoto K, Encarnación-Rosado J, Rose RE, LaRue MM, Sohn ASW, Biancur DE, Paulo JA, Gygi SP, Jones DR, Wang H, Philips MR, Bar-Sagi D, Mancias JD, Kimmelman AC. Selective Alanine Transporter Utilization Creates a Targetable Metabolic Niche in Pancreatic Cancer. Cancer Discov 2020; 10:1018-1037. [PMID: 32341021 DOI: 10.1158/2159-8290.cd-19-0959] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 02/21/2020] [Accepted: 04/22/2020] [Indexed: 11/16/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) evolves a complex microenvironment comprised of multiple cell types, including pancreatic stellate cells (PSC). Previous studies have demonstrated that stromal supply of alanine, lipids, and nucleotides supports the metabolism, growth, and therapeutic resistance of PDAC. Here we demonstrate that alanine cross-talk between PSCs and PDAC is orchestrated by the utilization of specific transporters. PSCs utilize SLC1A4 and other transporters to rapidly exchange and maintain environmental alanine concentrations. Moreover, PDAC cells upregulate SLC38A2 to supply their increased alanine demand. Cells lacking SLC38A2 fail to concentrate intracellular alanine and undergo a profound metabolic crisis resulting in markedly impaired tumor growth. Our results demonstrate that stromal-cancer metabolic niches can form through differential transporter expression, creating unique therapeutic opportunities to target metabolic demands of cancer. SIGNIFICANCE: This work identifies critical neutral amino acid transporters involved in channeling alanine between pancreatic stellate and PDAC cells. Targeting PDAC-specific alanine uptake results in a metabolic crisis impairing metabolism, proliferation, and tumor growth. PDAC cells specifically activate and require SLC38A2 to fuel their alanine demands that may be exploited therapeutically.This article is highlighted in the In This Issue feature, p. 890.
Collapse
Affiliation(s)
- Seth J Parker
- Department of Radiation Oncology, New York University School of Medicine, New York, New York
- Perlmutter Cancer Center, New York University School of Medicine, New York, New York
| | - Caroline R Amendola
- Perlmutter Cancer Center, New York University School of Medicine, New York, New York
| | - Kate E R Hollinshead
- Department of Radiation Oncology, New York University School of Medicine, New York, New York
- Perlmutter Cancer Center, New York University School of Medicine, New York, New York
| | - Qijia Yu
- Division of Genomic Stability and DNA Repair, Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Keisuke Yamamoto
- Department of Radiation Oncology, New York University School of Medicine, New York, New York
- Perlmutter Cancer Center, New York University School of Medicine, New York, New York
| | - Joel Encarnación-Rosado
- Department of Radiation Oncology, New York University School of Medicine, New York, New York
- Perlmutter Cancer Center, New York University School of Medicine, New York, New York
| | - Rebecca E Rose
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York
| | - Madeleine M LaRue
- Perlmutter Cancer Center, New York University School of Medicine, New York, New York
| | - Albert S W Sohn
- Department of Radiation Oncology, New York University School of Medicine, New York, New York
- Perlmutter Cancer Center, New York University School of Medicine, New York, New York
| | - Doug E Biancur
- Department of Radiation Oncology, New York University School of Medicine, New York, New York
- Perlmutter Cancer Center, New York University School of Medicine, New York, New York
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts
| | - Drew R Jones
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York
| | - Huamin Wang
- Department of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mark R Philips
- Perlmutter Cancer Center, New York University School of Medicine, New York, New York
| | - Dafna Bar-Sagi
- Perlmutter Cancer Center, New York University School of Medicine, New York, New York
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York
- Department of Medicine, New York University School of Medicine, New York, New York
| | - Joseph D Mancias
- Division of Genomic Stability and DNA Repair, Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Alec C Kimmelman
- Department of Radiation Oncology, New York University School of Medicine, New York, New York.
- Perlmutter Cancer Center, New York University School of Medicine, New York, New York
| |
Collapse
|
16
|
Curnock R, Calcagni A, Ballabio A, Cullen PJ. TFEB controls retromer expression in response to nutrient availability. J Cell Biol 2019; 218:3954-3966. [PMID: 31694921 PMCID: PMC6891082 DOI: 10.1083/jcb.201903006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/05/2019] [Accepted: 09/23/2019] [Indexed: 12/13/2022] Open
Abstract
Endosomal recycling maintains the cell surface abundance of nutrient transporters for nutrient uptake, but how the cell integrates nutrient availability with recycling is less well understood. Here, in studying the recycling of human glutamine transporters ASCT2 (SLC1A5), LAT1 (SLC7A5), SNAT1 (SLC38A1), and SNAT2 (SLC38A2), we establish that following amino acid restriction, the adaptive delivery of SNAT2 to the cell surface relies on retromer, a master conductor of endosomal recycling. Upon complete amino acid starvation or selective glutamine depletion, we establish that retromer expression is upregulated by transcription factor EB (TFEB) and other members of the MiTF/TFE family of transcription factors through association with CLEAR elements in the promoters of the retromer genes VPS35 and VPS26A TFEB regulation of retromer expression therefore supports adaptive nutrient acquisition through endosomal recycling.
Collapse
Affiliation(s)
- Rachel Curnock
- School of Biochemistry, University of Bristol, Bristol, UK
| | | | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine, Naples, Italy.,Department of Molecular and Human Genetics and Neurological Research Institute, Baylor College of Medicine, Houston, TX.,Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, Naples, Italy
| | - Peter J Cullen
- School of Biochemistry, University of Bristol, Bristol, UK
| |
Collapse
|
17
|
Menchini RJ, Chaudhry FA. Multifaceted regulation of the system A transporter Slc38a2 suggests nanoscale regulation of amino acid metabolism and cellular signaling. Neuropharmacology 2019; 161:107789. [PMID: 31574264 DOI: 10.1016/j.neuropharm.2019.107789] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 09/16/2019] [Accepted: 09/20/2019] [Indexed: 02/07/2023]
Abstract
Amino acids are essential for cellular protein synthesis, growth, metabolism, signaling and in stress responses. Cell plasma membranes harbor specialized transporters accumulating amino acids to support a variety of cellular biochemical pathways. Several transporters for neutral amino acids have been characterized. However, Slc38a2 (also known as SA1, SAT2, ATA2, SNAT2) representing the classical transport system A activity stands in a unique position: Being a secondarily active transporter energized by the electrochemical gradient of Na+, it creates steep concentration gradients for amino acids such as glutamine: this may subsequently drive the accumulation of additional neutral amino acids through exchange via transport systems ASC and L. Slc38a2 is ubiquitously expressed, yet in a cell-specific manner. In this review, we show that Slc38a2 is regulated at the transcriptional and translational levels as well as by ions and proteins through direct interactions. We describe how Slc38a2 senses amino acid availability and passes this onto intracellular signaling pathways and how it regulates protein synthesis, cellular proliferation and apoptosis through the mechanistic (mammalian) target of rapamycin (mTOR) and general control nonderepressible 2 (GCN2) pathways. Furthermore, we review how this extensively regulated transporter contributes to cellular osmoadaptation and how it is regulated by endoplasmic reticulum stress and various hormonal stimuli to promote cellular metabolism, cellular signaling and cell survival. This article is part of the issue entitled 'Special Issue on Neurotransmitter Transporters'.
Collapse
Affiliation(s)
| | - Farrukh Abbas Chaudhry
- Department of Molecular Medicine, University of Oslo, Oslo, Norway; Department of Plastic and Reconstructive Surgery, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
18
|
Abstract
Amino acids perform a variety of functions in cells and organisms, particularly in the synthesis of proteins, as energy metabolites, neurotransmitters, and precursors for many other molecules. Amino acid transport plays a key role in all these functions. Inhibition of amino acid transport is pursued as a therapeutic strategy in several areas, such as diabetes and related metabolic disorders, neurological disorders, cancer, and stem cell biology. The role of amino acid transporters in these disorders and processes is well established, but the implementation of amino acid transporters as drug targets is still in its infancy. This is at least in part due to the underdeveloped pharmacology of this group of membrane proteins. Recent advances in structural biology, membrane protein expression, and inhibitor screening methodology will see an increased number of improved and selective inhibitors of amino acid transporters that can serve as tool compounds for further studies.
Collapse
Affiliation(s)
- Stefan Bröer
- 1 Research School of Biology, College of Science, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
19
|
Morotti M, Bridges E, Valli A, Choudhry H, Sheldon H, Wigfield S, Gray N, Zois CE, Grimm F, Jones D, Teoh EJ, Cheng WC, Lord S, Anastasiou D, Haider S, McIntyre A, Goberdhan DCI, Buffa F, Harris AL. Hypoxia-induced switch in SNAT2/SLC38A2 regulation generates endocrine resistance in breast cancer. Proc Natl Acad Sci U S A 2019; 116:12452-12461. [PMID: 31152137 PMCID: PMC6589752 DOI: 10.1073/pnas.1818521116] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Tumor hypoxia is associated with poor patient outcomes in estrogen receptor-α-positive (ERα+) breast cancer. Hypoxia is known to affect tumor growth by reprogramming metabolism and regulating amino acid (AA) uptake. Here, we show that the glutamine transporter, SNAT2, is the AA transporter most frequently induced by hypoxia in breast cancer, and is regulated by hypoxia both in vitro and in vivo in xenografts. SNAT2 induction in MCF7 cells was also regulated by ERα, but it became predominantly a hypoxia-inducible factor 1α (HIF-1α)-dependent gene under hypoxia. Relevant to this, binding sites for both HIF-1α and ERα overlap in SNAT2's cis-regulatory elements. In addition, the down-regulation of SNAT2 by the ER antagonist fulvestrant was reverted in hypoxia. Overexpression of SNAT2 in vitro to recapitulate the levels induced by hypoxia caused enhanced growth, particularly after ERα inhibition, in hypoxia, or when glutamine levels were low. SNAT2 up-regulation in vivo caused complete resistance to antiestrogen and, partially, anti-VEGF therapies. Finally, high SNAT2 expression levels correlated with hypoxia profiles and worse outcome in patients given antiestrogen therapies. Our findings show a switch in the regulation of SNAT2 between ERα and HIF-1α, leading to endocrine resistance in hypoxia. Development of drugs targeting SNAT2 may be of value for a subset of hormone-resistant breast cancer.
Collapse
Affiliation(s)
- Matteo Morotti
- Hypoxia and Angiogenesis Group, Cancer Research UK Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, Department of Oncology, University of Oxford, Oxford OX3 9DS, United Kingdom;
| | - Esther Bridges
- Hypoxia and Angiogenesis Group, Cancer Research UK Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, Department of Oncology, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Alessandro Valli
- Hypoxia and Angiogenesis Group, Cancer Research UK Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, Department of Oncology, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Hani Choudhry
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah F6VM+J2, Saudi Arabia
| | - Helen Sheldon
- Hypoxia and Angiogenesis Group, Cancer Research UK Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, Department of Oncology, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Simon Wigfield
- Hypoxia and Angiogenesis Group, Cancer Research UK Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, Department of Oncology, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Nicki Gray
- Computational Biology Research Group, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Christos E Zois
- Hypoxia and Angiogenesis Group, Cancer Research UK Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, Department of Oncology, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Fiona Grimm
- Cancer Metabolism Laboratory, Francis Crick Institute, London NW1 1ST, United Kingdom
| | - Dylan Jones
- Hypoxia and Angiogenesis Group, Cancer Research UK Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, Department of Oncology, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Eugene J Teoh
- Hypoxia and Angiogenesis Group, Cancer Research UK Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, Department of Oncology, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Wei-Chen Cheng
- Computational Biology and Integrative Genomics, Department of Oncology, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Simon Lord
- Hypoxia and Angiogenesis Group, Cancer Research UK Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, Department of Oncology, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Dimitrios Anastasiou
- Cancer Metabolism Laboratory, Francis Crick Institute, London NW1 1ST, United Kingdom
| | - Syed Haider
- Computational Biology and Integrative Genomics, Department of Oncology, University of Oxford, Oxford OX3 7DQ, United Kingdom
- The Breast Cancer Now Toby Robins Research Centre, Division of Breast Cancer Research, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Alan McIntyre
- Hypoxia and Angiogenesis Group, Cancer Research UK Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, Department of Oncology, University of Oxford, Oxford OX3 9DS, United Kingdom
- Cancer Biology, Division of Cancer and Stem Cells, The University of Nottingham, Nottingham NG7 2UH, United Kingdom
| | - Deborah C I Goberdhan
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, United Kingdom
| | - Francesca Buffa
- Computational Biology and Integrative Genomics, Department of Oncology, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Adrian L Harris
- Hypoxia and Angiogenesis Group, Cancer Research UK Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, Department of Oncology, University of Oxford, Oxford OX3 9DS, United Kingdom;
| |
Collapse
|
20
|
Dietary supplementation with arginine and glutamic acid alters the expression of amino acid transporters in skeletal muscle of growing pigs. Amino Acids 2019; 51:1081-1092. [DOI: 10.1007/s00726-019-02748-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 05/26/2019] [Indexed: 01/06/2023]
|
21
|
Baek JH, Vignesh A, Son H, Lee DH, Roh GS, Kang SS, Cho GJ, Choi WS, Kim HJ. Glutamine Supplementation Ameliorates Chronic Stress-induced Reductions in Glutamate and Glutamine Transporters in the Mouse Prefrontal Cortex. Exp Neurobiol 2019; 28:270-278. [PMID: 31138994 PMCID: PMC6526116 DOI: 10.5607/en.2019.28.2.270] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 03/28/2019] [Accepted: 04/07/2019] [Indexed: 12/14/2022] Open
Abstract
Chronic immobilization stress (CIS) induces low levels of glutamate (Glu) and glutamine (Gln) and hypoactive glutamatergic signaling in the mouse prefrontal cortex (PFC), which is closely related to the Glu-Gln cycle. A Gln-supplemented diet ameliorates CIS-induced deleterious changes. Here, we investigated the effects of CIS and Gln supplementation on Glu-Gln cycle-related proteins to characterize the underlying mechanisms. Using the CIS-induced depression mouse model, we examined the expression of 11 proteins involved in the Glu-Gln cycle in the PFC. CIS decreased levels of glutamate transporter 1 (GLT1) and sodium-coupled neutral amino acid transporter (SNAT) 1, SANT2, SNAT3, and SNAT5. Gln supplementation did not affect the non-stressed group but significantly increased GLT1 and SNATs of the stressed group. By immunohistochemical analysis, we confirmed that SNAT1 and SNAT2 were decreased in neurons and GLT1, SNAT3, and SNAT5 were decreased in astrocytes in the medial PFC of the stressed group, but Gln-supplemented diet ameliorated these decrements. Collectively, these results suggest that CIS may cause depressive-like behaviors by decreasing Glu and Gln transportation in the PFC and that a Gln-supplemented diet could prevent the deleterious effects of CIS.
Collapse
Affiliation(s)
- Ji Hyeong Baek
- Department of Anatomy and Convergence Medical Sciences, Institute of Health Sciences, Bio Anti-aging Medical Research Center, Gyeongsang National University School of Medicine, Jinju 52727, Korea
| | - Arul Vignesh
- Department of Anatomy and Convergence Medical Sciences, Institute of Health Sciences, Bio Anti-aging Medical Research Center, Gyeongsang National University School of Medicine, Jinju 52727, Korea
| | - Hyeonwi Son
- Department of Anatomy and Convergence Medical Sciences, Institute of Health Sciences, Bio Anti-aging Medical Research Center, Gyeongsang National University School of Medicine, Jinju 52727, Korea
| | - Dong Hoon Lee
- Department of Anatomy and Convergence Medical Sciences, Institute of Health Sciences, Bio Anti-aging Medical Research Center, Gyeongsang National University School of Medicine, Jinju 52727, Korea
| | - Gu Seob Roh
- Department of Anatomy and Convergence Medical Sciences, Institute of Health Sciences, Bio Anti-aging Medical Research Center, Gyeongsang National University School of Medicine, Jinju 52727, Korea
| | - Sang Soo Kang
- Department of Anatomy and Convergence Medical Sciences, Institute of Health Sciences, Bio Anti-aging Medical Research Center, Gyeongsang National University School of Medicine, Jinju 52727, Korea
| | - Gyeong Jae Cho
- Department of Anatomy and Convergence Medical Sciences, Institute of Health Sciences, Bio Anti-aging Medical Research Center, Gyeongsang National University School of Medicine, Jinju 52727, Korea
| | - Wan Sung Choi
- Department of Anatomy and Convergence Medical Sciences, Institute of Health Sciences, Bio Anti-aging Medical Research Center, Gyeongsang National University School of Medicine, Jinju 52727, Korea
| | - Hyun Joon Kim
- Department of Anatomy and Convergence Medical Sciences, Institute of Health Sciences, Bio Anti-aging Medical Research Center, Gyeongsang National University School of Medicine, Jinju 52727, Korea
| |
Collapse
|
22
|
Ma YF, Batistel F, Xu TL, Han LQ, Bucktrout R, Liang Y, Coleman DN, Parys C, Loor JJ. Phosphorylation of AKT serine/threonine kinase and abundance of milk protein synthesis gene networks in mammary tissue in response to supply of methionine in periparturient Holstein cows. J Dairy Sci 2019; 102:4264-4274. [PMID: 30879806 DOI: 10.3168/jds.2018-15451] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 01/25/2019] [Indexed: 12/17/2022]
Abstract
The main objective was to evaluate the effect of increasing the supply of Met around parturition on abundance and phosphorylation of insulin- and mechanistic target of rapamycin complex 1 (mTORC1)-related signaling proteins along with mRNA abundance of milk protein and fat synthesis-related genes in postpartal mammary tissue. A basal control diet (control) or the basal diet plus ethyl-cellulose rumen-protected Met (0.9 g/kg of dry matter intake; Mepron, Evonik Nutrition & Care GmbH, Hanau-Wolfgang, Germany) were fed (n = 30 cows/diet) from d -28 to 60 relative to parturition. Mammary tissue and blood plasma were harvested from the same cows (n = 5/diet) in the control and Met groups at d 21 postpartum for mRNA, protein, and AA analysis. Increasing the supply of Met led to greater milk protein percentage and milk yield along with greater ratio of phosphorylated (p-)AKT to total AKT. The ratio of p-mTORC1 to total mTORC1 did not differ, but ratio of p-RPS6 to total ribosomal protein S6 (RPS6) was lower in response to Met supply. These responses were associated with greater mRNA abundance of the signaling proteins Janus kinase 2 (JAK2) and insulin receptor substrate 1 (IRS1). Greater Met supply also upregulated mRNA abundance of high-affinity cationic (SLC7A1) and sodium-coupled AA transporters (SLC38A1, SLC38A2); leucyl-tRNA (LARS), valyl-tRNA (VARS), and isoleucyl-tRNA synthetases (IARS); glucose transport solute carrier family 2 member 3 (SLC2A1); glucose transport solute carrier family 2 member 3 (SLC2A3); and casein α-s1 (CSN1S1). The mRNA abundance of components of the unfolded protein response, such as x-box binding protein 1 (XBP1) and activating transcription factor 6 (ATF6), were upregulated, and protein phosphatase 1, regulatory subunit 15A (PPP1R15A) was downregulated in response to greater Met supply. Overall, the data suggest that increased dry matter intake, greater phosphorylation status of AKT, upregulation of glucose and AA transporters, and transcripts of tRNases in response to enhanced Met supply might have compensated for a reduction in ribosome biogenesis due to a lower ratio of p-RPS6 to total RPS6. Together, these cellular responses constitute a mechanism whereby Met supply can regulate milk protein synthesis in early lactation.
Collapse
Affiliation(s)
- Y F Ma
- Institute of Animal Nutrition and Feed, Inner Mongolia Academy of Agriculture and Animal Husbandry Sciences, Hohhot 010031, P. R. China; Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - F Batistel
- Department of Animal, Dairy & Veterinary Science, Utah State University, Logan 84322
| | - T L Xu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, P. R. China
| | - L Q Han
- Department of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, P. R. China
| | - R Bucktrout
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Y Liang
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - D N Coleman
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - C Parys
- Evonik Nutrition & Care GmbH, Hanau-Wolfgang, 63457, Germany
| | - J J Loor
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801.
| |
Collapse
|
23
|
Shimamoto A, Rappeneau V, Munjal H, Farris T, Davis C, Wilson A, Edwards M, Moore C, Reynolds C, Meshul CK. Glutamate-Glutamine Transfer and Chronic Stress-Induced Sex Differences in Cocaine Responses. Neuroscience 2018; 391:104-119. [DOI: 10.1016/j.neuroscience.2018.09.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 09/05/2018] [Accepted: 09/06/2018] [Indexed: 01/16/2023]
|
24
|
Kandasamy P, Gyimesi G, Kanai Y, Hediger MA. Amino acid transporters revisited: New views in health and disease. Trends Biochem Sci 2018; 43:752-789. [PMID: 30177408 DOI: 10.1016/j.tibs.2018.05.003] [Citation(s) in RCA: 284] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 05/23/2018] [Accepted: 05/25/2018] [Indexed: 02/09/2023]
Abstract
Amino acid transporters (AATs) are membrane-bound transport proteins that mediate transfer of amino acids into and out of cells or cellular organelles. AATs have diverse functional roles ranging from neurotransmission to acid-base balance, intracellular energy metabolism, and anabolic and catabolic reactions. In cancer cells and diabetes, dysregulation of AATs leads to metabolic reprogramming, which changes intracellular amino acid levels, contributing to the pathogenesis of cancer, obesity and diabetes. Indeed, the neutral amino acid transporters (NATs) SLC7A5/LAT1 and SLC1A5/ASCT2 are likely involved in several human malignancies. However, a clinical therapy that directly targets AATs has not yet been developed. The purpose of this review is to highlight the structural and functional diversity of AATs, their diverse physiological roles in different tissues and organs, their wide-ranging implications in human diseases and the emerging strategies and tools that will be necessary to target AATs therapeutically.
Collapse
Affiliation(s)
- Palanivel Kandasamy
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, CH-3012 Bern, Switzerland
| | - Gergely Gyimesi
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, CH-3012 Bern, Switzerland
| | - Yoshikatsu Kanai
- Division of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Osaka, Japan.
| | - Matthias A Hediger
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, CH-3012 Bern, Switzerland.
| |
Collapse
|
25
|
Araya S, Kuster E, Gluch D, Mariotta L, Lutz C, Reding TV, Graf R, Verrey F, Camargo SMR. Exocrine pancreas glutamate secretion help to sustain enterocyte nutritional needs under protein restriction. Am J Physiol Gastrointest Liver Physiol 2018; 314:G517-G536. [PMID: 29167114 DOI: 10.1152/ajpgi.00135.2017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Glutamine (Gln) is the most concentrated amino acid in blood and considered conditionally essential. Its requirement is increased during physiological stress, such as malnutrition or illness, despite its production by muscle and other organs. In the malnourished state, Gln has been suggested to have a trophic effect on the exocrine pancreas and small intestine. However, the Gln transport capacity, the functional relationship of these two organs, and the potential role of the Gln-glutamate (Glu) cycle are unknown. We observed that pancreatic acinar cells express lower levels of Glu than Gln transporters. Consistent with this expression pattern, the rate of Glu influx into acinar cells was approximately sixfold lower than that of Gln. During protein restriction, acinar cell glutaminase expression was increased and Gln accumulation was maintained. Moreover, Glu secretion by acinar cells into pancreatic juice and thus into the lumen of the small intestine was maintained. In the intestinal lumen, Glu absorption was preserved and Glu dehydrogenase expression was augmented, potentially providing the substrates for increasing energy production via the TCA cycle. Our findings suggest that one mechanism by which Gln exerts a positive effect on exocrine pancreas and small intestine involves the Gln metabolism in acinar cells and the secretion of Glu into the small intestine lumen. The exocrine pancreas acinar cells not only avidly accumulate Gln but metabolize Gln to generate energy and to synthesize Glu for secretion in the pancreatic juice. Secreted Glu is suggested to play an important role during malnourishment in sustaining small intestinal homeostasis. NEW & NOTEWORTHY Glutamine (Gln) has been suggested to have a trophic effect on exocrine pancreas and small intestine in malnourished states, but the mechanism is unknown. In this study, we suggest that this trophic effect derives from an interorgan relationship between exocrine pancreas and small intestine for Gln-glutamate (Glu) utilization involving the uptake and metabolism of Gln in acinar cells and secretion of Glu into the lumen of the small intestine.
Collapse
Affiliation(s)
- S Araya
- Institute of Physiology and Zurich Center for Integrative Human Physiology, University of Zurich , Zurich , Switzerland
| | - E Kuster
- Institute of Physiology and Zurich Center for Integrative Human Physiology, University of Zurich , Zurich , Switzerland
| | - D Gluch
- Institute of Physiology and Zurich Center for Integrative Human Physiology, University of Zurich , Zurich , Switzerland
| | - L Mariotta
- Institute of Physiology and Zurich Center for Integrative Human Physiology, University of Zurich , Zurich , Switzerland
| | - C Lutz
- Institute of Physiology and Zurich Center for Integrative Human Physiology, University of Zurich , Zurich , Switzerland
| | - T V Reding
- Department of Surgery, University Hospital Zurich , Zurich , Switzerland
| | - R Graf
- Department of Surgery, University Hospital Zurich , Zurich , Switzerland
| | - F Verrey
- Institute of Physiology and Zurich Center for Integrative Human Physiology, University of Zurich , Zurich , Switzerland
| | - S M R Camargo
- Institute of Physiology and Zurich Center for Integrative Human Physiology, University of Zurich , Zurich , Switzerland
| |
Collapse
|
26
|
Steyfkens F, Zhang Z, Van Zeebroeck G, Thevelein JM. Multiple Transceptors for Macro- and Micro-Nutrients Control Diverse Cellular Properties Through the PKA Pathway in Yeast: A Paradigm for the Rapidly Expanding World of Eukaryotic Nutrient Transceptors Up to Those in Human Cells. Front Pharmacol 2018; 9:191. [PMID: 29662449 PMCID: PMC5890159 DOI: 10.3389/fphar.2018.00191] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 02/20/2018] [Indexed: 12/17/2022] Open
Abstract
The nutrient composition of the medium has dramatic effects on many cellular properties in the yeast Saccharomyces cerevisiae. In addition to the well-known specific responses to starvation for an essential nutrient, like nitrogen or phosphate, the presence of fermentable sugar or a respirative carbon source leads to predominance of fermentation or respiration, respectively. Fermenting and respiring cells also show strong differences in other properties, like storage carbohydrate levels, general stress tolerance and cellular growth rate. However, the main glucose repression pathway, which controls the switch between respiration and fermentation, is not involved in control of these properties. They are controlled by the protein kinase A (PKA) pathway. Addition of glucose to respiring yeast cells triggers cAMP synthesis, activation of PKA and rapid modification of its targets, like storage carbohydrate levels, general stress tolerance and growth rate. However, starvation of fermenting cells in a glucose medium for any essential macro- or micro-nutrient counteracts this effect, leading to downregulation of PKA and its targets concomitant with growth arrest and entrance into G0. Re-addition of the lacking nutrient triggers rapid activation of the PKA pathway, without involvement of cAMP as second messenger. Investigation of the sensing mechanism has revealed that the specific high-affinity nutrient transporter(s) induced during starvation function as transporter-receptors or transceptors for rapid activation of PKA upon re-addition of the missing substrate. In this way, transceptors have been identified for amino acids, ammonium, phosphate, sulfate, iron, and zinc. We propose a hypothesis for regulation of PKA activity by nutrient transceptors to serve as a conceptual framework for future experimentation. Many properties of transceptors appear to be similar to those of classical receptors and nutrient transceptors may constitute intermediate forms in the development of receptors from nutrient transporters during evolution. The nutrient-sensing transceptor system in yeast for activation of the PKA pathway has served as a paradigm for similar studies on candidate nutrient transceptors in other eukaryotes and we succinctly discuss the many examples of transceptors that have already been documented in other yeast species, filamentous fungi, plants, and animals, including the examples in human cells.
Collapse
Affiliation(s)
- Fenella Steyfkens
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, Leuven, Belgium.,Center for Microbiology, VIB, Flanders, Belgium
| | - Zhiqiang Zhang
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, Leuven, Belgium.,Center for Microbiology, VIB, Flanders, Belgium
| | - Griet Van Zeebroeck
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, Leuven, Belgium.,Center for Microbiology, VIB, Flanders, Belgium
| | - Johan M Thevelein
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, Leuven, Belgium.,Center for Microbiology, VIB, Flanders, Belgium
| |
Collapse
|
27
|
Hoffmann TM, Cwiklinski E, Shah DS, Stretton C, Hyde R, Taylor PM, Hundal HS. Effects of Sodium and Amino Acid Substrate Availability upon the Expression and Stability of the SNAT2 (SLC38A2) Amino Acid Transporter. Front Pharmacol 2018; 9:63. [PMID: 29467657 PMCID: PMC5808304 DOI: 10.3389/fphar.2018.00063] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 01/17/2018] [Indexed: 12/14/2022] Open
Abstract
The SNAT2 (SLC38A2) System A amino acid transporter mediates Na+-coupled cellular uptake of small neutral α-amino acids (AAs) and is extensively regulated in response to humoral and nutritional cues. Understanding the basis of such regulation is important given that AA uptake via SNAT2 has been linked to activation of mTORC1; a major controller of many important cellular processes including, for example, mRNA translation, lipid synthesis, and autophagy and whose dysregulation has been implicated in the development of cancer and conditions such as obesity and type 2 diabetes. Extracellular AA withdrawal induces an adaptive upregulation of SNAT2 gene transcription and SNAT2 protein stability but, as yet, the sensing mechanism(s) that initiate this response remain poorly understood although interactions between SNAT2 and its substrates may play a vital role. Herein, we have explored how changes in substrate (AA and Na+) availability impact upon the adaptive regulation of SNAT2 in HeLa cells. We show that while AA deprivation induces SNAT2 gene expression, this induction was not apparent if extracellular Na+ was removed during the AA withdrawal period. Furthermore, we show that the increase in SNAT2 protein stability associated with AA withdrawal is selectively repressed by provision of SNAT2 AA substrates (N-methylaminoisobutyric acid and glutamine), but not non-substrates. This stabilization and substrate-induced repression were critically dependent upon the cytoplasmic N-terminal tail of SNAT2 (containing lysyl residues which are putative targets of the ubiquitin-proteasome system), because “grafting” this tail onto SNAT5, a related SLC38 family member that does not exhibit adaptive regulation, confers substrate-induced changes in stability of the SNAT2-5 chimeric transporter. In contrast, expression of SNAT2 in which the N-terminal lysyl residues were mutated to alanine rendered the transporter stable and insensitive to substrate-induced changes in protein stability. Intriguingly, SNAT2 protein stability was dramatically reduced in the absence of extracellular Na+ irrespective of whether substrate AAs were present or absent. Our findings indicate that the presence of extracellular Na+ (and potentially its binding to SNAT2) may be crucial for not only sensing SNAT2 AA occupancy and consequently for initiating the adaptive response under AA insufficient conditions, but for enabling substrate-induced changes in SNAT2 protein stability.
Collapse
Affiliation(s)
- Thorsten M Hoffmann
- Division of Cell Signalling and Immunology, Sir James Black Centre, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Emma Cwiklinski
- Division of Cell Signalling and Immunology, Sir James Black Centre, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Dinesh S Shah
- Division of Cell Signalling and Immunology, Sir James Black Centre, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Clare Stretton
- Division of Cell Signalling and Immunology, Sir James Black Centre, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Russell Hyde
- Division of Cell Signalling and Immunology, Sir James Black Centre, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Peter M Taylor
- Division of Cell Signalling and Immunology, Sir James Black Centre, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Harinder S Hundal
- Division of Cell Signalling and Immunology, Sir James Black Centre, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
28
|
Lipina C, Hundal HS. Lipid modulation of skeletal muscle mass and function. J Cachexia Sarcopenia Muscle 2017; 8:190-201. [PMID: 27897400 PMCID: PMC5377414 DOI: 10.1002/jcsm.12144] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 07/15/2016] [Accepted: 07/25/2016] [Indexed: 12/22/2022] Open
Abstract
Loss of skeletal muscle mass is a characteristic feature of various pathologies including cancer, diabetes, and obesity, as well as being a general feature of ageing. However, the processes underlying its pathogenesis are not fully understood and may involve multiple factors. Importantly, there is growing evidence which supports a role for fatty acids and their derived lipid intermediates in the regulation of skeletal muscle mass and function. In this review, we discuss evidence pertaining to those pathways which are involved in the reduction, increase and/or preservation of skeletal muscle mass by such lipids under various pathological conditions, and highlight studies investigating how these processes may be influenced by dietary supplementation as well as genetic and/or pharmacological intervention.
Collapse
Affiliation(s)
- Christopher Lipina
- Division of Cell Signalling and Immunology, Sir James Black Centre, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Harinder S Hundal
- Division of Cell Signalling and Immunology, Sir James Black Centre, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| |
Collapse
|
29
|
Abbas A, Beamish C, McGirr R, Demarco J, Cockburn N, Krokowski D, Lee TY, Kovacs M, Hatzoglou M, Dhanvantari S. Characterization of 5-(2- 18F-fluoroethoxy)-L-tryptophan for PET imaging of the pancreas. F1000Res 2016; 5:1851. [PMID: 27909574 PMCID: PMC5112576 DOI: 10.12688/f1000research.9129.2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/04/2016] [Indexed: 12/23/2022] Open
Abstract
Purpose: In diabetes, pancreatic beta cell mass declines significantly prior to onset of fasting hyperglycemia. This decline may be due to endoplasmic reticulum (ER) stress, and the system L amino acid transporter LAT1 may be a biomarker of this process. In this study, we used 5-(2-
18F-fluoroethoxy)-L-tryptophan (
18F-L-FEHTP) to target LAT1 as a potential biomarker of beta cell function in diabetes. Procedures: Uptake of
18F-L-FEHTP was determined in wild-type C57BL/6 mice by
ex vivo biodistribution. Both dynamic and static positron emission tomography (PET) images were acquired in wild-type and Akita mice, a model of ER stress-induced diabetes, as well as in mice treated with streptozotocin (STZ). LAT1 expression in both groups of mice was evaluated by immunofluorescence microscopy. Results: Uptake of
18F-L-FEHTP was highest in the pancreas, and static PET images showed highly specific pancreatic signal. Time-activity curves showed significantly reduced
18F-L-FEHTP uptake in Akita mice, and LAT1 expression was also reduced. However, mice treated with STZ, in which beta cell mass was reduced by 62%, showed no differences in
18F-L-FEHTP uptake in the pancreas, and there was no significant correlation of
18F-L-FEHTP uptake with beta cell mass. Conclusions: 18F-L-FEHTP is highly specific for the pancreas with little background uptake in kidney or liver. We were able to detect changes in LAT1 in a mouse model of diabetes, but these changes did not correlate with beta cell function or mass. Therefore,
18F-L-FEHTP PET is not a suitable method for the noninvasive imaging of changes in beta cell function during the progression of diabetes.
Collapse
Affiliation(s)
- Ahmed Abbas
- Department of Medical Biophysics, Western University, London, ON, N6A 5C1, Canada
| | - Christine Beamish
- Metabolism and Diabetes Program, Lawson Health Research Institute, London, ON, N6A 4V2, Canada
| | - Rebecca McGirr
- Metabolism and Diabetes Program, Lawson Health Research Institute, London, ON, N6A 4V2, Canada; Imaging Program, Lawson Health Research Institute, London, ON, N6A 4V2, Canada
| | - John Demarco
- Imaging Program, Lawson Health Research Institute, London, ON, N6A 4V2, Canada
| | - Neil Cockburn
- Imaging Program, Lawson Health Research Institute, London, ON, N6A 4V2, Canada
| | - Dawid Krokowski
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Ting-Yim Lee
- Department of Medical Biophysics, Western University, London, ON, N6A 5C1, Canada; Imaging Program, Lawson Health Research Institute, London, ON, N6A 4V2, Canada
| | - Michael Kovacs
- Department of Medical Biophysics, Western University, London, ON, N6A 5C1, Canada; Imaging Program, Lawson Health Research Institute, London, ON, N6A 4V2, Canada
| | - Maria Hatzoglou
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Savita Dhanvantari
- Department of Medical Biophysics, Western University, London, ON, N6A 5C1, Canada; Metabolism and Diabetes Program, Lawson Health Research Institute, London, ON, N6A 4V2, Canada; Imaging Program, Lawson Health Research Institute, London, ON, N6A 4V2, Canada; Department of Pathology and Laboratory Medicine, Western University, London, ON, N6A 5C1, Canada
| |
Collapse
|
30
|
Abbas A, Beamish C, McGirr R, Demarco J, Cockburn N, Krokowski D, Lee TY, Kovacs M, Hatzoglou M, Dhanvantari S. Characterization of 5-(2- 18F-fluoroethoxy)-L-tryptophan for PET imaging of the pancreas. F1000Res 2016; 5:1851. [PMID: 27909574 PMCID: PMC5112576 DOI: 10.12688/f1000research.9129.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/04/2016] [Indexed: 10/29/2023] Open
Abstract
Purpose: In diabetes, pancreatic beta cell mass declines significantly prior to onset of fasting hyperglycemia. This decline may be due to endoplasmic reticulum (ER) stress, and the system L amino acid transporter LAT1 may be a biomarker of this process. In this study, we used 5-(2- 18F-fluoroethoxy)-L-tryptophan ( 18F-L-FEHTP) to target LAT1 as a potential biomarker of beta cell function in diabetes. Procedures: Uptake of 18F-L-FEHTP was determined in wild-type C57BL/6 mice by ex vivo biodistribution. Both dynamic and static positron emission tomography (PET) images were acquired in wild-type and Akita mice, a model of ER stress-induced diabetes, as well as in mice treated with streptozotocin (STZ). LAT1 expression in both groups of mice was evaluated by immunofluorescence microscopy. Results: Uptake of 18F-L-FEHTP was highest in the pancreas, and static PET images showed highly specific pancreatic signal. Time-activity curves showed significantly reduced 18F-L-FEHTP uptake in Akita mice, and LAT1 expression was also reduced. However, mice treated with STZ, in which beta cell mass was reduced by 62%, showed no differences in 18F-L-FEHTP uptake in the pancreas, and there was no significant correlation of 18F-L-FEHTP uptake with beta cell mass. Conclusions:18F-L-FEHTP is highly specific for the pancreas with little background uptake in kidney or liver. We were able to detect changes in LAT1 in a mouse model of diabetes, but these changes did not correlate with beta cell function or mass. Therefore, 18F-L-FEHTP PET is not a suitable method for the noninvasive imaging of changes in beta cell function during the progression of diabetes.
Collapse
Affiliation(s)
- Ahmed Abbas
- Department of Medical Biophysics, Western University, London, ON, N6A 5C1, Canada
| | - Christine Beamish
- Metabolism and Diabetes Program, Lawson Health Research Institute, London, ON, N6A 4V2, Canada
| | - Rebecca McGirr
- Metabolism and Diabetes Program, Lawson Health Research Institute, London, ON, N6A 4V2, Canada
- Imaging Program, Lawson Health Research Institute, London, ON, N6A 4V2, Canada
| | - John Demarco
- Imaging Program, Lawson Health Research Institute, London, ON, N6A 4V2, Canada
| | - Neil Cockburn
- Imaging Program, Lawson Health Research Institute, London, ON, N6A 4V2, Canada
| | - Dawid Krokowski
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Ting-Yim Lee
- Department of Medical Biophysics, Western University, London, ON, N6A 5C1, Canada
- Imaging Program, Lawson Health Research Institute, London, ON, N6A 4V2, Canada
| | - Michael Kovacs
- Department of Medical Biophysics, Western University, London, ON, N6A 5C1, Canada
- Imaging Program, Lawson Health Research Institute, London, ON, N6A 4V2, Canada
| | - Maria Hatzoglou
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Savita Dhanvantari
- Department of Medical Biophysics, Western University, London, ON, N6A 5C1, Canada
- Metabolism and Diabetes Program, Lawson Health Research Institute, London, ON, N6A 4V2, Canada
- Imaging Program, Lawson Health Research Institute, London, ON, N6A 4V2, Canada
- Department of Pathology and Laboratory Medicine, Western University, London, ON, N6A 5C1, Canada
| |
Collapse
|
31
|
Fatty acid biosynthesis is involved in the production of hepatitis B virus particles. Biochem Biophys Res Commun 2016; 475:87-92. [PMID: 27178211 DOI: 10.1016/j.bbrc.2016.05.043] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 05/03/2016] [Indexed: 01/12/2023]
Abstract
Hepatitis B virus (HBV) proliferates in hepatocytes after infection, but the host factors that contribute to the HBV lifecycle are poorly understood at the molecular level. We investigated whether fatty acid biosynthesis (FABS), which was recently reported to contribute to the genomic replication of hepatitis C virus, plays a role in HBV proliferation. We examined the effects of inhibitors of the enzymes in the FABS pathway on the HBV lifecycle by using recombinant HBV-producing cultured cells and found that the extracellular HBV DNA level, reflecting HBV particle production, was decreased by treatment with inhibitors suppressed the synthesis of long-chain saturated fatty acids with little cytotoxicity. The reduced HBV DNA level was reversed when palmitic acid, which is the product of fatty acid synthase (FAS) during FABS, was used simultaneously with the inhibitor. We also observed that the amount of intracellular HBV DNA in the cells was increased by FAS inhibitor treatment, suggesting that FABS is associated with HBV particle production but not its genome replication. This suggests that FABS might be a potent target for anti-HBV drug with a mode of action different from current HBV therapy.
Collapse
|
32
|
Regulation of amino acid transporter trafficking by mTORC1 in primary human trophoblast cells is mediated by the ubiquitin ligase Nedd4-2. Clin Sci (Lond) 2015; 130:499-512. [PMID: 26608079 DOI: 10.1042/cs20150554] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 11/25/2015] [Indexed: 12/11/2022]
Abstract
Changes in placental amino acid transfer directly contribute to altered fetal growth, which increases the risk for perinatal complications and predisposes for the development of obesity, diabetes and cardiovascular disease later in life. Placental amino acid transfer is critically dependent on the expression of specific transporters in the plasma membrane of the trophoblast, the transporting epithelium of the human placenta. However, the molecular mechanisms regulating this process are largely unknown. Nedd4-2 is an ubiquitin ligase that catalyses the ubiquitination of proteins, resulting in proteasomal degradation. We hypothesized that inhibition of mechanistic target of rapamycin complex 1 (mTORC1) decreases amino acid uptake in primary human trophoblast (PHT) cells by activation of Nedd4-2, which increases transporter ubiquitination resulting in decreased transporter expression in the plasma membrane. mTORC 1 inhibition increased the expression of Nedd4-2, promoted ubiquitination and decreased the plasma membrane expression of SNAT2 (an isoform of the System A amino acid transporter) and LAT1 (a System L amino acid transporter isoform), resulting in decreased cellular amino acid uptake. Nedd4-2 silencing markedly increased the trafficking of SNAT2 and LAT1 to the plasma membrane, which stimulated cellular amino acid uptake. mTORC1 inhibition by silencing of raptor failed to decrease amino acid transport following Nedd4-2 silencing. In conclusion, we have identified a novel link between mTORC1 signalling and ubiquitination, a common posttranslational modification. Because placental mTORC1 is inhibited in fetal growth restriction and activated in fetal overgrowth, we propose that regulation of placental amino acid transporter ubiquitination by mTORC1 and Nedd4-2 constitutes a molecular mechanisms underlying abnormal fetal growth.
Collapse
|
33
|
Aye ILMH, Jansson T, Powell TL. TNF-α stimulates System A amino acid transport in primary human trophoblast cells mediated by p38 MAPK signaling. Physiol Rep 2015; 3:3/10/e12594. [PMID: 26508738 PMCID: PMC4632960 DOI: 10.14814/phy2.12594] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Maternal obesity and gestational diabetes mellitus (GDM) increase the risk of delivering infants that are large for gestational age with greater adiposity, who are prone to the development of metabolic disease in childhood and beyond. These maternal conditions are also associated with increased levels of the proinflammatory cytokine TNF-α in maternal tissues and the placenta. Recent evidence suggests that changes in placental amino acid transport contribute to altered fetal growth. TNF-α was previously shown to stimulate System A amino acid transport in primary human trophoblasts (PHTs), however the molecular mechanisms remain unknown. In this study, we tested the hypothesis that TNF-α regulates amino acid uptake in cultured PHTs by a mitogen-activated protein kinase (MAPK)-dependent mechanism. Treatment of PHTs with TNF-α significantly increased System A amino acid transport, as well as Erk and p38 MAPK signaling. Pharmacological antagonism of p38, but not Erk MAPK activity, inhibited TNF-α stimulated System A activity. Silencing of p38 MAPK using siRNA transfections prevented TNF-α stimulated System A transport in PHTs. TNF-α significantly increased the protein expression of System A transporters SNAT1 and SNAT2, but did not affect their mRNA expression. The effects of TNF-α on SNAT1 and SNAT2 protein expression were reversed by p38 MAPK siRNA silencing. In conclusion, TNF-α regulates System A activity through increased SNAT1 and SNAT2 transporter protein expression in PHTs. These findings suggest that p38 MAPK may represent a critical mechanistic link between elevated proinflammatory cytokines and increased placental amino acid transport in obese and GDM pregnancies associated with fetal overgrowth.
Collapse
Affiliation(s)
- Irving L M H Aye
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Thomas Jansson
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Theresa L Powell
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado Section of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
34
|
Krokowski D, Jobava R, Guan BJ, Farabaugh K, Wu J, Majumder M, Bianchi MG, Snider MD, Bussolati O, Hatzoglou M. Coordinated Regulation of the Neutral Amino Acid Transporter SNAT2 and the Protein Phosphatase Subunit GADD34 Promotes Adaptation to Increased Extracellular Osmolarity. J Biol Chem 2015; 290:17822-17837. [PMID: 26041779 DOI: 10.1074/jbc.m114.636217] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Indexed: 02/04/2023] Open
Abstract
Cells respond to shrinkage induced by increased extracellular osmolarity via programmed changes in gene transcription and mRNA translation. The immediate response to this stress includes the induction of expression of the neutral amino acid transporter SNAT2. Increased SNAT2-mediated uptake of neutral amino acids is an essential adaptive mechanism for restoring cell volume. In contrast, stress-induced phosphorylation of the α subunit of the translation initiation factor eIF2 (eIF2α) can promote apoptosis. Here we show that the response to mild hyperosmotic stress involves regulation of the phosphorylation of eIF2α by increased levels of GADD34, a regulatory subunit of protein phosphatase 1 (PP1). The induction of GADD34 was dependent on transcriptional control by the c-Jun-binding cAMP response element in the GADD34 gene promoter and posttranscriptional stabilization of its mRNA. This mechanism differs from the regulation of GADD34 expression by other stresses that involve activating transcription factor 4 (ATF4). ATF4 was not translated during hyperosmotic stress despite an increase in eIF2α phosphorylation. The SNAT2-mediated increase in amino acid uptake was enhanced by increased GADD34 levels in a manner involving decreased eIF2α phosphorylation. It is proposed that the induction of the SNAT2/GADD34 axis enhances cell survival by promoting the immediate adaptive response to stress.
Collapse
Affiliation(s)
- Dawid Krokowski
- Departments of Pharmacology, Case Western Reserve University, School of Medicine, Cleveland, Ohio 44106.
| | - Raul Jobava
- Departments of Pharmacology, Case Western Reserve University, School of Medicine, Cleveland, Ohio 44106
| | - Bo-Jhih Guan
- Departments of Pharmacology, Case Western Reserve University, School of Medicine, Cleveland, Ohio 44106
| | - Kenneth Farabaugh
- Departments of Pharmacology, Case Western Reserve University, School of Medicine, Cleveland, Ohio 44106
| | - Jing Wu
- Departments of Pharmacology, Case Western Reserve University, School of Medicine, Cleveland, Ohio 44106
| | - Mithu Majumder
- Departments of Pharmacology, Case Western Reserve University, School of Medicine, Cleveland, Ohio 44106
| | - Massimiliano G Bianchi
- Department of Biomedical, Biotechnological, and Translational Sciences, University of Parma, 43100 Parma, Italy
| | - Martin D Snider
- Departments of Biochemistry, Case Western Reserve University, School of Medicine, Cleveland, Ohio 44106
| | - Ovidio Bussolati
- Department of Biomedical, Biotechnological, and Translational Sciences, University of Parma, 43100 Parma, Italy
| | - Maria Hatzoglou
- Departments of Pharmacology, Case Western Reserve University, School of Medicine, Cleveland, Ohio 44106.
| |
Collapse
|