1
|
Aguayo-Cerón KA, Sánchez-Muñoz F, Gutierrez-Rojas RA, Acevedo-Villavicencio LN, Flores-Zarate AV, Huang F, Giacoman-Martinez A, Villafaña S, Romero-Nava R. Glycine: The Smallest Anti-Inflammatory Micronutrient. Int J Mol Sci 2023; 24:11236. [PMID: 37510995 PMCID: PMC10379184 DOI: 10.3390/ijms241411236] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Glycine is a non-essential amino acid with many functions and effects. Glycine can bind to specific receptors and transporters that are expressed in many types of cells throughout an organism to exert its effects. There have been many studies focused on the anti-inflammatory effects of glycine, including its abilities to decrease pro-inflammatory cytokines and the concentration of free fatty acids, to improve the insulin response, and to mediate other changes. However, the mechanism through which glycine acts is not clear. In this review, we emphasize that glycine exerts its anti-inflammatory effects throughout the modulation of the expression of nuclear factor kappa B (NF-κB) in many cells. Although glycine is a non-essential amino acid, we highlight how dietary glycine supplementation is important in avoiding the development of chronic inflammation.
Collapse
Affiliation(s)
- Karla Aidee Aguayo-Cerón
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Sección de Estudios de Posgrado e Investigación, Ciudad de Mexico 11340, Mexico
| | - Fausto Sánchez-Muñoz
- Departamento de Inmunología, Instituto Nacional de Cardiología "Ignacio Chávez", Ciudad de Mexico 14080, Mexico
| | | | | | - Aurora Vanessa Flores-Zarate
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Sección de Estudios de Posgrado e Investigación, Ciudad de Mexico 11340, Mexico
| | - Fengyang Huang
- Laboratorio de Investigación en Obesidad y Asma, Hospital Infantil de México Federico Gómez, Ciudad de Mexico 06720, Mexico
| | - Abraham Giacoman-Martinez
- Laboratorio de Framacología, Departamaneto de Ciencias de la Salud, DCBS, Universidad Autónoma Mteropolitana-Iztapalapa (UAM-I), Ciudad de Mexico 09340, Mexico
| | - Santiago Villafaña
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Sección de Estudios de Posgrado e Investigación, Ciudad de Mexico 11340, Mexico
| | - Rodrigo Romero-Nava
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Sección de Estudios de Posgrado e Investigación, Ciudad de Mexico 11340, Mexico
| |
Collapse
|
2
|
Zhang X, Deng Y, Hu S, Hu X, Ma J, Hu J, Hu B, He H, Li L, Liu H, Wang J. Comparative analysis of amino acid content and protein synthesis-related genes expression levels in breast muscle among different duck breeds/strains. Poult Sci 2022; 102:102277. [PMID: 36410066 PMCID: PMC9678761 DOI: 10.1016/j.psj.2022.102277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 10/14/2022] [Accepted: 10/18/2022] [Indexed: 11/15/2022] Open
Abstract
Evidences have found important effects of breeds/strains on the content of amino acids (AAs) which is an important substrate for protein synthesis and contributes greatly to meat quality. Therefore, the objective of the present study was to compare the AAs content and protein synthesis-related genes expression levels in breast muscle of native breed (Jianchang duck (J)), hybrid strains (BH1, BH2, and MC♂ × (BGF2♂ × GF2♀)♀ (MC)), and commercial breed (Cherry Verry duck). Results showed that a total of 17 AAs (TAAs) was detected from breast muscle among 5 duck breeds/strains including 11 essential AAs (EAAs). Among these AAs, the contents of Proline, Threonine, Glutamine, Serine, Methionine, Phenylalanine, Histidine, and Cysteine were significant difference among 5 duck breeds/strains. The contents of EAAs, TAAs, and flavor AAs were higher in breast muscle of J and BH2 than those in other duck breeds/strains, and the ratio of EAAs/TAAs was higher in breast muscle of BH2. Furthermore, the expression levels of eukaryotic translation initiation factor 4E-binding protein 1, mammalian target of rapamycin, and proton-coupled amino acid transporter 1 were the highest in breast muscle of BH2, and that of solute carrier family 38 member 2 was the highest in breast muscle of J. Meanwhile, principal component analysis results showed that principal component 1 of BH1, principal component 3 of BH2, and principal component 2 of MC were positively corelated with EAAs/TAAs, and principal component 1 was positively correlated with flavor AAs and EAAs. In conclusion, compared to BH1, MC, and Cherry Verry duck, AA content was higher in breast muscle of BH2 and J, which might be associated with the higher expression levels of mammalian target of rapamycin, eukaryotic translation initiation factor 4E-binding protein 1, and proton-coupled amino acid transporter 1 in breast muscle of BH2 and solute carrier family 38 member 2 in breast muscle of J. The comparative analysis of AA content in breast muscle among different duck breeds/strains could provide an important basis for improving the nutritional value of duck meat in the breeding process.
Collapse
Affiliation(s)
- Xin Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130, Chengdu, Sichuan, China
| | - Yan Deng
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130, Chengdu, Sichuan, China
| | - Shenqiang Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130, Chengdu, Sichuan, China
| | - Xinyue Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130, Chengdu, Sichuan, China
| | - Jiaming Ma
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130, Chengdu, Sichuan, China
| | - Jiwei Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130, Chengdu, Sichuan, China
| | - Bo Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130, Chengdu, Sichuan, China
| | - Hua He
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130, Chengdu, Sichuan, China
| | - Liang Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130, Chengdu, Sichuan, China
| | - Hehe Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130, Chengdu, Sichuan, China
| | - Jiwen Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, 611130, Chengdu, Sichuan, China.
| |
Collapse
|
3
|
Barguil Nepomuceno D, DÁvila Pessoa GC, Nascimento Araújo R, Barbosa Koerich L, Viana Sant’Anna MR, Horácio Pereira M, Figueiredo Gontijo N. Na+/K+-ATPase Activation by cAMP in the Midgut of Lutzomyia longipalpis (Lutz & Neiva, 1912; Diptera: Psychodidae). JOURNAL OF INSECT SCIENCE (ONLINE) 2022; 22:1. [PMID: 35271719 PMCID: PMC8912928 DOI: 10.1093/jisesa/ieac008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Indexed: 06/14/2023]
Abstract
Lutzomyia longipalpis (Lutz & Neiva, 1912) females have been intensively studied regarding the regulation of midgut pH. The mechanisms involved in pH regulation are complex, and some aspects remain to be clarified. Here, we investigated the role of the Na+/K+-ATPase pump as an electrochemical potential generator and its modulation by the second messenger cAMP in the midgut of female L. longipalpis. Our results suggest that not only may Na+/K+-ATPase be the main generator of an electrochemical potential across membranes in the midgut of female L. longipalpis, but also its activity is positively regulated by cAMP. cAMP-mediated Na+/K+-ATPase pump activity might be necessary to maintain the transport of the nutrients produced during blood digestion.
Collapse
Affiliation(s)
- Denise Barguil Nepomuceno
- Laboratório de Fisiologia de Insetos Hematófagos, Departamento de Parasitologia—ICB, Universidade Federal de Minas Gerais—UFMG, Belo Horizonte, MG, Brazil
| | - Grasielle Caldas DÁvila Pessoa
- Laboratório de Fisiologia de Insetos Hematófagos, Departamento de Parasitologia—ICB, Universidade Federal de Minas Gerais—UFMG, Belo Horizonte, MG, Brazil
| | - Ricardo Nascimento Araújo
- Laboratório de Fisiologia de Insetos Hematófagos, Departamento de Parasitologia—ICB, Universidade Federal de Minas Gerais—UFMG, Belo Horizonte, MG, Brazil
| | - Leonardo Barbosa Koerich
- Laboratório de Fisiologia de Insetos Hematófagos, Departamento de Parasitologia—ICB, Universidade Federal de Minas Gerais—UFMG, Belo Horizonte, MG, Brazil
| | - Maurício Roberto Viana Sant’Anna
- Laboratório de Fisiologia de Insetos Hematófagos, Departamento de Parasitologia—ICB, Universidade Federal de Minas Gerais—UFMG, Belo Horizonte, MG, Brazil
| | - Marcos Horácio Pereira
- Laboratório de Fisiologia de Insetos Hematófagos, Departamento de Parasitologia—ICB, Universidade Federal de Minas Gerais—UFMG, Belo Horizonte, MG, Brazil
| | - Nelder Figueiredo Gontijo
- Laboratório de Fisiologia de Insetos Hematófagos, Departamento de Parasitologia—ICB, Universidade Federal de Minas Gerais—UFMG, Belo Horizonte, MG, Brazil
| |
Collapse
|
4
|
Nielsen MMK, Aryal E, Safari E, Mojsoska B, Jenssen H, Prabhala BK. Current State of SLC and ABC Transporters in the Skin and Their Relation to Sweat Metabolites and Skin Diseases. Proteomes 2021; 9:proteomes9020023. [PMID: 34065737 PMCID: PMC8163169 DOI: 10.3390/proteomes9020023] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 01/02/2023] Open
Abstract
With a relatively large surface area (2 m2) and 15% of total body mass, the skin forms the largest organ of the human body. The main functions of the skin include regulation of body temperature by insulation or sweating, regulation of the nervous system, regulation of water content, and protection against external injury. To perform these critical functions, the skin encodes genes for transporters responsible for the cellular trafficking of essential nutrients and metabolites to maintain cellular hemostasis. However, the knowledge on the expression, regulation, and function of these transporters is very limited and needs more work to elucidate how these transporters play a role both in disease progression and in healing. Furthermore, SLC and ABC transporters are understudied, and even less studied in skin. There are sparse reports on relation between transporters in skin and sweat metabolites. This mini review focuses on the current state of SLC and ABC transporters in the skin and their relation to sweat metabolites and skin diseases.
Collapse
Affiliation(s)
- Marcus M. K. Nielsen
- Institute of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark; (M.M.K.N.); (E.A.)
| | - Eva Aryal
- Institute of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark; (M.M.K.N.); (E.A.)
| | - Elnaz Safari
- Department of Organic Chemistry, Faculty of Chemistry, Urmia University, Urmia 5756151818, Iran;
| | - Biljana Mojsoska
- Institute of Science and Environment, Roskilde University, Universitetsvej 1, 4000 Roskilde, Denmark; (B.M.); (H.J.)
| | - Håvard Jenssen
- Institute of Science and Environment, Roskilde University, Universitetsvej 1, 4000 Roskilde, Denmark; (B.M.); (H.J.)
| | - Bala Krishna Prabhala
- Institute of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark; (M.M.K.N.); (E.A.)
- Correspondence:
| |
Collapse
|
5
|
Wei Y, Li B, Xu H, Liang M. Effects of lysine and leucine in free and different dipeptide forms on the growth, amino acid profile and transcription of intestinal peptide, and amino acid transporters in turbot (Scophthalmus maximus). FISH PHYSIOLOGY AND BIOCHEMISTRY 2020; 46:1795-1807. [PMID: 32514852 DOI: 10.1007/s10695-020-00828-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 05/26/2020] [Indexed: 06/11/2023]
Abstract
This study was conducted to evaluate the effects of different dipeptides (lysine-leucine, lysine-glycine, and leucine-glycine) and free amino acids (lysine and leucine) on the growth, gene expression of intestinal peptide and amino acid transporters, and serum free amino acid concentrations in turbot. Fish (11.98 ± 0.03 g) were fed four experimental diets supplementing with crystalline amino acids (CAA), lysine-leucine (Lys-Leu), lysine-glycine (Lys-Gly), and leucine-glycine (Gly-Leu). Fish protein hydrolysate (FPH) containing a mixture of free amino acids and small peptides was designed as a positive control diet. There was no significant difference in the growth and feed utilization among three dipeptide diets (Lys-Leu, Lys-Gly, and Gly-Leu). Compared with the CAA group, feed efficiency ratio was significantly higher in the Lys-Leu and Lys-Gly groups, and protein efficiency ratio was significantly higher in the Lys-Leu group. For peptide transporter, oligopeptide transporter 1 (PepT1) mRNA level was not affected by dietary treatments. For amino acid transporters, lower expression of B0 neutral amino acid transporter 1 (B0AT1) and proton-coupled amino acid transporter 1 (PAT1) were observed in fish fed the dipeptide and FPH diets compared with the CAA diet. In conclusion, juvenile turbot fed Lys-Leu, Gly-Leu, and Lys-Gly had a similar growth performance, whereas lysine and leucine in the Lys-Leu form can be utilized more efficiently for feed utilization than those in free amino acid from. In addition, compared to free amino acids, dipeptides and fish protein hydrolysate in diets may down-regulate the expression of amino acid transporters but did not affect the expression of PepT1.
Collapse
Affiliation(s)
- Yuliang Wei
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao, 266071, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, 266237, Shandong, China
| | - Benxiang Li
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao, 266071, China
| | - Houguo Xu
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao, 266071, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, 266237, Shandong, China
| | - Mengqing Liang
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao, 266071, China.
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, 266237, Shandong, China.
| |
Collapse
|
6
|
Liu H, Tan B, Kong X, Li J, Li G, He L, Bai M, Yin Y. Dietary Insect Powder Protein Sources Improve Protein Utilization by Regulation on Intestinal Amino Acid-Chemosensing System. Animals (Basel) 2020; 10:ani10091590. [PMID: 32906579 PMCID: PMC7552256 DOI: 10.3390/ani10091590] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/24/2020] [Accepted: 09/01/2020] [Indexed: 12/27/2022] Open
Abstract
Simple Summary Insect powders, including Tenebrio molitor (TM), Musca domestica larvae (MDL) and Zophobas morio (ZM), as high-quality and renewable protein sources are commonly applied in livestock and poultry feed production. The molecular effect of insect protein on amino acid metabolism in pigs needs to be explored. We found that insect powder as a protein source in feed regulated the mTOR signal pathway and improved amino acid transportation in the intestine for growth promotion. Insect powder may be a potentially promising protein source for pig production. Abstract This study was conducted to evaluate the effects of dietary insect powder supplementation as a protein source on plasma amino acid profiles, intestinal amino acid transport and sensing in a piglet model. A total of 144 weanling piglets were randomly assigned to four experimental diets for two phases (Days 1–28 and Days 29–56), to assess the effects on amino acid profiles and transportation in the segments of the intestine. The groups were basal diet (control), control diet plus Tenebrio molitor (TM), control diet plus Musca domestica larvae (MDL) and control diet plus Zophobas morio (ZM). The plasma free amino acid levels were stable comparable among treatments, except that the lysine level was significantly reduced by dietary MDL and ZM supplementation in the first phase (p < 0.05). In the 1st phase, the sensitivity of intestinal segments to the regulation of the amino acid level by insect powder supplementation follows sequence: colon > ileum > jejunum, while the order switched to jejunum > colon > ileum in the 2nd phase. The relative RNA expressions of mitogen-activated protein 4 kinase 3 (MAP4K3), sodium dependent neutral amino acid transporter2 (SNAT2), the transient receptor potential cation channel subfamily V member 1 (TRPV1) and taste 1 receptor member 1/3 (T1R3) in the segments of the intestine were affected by different dietary insect powder supplementation. G protein-coupled receptor family C group 6 member A (GPRC6A) level in the jejunal and colonic mucosa was upregulated by MDL supplementation (p < 0.05). These results indicated that dietary insects improved the metabolism of the amino acid in the prophase (the 1st phase) through regulating the sensing gene and mTOR signal pathway in intestinal mucosa by targeting different receptors. The finding demonstrates that the insect powder is a potentially promising source for protein deposition.
Collapse
Affiliation(s)
| | - Bie Tan
- Correspondence: (B.T.); (X.K.)
| | | | | | | | | | | | | |
Collapse
|
7
|
Li X, Zheng S, Wu G. Amino Acid Metabolism in the Kidneys: Nutritional and Physiological Significance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1265:71-95. [DOI: 10.1007/978-3-030-45328-2_5] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
8
|
Nepomuceno DB, Paim RMM, Araújo RN, Pereira MH, Pessoa GCD, Koerich LB, Sant'Anna MRV, Gontijo NF. The role of LuloPAT amino acid/proton symporters in midgut alkalinization in the sandfly Lutzomyia longipalpis (Diptera - Psychodidae). JOURNAL OF INSECT PHYSIOLOGY 2020; 120:103973. [PMID: 31715141 DOI: 10.1016/j.jinsphys.2019.103973] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 11/07/2019] [Accepted: 11/08/2019] [Indexed: 06/10/2023]
Abstract
In Lutzomyia longipalpis females, which are the main vectors of Leishmania infantum in the Americas, hematophagy is crucial for ovary development. The control of pH in the midgut during blood digestion is important to the functioning of the digestive enzymes, which release amino acids in the luminal compartment that are then transported through the enterocytes to the hemolymph for delivery to the ovary and other organs. In the present work, we investigated transport systems known as LuloPATs that are present in the midgut of L. longipalpis but not in other organs. These transporters achieve symport of amino acids with H+ ions, and one of them (LuloPAT1) is orthologous to a transporter described in Aedes aegypti. According to our results, the transcription levels of LuloPAT1 increased significantly immediately after a blood meal. Based on the variation of the fluorescence of fluorescein with the pH of the medium, we developed a technique that shows the acidification of the cytoplasm of gut cells when amino acids are cotransported with H+ from the lumen into the enterocytes. In our experiments, the midguts of the sandflies were dissected and opened longitudinally so that added amino acids could enter the enterocytes via the lumen (PAT carriers are apical). LuloPAT1 transporters are part of a complex of mechanisms that act synergistically to promote gut alkalinization as soon as blood intake by the vector occurs. In dissected but not longitudinally opened midguts, added amino acids could only enter through the basolateral region of enterocytes. However, alkalinization of the lumen was observed because the entry of some amino acids into the cytoplasm of enterocytes triggers a luminal alkalinization mechanism independent of LuloPATs. These findings provide new perspectives that will enable the characterization of the set of signaling pathways involved in pH regulation within the L. longipalpis midgut.
Collapse
Affiliation(s)
- Denise Barguil Nepomuceno
- Laboratório de Fisiologia de Insetos Hematófagos, Departamento de Parasitologia/ICB, Universidade Federal de Minas Gerais, Caixa Postal 486, 31270-901 Belo Horizonte, MG, Brazil
| | - Rafaela Magalhães Macedo Paim
- Laboratório de Fisiologia de Insetos Hematófagos, Departamento de Parasitologia/ICB, Universidade Federal de Minas Gerais, Caixa Postal 486, 31270-901 Belo Horizonte, MG, Brazil
| | - Ricardo Nascimento Araújo
- Laboratório de Fisiologia de Insetos Hematófagos, Departamento de Parasitologia/ICB, Universidade Federal de Minas Gerais, Caixa Postal 486, 31270-901 Belo Horizonte, MG, Brazil
| | - Marcos Horácio Pereira
- Laboratório de Fisiologia de Insetos Hematófagos, Departamento de Parasitologia/ICB, Universidade Federal de Minas Gerais, Caixa Postal 486, 31270-901 Belo Horizonte, MG, Brazil.
| | - Grasielle Caldas D'Ávila Pessoa
- Laboratório de Fisiologia de Insetos Hematófagos, Departamento de Parasitologia/ICB, Universidade Federal de Minas Gerais, Caixa Postal 486, 31270-901 Belo Horizonte, MG, Brazil
| | - Leonardo Barbosa Koerich
- Laboratório de Fisiologia de Insetos Hematófagos, Departamento de Parasitologia/ICB, Universidade Federal de Minas Gerais, Caixa Postal 486, 31270-901 Belo Horizonte, MG, Brazil
| | - Mauricio Roberto Viana Sant'Anna
- Laboratório de Fisiologia de Insetos Hematófagos, Departamento de Parasitologia/ICB, Universidade Federal de Minas Gerais, Caixa Postal 486, 31270-901 Belo Horizonte, MG, Brazil
| | - Nelder Figueiredo Gontijo
- Laboratório de Fisiologia de Insetos Hematófagos, Departamento de Parasitologia/ICB, Universidade Federal de Minas Gerais, Caixa Postal 486, 31270-901 Belo Horizonte, MG, Brazil.
| |
Collapse
|
9
|
Glycine Metabolism and Its Alterations in Obesity and Metabolic Diseases. Nutrients 2019; 11:nu11061356. [PMID: 31208147 PMCID: PMC6627940 DOI: 10.3390/nu11061356] [Citation(s) in RCA: 225] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/07/2019] [Accepted: 06/12/2019] [Indexed: 12/11/2022] Open
Abstract
Glycine is the proteinogenic amino-acid of lowest molecular weight, harboring a hydrogen atom as a side-chain. In addition to being a building-block for proteins, glycine is also required for multiple metabolic pathways, such as glutathione synthesis and regulation of one-carbon metabolism. Although generally viewed as a non-essential amino-acid, because it can be endogenously synthesized to a certain extent, glycine has also been suggested as a conditionally essential amino acid. In metabolic disorders associated with obesity, type 2 diabetes (T2DM), and non-alcoholic fatty liver disease (NAFLDs), lower circulating glycine levels have been consistently observed, and clinical studies suggest the existence of beneficial effects induced by glycine supplementation. The present review aims at synthesizing the recent advances in glycine metabolism, pinpointing its main metabolic pathways, identifying the causes leading to glycine deficiency-especially in obesity and associated metabolic disorders-and evaluating the potential benefits of increasing glycine availability to curb the progression of obesity and obesity-related metabolic disturbances. This study focuses on the importance of diet, gut microbiota, and liver metabolism in determining glycine availability in obesity and associated metabolic disorders.
Collapse
|
10
|
Zeng N, Prodhan U, D'Souza RF, Ramzan F, Mitchell SM, Sharma P, Knowles SO, Roy NC, Sjödin A, Wagner KH, Milan AM, Cameron-Smith D, Mitchell CJ. Regulation of Amino Acid Transporters and Sensors in Response to a High protein Diet: A Randomized Controlled Trial in Elderly Men. J Nutr Health Aging 2019; 23:354-363. [PMID: 30932134 DOI: 10.1007/s12603-019-1171-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND The mammalian target of rapamycin complex 1 (mTORC1) is fundamental for many cellular processes, yet it is often dysregulated with aging. Increased amino acid (AA) availability is correlated with the expression of AA transporters (AAT) and mTORC1 activity. Although many AA sensors and mediators have been proposed to relay the AA signal to mTORC1, it has not yet been determined if chronic dietary intervention affects the expression of AAT, sensors and mediators and their relationships with mTORC1 activity. OBJECTIVE AND DESIGN This study investigated whether the consumption of a diet containing either the current recommended daily allowance (RDA) of protein intake (0.8 g/kg/d) or twice the RDA (2RDA) for ten weeks affected the expression of targets associated with AA transport, sensing and mTORC1 regulation in 26 older men (70-81 years). METHOD Muscle biopsies were collected before and after the intervention under fasting conditions. Diets were controlled by providing fully prepared meals and snacks. Western blot and quantitative polymerase chain reaction were used to measure protein and gene expression respectively. RESULTS Consumption of 2RDA reduced the protein expression of L-type amino acid transporter 1 (LAT1). However, plasma leucine concentration and basal mTORC1 activity were unaltered. The downregulation of LAT1 did not affect the expression of AA sensors and mediators, including leucyl tRNA synthetase (LRS), cytosolic arginine sensor for mTORC1 (CASTOR1), Sestrin2 and Rag proteins. Instead, total ribosomal protein S6 (RPS6) was upregulated with 2RDA. CONCLUSION Ten weeks of 2RDA diet did not affect the fasting mTORC1 signaling, but increased total RPS6 might suggest improved muscular translational capacity to maintain muscular mass.
Collapse
Affiliation(s)
- N Zeng
- Dr. Cameron Mitchell Faculty of Education | School of Kinesiology, The University of British Columbia | Vancouver Campus, 2553 Wesbrook Mall | Vancouver British Columbia | V6T 1Z3 Canada, Phone 604 827 2072| Cell 604 790 3815,
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Zhang X, Zhang N, Wan X, Li L, Zou X. Gene expression of amino acid transporter in pigeon (Columbia livia) intestine during post-hatch development and its correlation with amino acid in pigeon milk. Poult Sci 2017; 96:1120-1131. [DOI: 10.3382/ps/pew320] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 08/02/2016] [Indexed: 12/16/2022] Open
|
12
|
Piert M, Shao X, Raffel D, Davenport MS, Montgomery J, Kunju LP, Hockley BG, Siddiqui J, Scott PJH, Chinnaiyan AM, Rajendiran T. Preclinical Evaluation of 11C-Sarcosine as a Substrate of Proton-Coupled Amino Acid Transporters and First Human Application in Prostate Cancer. J Nucl Med 2017; 58:1216-1223. [PMID: 28302759 DOI: 10.2967/jnumed.116.173179] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 02/14/2017] [Indexed: 12/14/2022] Open
Abstract
Sarcosine is a known substrate of proton-coupled amino acid transporters (PATs), which are overexpressed in selected tissues and solid tumors. Sarcosine, an N-methyl derivative of the amino acid glycine and a metabolic product of choline, plays an important role for prostate cancer aggressiveness and progression. Methods:11C-radiolabeled sarcosine was tested as a new PET imaging probe in comparison with 11C-choline in 2 prostate cancer tumor xenograft models (DU-145 and PC-3). We characterized 11C-sarcosine transport in PC-3 and LNCaP tumor cells and performed 11C-sarcosine PET with CT in the first human subject with localized Gleason 4 + 3 prostate cancer. Target metabolite analyses of sarcosine and its natural precursors, glycine and choline, were performed from independent human prostate tissues. Results: In vitro assays indicated blockage of 11C-sarcosine uptake into PC-3 and LNCaP tumor cells by excess unlabeled (cold) sarcosine. 5-hydroxy-l-tryptophan, but not 2-aminobicyclo-(2,2,1)-heptane-2-carboxylic acid, competitively inhibited 11C-sarcosine tumor cell uptake, confirming PAT-mediated transport. In vivo tumor-to-background ratios (TBRs) obtained from 11C-sarcosine PET were significantly elevated compared with 11C-choline in DU-145 (TBR: 1.92 ± 0.11 for 11C-sarcosine vs. 1.41 ± 0.13 for 11C-choline [n = 10; P < 0.002]) and PC-3 tumors (TBR: 1.89 ± 0.2 for 11C-sarcosine vs. 1.34 ± 0.16 for 11C-choline [n = 7; P < 0.002]). 11C-sarcosine produced high-contrast images in 1 case of localized clinically significant prostate cancer. Target metabolite analyses revealed significant stepwise increases of sarcosine, glycine, and choline tissue levels from benign prostate tissue to localized prostate cancer and subsequently metastatic disease. 11C-sarcosine showed a favorable radiation dosimetry with an effective dose estimate of 0.0045 mSv/MBq, resulting in 2.68 mSv for a human subject (600-MBq dose). Conclusion:11C-sarcosine is a novel radiotracer for PATs and shows initial utility for prostate cancer imaging, with potential benefit over commonly used 11C-choline.
Collapse
Affiliation(s)
- Morand Piert
- Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - Xia Shao
- Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - David Raffel
- Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | | | | | | | - Brian G Hockley
- Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - Javed Siddiqui
- Pathology Department, University of Michigan, Ann Arbor, Michigan; and.,Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
| | - Peter J H Scott
- Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - Arul M Chinnaiyan
- Pathology Department, University of Michigan, Ann Arbor, Michigan; and.,Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
| | - Thekkelnaycke Rajendiran
- Pathology Department, University of Michigan, Ann Arbor, Michigan; and.,Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
13
|
Hellsten SV, Eriksson MM, Lekholm E, Arapi V, Perland E, Fredriksson R. The gene expression of the neuronal protein, SLC38A9, changes in mouse brain after in vivo starvation and high-fat diet. PLoS One 2017; 12:e0172917. [PMID: 28235079 PMCID: PMC5325605 DOI: 10.1371/journal.pone.0172917] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 02/03/2017] [Indexed: 11/18/2022] Open
Abstract
SLC38A9 is characterized as a lysosomal component of the amino acid sensing Ragulator-RAG GTPase complex, controlling the mechanistic target of rapamycin complex 1 (mTORC1). Here, immunohistochemistry was used to map SLC38A9 in mouse brain and staining was detected throughout the brain, in cortex, hypothalamus, thalamus, hippocampus, brainstem and cerebellum. More specifically, immunostaining was found in areas known to be involved in amino acid sensing and signaling pathways e.g. piriform cortex and hypothalamus. SLC38A9 immunoreactivity co-localized with both GABAergic and glutamatergic neurons, but not with astrocytes. SLC38A9 play a key role in the mTORC1 pathway, and therefore we performed in vivo starvation and high-fat diet studies, to measure gene expression alterations in specific brain tissues and in larger brain regions. Following starvation, Slc38a9 was upregulated in brainstem and cortex, and in anterior parts of the brain (Bregma 3.2 to -2.1mm). After high-fat diet, Slc38a9 was specifically upregulated in hypothalamus, while overall downregulation was noticed throughout the brain (Bregma 3.2 to -8.6mm).
Collapse
Affiliation(s)
- Sofie V. Hellsten
- Department of Pharmaceutical Bioscience, Molecular Neuropharmacology, Uppsala University, Uppsala SE, Sweden
- * E-mail:
| | - Mikaela M. Eriksson
- Department of Pharmaceutical Bioscience, Molecular Neuropharmacology, Uppsala University, Uppsala SE, Sweden
| | - Emilia Lekholm
- Department of Pharmaceutical Bioscience, Molecular Neuropharmacology, Uppsala University, Uppsala SE, Sweden
| | - Vasiliki Arapi
- Department of Pharmaceutical Bioscience, Molecular Neuropharmacology, Uppsala University, Uppsala SE, Sweden
| | - Emelie Perland
- Department of Pharmaceutical Bioscience, Molecular Neuropharmacology, Uppsala University, Uppsala SE, Sweden
| | - Robert Fredriksson
- Department of Pharmaceutical Bioscience, Molecular Neuropharmacology, Uppsala University, Uppsala SE, Sweden
| |
Collapse
|
14
|
Lin WY, Williams CR, Yan C, Parrish JZ. Functions of the SLC36 transporter Pathetic in growth control. Fly (Austin) 2016; 9:99-106. [PMID: 26735916 DOI: 10.1080/19336934.2015.1129089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Neurons exhibit extreme diversity in size, but whether large neurons have specialized mechanisms to support their growth is largely unknown. Recently, we identified the SLC36 transporter Pathetic (Path) as a factor required for extreme dendrite growth in neurons. Path is broadly expressed, but only neurons with large dendrite arbors or small neurons that are forced to grow large require path for their growth. To gain insight into the basis of growth control by path, we generated additional alleles of path and further examined the apparent specificity of growth defects in path mutants. Here, we confirm our prior finding that loss of path function imposes an upper limit on neuron growth, and additionally report that path likely limits overall neurite length rather than dendrite length alone. Using a GFP knock-in allele of path, we identify additional tissues where path likely functions in nutrient sensing and possibly growth control. Finally, we demonstrate that path regulates translational capacity in a cell type that does not normally require path for growth, suggesting that path may confer robustness on growth programs by buffering translational output. Altogether, these studies suggest that Path is a nutrient sensor with widespread function in Drosophila.
Collapse
Affiliation(s)
- Wen-Yang Lin
- a Department of Biology ; University of Washington ; Seattle , WA USA
| | - Claire R Williams
- a Department of Biology ; University of Washington ; Seattle , WA USA
| | - Connie Yan
- a Department of Biology ; University of Washington ; Seattle , WA USA
| | - Jay Z Parrish
- a Department of Biology ; University of Washington ; Seattle , WA USA
| |
Collapse
|
15
|
Lu HL, Chang CC, Wilson ACC. Amino acid transporters implicated in endocytosis of Buchnera during symbiont transmission in the pea aphid. EvoDevo 2016; 7:24. [PMID: 27895889 PMCID: PMC5117694 DOI: 10.1186/s13227-016-0061-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Accepted: 11/04/2016] [Indexed: 12/29/2022] Open
Abstract
Background Many insects host their obligate, maternally transmitted symbiotic bacteria in specialized cells called bacteriocytes. One of the best-studied insect nutritional endosymbioses is that of the aphid and its endosymbiont, Buchnera aphidicola. Aphids and Buchnera are metabolically and developmentally integrated, but the molecular mechanisms underlying Buchnera transmission and coordination with aphid development remain largely unknown. Previous work using electron microscopy to study aphid asexual embryogenesis has revealed that Buchnera transmission involves exocytosis from a maternal bacteriocyte followed by endocytotic uptake by a blastula. While the importance of exo- and endocytic cellular processes for symbiont transmission is clear, the molecular mechanisms that regulate these processes are not known. Here, we shed light on the molecular mechanisms that regulate Buchnera transmission and developmental integration. Results We present the developmental atlas of ACYPI000536 and ACYPI008904 mRNAs during asexual embryogenesis in the pea aphid, Acyrthosiphon pisum. Immediately before Buchnera invasion, transcripts of both genes were detected by whole-mount in situ hybridization in the posterior syncytial nuclei of late blastula embryos. Following Buchnera invasion, expression of both genes was identified in the region occupied by Buchnera throughout embryogenesis. Notably during Buchnera migration, expression of both genes was not concomitant with the entirety of the bacterial mass but rather expression colocalized with Buchnera in the anterior region of the bacterial mass. In addition, we found that ACYPI000536 was expressed in nuclei at the leading edge of the bacterial mass, joining the bacterial mass in subsequent developmental stages. Finally, quantitative reverse transcription real-time PCR suggested that early in development both transcripts were maternally provisioned to embryos. Conclusions We venture that ACYPI000536 and ACYPI008904 function as nutrient sensors at the site of symbiont invasion to facilitate TOR-pathway-mediated endocytosis of Buchnera by the aphid blastula. Our data support earlier reports of bacteriocyte determination involving a two-step recruitment process but suggest that the second wave of recruitment occurs earlier than previously described. Finally, our work highlights that bacteriocyte-enriched amino acid transporter paralogs have additionally been retained to play novel developmental roles in both symbiont recruitment and bacteriome development. Electronic supplementary material The online version of this article (doi:10.1186/s13227-016-0061-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hsiao-Ling Lu
- Department of Biology, University of Miami, Coral Gables, FL 33146 USA ; Department of Entomology, College of Bioresources and Agriculture, National Taiwan University, Taipei, 10617 Taiwan ; Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, 10617 Taiwan
| | - Chun-Che Chang
- Department of Entomology, College of Bioresources and Agriculture, National Taiwan University, Taipei, 10617 Taiwan ; Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, 10617 Taiwan ; Institute of Biotechnology, College of Bioresources and Agriculture, National Taiwan University, Taipei, 10617 Taiwan
| | - Alex C C Wilson
- Department of Biology, University of Miami, Coral Gables, FL 33146 USA
| |
Collapse
|
16
|
The Glutamine Transporters and Their Role in the Glutamate/GABA-Glutamine Cycle. ADVANCES IN NEUROBIOLOGY 2016; 13:223-257. [PMID: 27885631 DOI: 10.1007/978-3-319-45096-4_8] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Glutamine is a key amino acid in the CNS, playing an important role in the glutamate/GABA-glutamine cycle (GGC). In the GGC, glutamine is transferred from astrocytes to neurons, where it will replenish the inhibitory and excitatory neurotransmitter pools. Different transporters participate in this neural communication, i.e., the transporters responsible for glutamine efflux from astrocytes and influx into the neurons, such as the members of the SNAT, LAT, y+LAT, and ASC families of transporters. The SNAT family consists of the transporter isoforms SNAT3 and SNAT5 that are related to efflux from the astrocytic compartment, and SNAT1 and SNAT2 that are associated with glutamine uptake into the neuronal compartment. The isoforms SNAT7 and SNAT8 do not have their role completely understood, but they likely also participate in the GGC. The isoforms LAT2 and y+LAT2 facilitate the exchange of neutral amino acids and cationic amino acids (y+LAT2 isoform) and have been associated with glutamine efflux from astrocytes. ASCT2 is a Na+-dependent antiporter, the participation of which in the GGC also remains to be better characterized. All these isoforms are tightly regulated by transcriptional and translational mechanisms, which are induced by several determinants such as amino acid deprivation, hormones, pH, and the activity of different signaling pathways. Dysfunctional glutamine transporter activity has been associated with the pathophysiological mechanisms of certain neurologic diseases, such as Hepatic Encephalopathy and Manganism. However, there might also be other neuropathological conditions associated with an altered GGC, in which glutamine transporters are dysfunctional. Hence, it appears to be of critical importance that the physiological and pathological aspects of glutamine transporters are thoroughly investigated.
Collapse
|
17
|
Bagchi S, Baomar HA, Al-Walai S, Al-Sadi S, Fredriksson R. Histological analysis of SLC38A6 (SNAT6) expression in mouse brain shows selective expression in excitatory neurons with high expression in the synapses. PLoS One 2014; 9:e95438. [PMID: 24752331 PMCID: PMC3994050 DOI: 10.1371/journal.pone.0095438] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 03/27/2014] [Indexed: 11/18/2022] Open
Abstract
SLC38A6 is one of the newly found members of the solute carrier 38 family consisting of total 11 members, of which only 6 have been characterized so far. Being the only glutamine transporter family expressed in the brain, this family of proteins are most probably involved in the regulation of the glutamate-glutamine cycle, responsible for preventing excitotoxicity. We used immunohistochemistry to show that SLC38A6 is primarily expressed in excitatory neurons and is not expressed in the astrocytes. Using proximity ligation assay, we have quantified the interactions of this SLC38 family protein with other proteins with known localization in the cells, showing that this transporter is expressed at the synapses. Moreover, this study has enabled us to come up with a model suggesting sub-cellular localization of SLC38A6 at the synaptic membrane of the excitatory neurons.
Collapse
Affiliation(s)
- Sonchita Bagchi
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden
| | - Hajar Ali Baomar
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden
| | - Somar Al-Walai
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden
| | - Saifaddin Al-Sadi
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden
| | - Robert Fredriksson
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden
- * E-mail:
| |
Collapse
|
18
|
PAT4 is abundantly expressed in excitatory and inhibitory neurons as well as epithelial cells. Brain Res 2014; 1557:12-25. [PMID: 24530433 DOI: 10.1016/j.brainres.2014.02.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 02/05/2014] [Indexed: 11/23/2022]
Abstract
PAT4, the fourth member of the SLC36/proton dependent amino acid transporter (PAT) family, is a high-affinity, low capacity electroneutral transporter of neutral amino acids like proline and tryptophan. It has also been associated with the function of mTORC1, a complex in the mammalian target of rapamycin (mTOR) pathway. We performed in situ hybridization and immunohistological analysis to determine the expression profile of PAT4, as well as an RT-PCR study on tissue from mice exposed to leucine. We performed a phylogenetic analysis to determine the evolutionary origin of PAT4. The in situ hybridization and the immunohistochemistry on mouse brain sections and hypothalamic cells showed abundant PAT4 expression in the mouse brain intracellularly in both inhibitory and excitatory neurons, partially co-localizing with lysosomal markers and epithelial cells lining the ventricles. Its location in epithelial cells around the ventricles indicates a transport of substrates across the blood brain barrier. Phylogenetic analysis showed that PAT4 belongs to an evolutionary old family most likely predating animals, and PAT4 is the oldest member of that family.
Collapse
|
19
|
|
20
|
Li X, Garrity AG, Xu H. Regulation of membrane trafficking by signalling on endosomal and lysosomal membranes. J Physiol 2013; 591:4389-401. [PMID: 23878375 DOI: 10.1113/jphysiol.2013.258301] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Endosomal and lysosomal membrane trafficking requires the coordination of multiple signalling events to control cargo sorting and processing, and endosome maturation. The initiation and termination of signalling events in endosomes and lysosomes is not well understood, but several key regulators have been identified, which include small GTPases, phosphoinositides, and Ca2+. Small GTPases act as master regulators and molecular switches in a GTP-dependent manner, initiating signalling cascades to regulate the direction and specificity of endosomal trafficking. Phosphoinositides are membrane-bound lipids that indicate vesicular identities for recruiting specific cytoplasmic proteins to endosomal membranes, thus allowing specificity of membrane fusion, fission, and cargo sorting to occur within and between specific vesicle compartments. In addition, phosphoinositides regulate the function of membrane proteins such as ion channels and transporters in a compartment-specific manner to mediate transport and signalling. Finally, Ca2+, a locally acting second messenger released from intracellular ion channels, may provide precise spatiotemporal regulation of endosomal signalling and trafficking events. Small GTPase signalling can regulate phosphoinositide conversion during endosome maturation, and electrophysiological studies on isolated endosomes have shown that endosomal and lysosomal Ca2+ channels are directly modulated by endosomal lipids. Thus trafficking and maturation of endosomes and lysosomes can be precisely regulated by dynamic changes in GTPases and membrane lipids, as well as Ca2+ signalling. Importantly, impaired phosphoinositide and Ca2+ signalling can cause endosomal and lysosomal trafficking defects at the cellular level, and a spectrum of lysosome storage diseases.
Collapse
Affiliation(s)
- Xinran Li
- H. Xu: University of Michigan, MCDB, 3089 Natural Science Building (Kraus), 830 North University, Ann Arbor, MI 48109, USA.
| | | | | |
Collapse
|
21
|
Schweikhard ES, Ziegler CM. Amino acid secondary transporters: toward a common transport mechanism. CURRENT TOPICS IN MEMBRANES 2013. [PMID: 23177982 DOI: 10.1016/b978-0-12-394316-3.00001-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Solute carriers (SLC) that transport amino acids are key players in health and diseases in humans. Their prokaryotic relatives are often involved in essential physiological processes in microorganisms, e.g. in homeostasis and acidic/osmotic stress response. High-resolution X-ray structures of the sequence-unrelated amino acid transporters unraveled a striking structural similarity between carriers, which were formerly assigned to different families. The highly conserved fold is characterized by two inverted structural repeats of five transmembrane helices each and indicates common mechanistic transport concepts if not an evolutionary link among a large number of amino acid transporters. Therefore, these transporters are classified now into the structural amino acid-polyamine-organocation superfamily (APCS). The APCS includes among others the mammalian SLC6 transporters and the heterodimeric SLC7/SLC3 transporters. However, it has to be noted that the APCS is not limited entirely to amino acid transporters but contains also transporters for, e.g. amino acid derivatives and sugars. For instance, the betaine-choline-carnitine transporter family of bacterial activity-regulated Na(+)- and H(+)-coupled symporters for glycine betaine and choline is also part of this second largest structural superfamily. The APCS fold provides different possibilities to transport the same amino acid. Arginine can be transported by an H(+)-coupled symport or by antiport mechanism in exchange against agmatine for example. The convergence of the mechanistic concept of transport under comparable physiological conditions allows speculating if structurally unexplored amino acid transporters, e.g. the members of the SLC36 and SLC38 family, belong to the APCS, too. In the kidney, which is an organ that depends critically on the regulated amino acid transport, these different SLC transporters have to work together to account for proper function. Here, we will summarize the basic concepts of Na(+)- and H(+)-coupled amino acid symport and amino acid-product antiport in the light of the respective physiological requirements.
Collapse
Affiliation(s)
- Eva S Schweikhard
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt, Germany
| | | |
Collapse
|
22
|
Metzler R, Meleshkevitch EA, Fox J, Kim H, Boudko DY. An SLC6 transporter of the novel B(0,)- system aids in absorption and detection of nutrient amino acids in Caenorhabditis elegans. ACTA ACUST UNITED AC 2013; 216:2843-57. [PMID: 23580723 DOI: 10.1242/jeb.081497] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Nutrient amino acid transporters (NATs) of solute carrier family 6 (SLC6) mediate uptake of essential amino acids in mammals and insects. Phylogenomic analysis of the Caenorhabditis elegans (Ce) SLC6 family identifies five genes paralogous to an insect-specific NAT subfamily. Here we cloned and characterized the first representative of the identified nematode-specific transporters, SNF-5. SNF-5 mediates broad spectrum cation-coupled transport of neutral amino acids with submillimolar affinities and stoichiometry of 1 AA:1 Na(+), except for 1 l-Pro:2 Na(+). Unexpectedly, it transports acidic l-Glu(-) and l-Asp(-) (1 AA(-):3 Na(+)), revealing it to be the first member of a new B(0,-) system among characterized SLC6 transporters. This activity correlates with a unique positively charged His(+) 377 in the substrate-binding pocket. snf-5 promoter-driven enhanced green fluorescent protein labels intestinal cells INT1-9 and three pairs of amphid sensory neurons: ASI, ADF and ASK. These cells are intimately involved in control of dauer diapause, development, metabolism and longevity. The snf-5 deletion mutants do not show apparent morphological disorders, but increase dauer formation while reducing dauer maintenance upon starvation. Overall, the present study characterized the first nematode-specific NAT and revealed important structural and functional aspects of this transporter. In addition to the predictable role in alimentary amino acid absorption, our results indicate possible neuronal roles of SNF-5 as an amino acid provider to specific neuronal functions, including sensing of amino acid availability.
Collapse
Affiliation(s)
- Ryan Metzler
- The Department of Physiology and Biophysics of the Rosalind Franklin University of Medicine and Science, Chicago Medical School, North Chicago, IL 60064, USA
| | | | | | | | | |
Collapse
|
23
|
Parker MD, Boron WF. The divergence, actions, roles, and relatives of sodium-coupled bicarbonate transporters. Physiol Rev 2013; 93:803-959. [PMID: 23589833 PMCID: PMC3768104 DOI: 10.1152/physrev.00023.2012] [Citation(s) in RCA: 208] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The mammalian Slc4 (Solute carrier 4) family of transporters is a functionally diverse group of 10 multi-spanning membrane proteins that includes three Cl-HCO3 exchangers (AE1-3), five Na(+)-coupled HCO3(-) transporters (NCBTs), and two other unusual members (AE4, BTR1). In this review, we mainly focus on the five mammalian NCBTs-NBCe1, NBCe2, NBCn1, NDCBE, and NBCn2. Each plays a specialized role in maintaining intracellular pH and, by contributing to the movement of HCO3(-) across epithelia, in maintaining whole-body pH and otherwise contributing to epithelial transport. Disruptions involving NCBT genes are linked to blindness, deafness, proximal renal tubular acidosis, mental retardation, and epilepsy. We also review AE1-3, AE4, and BTR1, addressing their relevance to the study of NCBTs. This review draws together recent advances in our understanding of the phylogenetic origins and physiological relevance of NCBTs and their progenitors. Underlying these advances is progress in such diverse disciplines as physiology, molecular biology, genetics, immunocytochemistry, proteomics, and structural biology. This review highlights the key similarities and differences between individual NCBTs and the genes that encode them and also clarifies the sometimes confusing NCBT nomenclature.
Collapse
Affiliation(s)
- Mark D Parker
- Dept. of Physiology and Biophysics, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106-4970, USA.
| | | |
Collapse
|
24
|
Matsui T, Fukuda M. Rab12 regulates mTORC1 activity and autophagy through controlling the degradation of amino-acid transporter PAT4. EMBO Rep 2013; 14:450-7. [PMID: 23478338 DOI: 10.1038/embor.2013.32] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 02/16/2013] [Accepted: 02/19/2013] [Indexed: 12/22/2022] Open
Abstract
Autophagy is an evolutionarily conserved catabolic mechanism that targets intracellular molecules and damaged organelles to lysosomes. Autophagy is achieved by a series of membrane trafficking events, but their regulatory mechanisms are poorly understood. Here, we report small GTPase Rab12 as a new type of autophagic regulator that controls the degradation of an amino-acid transporter. Knockdown of Rab12 results in inhibition of autophagy and in increased activity of mTORC1 (mammalian/mechanistic target of rapamycin complex 1), an upstream regulator of autophagy. We also found that Rab12 promotes constitutive degradation of PAT4 (proton-coupled amino-acid transporter 4), whose accumulation in Rab12-knockdown cells modulates mTORC1 activity and autophagy. Our findings reveal a new mechanism of regulation of mTORC1 signalling and autophagy, that is, quality control of PAT4 by Rab12.
Collapse
Affiliation(s)
- Takahide Matsui
- Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | | |
Collapse
|
25
|
Voigt V, Laug L, Zebisch K, Thondorf I, Markwardt F, Brandsch M. Transport of the areca nut alkaloid arecaidine by the human proton-coupled amino acid transporter 1 (hPAT1). J Pharm Pharmacol 2012; 65:582-90. [DOI: 10.1111/jphp.12006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Accepted: 10/18/2012] [Indexed: 11/27/2022]
Abstract
Abstract
Objectives
The pyridine alkaloid arecaidine is an ingredient of areca nut preparations. It is responsible for many physiological effects observed during areca nut chewing. However, the mechanism underlying its oral bioavailability has not yet been studied. We investigated whether the H+-coupled amino acid transporter 1 (PAT1, SLC36A1), which is expressed in the intestinal epithelium, accepts arecaidine, arecoline, isoguvacine and other derivatives as substrates.
Methods
Inhibition of l-[3H]proline uptake by arecaidine and derivatives was determined in Caco-2 cells expressing hPAT1 constitutively and in HeLa cells transiently transfected with hPAT1-cDNA. Transmembrane transport of arecaidine and derivatives was measured electrophysiologically in Xenopus laevis oocytes.
Key findings
Arecaidine, guvacine and isoguvacine but not arecoline strongly inhibited the uptake of l-[3H]proline into Caco-2 cells. Kinetic analyses revealed the competitive manner of l-proline uptake inhibition by arecaidine. In HeLa cells transfected with hPAT1-cDNA an affinity constant of 3.8 mm was obtained for arecaidine. Electrophysiological measurements at hPAT1-expressing X. laevis oocytes demonstrated that arecaidine, guvacine and isoguvacine are transported by hPAT1 in an electrogenic manner.
Conclusion
We conclude that hPAT1 transports arecaidine, guvacine and isoguvacine across the apical membrane of enterocytes and that hPAT1 might be responsible for the intestinal absorption of these drug candidates.
Collapse
Affiliation(s)
- Valerie Voigt
- Biozentrum of the Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Linda Laug
- Biozentrum of the Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Katja Zebisch
- Biozentrum of the Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Iris Thondorf
- Institute of Biochemistry and Biotechnology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Fritz Markwardt
- Julius-Bernstein-Institute for Physiology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Matthias Brandsch
- Biozentrum of the Martin-Luther-University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
26
|
Singer D, Camargo SMR, Ramadan T, Schäfer M, Mariotta L, Herzog B, Huggel K, Wolfer D, Werner S, Penninger JM, Verrey F. Defective intestinal amino acid absorption in Ace2 null mice. Am J Physiol Gastrointest Liver Physiol 2012; 303:G686-95. [PMID: 22790597 DOI: 10.1152/ajpgi.00140.2012] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Mutations in the main intestinal and kidney luminal neutral amino acid transporter B(0)AT1 (Slc6a19) lead to Hartnup disorder, a condition that is characterized by neutral aminoaciduria and in some cases pellagra-like symptoms. These latter symptoms caused by low-niacin are thought to result from defective intestinal absorption of its precursor L-tryptophan. Since Ace2 is necessary for intestinal B(0)AT1 expression, we tested the impact of intestinal B(0)AT1 absence in ace2 null mice. Their weight gain following weaning was decreased, and Na(+)-dependent uptake of B(0)AT1 substrates measured in everted intestinal rings was defective. Additionally, high-affinity Na(+)-dependent transport of L-proline, presumably via SIT1 (Slc6a20), was absent, whereas glucose uptake via SGLT1 (Slc5a1) was not affected. Measurements of small intestine luminal amino acid content following gavage showed that more L-tryptophan than other B(0)AT1 substrates reach the ileum in wild-type mice, which is in line with its known lower apparent affinity. In ace2 null mice, the absorption defect was confirmed by a severalfold increase of L-tryptophan and of other neutral amino acids reaching the ileum lumen. Furthermore, plasma and muscle levels of glycine and L-tryptophan were significantly decreased in ace2 null mice, with other neutral amino acids displaying a similar trend. A low-protein/low-niacin diet challenge led to differential changes in plasma amino acid levels in both wild-type and ace2 null mice, but only in ace2 null mice to a stop in weight gain. Despite the combination of low-niacin with a low-protein diet, plasma niacin concentrations remained normal in ace2 null mice and no pellagra symptoms, such as photosensitive skin rash or ataxia, were observed. In summary, mice lacking Ace2-dependent intestinal amino acid transport display no total niacin deficiency nor clear pellagra symptoms, even under a low-protein and low-niacin diet, despite gross amino acid homeostasis alterations.
Collapse
Affiliation(s)
- Dustin Singer
- Institute of Physiology, University of Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Petrat F, Boengler K, Schulz R, de Groot H. Glycine, a simple physiological compound protecting by yet puzzling mechanism(s) against ischaemia-reperfusion injury: current knowledge. Br J Pharmacol 2012; 165:2059-72. [PMID: 22044190 DOI: 10.1111/j.1476-5381.2011.01711.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Ischaemia is amongst the leading causes of death. Despite this importance, there are only a few therapeutic approaches to protect from ischaemia-reperfusion injury (IRI). In experimental studies, the amino acid glycine effectively protected from IRI. In the prevention of IRI by glycine in cells and isolated perfused or cold-stored organs (tissues), direct cytoprotection plays a crucial role, most likely by prevention of the formation of pathological plasma membrane pores. Under in vivo conditions, the mechanism of protection by glycine is less clear, partly due to the physiological presence of the amino acid. Here, inhibition of the inflammatory response in the injured tissue is considered to contribute decisively to the glycine-induced reduction of IRI. However, attenuation of IRI recently achieved in experimental animals by low-dose glycine treatment regimens suggests additional/other (unknown) protective mechanisms. Despite the convincing experimental evidence and the large therapeutic width of glycine, there are only a few clinical trials on the protection from IRI by glycine with ambivalent results. Thus, both the mechanism(s) behind the protection of glycine against IRI in vivo and its true clinical potential remain to be addressed in future experimental studies/clinical trials.
Collapse
Affiliation(s)
- Frank Petrat
- Institut für Physiologische Chemie, Universitätsklinikum Essen, Essen, Germany
| | | | | | | |
Collapse
|
28
|
|
29
|
Zebisch K, Brandsch M. Transport of L-proline by the proton-coupled amino acid transporter PAT2 in differentiated 3T3-L1 cells. Amino Acids 2012; 44:373-81. [PMID: 22711289 DOI: 10.1007/s00726-012-1340-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 06/04/2012] [Indexed: 10/28/2022]
Abstract
Mechanism and substrate specificity of the proton-coupled amino acid transporter 2 (PAT2, SLC36A2) have been studied so far only in heterologous expression systems such as HeLa cells and Xenopus laevis oocytes. In this study, we describe the identification of the first cell line that expresses PAT2. We cultured 3T3-L1 cells for up to 2 weeks and differentiated the cells into adipocytes in supplemented media containing 2 μM rosiglitazone. During the 14 day differentiation period the uptake of the prototype PAT2 substrate L-[(3)H]proline increased ~5-fold. The macro- and microscopically apparent differentiation of 3T3-L1 cells coincided with their H(+) gradient-stimulated uptake of L-[(3)H]proline. Uptake was rapid, independent of a Na(+) gradient but stimulated by an inwardly directed H(+) gradient with maximal uptake occurring at pH 6.0. L-Proline uptake was found to be mediated by a transport system with a Michaelis constant (K(t)) of 130 ± 10 μM and a maximal transport velocity of 4.9 ± 0.2 nmol × 5 min(-1 )mg of protein(-1). Glycine, L-alanine, and L-tryptophan strongly inhibited L-proline uptake indicating that these amino acids also interact with the transport system. It is concluded that 3T3-L1 adipocytes express the H(+)-amino acid cotransport system PAT2.
Collapse
Affiliation(s)
- Katja Zebisch
- Biozentrum of the Martin-Luther-University Halle-Wittenberg, Weinbergweg 22, 06120 Halle, Germany
| | | |
Collapse
|
30
|
Proton-assisted amino acid transporter PAT1 complexes with Rag GTPases and activates TORC1 on late endosomal and lysosomal membranes. PLoS One 2012; 7:e36616. [PMID: 22574197 PMCID: PMC3344915 DOI: 10.1371/journal.pone.0036616] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Accepted: 04/03/2012] [Indexed: 01/19/2023] Open
Abstract
Mammalian Target of Rapamycin Complex 1 (mTORC1) is activated by growth factor-regulated phosphoinositide 3-kinase (PI3K)/Akt/Rheb signalling and extracellular amino acids (AAs) to promote growth and proliferation. These AAs induce translocation of mTOR to late endosomes and lysosomes (LELs), subsequent activation via mechanisms involving the presence of intralumenal AAs, and interaction between mTORC1 and a multiprotein assembly containing Rag GTPases and the heterotrimeric Ragulator complex. However, the mechanisms by which AAs control these different aspects of mTORC1 activation are not well understood. We have recently shown that intracellular Proton-assisted Amino acid Transporter 1 (PAT1)/SLC36A1 is an essential mediator of AA-dependent mTORC1 activation. Here we demonstrate in Human Embryonic Kidney (HEK-293) cells that PAT1 is primarily located on LELs, physically interacts with the Rag GTPases and is required for normal AA-dependent mTOR relocalisation. We also use the powerful in vivo genetic methodologies available in Drosophila to investigate the regulation of the PAT1/Rag/Ragulator complex. We show that GFP-tagged PATs reside at both the cell surface and LELs in vivo, mirroring PAT1 distribution in several normal mammalian cell types. Elevated PI3K/Akt/Rheb signalling increases intracellular levels of PATs and synergistically enhances PAT-induced growth via a mechanism requiring endocytosis. In light of the recent identification of the vacuolar H+-ATPase as another Rag-interacting component, we propose a model in which PATs function as part of an AA-sensing engine that drives mTORC1 activation from LEL compartments.
Collapse
|
31
|
Boudko DY. Molecular basis of essential amino acid transport from studies of insect nutrient amino acid transporters of the SLC6 family (NAT-SLC6). JOURNAL OF INSECT PHYSIOLOGY 2012; 58:433-49. [PMID: 22230793 PMCID: PMC3397479 DOI: 10.1016/j.jinsphys.2011.12.018] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Revised: 12/21/2011] [Accepted: 12/23/2011] [Indexed: 05/03/2023]
Abstract
Two protein families that represent major components of essential amino acid transport in insects have been identified. They are annotated as the SLC6 and SLC7 families of transporters according to phylogenetic proximity to characterized amino acid transporters (HUGO nomenclature). Members of these families have been identified as important apical and basolateral parts of transepithelial essential amino acid absorption in the metazoan alimentary canal. Synergistically, they play critical physiological roles as essential substrate providers to diverse metabolic processes, including generic protein synthesis. This review briefly clarifies the requirements for amino acid transport and a variety of amino acid transport mechanisms, including the aforementioned families. Further it focuses on the large group of Nutrient Amino acid Transporters (NATs), which comprise a recently identified subfamily of the Neurotransmitter Sodium Symporter family (NSS or SLC6). The first insect NAT, cloned from the caterpillar gut, has a broad substrate spectrum similar to mammalian B(0) transporters. Several new NAT-SLC6 members have been characterized in an effort to explore mechanisms for the essential amino acid absorption in model dipteran insects. The identification and functional characterization of new B(0)-like and narrow specificity transporters of essential amino acids in fruit fly and mosquitoes leads to a fundamentally important insight: that NATs evolved and act together as the integrated active core of a transport network that mediates active alimentary absorption and systemic distribution of essential amino acids. This role of NATs is projected from the most primitive prokaryotes to the most complex metazoan organisms, and represents an interesting platform for unraveling the molecular evolution of amino acid transport and modeling amino acid transport disorders. The comparative study of NATs elucidates important adaptive differences between essential amino acid transportomes of invertebrate and vertebrate organisms, outlining a new possibility for selective targeting of essential amino acid absorption mechanisms to control medically and economically important arthropods and other invertebrate organisms.
Collapse
Affiliation(s)
- Dmitri Y Boudko
- Department of Physiology and Biophysics of Rosalind Franklin University, Chicago Medical School, North Chicago, IL 60064, USA.
| |
Collapse
|
32
|
Kiuchi T, Banno Y, Katsuma S, Shimada T. Mutations in an amino acid transporter gene are responsible for sex-linked translucent larval skin of the silkworm, Bombyx mori. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2011; 41:680-687. [PMID: 21619931 DOI: 10.1016/j.ibmb.2011.04.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 04/20/2011] [Accepted: 04/27/2011] [Indexed: 05/30/2023]
Abstract
The sex-linked translucent (os) mutation in the silkworm, Bombyx mori, confers slightly translucent larval skin resulting from a decrease in the incorporation of uric acid into epidermal cells. By positional cloning, we narrowed a region linked to the os phenotype to approximately 157 kb located on scaffold Bm_scaf72 on the Z chromosome (chromosome 1). The region contained four gene models. Sequencing analysis revealed that one of the candidate genes had a 7-bp deletion in the coding region. We also found a 111-bp deletion or single-nucleotide substitution in the same gene using independent os mutant strains. Because all the mutations caused the generation of abnormal transcripts followed by translation of a truncated protein, we conclude that the mutation of this candidate gene is responsible for the translucent larval skin of the os mutant. Sequence analysis indicated that the gene responsible for the os mutation had homology to amino acid transporters of the solute carrier family of proteins. Our results suggest that solute carrier proteins are involved in uric acid transport in insects and other invertebrates.
Collapse
Affiliation(s)
- Takashi Kiuchi
- Department of Agricultural and Environmental Biology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Yayoi 1-1-1, Bunkyo-ku, Tokyo 113-8657, Japan
| | | | | | | |
Collapse
|
33
|
Dhaoui M, Auchère F, Blaiseau PL, Lesuisse E, Landoulsi A, Camadro JM, Haguenauer-Tsapis R, Belgareh-Touzé N. Gex1 is a yeast glutathione exchanger that interferes with pH and redox homeostasis. Mol Biol Cell 2011; 22:2054-67. [PMID: 21490148 PMCID: PMC3113770 DOI: 10.1091/mbc.e10-11-0906] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Revised: 11/18/2011] [Accepted: 04/07/2011] [Indexed: 01/20/2023] Open
Abstract
In the yeast Saccharomyces cerevisiae, glutathione plays a major role in heavy metal detoxification and protection of cells against oxidative stress. We show that Gex1 is a new glutathione exchanger. Gex1 and its paralogue Gex2 belong to the major facilitator superfamily of transporters and display similarities to the Aft1-regulon family of siderophore transporters. Gex1 was found mostly at the vacuolar membrane and, to a lesser extent, at the plasma membrane. Gex1 expression was induced under conditions of iron depletion and was principally dependent on the iron-responsive transcription factor Aft2. However, a gex1Δ gex2Δ strain displayed no defect in known siderophore uptake. The deletion mutant accumulated intracellular glutathione, and cells overproducing Gex1 had low intracellular glutathione contents, with glutathione excreted into the extracellular medium. Furthermore, the strain overproducing Gex1 induced acidification of the cytosol, confirming the involvement of Gex1 in proton transport as a probable glutathione/proton antiporter. Finally, the imbalance of pH and glutathione homeostasis in the gex1Δ gex2Δ and Gex1-overproducing strains led to modulations of the cAMP/protein kinase A and protein kinase C1 mitogen-activated protein kinase signaling pathways.
Collapse
Affiliation(s)
- Manel Dhaoui
- Laboratoire Ubiquitine et Trafic Intracellulaire, Institut Jacques Monod, UMR 7592 CNRS-Université Paris-Diderot, Paris, France
- Laboratoire de Biochimie et Biologie Moléculaire 03/UR/0902, Faculté des Sciences de Bizerte, Zarzouna, Tunisia
| | - Françoise Auchère
- Laboratoire Mitochondries, Métaux et Stress Oxydatif, Institut Jacques Monod, UMR 7592 CNRS-Université Paris-Diderot, Paris, France
| | - Pierre-Louis Blaiseau
- Laboratoire Mitochondries, Métaux et Stress Oxydatif, Institut Jacques Monod, UMR 7592 CNRS-Université Paris-Diderot, Paris, France
| | - Emmanuel Lesuisse
- Laboratoire Mitochondries, Métaux et Stress Oxydatif, Institut Jacques Monod, UMR 7592 CNRS-Université Paris-Diderot, Paris, France
| | - Ahmed Landoulsi
- Laboratoire de Biochimie et Biologie Moléculaire 03/UR/0902, Faculté des Sciences de Bizerte, Zarzouna, Tunisia
| | - Jean-Michel Camadro
- Laboratoire Mitochondries, Métaux et Stress Oxydatif, Institut Jacques Monod, UMR 7592 CNRS-Université Paris-Diderot, Paris, France
| | - Rosine Haguenauer-Tsapis
- Laboratoire Ubiquitine et Trafic Intracellulaire, Institut Jacques Monod, UMR 7592 CNRS-Université Paris-Diderot, Paris, France
| | - Naïma Belgareh-Touzé
- Laboratoire Ubiquitine et Trafic Intracellulaire, Institut Jacques Monod, UMR 7592 CNRS-Université Paris-Diderot, Paris, France
| |
Collapse
|
34
|
Spinster is required for autophagic lysosome reformation and mTOR reactivation following starvation. Proc Natl Acad Sci U S A 2011; 108:7826-31. [PMID: 21518918 DOI: 10.1073/pnas.1013800108] [Citation(s) in RCA: 224] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Autophagy is a conserved cellular process to degrade and recycle cytoplasmic components. During autophagy, lysosomes fuse with an autophagosome to form an autolysosome. Sequestered components are degraded by lysosomal hydrolases and presumably released into the cytosol by lysosomal efflux permeases. Following starvation-induced autophagy, lysosome homeostasis is restored by autophagic lysosome reformation (ALR) requiring activation of the "target of rapamycin" (TOR) kinase. Spinster (Spin) encodes a putative lysosomal efflux permease with the hallmarks of a sugar transporter. Drosophila spin mutants accumulate lysosomal carbohydrates and enlarged lysosomes. Here we show that defects in spin lead to the accumulation of enlarged autolysosomes. We find that spin is essential for mTOR reactivation and lysosome reformation following prolonged starvation. Further, we demonstrate that the sugar transporter activity of Spin is essential for ALR.
Collapse
|
35
|
Pillai SM, Meredith D. SLC36A4 (hPAT4) is a high affinity amino acid transporter when expressed in Xenopus laevis oocytes. J Biol Chem 2010; 286:2455-60. [PMID: 21097500 DOI: 10.1074/jbc.m110.172403] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The SLC36 family of transporters consists of four genes, two of which, SLC36A1 and SLC36A2, have been demonstrated to code for human proton-coupled amino acid transporters or hPATs. Here we report the characterization of the fourth member of the family, SLC36A4 or hPAT4, which when expressed in Xenopus laevis oocytes also encodes a plasma membrane amino acid transporter, but one that is not proton-coupled and has a very high substrate affinity for the amino acids proline and tryptophan. hPAT4 in Xenopus oocytes mediated sodium-independent, electroneutral uptake of [(3)H]proline, with the highest rate of uptake when the uptake medium pH was 7.4 and an affinity of 3.13 μM. Tryptophan was also an excellently transported substrate with a similarly high affinity (1.72 μM). Other amino acids that inhibited [(3)H]proline were isoleucine (K(i) 0.23 mM), glutamine (0.43 mM), methionine (0.44 mM), and alanine (1.48 mM), and with lower affinity, glycine, threonine, and cysteine (K(i) >5 mM for all). Of the amino acids directly tested for transport, only proline, tryptophan, and alanine showed significant uptake, whereas glycine and cysteine did not. Of the non-proteogenic amino acids and drugs tested, only sarcosine produced inhibition (K(i) 1.09 mM), whereas γ-aminobutyric acid (GABA), β-alanine, L-Dopa, D-serine, and δ-aminolevulinic acid were without effect on [(3)H]proline uptake. This characterization of hPAT4 as a very high affinity/low capacity non-proton-coupled amino acid transporter raises questions about its physiological role, especially as the transport characteristics of hPAT4 are very similar to the Drosophila orthologue PATH, an amino acid "transceptor" that plays a role in nutrient sensing.
Collapse
|
36
|
Heublein S, Kazi S, Ogmundsdóttir MH, Attwood EV, Kala S, Boyd CAR, Wilson C, Goberdhan DCI. Proton-assisted amino-acid transporters are conserved regulators of proliferation and amino-acid-dependent mTORC1 activation. Oncogene 2010; 29:4068-79. [PMID: 20498635 PMCID: PMC3018277 DOI: 10.1038/onc.2010.177] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2009] [Revised: 04/12/2010] [Accepted: 04/19/2010] [Indexed: 12/18/2022]
Abstract
The phosphoinositide3-kinase (PI3K)/Akt and downstream mammalian target of rapamycin complex 1 (mTORC1) signalling cascades promote normal growth and are frequently hyperactivated in tumour cells. mTORC1 is also regulated by local nutrients, particularly amino acids, but the mechanisms involved are poorly understood. Unexpectedly, members of the proton-assisted amino-acid transporter (PAT or SLC36) family emerged from in vivo genetic screens in Drosophila as transporters with uniquely potent effects on mTORC1-mediated growth. In this study, we show the two human PATs that are widely expressed in normal tissues and cancer cell lines, namely PAT1 and PAT4, behave similarly to fly PATs when expressed in Drosophila. Small interfering RNA knockdown shows that these molecules are required for the activation of mTORC1 targets and for proliferation in human MCF-7 breast cancer and HEK-293 embryonic kidney cell lines. Furthermore, activation of mTORC1 in starved HEK-293 cells stimulated by amino acids requires PAT1 and PAT4, and is elevated in PAT1-overexpressing cells. Importantly, in HEK-293 cells, PAT1 is highly concentrated in intracellular compartments, including endosomes, wherein mTOR shuttles upon amino-acid stimulation. Therefore our data are consistent with a model in which PATs modulate the activity of mTORC1 not by transporting amino acids into the cell but by modulating the intracellular response to amino acids.
Collapse
Affiliation(s)
- S Heublein
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Mah AK, Tu DK, Johnsen RC, Chu JS, Chen N, Baillie DL. Characterization of the octamer, a cis-regulatory element that modulates excretory cell gene-expression in Caenorhabditis elegans. BMC Mol Biol 2010; 11:19. [PMID: 20211011 PMCID: PMC2841177 DOI: 10.1186/1471-2199-11-19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2009] [Accepted: 03/08/2010] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND We have previously demonstrated that the POU transcription factor CEH-6 is required for driving aqp-8 expression in the C. elegans excretory (canal) cell, an osmotic regulatory organ that is functionally analogous to the kidney. This transcriptional regulation occurs through a CEH-6 binding to a cis-regulatory element called the octamer (ATTTGCAT), which is located in the aqp-8 promoter. RESULTS Here, we further characterize octamer driven transcription in C. elegans. First, we analyzed the positional requirements of the octamer. To do so, we assayed the effects on excretory cell expression by placing the octamer within the well-characterized promoter of vit-2. Second, using phylogenetic footprinting between three Caenorhabditis species, we identified a set of 165 genes that contain conserved upstream octamers in their promoters. Third, we used promoter::GFP fusions to examine the expression patterns of 107 of the 165 genes. This analysis demonstrated that conservation of octamers in promoters increases the likelihood that the gene is expressed in the excretory cell. Furthermore, we found that the sequences flanking the octamers may have functional importance. Finally, we altered the octamer using site-directed mutagenesis. Thus, we demonstrated that some nucleotide substitutions within the octamer do not affect the expression pattern of nearby genes, but change their overall expression was changed. Therefore, we have expanded the core octamer to include flanking regions and variants of the motif. CONCLUSIONS Taken together, we have demonstrated that octamer-containing regions are associated with excretory cell expression of several genes that have putative roles in osmoregulation. Moreover, our analysis of the octamer sequence and its sequence variants could aid in the identification of additional genes that are expressed in the excretory cell and that may also be regulated by CEH-6.
Collapse
Affiliation(s)
- Allan K Mah
- Department Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia, Canada, V5A 1S6
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, 950 West 28th Avenue, Vancouver, British Columbia, Canada V5Z H4H
| | - Domena K Tu
- Department Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia, Canada, V5A 1S6
| | - Robert C Johnsen
- Department Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia, Canada, V5A 1S6
| | - Jeffrey S Chu
- Department Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia, Canada, V5A 1S6
| | - Nansheng Chen
- Department Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia, Canada, V5A 1S6
| | - David L Baillie
- Department Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia, Canada, V5A 1S6
| |
Collapse
|
38
|
Bröer S, Schneider HP, Bröer A, Deitmer JW. Mutation of asparagine 76 in the center of glutamine transporter SNAT3 modulates substrate-induced conductances and Na+ binding. J Biol Chem 2009; 284:25823-31. [PMID: 19596860 PMCID: PMC2757984 DOI: 10.1074/jbc.m109.031013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2009] [Revised: 07/08/2009] [Indexed: 11/06/2022] Open
Abstract
The glutamine transporter SLC38A3 (SNAT3) plays an important role in the release of glutamine from brain astrocytes and the uptake of glutamine into hepatocytes. It is related to the vesicular GABA (gamma-aminobutyric acid) transporter and the SLC36 family of proton-amino acid cotransporters. The transporter carries out electroneutral Na+-glutamine cotransport-H+ antiport. In addition, substrate-induced uncoupled cation currents are observed. Mutation of asparagine 76 to glutamine or histidine in predicted transmembrane helix 1 abolished all substrate-induced currents. Mutation of asparagine 76 to aspartate rendered the transporter Na+-independent and resulted in a gain of a large substrate-induced chloride conductance in the absence of Na+. Thus, a single residue is critical for coupled and uncoupled ion flows in the glutamine transporter SNAT3. Homology modeling of SNAT3 along the structure of the related benzyl-hydantoin permease from Microbacterium liquefaciens reveals that Asn-76 is likely to be located in the center of the membrane close to the translocation pore and forms part of the predicted Na+ -binding site.
Collapse
Affiliation(s)
- Stefan Bröer
- From the Research School of Biology, The Australian National University, Canberra, ACT 0200, Australia and
| | - Hans-Peter Schneider
- the Abteilung für Allgemeine Zoologie, Fachbereich Biologie, Technische Universität Kaiserslautern, Erwin-Schrödinger Strasse, D-67653 Kaiserslautern, Germany
| | - Angelika Bröer
- From the Research School of Biology, The Australian National University, Canberra, ACT 0200, Australia and
| | - Joachim W. Deitmer
- the Abteilung für Allgemeine Zoologie, Fachbereich Biologie, Technische Universität Kaiserslautern, Erwin-Schrödinger Strasse, D-67653 Kaiserslautern, Germany
| |
Collapse
|
39
|
Shaffer PL, Goehring A, Shankaranarayanan A, Gouaux E. Structure and mechanism of a Na+-independent amino acid transporter. Science 2009; 325:1010-4. [PMID: 19608859 PMCID: PMC2851542 DOI: 10.1126/science.1176088] [Citation(s) in RCA: 259] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Amino acid, polyamine, and organocation (APC) transporters are secondary transporters that play essential roles in nutrient uptake, neurotransmitter recycling, ionic homeostasis, and regulation of cell volume. Here, we present the crystal structure of apo-ApcT, a proton-coupled broad-specificity amino acid transporter, at 2.35 angstrom resolution. The structure contains 12 transmembrane helices, with the first 10 consisting of an inverted structural repeat of 5 transmembrane helices like the leucine transporter LeuT. The ApcT structure reveals an inward-facing, apo state and an amine moiety of lysine-158 located in a position equivalent to the sodium ion site Na2 of LeuT. We propose that lysine-158 is central to proton-coupled transport and that the amine group serves the same functional role as the Na2 ion in LeuT, thus demonstrating common principles among proton- and sodium-coupled transporters.
Collapse
Affiliation(s)
- Paul L Shaffer
- Vollum Institute, Oregon Health and Science University, 3181 Southwest Sam Jackson Park Road, Portland, OR 97239, USA
| | | | | | | |
Collapse
|
40
|
Hundal HS, Taylor PM. Amino acid transceptors: gate keepers of nutrient exchange and regulators of nutrient signaling. Am J Physiol Endocrinol Metab 2009; 296:E603-13. [PMID: 19158318 PMCID: PMC2670634 DOI: 10.1152/ajpendo.91002.2008] [Citation(s) in RCA: 230] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2008] [Accepted: 01/13/2009] [Indexed: 11/22/2022]
Abstract
Amino acid transporters at the surface of cells are in an ideal location to relay nutritional information, as well as nutrients themselves, to the cell interior. These transporters are able to modulate signaling downstream of intracellular amino acid receptors by regulating intracellular amino acid concentrations through processes of coupled transport. The concept of dual-function amino acid transporter/receptor (or "transceptor") proteins is well established in primitive eukaryotes such as yeast, where detection of extracellular amino acid deficiency leads to upregulation of proteins involved in biosynthesis and transport of the deficient amino acid(s). The evolution of the "extracellular milieu" and nutrient-regulated endocrine controls in higher eukaryotes, alongside their frequent inability to synthesize all proteinaceous amino acids (and, hence, the requirement for indispensable amino acids in their diet), appears to have lessened the priority of extracellular amino acid sensing as a stimulus for metabolic signals. Nevertheless, recent studies of amino acid transporters in flies and mammalian cell lines have revealed perhaps unanticipated "echoes" of these transceptor functions, which are revealed by cellular stresses (notably starvation) or gene modification/silencing. APC-transporter superfamily members, including slimfast, path, and SNAT2 all appear capable of sensing and signaling amino acid availability to the target of rapamycin (TOR) pathway, possibly through PI 3-kinase-dependent mechanisms. We hypothesize (by extrapolation from knowledge of the yeast Ssy1 transceptor) that, at least for SNAT2, the transceptor discriminates between extracellular and intracellular amino acid stimuli when evoking a signal.
Collapse
Affiliation(s)
- Harinder S Hundal
- Division of Molecular Physiology, Sir James Black Centre, College of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK.
| | | |
Collapse
|
41
|
Nickel A, Kottra G, Schmidt G, Danier J, Hofmann T, Daniel H. Characteristics of transport of selenoamino acids by epithelial amino acid transporters. Chem Biol Interact 2009; 177:234-41. [DOI: 10.1016/j.cbi.2008.09.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2008] [Revised: 09/10/2008] [Accepted: 09/11/2008] [Indexed: 11/26/2022]
|
42
|
Abstract
Near complete reabsorption of filtered amino acids is a main specialized transport function of the kidney proximal tubule. This evolutionary conserved task is carried out by a subset of luminal and basolateral transporters that together form the transcellular amino acid transport machinery similar to that of small intestine. A number of other amino acid transporters expressed in the basolateral membrane of proximal kidney tubule cells subserve either specialized metabolic functions, such as the production of ammonium, or are part of the cellular housekeeping equipment. A new finding is that the luminal Na(+)-dependent neutral amino acid transporters of the SLC6 family require an associated protein for their surface expression as shown for the Hartnup transporter B(0)AT1 (SLC6A19) and suggested for the L: -proline transporter SIT1 (IMINO(B), SLC6A20) and for B(0)AT3 (XT2, SLC6A18). This accessory subunit called collectrin (TMEM27) is homologous to the transmembrane anchor region of the renin-angiotensin system enzyme ACE2 that we have shown to function in small intestine as associated subunit of the luminal SLC6 transporters B(0)AT1 and SIT1. Some mutations of B(0)AT1 differentially interact with these accessory subunits, providing an explanation for differential intestinal phenotypes among Hartnup patients. The basolateral efflux of numerous amino acids from kidney tubular cells is mediated by heteromeric amino acid transporters that function as obligatory exchangers. Thus, other transporters within the same membrane need to mediate the net efflux of exchange substrates, controlling thereby the net basolateral amino transport and thus the intracellular amino acid concentration.
Collapse
|
43
|
He L, Vasiliou K, Nebert DW. Analysis and update of the human solute carrier (SLC) gene superfamily. Hum Genomics 2009; 3:195-206. [PMID: 19164095 PMCID: PMC2752037 DOI: 10.1186/1479-7364-3-2-195] [Citation(s) in RCA: 247] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2008] [Accepted: 10/02/2008] [Indexed: 12/20/2022] Open
Abstract
The solute-carrier gene (SLC) superfamily encodes membrane-bound transporters. The SLC superfamily comprises 55 gene families having at least 362 putatively functional protein-coding genes. The gene products include passive transporters, symporters and antiporters, located in all cellular and organelle membranes, except, perhaps, the nuclear membrane. Transport substrates include amino acids and oligopeptides, glucose and other sugars, inorganic cations and anions (H(+), HCO(3)(-), Cl(-), Na(+), K(+), Ca(2+), Mg(2+), PO(4)(3-), HPO(4)(2-), H(2)PO(4)(-), SO(4)(2-), C(2)O(4)(2-), OH(-), CO(3)(2-)), bile salts, carboxylate and other organic anions, acetyl coenzyme A, essential metals, biogenic amines, neurotransmitters, vitamins, fatty acids and lipids, nucleosides, ammonium, choline, thyroid hormone and urea. Contrary to gene nomenclature commonly assigned on the basis of evolutionary divergence (http://www.genenames.org/), the SLC gene superfamily has been named based largely on transporter function by proteins having multiple transmembrane domains. Whereas all the transporters exist for endogenous substrates, it is likely that drugs, non-essential metals and many other environmental toxicants are able to 'hitch-hike' on one or another of these transporters, thereby enabling these moieties to enter (or leave) the cell. Understanding and characterising the functions of these transporters is relevant to medicine, genetics, developmental biology, pharmacology and cancer chemotherapy.
Collapse
Affiliation(s)
- Lei He
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA 02114, USA
| | - Konstandinos Vasiliou
- Molecular Toxicology and Environmental Health Sciences Program, Department of Pharmaceutical Sciences, University of Colorado Health Sciences Center, Denver, CO 80262, USA
| | - Daniel W Nebert
- Department of Environmental Health and Center for Environmental Genetics (CEG), University of Cincinnati Medical Center, Cincinnati, OH 45267-0056, USA
| |
Collapse
|
44
|
Bröer S, Bailey CG, Kowalczuk S, Ng C, Vanslambrouck JM, Rodgers H, Auray-Blais C, Cavanaugh JA, Bröer A, Rasko JEJ. Iminoglycinuria and hyperglycinuria are discrete human phenotypes resulting from complex mutations in proline and glycine transporters. J Clin Invest 2008. [PMID: 19033659 DOI: 10.1172/jci3662536625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023] Open
Abstract
Iminoglycinuria (IG) is an autosomal recessive abnormality of renal transport of glycine and the imino acids proline and hydroxyproline, but the specific genetic defect(s) have not been determined. Similarly, although the related disorder hyperglycinuria (HG) without iminoaciduria has been attributed to heterozygosity of a putative defective glycine, proline, and hydroxyproline transporter, confirming the underlying genetic defect(s) has been difficult. Here we applied a candidate gene sequencing approach in 7 families first identified through newborn IG screening programs. Both inheritance and functional studies identified the gene encoding the proton amino acid transporter SLC36A2 (PAT2) as the major gene responsible for IG in these families, and its inheritance was consistent with a classical semidominant pattern in which 2 inherited nonfunctional alleles conferred the IG phenotype, while 1 nonfunctional allele was sufficient to confer the HG phenotype. Mutations in SLC36A2 that retained residual transport activity resulted in the IG phenotype when combined with mutations in the gene encoding the imino acid transporter SLC6A20 (IMINO). Additional mutations were identified in the genes encoding the putative glycine transporter SLC6A18 (XT2) and the neutral amino acid transporter SLC6A19 (B0AT1) in families with either IG or HG, suggesting that mutations in the genes encoding these transporters may also contribute to these phenotypes. In summary, although recognized as apparently simple Mendelian disorders, IG and HG exhibit complex molecular explanations depending on a major gene and accompanying modifier genes.
Collapse
Affiliation(s)
- Stefan Bröer
- School of Biochemistry and Molecular Biology, Australian National University, Canberra, Australian Capital Territory, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Bröer S, Bailey CG, Kowalczuk S, Ng C, Vanslambrouck JM, Rodgers H, Auray-Blais C, Cavanaugh JA, Bröer A, Rasko JEJ. Iminoglycinuria and hyperglycinuria are discrete human phenotypes resulting from complex mutations in proline and glycine transporters. J Clin Invest 2008; 118:3881-92. [PMID: 19033659 DOI: 10.1172/jci36625] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2008] [Accepted: 10/01/2008] [Indexed: 11/17/2022] Open
Abstract
Iminoglycinuria (IG) is an autosomal recessive abnormality of renal transport of glycine and the imino acids proline and hydroxyproline, but the specific genetic defect(s) have not been determined. Similarly, although the related disorder hyperglycinuria (HG) without iminoaciduria has been attributed to heterozygosity of a putative defective glycine, proline, and hydroxyproline transporter, confirming the underlying genetic defect(s) has been difficult. Here we applied a candidate gene sequencing approach in 7 families first identified through newborn IG screening programs. Both inheritance and functional studies identified the gene encoding the proton amino acid transporter SLC36A2 (PAT2) as the major gene responsible for IG in these families, and its inheritance was consistent with a classical semidominant pattern in which 2 inherited nonfunctional alleles conferred the IG phenotype, while 1 nonfunctional allele was sufficient to confer the HG phenotype. Mutations in SLC36A2 that retained residual transport activity resulted in the IG phenotype when combined with mutations in the gene encoding the imino acid transporter SLC6A20 (IMINO). Additional mutations were identified in the genes encoding the putative glycine transporter SLC6A18 (XT2) and the neutral amino acid transporter SLC6A19 (B0AT1) in families with either IG or HG, suggesting that mutations in the genes encoding these transporters may also contribute to these phenotypes. In summary, although recognized as apparently simple Mendelian disorders, IG and HG exhibit complex molecular explanations depending on a major gene and accompanying modifier genes.
Collapse
Affiliation(s)
- Stefan Bröer
- School of Biochemistry and Molecular Biology, Australian National University, Canberra, Australian Capital Territory, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Gosset G, Satre M, Blaive B, Clément JL, Martin JB, Culcasi M, Pietri S. Investigation of subcellular acidic compartments using α-aminophosphonate 31P nuclear magnetic resonance probes. Anal Biochem 2008; 380:184-94. [DOI: 10.1016/j.ab.2008.05.052] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2008] [Revised: 05/19/2008] [Accepted: 05/30/2008] [Indexed: 10/22/2022]
|
47
|
Martín-Venegas R, Geraert P, Ferrer R. Partial Na+ Dependence of dl-2-Hydroxy-4-(Methylthio)Butanoic Acid Uptake in the Chicken Small Intestine. Poult Sci 2008; 87:1392-4. [DOI: 10.3382/ps.2007-00218] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
48
|
Bröer S. Apical transporters for neutral amino acids: physiology and pathophysiology. Physiology (Bethesda) 2008; 23:95-103. [PMID: 18400692 DOI: 10.1152/physiol.00045.2007] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Absorption of amino acids in kidney and intestine involves a variety of transporters for different groups of amino acids. This is illustrated by inherited disorders of amino acid absorption, such as Hartnup disorder, cystinuria, iminoglycinuria, dicarboxylic aminoaciduria, and lysinuric protein intolerance, affecting separate groups of amino acids. Recent advances in the molecular identification of apical neutral amino acid transporters has shed a light on the molecular basis of Hartnup disorder and iminoglycinuria.
Collapse
Affiliation(s)
- Stefan Bröer
- School of Biochemistry and Molecular Biology, Australian National University, Canberra, Australia.
| |
Collapse
|
49
|
de Vogel-van den Bosch HM, Bünger M, de Groot PJ, Bosch-Vermeulen H, Hooiveld GJEJ, Müller M. PPARalpha-mediated effects of dietary lipids on intestinal barrier gene expression. BMC Genomics 2008; 9:231. [PMID: 18489776 PMCID: PMC2408604 DOI: 10.1186/1471-2164-9-231] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2007] [Accepted: 05/19/2008] [Indexed: 12/31/2022] Open
Abstract
Background The selective absorption of nutrients and other food constituents in the small intestine is mediated by a group of transport proteins and metabolic enzymes, often collectively called 'intestinal barrier proteins'. An important receptor that mediates the effects of dietary lipids on gene expression is the peroxisome proliferator-activated receptor alpha (PPARα), which is abundantly expressed in enterocytes. In this study we examined the effects of acute nutritional activation of PPARα on expression of genes encoding intestinal barrier proteins. To this end we used triacylglycerols composed of identical fatty acids in combination with gene expression profiling in wild-type and PPARα-null mice. Treatment with the synthetic PPARα agonist WY14643 served as reference. Results We identified 74 barrier genes that were PPARα-dependently regulated 6 hours after activation with WY14643. For eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA) and oleic acid (OA) these numbers were 46, 41, and 19, respectively. The overlap between EPA-, DHA-, and WY14643-regulated genes was considerable, whereas OA treatment showed limited overlap. Functional implications inferred form our data suggested that nutrient-activated PPARα regulated transporters and phase I/II metabolic enzymes were involved in a) fatty acid oxidation, b) cholesterol, glucose, and amino acid transport and metabolism, c) intestinal motility, and d) oxidative stress defense. Conclusion We identified intestinal barrier genes that were PPARα-dependently regulated after acute activation by fatty acids. This knowledge provides a better understanding of the impact dietary fat has on the barrier function of the gut, identifies PPARα as an important factor controlling this key function, and underscores the importance of PPARα for nutrient-mediated gene regulation in intestine.
Collapse
Affiliation(s)
- Heleen M de Vogel-van den Bosch
- Nutrition, Metabolism and Genomics group, Division of Human Nutrition, Wageningen University, PO Box 8129, NL-6700EV, Wageningen, the Netherlands.
| | | | | | | | | | | |
Collapse
|
50
|
Zhang Z, Gameiro A, Grewer C. Highly conserved asparagine 82 controls the interaction of Na+ with the sodium-coupled neutral amino acid transporter SNAT2. J Biol Chem 2008; 283:12284-92. [PMID: 18319257 PMCID: PMC2430088 DOI: 10.1074/jbc.m706774200] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2007] [Revised: 03/03/2008] [Indexed: 11/06/2022] Open
Abstract
The neutral amino acid transporter 2 (SNAT2), which belongs to the SLC38 family of solute transporters, couples the transport of amino acid to the cotransport of one Na(+) ion into the cell. Several polar amino acids are highly conserved within the SLC38 family. Here, we mutated three of these conserved amino acids, Asn(82) in the predicted transmembrane domain 1 (TMD1), Tyr(337) in TMD7, and Arg(374) in TMD8; and we studied the functional consequences of these modifications. The mutation of N82A virtually eliminated the alanine-induced transport current, as well as amino acid uptake by SNAT2. In contrast, the mutations Y337A and R374Q did not abolish amino acid transport. The K(m) of SNAT2 for its interaction with Na(+), K(Na(+)), was dramatically reduced by the N82A mutation, whereas the more conservative mutation N82S resulted in a K(Na(+)) that was in between SNAT2(N82A) and SNAT2(WT). These results were interpreted as a reduction of Na(+) affinity caused by the Asn(82) mutations, suggesting that these mutations interfere with the interaction of SNAT2 with the sodium ion. As a consequence of this dramatic reduction in Na(+) affinity, the apparent K(m) of SNAT2(N82A) for alanine was increased 27-fold compared with that of SNAT2(WT). Our results demonstrate a direct or indirect involvement of Asn(82) in Na(+) coordination by SNAT2. Therefore, we predict that TMD1 is crucial for the function of SLC38 transporters and that of related families.
Collapse
Affiliation(s)
- Zhou Zhang
- Department of Physiology and Biophysics, University of Miami School of Medicine, Miami, Florida 33136, USA
| | | | | |
Collapse
|