1
|
Oh J, Cha J, Choi S. Identification of Novel Genetic Variants and Food Intake Factors Associated with Type 2 Diabetes in South Korean Adults, Using an Illness-Death Model. Int J Mol Sci 2025; 26:2597. [PMID: 40141237 PMCID: PMC11942363 DOI: 10.3390/ijms26062597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
Type 2 diabetes (T2D) is a prevalent chronic disease in the Korean population, influenced by lifestyle, dietary habits, and genetics. This study aimed to identify the effects of food intake and genetic factors on T2D progression in Korean adults using a multi-state illness-death model. We analyzed three transition models: normal glucose tolerance (NGT) to prediabetes (PD), NGT to T2D, and PD to T2D. We first identified dietary patterns significantly associated with each transition, using multivariate Cox proportional hazards models. Then, we assessed the impact of single-nucleotide polymorphisms (SNPs) on each transition, incorporating these dietary patterns as covariates. Our analysis revealed significant associations between the identified dietary patterns and the risk of PD and T2D incidence among individuals with NGT. We also identified novel genetic variants associated with disease progression: two SNPs (rs4607517 in Glucokinase [GCK] and rs758982 in Calcium/Calmodulin-Dependent Protein Kinase II Beta [CAMK2B]) in the NGT to PD model, and eight SNPs in the NGT to T2D model, including variants in the Zinc Finger Protein 106 (ZNF106), PTOV1 Extended AT-Hook Containing Adaptor Protein (PTOV1), Proprotein Convertase Subtilisin/Kexin Type 2 (PCSK2), Forkhead Box D2 (FOXD2), Solute Carrier Family 38 Member 7 (SLC38A7), and Neuronal Growth Regulator 1 (NEGR1) genes. Functional annotation analysis using ANNOVAR revealed that rs4607517 (GCK) and rs59595912 (PTOV1) exhibited high Combined Annotation-Dependent Depletion (CADD) and Deleterious Annotation of Genetic Variants using Neural Networks (DANN) scores, suggesting potential pathogenicity and providing a functional basis for their association with T2D progression. Integrating dietary and genetic factors with a multi-state model, this comprehensive approach offers valuable insights into T2D development and highlights potential targets for prevention and personalized interventions.
Collapse
Affiliation(s)
- Jeongmin Oh
- Department of Applied Mathematics, College of Science and Convergence Technology, Hanyang University, 55 Hanyang-daehak-ro, Sangnok-gu, Ansan 15588, Republic of Korea;
| | - Junho Cha
- Department of Applied Artificial Intelligence, College of Computing, Hanyang University, 55 Hanyang-daehak-ro, Sangnok-gu, Ansan 15588, Republic of Korea;
| | - Sungkyoung Choi
- Department of Applied Mathematics, College of Science and Convergence Technology, Hanyang University, 55 Hanyang-daehak-ro, Sangnok-gu, Ansan 15588, Republic of Korea;
- Department of Applied Artificial Intelligence, College of Computing, Hanyang University, 55 Hanyang-daehak-ro, Sangnok-gu, Ansan 15588, Republic of Korea;
- Department of Mathematical Data Science, College of Science and Convergence Technology, Hanyang University, 55 Hanyang-daehak-ro, Sangnok-gu, Ansan 15588, Republic of Korea
| |
Collapse
|
2
|
Dzyubenko E, Hermann DM. Neuroglia and extracellular matrix molecules. HANDBOOK OF CLINICAL NEUROLOGY 2025; 209:197-211. [PMID: 40122625 DOI: 10.1016/b978-0-443-19104-6.00010-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
This chapter provides a comprehensive overview of the roles of astrocytes, microglia, and the extracellular matrix (ECM) in regulating neuroplasticity and maintaining brain homeostasis. Astrocytes provide essential metabolic support to neurons, regulate synapse development, support neuroplasticity mechanisms, and modulate neurotransmission. Microglia, the resident immune cells of the brain, play a critical role in neuroinflammatory responses and homeostatic brain regulation by modulating synapse formation and pruning. The extracellular space (ECS) mediates intercellular interactions, provides a highly regulated environment for intercellular communication, and is filled with ECM molecules. Proteoglycans and polysaccharides of the ECM play a vital role not only in brain development but also in brain function throughout life. In the injured brain, neuroplasticity and regeneration can be bidirectionally regulated as a result of the interplay between ECM, astrocytes, and microglia. The modulation of synaptic strength, structural remodeling, and modification of intrinsic neuronal properties are among the central mechanisms that contribute to neuronal plasticity in health and disease. We believe that the understanding of ECM-glia interactions and their role in neuroplasticity regulation is key to the development of novel therapeutic strategies in neurologic disorders.
Collapse
Affiliation(s)
- Egor Dzyubenko
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| | - Dirk M Hermann
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
3
|
Chakraborty A, Patnaik J, Sinha A, Parida N, Parija T, Patnaik S. In silico analysis shows slc1a4 as a potential target of hsa-mir-133a for regulating glutamine metabolism in gastric cancer. Int J Biol Macromol 2024; 282:136974. [PMID: 39486714 DOI: 10.1016/j.ijbiomac.2024.136974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 10/24/2024] [Accepted: 10/25/2024] [Indexed: 11/04/2024]
Abstract
Cutting-edge research has spotlighted glutamine metabolism as a promising therapeutic target in managing gastric cancer. This investigation highlights the upregulated glutamine transporters by leveraging clinical data from the TCGA Database and the expression analysis of the transcriptome profile of stomach adenocarcinoma (STAD) patients. Notably, it identifies SLC1A4 as a potential glutamine transporter in STAD. The screening of human miRNAs conducted using the TargetScan database, and the subsequent docking analysis present multiple miRNAs with the potential of being explored as therapeutic agents. By integrating transcriptome profiling, miRNA screening, and molecular docking, this study reveals, for the first time, the potential of hsa-mir-133a-1 in targeting slc1a4, along with its known target mTOR, in stomach cancer. The myriad interactions that can be regulated by this silencing mechanism are anticipated to ultimately reduce glutamine uptake in STAD. This study provides compelling evidence of glutamine transport via SLC1A4 in stomach cancer and delves into how it might impact mTOR and some of its pivotal downstream molecules. Considering these findings, novel therapeutic strategies can be devised to further enhance existing methods for combating gastric cancer.
Collapse
Affiliation(s)
- Averi Chakraborty
- School of Biotechnology, KIIT University, Campus-XI, Bhubaneswar, 751024, India
| | - Jayasree Patnaik
- School of Biotechnology, KIIT University, Campus-XI, Bhubaneswar, 751024, India
| | - Anupriya Sinha
- School of Biotechnology, KIIT University, Campus-XI, Bhubaneswar, 751024, India
| | - Nandita Parida
- School of Biotechnology, KIIT University, Campus-XI, Bhubaneswar, 751024, India
| | - Tithi Parija
- School of Biotechnology, KIIT University, Campus-XI, Bhubaneswar, 751024, India.
| | - Srinivas Patnaik
- School of Biotechnology, KIIT University, Campus-XI, Bhubaneswar, 751024, India.
| |
Collapse
|
4
|
Hosseini K, Cediel-Ulloa A, AL-Sabri MH, Forsby A, Fredriksson R. Assessing the Neurodevelopmental Impact of Fluoxetine, Citalopram, and Paroxetine on Neural Stem Cell-Derived Neurons. Pharmaceuticals (Basel) 2024; 17:1392. [PMID: 39459031 PMCID: PMC11510426 DOI: 10.3390/ph17101392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Many pregnant women globally suffer from depression and are routinely prescribed selective serotonin reuptake inhibitors (SSRIs). These drugs function by blocking the re-uptake of serotonin by the serotonin transporter (SERT) into neurons, resulting in its accumulation in the presynaptic cleft. Despite a large amount of research suggesting a potential link to neurodevelopmental disorders in children whose mothers took these drugs during pregnancy, their possible adverse effects are still debated, and results are contradictory. On the other hand, there is an immediate need for improved cell-based models for developmental neurotoxicity studies (DNT) to minimize the use of animals in research. METHODS In this study, we aimed to assess the effects of clinically relevant concentrations of paroxetine (PAR), fluoxetine (FLX), and citalopram (CIT)-on maturing neurons derived from human neural stem cells using multiple endpoints. RESULTS Although none of the tested concentrations of FLX, CIT, or PAR significantly affected cell viability, FLX (10 µM) exhibited the highest reduction in viability compared to the other drugs. Regarding neurite outgrowth, CIT did not have a significant effect. However, FLX (10 µM) significantly reduced both mean neurite outgrowth and mean processes, PAR significantly reduced mean processes, and showed a trend of dysregulation of multiple genes associated with neuronal development at therapeutic-relevant serum concentrations. CONCLUSIONS Transcriptomic data and uptake experiments found no SERT activity in the system, suggesting that the adverse effects of FLX and PAR are independent of SERT.
Collapse
Affiliation(s)
- Kimia Hosseini
- Department of Pharmaceutical Bioscience, Uppsala University, 751 24 Uppsala, Sweden (R.F.)
| | - Andrea Cediel-Ulloa
- Department of Organismal Biology, Uppsala University, 752 36 Uppsala, Sweden
| | - Mohamed H. AL-Sabri
- Department of Pharmaceutical Bioscience, Uppsala University, 751 24 Uppsala, Sweden (R.F.)
- Department of Surgical Science, Functional Pharmacology and Neuroscience, Uppsala University, 751 24 Uppsala, Sweden
| | - Anna Forsby
- Department of Biochemistry and Biophysics, Stockholm University, 106 91 Stockholm, Sweden
| | - Robert Fredriksson
- Department of Pharmaceutical Bioscience, Uppsala University, 751 24 Uppsala, Sweden (R.F.)
| |
Collapse
|
5
|
Xu J, Gu J, Pei W, Zhang Y, Wang L, Gao J. The role of lysosomal membrane proteins in autophagy and related diseases. FEBS J 2024; 291:3762-3785. [PMID: 37221945 DOI: 10.1111/febs.16820] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/12/2023] [Accepted: 05/15/2023] [Indexed: 05/25/2023]
Abstract
As a self-degrading and highly conserved survival mechanism, autophagy plays an important role in maintaining cell survival and recycling. The discovery of autophagy-related (ATG) genes has revolutionized our understanding of autophagy. Lysosomal membrane proteins (LMPs) are important executors of lysosomal function, and increasing evidence has demonstrated their role in the induction and regulation of autophagy. In addition, the functional dysregulation of the process mediated by LMPs at all stages of autophagy is closely related to neurodegenerative diseases and cancer. Here, we review the role of LMPs in autophagy, focusing on their roles in vesicle nucleation, vesicle elongation and completion, the fusion of autophagosomes and lysosomes, and degradation, as well as their broad association with related diseases.
Collapse
Affiliation(s)
- Jiahao Xu
- Department of Endocrinology and Genetic Metabolism, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- Department of Endocrinology and Genetic Metabolism, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- Anhui Province Key Laboratory of Biological Macro-Molecules Research, Wannan Medical College, Wuhu, China
- School of Clinical Medicine, Wannan Medical College, Wuhu, China
| | - Jing Gu
- Department of Endocrinology and Genetic Metabolism, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- Department of Endocrinology and Genetic Metabolism, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- Anhui Province Key Laboratory of Biological Macro-Molecules Research, Wannan Medical College, Wuhu, China
| | - Wenjun Pei
- Anhui Province Key Laboratory of Biological Macro-Molecules Research, Wannan Medical College, Wuhu, China
- Department of Biochemistry and Molecular Biology, Wannan Medical College, Wuhu, China
| | - Yao Zhang
- Anhui Province Key Laboratory of Biological Macro-Molecules Research, Wannan Medical College, Wuhu, China
- Department of Biochemistry and Molecular Biology, Wannan Medical College, Wuhu, China
| | - Lizhuo Wang
- Anhui Province Key Laboratory of Biological Macro-Molecules Research, Wannan Medical College, Wuhu, China
- Department of Biochemistry and Molecular Biology, Wannan Medical College, Wuhu, China
| | - Jialin Gao
- Department of Endocrinology and Genetic Metabolism, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- Department of Endocrinology and Genetic Metabolism, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- Anhui Province Key Laboratory of Biological Macro-Molecules Research, Wannan Medical College, Wuhu, China
- Anhui Provincial College Key Laboratory of Non-coding RNA Transformation Research on Critical Diseases, Wannan Medical College, Wuhu, China
| |
Collapse
|
6
|
Vázquez-Durán DL, Ortega A, Rodríguez A. Amino Acid Transporters Proteins Involved in the Glutamate-Glutamine Cycle and Their Alterations in Murine Models of Alzheimer's Disease. Mol Neurobiol 2024; 61:6077-6088. [PMID: 38273046 DOI: 10.1007/s12035-024-03966-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 01/16/2024] [Indexed: 01/27/2024]
Abstract
The brain's ability to integrate external stimuli and generate responses is highly complex. While these mechanisms are not completely understood, current evidence suggests that alterations in cellular metabolism and microenvironment are involved in some dysfunctions as complex as Alzheimer's disease. This pathology courses with defects in the establishment of chemical synapses, which is dependent on the production and supply of neurotransmitters like glutamate and its recycling through the glutamate-glutamine cycle. Alterations in the expression and function of the amino acid transporters proteins involved in this cycle have recently been reported in different stages of Alzheimer's disease. Most of these data come from patients in advanced stages of the disease or post-mortem, due to the ethical and technical limitations of human studies. Therefore, genetically modified mouse models have been an excellent tool to analyze metabolic and even behavioral parameters that are very similar to those that develop in Alzheimer's disease, even at presymptomatic stages. Hence, this paper analyzes the role of glutamate metabolism and its intercellular trafficking in excitatory synapses from different approaches using transgenic mouse models; such an analysis will contribute to our present understanding of AD.
Collapse
Affiliation(s)
| | - Arturo Ortega
- Departamento de Toxicología, Cinvestav- IPN, Mexico City, México
| | - Angelina Rodríguez
- Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Santiago de Querétaro, México.
| |
Collapse
|
7
|
Zhang D, Hua Z, Li Z. The role of glutamate and glutamine metabolism and related transporters in nerve cells. CNS Neurosci Ther 2024; 30:e14617. [PMID: 38358002 PMCID: PMC10867874 DOI: 10.1111/cns.14617] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/15/2023] [Accepted: 01/10/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND Glutamate and glutamine are the most abundant amino acids in the blood and play a crucial role in cell survival in the nervous system. Various transporters found in cell and mitochondrial membranes, such as the solute carriers (SLCs) superfamily, are responsible for maintaining the balance of glutamate and glutamine in the synaptic cleft and within cells. This balance affects the metabolism of glutamate and glutamine as non-essential amino acids. AIMS This review aims to provide an overview of the transporters and enzymes associated with glutamate and glutamine in neuronal cells. DISCUSSION We delve into the function of glutamate and glutamine in the nervous system by discussing the transporters involved in the glutamate-glutamine cycle and the key enzymes responsible for their mutual conversion. Additionally, we highlight the role of glutamate and glutamine as carbon and nitrogen donors, as well as their significance as precursors for the synthesis of reduced glutathione (GSH). CONCLUSION Glutamate and glutamine play a crucial role in the brain due to their special effects. It is essential to focus on understanding glutamate and glutamine metabolism to comprehend the physiological behavior of nerve cells and to treat nervous system disorders and cancer.
Collapse
Affiliation(s)
- Dongyang Zhang
- Department of PediatricsShengjing Hospital of China Medical UniversityShenyangLiaoningChina
- Medical Research Center, Liaoning Key Laboratory of Research and Application of Animal Models for Environment and Metabolic DiseasesShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Zhongyan Hua
- Department of PediatricsShengjing Hospital of China Medical UniversityShenyangLiaoningChina
- Medical Research Center, Liaoning Key Laboratory of Research and Application of Animal Models for Environment and Metabolic DiseasesShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Zhijie Li
- Department of PediatricsShengjing Hospital of China Medical UniversityShenyangLiaoningChina
- Medical Research Center, Liaoning Key Laboratory of Research and Application of Animal Models for Environment and Metabolic DiseasesShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| |
Collapse
|
8
|
Mylvaganam S, Freeman SA. The resolution of phagosomes. Immunol Rev 2023; 319:45-64. [PMID: 37551912 DOI: 10.1111/imr.13260] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/18/2023] [Indexed: 08/09/2023]
Abstract
Phagocytosis is a fundamental immunobiological process responsible for the removal of harmful particulates. While the number of phagocytic events achieved by a single phagocyte can be remarkable, exceeding hundreds per day, the same phagocytic cells are relatively long-lived. It should therefore be obvious that phagocytic meals must be resolved in order to maintain the responsiveness of the phagocyte and to avoid storage defects. In this article, we discuss the mechanisms involved in the resolution process, including solute transport pathways and membrane traffic. We describe how products liberated in phagolysosomes support phagocyte metabolism and the immune response. We also speculate on mechanisms involved in the redistribution of phagosomal metabolites back to circulation. Finally, we highlight the pathologies owed to impaired phagosome resolution, which range from storage disorders to neurodegenerative diseases.
Collapse
Affiliation(s)
- Sivakami Mylvaganam
- Program in Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Spencer A Freeman
- Program in Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
9
|
Taurino G, Chiu M, Bianchi MG, Griffini E, Bussolati O. The SLC38A5/SNAT5 amino acid transporter: from pathophysiology to pro-cancer roles in the tumor microenvironment. Am J Physiol Cell Physiol 2023; 325:C550-C562. [PMID: 37458433 DOI: 10.1152/ajpcell.00169.2023] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/11/2023] [Accepted: 07/11/2023] [Indexed: 08/09/2023]
Abstract
SLC38A5/SNAT5 is a system N transporter that can mediate net inward or outward transmembrane fluxes of neutral amino acids coupled with Na+ (symport) and H+ (antiport). Its preferential substrates are not only amino acids with side chains containing amide (glutamine and asparagine) or imidazole (histidine) groups, but also serine, glycine, and alanine are transported by the carrier. Expressed in the pancreas, intestinal tract, brain, liver, bone marrow, and placenta, it is regulated at mRNA and protein levels by mTORC1 and WNT/β-catenin pathways, and it is sensitive to pH, nutritional stress, inflammation, and hypoxia. SNAT5 expression has been found to be altered in pathological conditions such as chronic inflammatory diseases, gestational complications, chronic metabolic acidosis, and malnutrition. Growing experimental evidence shows that SNAT5 is overexpressed in several types of cancer cells. Moreover, recently published results indicate that SNAT5 expression in stromal cells can support the metabolic exchanges occurring in the tumor microenvironment of asparagine-auxotroph tumors. We review the functional role of the SNAT5 transporter in pathophysiology and propose that, due to its peculiar operational and regulatory features, SNAT5 may play important pro-cancer roles when expressed either in neoplastic or in stromal cells of glutamine-auxotroph tumors.NEW & NOTEWORTHY The transporter SLC38A5/SNAT5 provides net influx or efflux of glutamine, asparagine, and serine. These amino acids are of particular metabolic relevance in several conditions. Changes in transporter expression or activity have been described in selected types of human cancers, where SNAT5 can mediate amino acid exchanges between tumor and stromal cells, thus providing a potential therapeutic target. This is the first review that recapitulates the characteristics and roles of the transporter in physiology and pathology.
Collapse
Affiliation(s)
- Giuseppe Taurino
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Parma, Italy
- MRH-Microbiome Research Hub, University of Parma, Parma, Italy
| | - Martina Chiu
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Massimiliano G Bianchi
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Parma, Italy
- MRH-Microbiome Research Hub, University of Parma, Parma, Italy
| | - Erika Griffini
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Ovidio Bussolati
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Parma, Italy
- MRH-Microbiome Research Hub, University of Parma, Parma, Italy
| |
Collapse
|
10
|
Aboragah AA, Sherlock DN, Wichasit N, Loor JJ. Abundance of proteins and genes associated with nutrient signaling, protein turnover, and transport of amino acids and glucose in fetuses from lactating Holstein cows. Res Vet Sci 2023; 161:69-76. [PMID: 37321013 DOI: 10.1016/j.rvsc.2023.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/15/2023] [Accepted: 05/25/2023] [Indexed: 06/17/2023]
Abstract
Availability of nutrients in maternal circulation and abundance of nutrient transporters, metabolic enzymes, and nutrient-responsive proteins in fetal tissues coordinate growth. To begin characterizing these mechanisms, we evaluated the abundance of nutrient signaling genes and proteins in bovine fetal tissues. Liver, entire intestine, and semitendinosus muscle were harvested from fetuses (4 female, 2 male) collected at slaughter from 6 clinically-healthy multiparous Holstein dairy cows (167 ± 7 days in milk, 37 ± 6 kg milk/d, 100 ± 3 d gestation). Data were analyzed using PROC MIXED in SAS 9.4. Among proteins measured, abundance of the amino acid (AA) utilization and insulin signaling proteins p-AKT and p-mTOR was greater (P < 0.01) in liver and intestine. The abundance of p-EEF2 (translation elongation) and SLC2A4 (glucose uptake) was greater (P < 0.05) in liver relative to intestine and muscle suggesting this organ has a greater capacity for anabolic processes. In contrast, among mTOR signaling genes, the abundance of IRS1 was greatest (P < 0.01) in muscle and lowest in the intestine, whereas, abundance of AKT1 and mTOR was greater (P < 0.01) in intestine and muscle than liver. Abundance of the protein degradation-related genes UBA1, UBE2G1, and TRIM63 was greater (P < 0.01) in muscle than intestine and liver. Among nutrient transporters, abundance of glucose transporters SLC5A1 and SLC2A2 was greatest (P < 0.01) in the intestine than liver and muscle. Several AA transporters had greater (P < 0.01) abundance in the intestine or liver compared with muscle. Overall, these molecular analyses highlighted important biological differences on various aspects of metabolism in fetal tissues.
Collapse
Affiliation(s)
- Ahmad A Aboragah
- Department of Animal Sciences, University of Illinois, Urbana 61801, USA; Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | | | - Nithat Wichasit
- Department of Animal Sciences, University of Illinois, Urbana 61801, USA; Department of Agricultural Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Juan J Loor
- Department of Animal Sciences, University of Illinois, Urbana 61801, USA; Division of Nutritional Sciences, University of Illinois, Urbana 61801, USA.
| |
Collapse
|
11
|
Dzyubenko E, Hermann DM. Role of glia and extracellular matrix in controlling neuroplasticity in the central nervous system. Semin Immunopathol 2023:10.1007/s00281-023-00989-1. [PMID: 37052711 DOI: 10.1007/s00281-023-00989-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 03/24/2023] [Indexed: 04/14/2023]
Abstract
Neuronal plasticity is critical for the maintenance and modulation of brain activity. Emerging evidence indicates that glial cells actively shape neuroplasticity, allowing for highly flexible regulation of synaptic transmission, neuronal excitability, and network synchronization. Astrocytes regulate synaptogenesis, stabilize synaptic connectivity, and preserve the balance between excitation and inhibition in neuronal networks. Microglia, the brain-resident immune cells, continuously monitor and sculpt synapses, allowing for the remodeling of brain circuits. Glia-mediated neuroplasticity is driven by neuronal activity, controlled by a plethora of feedback signaling mechanisms and crucially involves extracellular matrix remodeling in the central nervous system. This review summarizes the key findings considering neurotransmission regulation and metabolic support by astrocyte-neuronal networks, and synaptic remodeling mediated by microglia. Novel data indicate that astrocytes and microglia are pivotal for controlling brain function, indicating the necessity to rethink neurocentric neuroplasticity views.
Collapse
Affiliation(s)
- Egor Dzyubenko
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany.
| | - Dirk M Hermann
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany.
| |
Collapse
|
12
|
Andersen JV, Schousboe A. Glial Glutamine Homeostasis in Health and Disease. Neurochem Res 2023; 48:1100-1128. [PMID: 36322369 DOI: 10.1007/s11064-022-03771-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 08/25/2022] [Accepted: 09/27/2022] [Indexed: 11/05/2022]
Abstract
Glutamine is an essential cerebral metabolite. Several critical brain processes are directly linked to glutamine, including ammonia homeostasis, energy metabolism and neurotransmitter recycling. Astrocytes synthesize and release large quantities of glutamine, which is taken up by neurons to replenish the glutamate and GABA neurotransmitter pools. Astrocyte glutamine hereby sustains the glutamate/GABA-glutamine cycle, synaptic transmission and general brain function. Cerebral glutamine homeostasis is linked to the metabolic coupling of neurons and astrocytes, and relies on multiple cellular processes, including TCA cycle function, synaptic transmission and neurotransmitter uptake. Dysregulations of processes related to glutamine homeostasis are associated with several neurological diseases and may mediate excitotoxicity and neurodegeneration. In particular, diminished astrocyte glutamine synthesis is a common neuropathological component, depriving neurons of an essential metabolic substrate and precursor for neurotransmitter synthesis, hereby leading to synaptic dysfunction. While astrocyte glutamine synthesis is quantitatively dominant in the brain, oligodendrocyte-derived glutamine may serve important functions in white matter structures. In this review, the crucial roles of glial glutamine homeostasis in the healthy and diseased brain are discussed. First, we provide an overview of cellular recycling, transport, synthesis and metabolism of glutamine in the brain. These cellular aspects are subsequently discussed in relation to pathological glutamine homeostasis of hepatic encephalopathy, epilepsy, Alzheimer's disease, Huntington's disease and amyotrophic lateral sclerosis. Further studies on the multifaceted roles of cerebral glutamine will not only increase our understanding of the metabolic collaboration between brain cells, but may also aid to reveal much needed therapeutic targets of several neurological pathologies.
Collapse
Affiliation(s)
- Jens V Andersen
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.
| | - Arne Schousboe
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
13
|
Lindberg FA, Nordenankar K, Forsberg EC, Fredriksson R. SLC38A10 Deficiency in Mice Affects Plasma Levels of Threonine and Histidine in Males but Not in Females: A Preliminary Characterization Study of SLC38A10−/− Mice. Genes (Basel) 2023; 14:genes14040835. [PMID: 37107593 PMCID: PMC10138244 DOI: 10.3390/genes14040835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/23/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Solute carriers belong to the biggest group of transporters in the human genome, but more knowledge is needed to fully understand their function and possible role as therapeutic targets. SLC38A10, a poorly characterized solute carrier, is preliminary characterized here. By using a knockout mouse model, we studied the biological effects of SLC38A10 deficiency in vivo. We performed a transcriptomic analysis of the whole brain and found seven differentially expressed genes in SLC38A10-deficient mice (Gm48159, Nr4a1, Tuba1c, Lrrc56, mt-Tp, Hbb-bt and Snord116/9). By measuring amino acids in plasma, we found lower levels of threonine and histidine in knockout males, whereas no amino acid levels were affected in females, suggesting that SLC38A10−/− might affect sexes differently. Using RT-qPCR, we investigated the effect of SLC38A10 deficiency on mRNA expression of other SLC38 members, Mtor and Rps6kb1 in the brain, liver, lung, muscle, and kidney, but no differences were found. Relative telomere length measurement was also taken, as a marker for cellular age, but no differences were found between the genotypes. We conclude that SLC38A10 might be important for keeping amino acid homeostasis in plasma, at least in males, but no major effects were seen on transcriptomic expression or telomere length in the whole brain.
Collapse
|
14
|
Xiong J, Luu TTT, Venkatachalam K, Du G, Zhu MX. Glutamine Produces Ammonium to Tune Lysosomal pH and Regulate Lysosomal Function. Cells 2022; 12:80. [PMID: 36611873 PMCID: PMC9819001 DOI: 10.3390/cells12010080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/21/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Glutamine is one of the most abundant amino acids in the cell. In mitochondria, glutaminases 1 and 2 (GLS1/2) hydrolyze glutamine to glutamate, which serves as the precursor of multiple metabolites. Here, we show that ammonium generated during GLS1/2-mediated glutaminolysis regulates lysosomal pH and in turn lysosomal degradation. In primary human skin fibroblasts BJ cells and mouse embryonic fibroblasts, deprivation of total amino acids for 1 h increased lysosomal degradation capacity as shown by the increased turnover of lipidated microtubule-associated proteins 1A/1B light chain 3B (LC3-II), several autophagic receptors, and endocytosed DQ-BSA. Removal of glutamine but not any other amino acids from the culture medium enhanced lysosomal degradation similarly as total amino acid starvation. The presence of glutamine in regular culture media increased lysosomal pH by >0.5 pH unit and the removal of glutamine caused lysosomal acidification. GLS1/2 knockdown, GLS1 antagonist, or ammonium scavengers reduced lysosomal pH in the presence of glutamine. The addition of glutamine or NH4Cl prevented the increase in lysosomal degradation and curtailed the extension of mTORC1 function during the early time period of amino acid starvation. Our findings suggest that glutamine tunes lysosomal pH by producing ammonium, which regulates lysosomal degradation to meet the demands of cellular activities. During the early stage of amino acid starvation, the glutamine-dependent mechanism allows more efficient use of internal reserves and endocytosed proteins to extend mTORC1 activation such that the normal anabolism is not easily interrupted by a brief disruption of the amino acid supply.
Collapse
Affiliation(s)
- Jian Xiong
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Program in Biochemistry and Cell Biology, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Thi Thu Trang Luu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Program in Biochemistry and Cell Biology, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Kartik Venkatachalam
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Program in Biochemistry and Cell Biology, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
- Program in Neuroscience, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Guangwei Du
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Program in Biochemistry and Cell Biology, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Michael X. Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Program in Biochemistry and Cell Biology, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
- Program in Neuroscience, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
15
|
Lindberg FA, Roman E, Fredriksson R. Behavioral profiling of SLC38A10 knockout mice using the multivariate concentric square field TM test. Front Behav Neurosci 2022; 16:987037. [PMID: 36620864 PMCID: PMC9815452 DOI: 10.3389/fnbeh.2022.987037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022] Open
Abstract
Introduction SLC38A10 is a gene that encodes the SLC38A10 protein, also known as SNAT10. The SLC38 family is evolutionary old, and SLC38A10 is one of the oldest members of the family. It is ubiquitously expressed, and its substrates are glutamine, glutamate, alanine, aspartate, and serine. However, little is known about its biological importance. Methods In the current study, an SLC38A10 knockout mouse was run in the multivariate concentric square field TM (MCSF) test. The MCSF test gives the mouse a choice of areas to explore; sheltered areas, elevated and illuminated areas, or open spaces, and a behavioral profile is obtained. The multivariate data obtained were analyzed (i) for each parameter, (ii) parameters grouped into functional categories, and (iii) with a principal component analysis. Results In the trend analysis, knockout mice had a decreased exploratory behavior compared to controls but did not show a distinct grouping in the principal component analysis. Discussion There was not a pronounced difference in the behavioral profile in SLC38A10 knockout mice compared to their wild-type controls, although subtle alterations in zones associated with exploratory behavior and risk assessment in female and male knockout mice, respectively, could be observed. These results imply that a loss of function of the SLC38A10 protein in mice does not drastically alter behavior in the MSCF test.
Collapse
Affiliation(s)
- Frida A. Lindberg
- Department of Pharmaceutical Biosciences, Molecular Neuropharmacology, Uppsala University, Uppsala, Sweden,*Correspondence: Frida A. Lindberg,
| | - Erika Roman
- Neuropharmacology and Addiction, Uppsala University, Uppsala, Sweden,Division of Anatomy and Physiology, Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Robert Fredriksson
- Department of Pharmaceutical Biosciences, Molecular Neuropharmacology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
16
|
The Genetic Variability of Members of the SLC38 Family of Amino Acid Transporters ( SLC38A3, SLC38A7 and SLC38A9) Affects Susceptibility to Type 2 Diabetes and Vascular Complications. Nutrients 2022; 14:nu14214440. [PMID: 36364703 PMCID: PMC9654215 DOI: 10.3390/nu14214440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 01/25/2023] Open
Abstract
Type 2 Diabetes (T2D) is a metabolic disease associated with long-term complications, with a multifactorial pathogenesis related to the interplay between genetic and modifiable risk factors, of which nutrition is the most relevant. In particular, the importance of proteins and constitutive amino acids (AAs) in disease susceptibility is emerging. The ability to sense and respond to changes in AA supplies is mediated by complex networks, of which AA transporters (AATs) are crucial components acting also as sensors of AA availability. This study explored the associations between polymorphisms in selected AATs genes and T2D and vascular complications in 433 patients and 506 healthy controls. Analyses revealed significant association of SLC38A3-rs1858828 with disease risk. Stratification of patients based on presence/absence of vascular complications highlighted significant associations of SLC7A8-rs3783436 and SLC38A7-rs9806843 with diabetic retinopathy. Additionally, the SLC38A9-rs4865615 resulted associated with chronic kidney disease. Notably, these genes function as AAs sensors, specifically glutamine, leucine, and arginine, linked to the main nutrient signaling pathway mammalian target of rapamycin complex 1 (mTORC1). Thus, their genetic variability may contribute to T2D by influencing the ability to properly transduce a signal activating mTORC1 in response to AA availability. In this scenario, the contribution of dietary AAs supply to disease risk may be relevant.
Collapse
|
17
|
Bian LH, Yao ZW, Wang ZY, Wang XM, Li QY, Yang X, Li JY, Wei XJ, Wan GH, Wang YQ, Shi JL, Guo JY. Nardosinone regulates the slc38a2 gene to alleviate Parkinson's symptoms in rats through the GABAergic synaptic and cAMP pathways. Biomed Pharmacother 2022; 153:113269. [PMID: 35728354 DOI: 10.1016/j.biopha.2022.113269] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/04/2022] [Accepted: 06/06/2022] [Indexed: 11/02/2022] Open
Abstract
In a rotenone-induced Parkinson's disease (PD) rat model, behavioral investigation, pathological examination, inflammatory factor analysis, and mitochondrial structure and function investigation verified the anti-PD efficacy of nardosinone. A combined transcriptome and proteome analysis proposed that the anti-PD target of nardosinone is the slc38a2 gene and may involve the GABAergic synaptic pathway and cAMP-signaling pathway. Analysis of targeted slc38a2 knockout cells and expression of key enzyme-encoding genes in both pathways verified the target and pathways proposed by the 'omics analysis. This further confirms that nardosinone can regulate the slc38a2 gene, a potential new target for the treatment of Parkinson's disease, and plays an anti-PD role through the GABAergic synaptic and cAMP pathways.
Collapse
Affiliation(s)
- Li-Hua Bian
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11A North Third Ring East Road, Chaoyang District, Beijing 100029, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Zi-Wei Yao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11A North Third Ring East Road, Chaoyang District, Beijing 100029, China.
| | - Zhe-Yi Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11A North Third Ring East Road, Chaoyang District, Beijing 100029, China; Qilu Hospital, Shandong University, Jinan 250012, Shandong, China.
| | - Xiao-Mei Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11A North Third Ring East Road, Chaoyang District, Beijing 100029, China.
| | - Qiu-Yu Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11A North Third Ring East Road, Chaoyang District, Beijing 100029, China.
| | - Xue Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11A North Third Ring East Road, Chaoyang District, Beijing 100029, China.
| | - Jia-Yuan Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11A North Third Ring East Road, Chaoyang District, Beijing 100029, China.
| | - Xiao-Jia Wei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11A North Third Ring East Road, Chaoyang District, Beijing 100029, China.
| | - Guo-Hui Wan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11A North Third Ring East Road, Chaoyang District, Beijing 100029, China.
| | - Yu-Qing Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11A North Third Ring East Road, Chaoyang District, Beijing 100029, China.
| | - Jin-Li Shi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11A North Third Ring East Road, Chaoyang District, Beijing 100029, China.
| | - Jian-You Guo
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, 4A DatunRoad, Chaoyang District, Beijing 100101, China.
| |
Collapse
|
18
|
Ghareeb AFA, Schneiders GH, Foutz JC, Milfort MC, Fuller AL, Yuan J, Rekaya R, Aggrey SE. Heat Stress Alters the Effect of Eimeria maxima Infection on Ileal Amino Acids Digestibility and Transporters Expression in Meat-Type Chickens. Animals (Basel) 2022; 12:ani12121554. [PMID: 35739890 PMCID: PMC9219439 DOI: 10.3390/ani12121554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 11/30/2022] Open
Abstract
Simple Summary Heat stress (HS) and Eimeria (E.) maxima infection are the most common physical and pathological stressors in chicken houses, and both affect intestinal digestibility and absorption leading to reduction in growth, morbidity, and mortality, causing massive economic losses. This study identifies the impact of each stressor and their combined effects on apparent amino acid digestibility and molecular transporters expression in the ileum of broiler chicken. Heat-stressed chickens showed no change in amino acids digestibility, despite the reduction in feed intake. Combining HS and E. maxima infection modulated the reduction in amino acids digestibility observed in the infected chickens. The expression of the ileal amino acid transporters was severely impacted by E. maxima infection but not by HS. Interestingly, the infected group reared under HS exhibited significantly higher expression levels in all the enterocytic apical and about half of the basolateral amino acid transporters than the infected birds raised in thermoneutral environment. Thus, HS putatively curtailed the maldigestion effects of E. maxima. Abstract Eimeria (E.) maxima invades the midgut of chickens and destroys the intestinal mucosa, impacting nutrient digestibility and absorption. Heat stress (HS) commonly affects the broiler chicken and contributes to inflammation and oxidative stress. We examined the independent and combined effects of HS and E. maxima infection on apparent amino acid ileal digestibility (AID) and mRNA expression of amino acid transporters in broiler chickens (Ross 708). There were four treatment groups: thermoneutral-control (TNc) and infected (TNi), heat-stress control (HSc) and infected (HSi), six replicates of 10 birds/treatment. Ileal content and tissue were sampled at 6 d post infection to determine AID and transporters expression. Surprisingly, the HSi chickens exposed to two critical stressors exhibited normal AID. Only the TNi group displayed reduction in AID. Using TNc as control, the HSc group showed upregulated CAT1, LAT4, TAT1, SNAT1, and SNAT7. The HSi group showed upregulated CAT1 and LAT1, and downregulated b0,+AT, rBAT, SNAT1, and SNAT2. The TNi group showed upregulated CAT1, LAT1, and SNAT1 and downregulated B0AT1, b0,+AT, rBAT, LAT4, and TAT1. The expression of all enterocytic-apical and about half of the basolateral transporters was higher in the HSi group than in the TNi group, indicating that HS can putatively alleviate the E. maxima adverse effect on ileal digestion and absorption.
Collapse
Affiliation(s)
- Ahmed F. A. Ghareeb
- Department of Poultry Science, University of Georgia, 110 Cedar St, Athens, GA 30602, USA; (A.F.A.G.); (G.H.S.); (J.C.F.); (M.C.M.); (A.L.F.)
| | - Gustavo H. Schneiders
- Department of Poultry Science, University of Georgia, 110 Cedar St, Athens, GA 30602, USA; (A.F.A.G.); (G.H.S.); (J.C.F.); (M.C.M.); (A.L.F.)
- Merck Animal Health, 2 Giralda Farms, Madison, NJ 07940, USA
| | - James C. Foutz
- Department of Poultry Science, University of Georgia, 110 Cedar St, Athens, GA 30602, USA; (A.F.A.G.); (G.H.S.); (J.C.F.); (M.C.M.); (A.L.F.)
- Boehringer Ingelheim Animal Health (BIAH), 1110 Airport Pkwy, Gainesville, GA 30501, USA
| | - Marie C. Milfort
- Department of Poultry Science, University of Georgia, 110 Cedar St, Athens, GA 30602, USA; (A.F.A.G.); (G.H.S.); (J.C.F.); (M.C.M.); (A.L.F.)
| | - Alberta L. Fuller
- Department of Poultry Science, University of Georgia, 110 Cedar St, Athens, GA 30602, USA; (A.F.A.G.); (G.H.S.); (J.C.F.); (M.C.M.); (A.L.F.)
| | - Jianmin Yuan
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China;
| | - Romdhane Rekaya
- Department of Animal and Dairy Science, University of Georgia, 425 River Rd, Athens, GA 30602, USA;
| | - Samuel E. Aggrey
- Department of Poultry Science, University of Georgia, 110 Cedar St, Athens, GA 30602, USA; (A.F.A.G.); (G.H.S.); (J.C.F.); (M.C.M.); (A.L.F.)
- Correspondence: ; Tel.: +1-706-542-1351
| |
Collapse
|
19
|
Huttunen J, Adla SK, Markowicz-Piasecka M, Huttunen KM. Increased/Targeted Brain (Pro)Drug Delivery via Utilization of Solute Carriers (SLCs). Pharmaceutics 2022; 14:pharmaceutics14061234. [PMID: 35745806 PMCID: PMC9228667 DOI: 10.3390/pharmaceutics14061234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 02/04/2023] Open
Abstract
Membrane transporters have a crucial role in compounds’ brain drug delivery. They allow not only the penetration of a wide variety of different compounds to cross the endothelial cells of the blood–brain barrier (BBB), but also the accumulation of them into the brain parenchymal cells. Solute carriers (SLCs), with nearly 500 family members, are the largest group of membrane transporters. Unfortunately, not all SLCs are fully characterized and used in rational drug design. However, if the structural features for transporter interactions (binding and translocation) are known, a prodrug approach can be utilized to temporarily change the pharmacokinetics and brain delivery properties of almost any compound. In this review, main transporter subtypes that are participating in brain drug disposition or have been used to improve brain drug delivery across the BBB via the prodrug approach, are introduced. Moreover, the ability of selected transporters to be utilized in intrabrain drug delivery is discussed. Thus, this comprehensive review will give insights into the methods, such as computational drug design, that should be utilized more effectively to understand the detailed transport mechanisms. Moreover, factors, such as transporter expression modulation pathways in diseases that should be taken into account in rational (pro)drug development, are considered to achieve successful clinical applications in the future.
Collapse
Affiliation(s)
- Johanna Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland; (J.H.); (S.K.A.)
| | - Santosh Kumar Adla
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland; (J.H.); (S.K.A.)
- Institute of Organic Chemistry and Biochemistry (IOCB), Czech Academy of Sciences, Flemingovo Namesti 542/2, 160 00 Prague, Czech Republic
| | - Magdalena Markowicz-Piasecka
- Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151 Lodz, Poland;
| | - Kristiina M. Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland; (J.H.); (S.K.A.)
- Correspondence:
| |
Collapse
|
20
|
Lindberg FA, Nordenankar K, Fredriksson R. SLC38A10 Knockout Mice Display a Decreased Body Weight and an Increased Risk-Taking Behavior in the Open Field Test. Front Behav Neurosci 2022; 16:840987. [PMID: 35677577 PMCID: PMC9169716 DOI: 10.3389/fnbeh.2022.840987] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/21/2022] [Indexed: 11/17/2022] Open
Abstract
The solute carrier 38 family (SLC38) is a family of 11 members. The most common substrate among these are alanine and glutamine, and members are present in a wide range of tissues with important functions for several biological processes, such as liver and brain function. Some of these transporters are better characterized than others and, in this paper, a behavioral characterization of SLC38A10−/− mice was carried out. A battery of tests for general activity, emotionality, motor function, and spatial memory was used. Among these tests, the elevated plus maze, Y-maze, marble burying and challenging beam walk have not been tested on the SLC38A10−/− mice previously, while the open field and the rotarod tests have been performed by the International Mouse Phenotyping Consortium (IMPC). Unlike the results from IMPC, the results from this study showed that SLC38A10−/− mice spend less time in the wall zone in the open field test than WT mice, implying that SLC38A10-deficient mice have an increased explorative behavior, which suggests an important function of SLC38A10 in brain. The present study also confirmed IMPC's data regarding rotarod performance and weight, showing that SLC38A10−/− mice do not have an affected motor coordination impairment and have a lower body weight than both SLC38A10+/− and SLC38A10+/+ mice. These results imply that a complete deficiency of the SLC38A10 protein might affect body weight homeostasis, but the underlying mechanisms needs to be studied further.
Collapse
|
21
|
Festa M, Minicozzi V, Boccaccio A, Lagostena L, Gradogna A, Qi T, Costa A, Larisch N, Hamamoto S, Pedrazzini E, Milenkovic S, Scholz-Starke J, Ceccarelli M, Vitale A, Dietrich P, Uozumi N, Gambale F, Carpaneto A. Current Methods to Unravel the Functional Properties of Lysosomal Ion Channels and Transporters. Cells 2022; 11:921. [PMID: 35326372 PMCID: PMC8946281 DOI: 10.3390/cells11060921] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/04/2022] [Accepted: 03/05/2022] [Indexed: 02/07/2023] Open
Abstract
A distinct set of channels and transporters regulates the ion fluxes across the lysosomal membrane. Malfunctioning of these transport proteins and the resulting ionic imbalance is involved in various human diseases, such as lysosomal storage disorders, cancer, as well as metabolic and neurodegenerative diseases. As a consequence, these proteins have stimulated strong interest for their suitability as possible drug targets. A detailed functional characterization of many lysosomal channels and transporters is lacking, mainly due to technical difficulties in applying the standard patch-clamp technique to these small intracellular compartments. In this review, we focus on current methods used to unravel the functional properties of lysosomal ion channels and transporters, stressing their advantages and disadvantages and evaluating their fields of applicability.
Collapse
Affiliation(s)
- Margherita Festa
- Department of Biology, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy;
| | - Velia Minicozzi
- INFN, Department of Physics, University of Rome Tor Vergata, Via della Ricerca Scientifica 1, 00133 Rome, Italy;
| | - Anna Boccaccio
- Institute of Biophysics, National Research Council, Via De Marini 6, 16149 Genoa, Italy; (A.B.); (L.L.); (A.G.); (J.S.-S.); (F.G.)
| | - Laura Lagostena
- Institute of Biophysics, National Research Council, Via De Marini 6, 16149 Genoa, Italy; (A.B.); (L.L.); (A.G.); (J.S.-S.); (F.G.)
| | - Antonella Gradogna
- Institute of Biophysics, National Research Council, Via De Marini 6, 16149 Genoa, Italy; (A.B.); (L.L.); (A.G.); (J.S.-S.); (F.G.)
| | - Tianwen Qi
- Department of Earth, Environment and Life Sciences (DISTAV), University of Genoa, Viale Benedetto XV 5, 16132 Genoa, Italy;
| | - Alex Costa
- Department of Biosciences, University of Milan, 20133 Milan, Italy;
| | - Nina Larisch
- Department of Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91058 Erlangen, Germany; (N.L.); (P.D.)
| | - Shin Hamamoto
- Collaborative Research Institute for Innovative Microbiology, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan;
| | - Emanuela Pedrazzini
- Institute of Agricultural Biology and Biotechnology, National Research Council, Via Bassini 15, 20133 Milan, Italy; (E.P.); (A.V.)
| | - Stefan Milenkovic
- Department of Physics, University of Cagliari, 09042 Monserrato, Italy; (S.M.); (M.C.)
- IOM-CNR Unità di Cagliari, Cittadella Universitaria, 09042 Monserrato, Italy
| | - Joachim Scholz-Starke
- Institute of Biophysics, National Research Council, Via De Marini 6, 16149 Genoa, Italy; (A.B.); (L.L.); (A.G.); (J.S.-S.); (F.G.)
| | - Matteo Ceccarelli
- Department of Physics, University of Cagliari, 09042 Monserrato, Italy; (S.M.); (M.C.)
- IOM-CNR Unità di Cagliari, Cittadella Universitaria, 09042 Monserrato, Italy
| | - Alessandro Vitale
- Institute of Agricultural Biology and Biotechnology, National Research Council, Via Bassini 15, 20133 Milan, Italy; (E.P.); (A.V.)
| | - Petra Dietrich
- Department of Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91058 Erlangen, Germany; (N.L.); (P.D.)
| | - Nobuyuki Uozumi
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Sendai 980-8579, Japan;
| | - Franco Gambale
- Institute of Biophysics, National Research Council, Via De Marini 6, 16149 Genoa, Italy; (A.B.); (L.L.); (A.G.); (J.S.-S.); (F.G.)
| | - Armando Carpaneto
- Institute of Biophysics, National Research Council, Via De Marini 6, 16149 Genoa, Italy; (A.B.); (L.L.); (A.G.); (J.S.-S.); (F.G.)
- Department of Earth, Environment and Life Sciences (DISTAV), University of Genoa, Viale Benedetto XV 5, 16132 Genoa, Italy;
| |
Collapse
|
22
|
Talebi M, Mohammadi Vadoud SA, Haratian A, Talebi M, Farkhondeh T, Pourbagher-Shahri AM, Samarghandian S. The interplay between oxidative stress and autophagy: focus on the development of neurological diseases. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2022; 18:3. [PMID: 35093121 PMCID: PMC8799983 DOI: 10.1186/s12993-022-00187-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 01/17/2022] [Indexed: 12/12/2022]
Abstract
Regarding the epidemiological studies, neurological dysfunctions caused by cerebral ischemia or neurodegenerative diseases (NDDs) have been considered a pointed matter. Mount-up shreds of evidence support that both autophagy and reactive oxygen species (ROS) are involved in the commencement and progression of neurological diseases. Remarkably, oxidative stress prompted by an increase of ROS threatens cerebral integrity and improves the severity of other pathogenic agents such as mitochondrial damage in neuronal disturbances. Autophagy is anticipated as a cellular defending mode to combat cytotoxic substances and damage. The recent document proposes that the interrelation of autophagy and ROS creates a crucial function in controlling neuronal homeostasis. This review aims to overview the cross-talk among autophagy and oxidative stress and its molecular mechanisms in various neurological diseases to prepare new perceptions into a new treatment for neurological disorders. Furthermore, natural/synthetic agents entailed in modulation/regulation of this ambitious cross-talk are described.
Collapse
Affiliation(s)
- Marjan Talebi
- Department of Pharmacognosy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyyed Ali Mohammadi Vadoud
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Haratian
- Department of Pharmacognosy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Talebi
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, TX, 76019, USA
- Viatris Pharmaceuticals Inc, 3300 Research Plaza, San Antonio, TX, 78235, USA
| | - Tahereh Farkhondeh
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences, Birjand, Iran
- Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | | | - Saeed Samarghandian
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| |
Collapse
|
23
|
Heden TD, Chow LS, Hughey CC, Mashek DG. Regulation and role of glycophagy in skeletal muscle energy metabolism. Autophagy 2021; 18:1078-1089. [PMID: 34506219 DOI: 10.1080/15548627.2021.1969633] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Glycophagy is the autophagic degradation of glycogen via the lysosomal enzyme GAA/alpha-acid glucosidase. Glycophagy is considered a housekeeping process to degrade poorly branched glycogen particles, but the regulation and role of glycophagy in skeletal muscle metabolism remains enigmatic. Herein, prior muscle contraction promoted glycogen supercompensation 24 and 48 h post contraction, an effect associated with reduced glycophagy. Moreover, NOTCH or cAMP signaling promoted glycophagy, whereas acute glycophagy deficiency rewired cell metabolism by reducing glycolysis and enhancing AMPK and PPAR signaling and fatty acid and glutamine metabolism. These metabolic adaptations were associated with reduced inflammation and triglyceride content but enhanced phosphoinositide 3-kinase (PI3K)-AKT/protein kinase B signaling and insulin action, the latter of which was abolished by exogenous oxidative stress. Collectively, these data suggest glycophagy is dynamically regulated, while the function of glycophagy can be extended beyond a housekeeping process to having an additional role in regulating energy metabolism and insulin action.Abbreviations: AMPK, AMP-activated protein kinase; ASM, acid soluble metabolites; cAMP, cyclic adenosine monophosphate; EPS, electrical pulse stimulation; FCCP, carbonyl cyanide-p-trifluoromethoxyphenylhydrazone; GAA, glucosidase, alpha, acid; mTOR, mechanistic target of rapamycin kinase; NAD, nicotinamide adenine dinucleotide; PARP, poly (ADP-ribose) polymerase family; PI3K, phosphoinositide 3-kinase; PPAR, peroxisome proliferator activated receptor ; PYGM, muscle glycogen phosphorylase; STBD1, starch binding domain 1; TFEB, transcription factor EB.
Collapse
Affiliation(s)
- Timothy D Heden
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Lisa S Chow
- Department of Medicine, Division of Endocrinology
| | - Curtis C Hughey
- Department of Medicine, Division of Molecular Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Douglas G Mashek
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA.,Department of Medicine, Division of Endocrinology
| |
Collapse
|
24
|
Rudnik S, Damme M. The lysosomal membrane-export of metabolites and beyond. FEBS J 2021; 288:4168-4182. [PMID: 33067905 DOI: 10.1111/febs.15602] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/01/2020] [Accepted: 10/15/2020] [Indexed: 01/01/2023]
Abstract
Lysosomes are degradative organelles in eukaryotic cells mediating the hydrolytic catabolism of various macromolecules to small basic building blocks. These low-molecular-weight metabolites are transported across the lysosomal membrane and reused in the cytoplasm and other organelles for biosynthetic pathways. Even though in the past 20 years our understanding of the lysosomal membrane regarding various transporters, other integral and peripheral membrane proteins, the lipid composition, but also its turnover has dramatically improved, there are still many unresolved questions concerning key aspects of the function of the lysosomal membrane. These include a possible function of lysosomes as a cellular storage compartment, yet unidentified transporters mediating the export such as various amino acids, mechanisms mediating the transport of lysosomal membrane proteins from the Golgi apparatus to lysosomes, and the turnover of lysosomal membrane proteins. Here, we review the current knowledge about the lysosomal membrane and identify some of the open questions that need to be solved in the future for a comprehensive and complete understanding of how lysosomes communicate with other organelles, cellular processes, and pathways.
Collapse
Affiliation(s)
- Sönke Rudnik
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Markus Damme
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| |
Collapse
|
25
|
Identification of SLC38A7 as a Prognostic Marker and Potential Therapeutic Target of Lung Squamous Cell Carcinoma. Ann Surg 2021; 274:500-507. [PMID: 34171866 DOI: 10.1097/sla.0000000000005001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND No effective molecular targeted therapy has been established for SCC. We conducted a comprehensive study of SCC patients using RNA-sequencing and TCGA dataset to clarify the driver oncogene of SCC. METHOD Forty-six samples of 23 patients were totally analyzed with RNA-sequencing. We then searched for candidate-oncogenes of SCC using the TCGA database. To identify candidate oncogenes, we used the following 2 criteria: (1) the genes of interest were overexpressed in tumor tissues of SCC patients in comparison to normal tissues; and (2) using an integrated mRNA expression and DNA copy number profiling analysis using the TCGA dataset, the DNA copy number of the genes was positively correlated with the mRNA expression. RESULT We identified 188 candidate-oncogenes. Among those, the high expression of SLC38A7 was a strong prognostic marker that was significantly associated with a poor prognosis in terms of both overall survival (OS) and recurrence-free survival in the TCGA dataset (P < 0.05). Additionally, 202 resected SCC specimens were also subjected to an immunohistochemical analysis. Patients with the high expression of SLC38A7 (alternative name is sodium-coupled amino acid transporters 7) protein showed significantly shorter OS in comparison to those with the low expression of SLC38A7 protein [median OS 3.9 years (95% confidence interval, 2.4-6.4 years) vs 2.2 years (95% confidence interval, 1.9-4.1 years); log rank test: P = 0.0021]. CONCLUSION SLC38A7, which is the primary lysosomal glutamine transporter required for the extracellular protein-dependent growth of cancer cells, was identified as a candidate therapeutic target of SCC.
Collapse
|
26
|
Effects of underwater and semi-aquatic environments on gut tissue and microbiota of the mudskipper Boleophthalmus pectinirostris. J Comp Physiol B 2021; 191:741-753. [PMID: 34057562 DOI: 10.1007/s00360-021-01380-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 05/04/2021] [Accepted: 05/16/2021] [Indexed: 01/16/2023]
Abstract
In both underwater and semi-aquatic environments, the gut microbiota is of particular physiological importance for amphibious animals, given that the gut tract is among those organs in direct communication with the external environment. In this study, we examined the effects of these contrasting environments on the dominant bacteria in the guts of the amphibious mudskipper Boleophthalmus pectinirostris. Compared with the guts of normal mudskippers, in which the dominant bacteria were identified as Vibrio and Faecalibacterium, we found that Acinetobacter, Shigella, and Bacillus predominated in their guts after exposure to the semi-aquatic environment, whereas Escherichia, Bacteroides, and Bacillus were more prevalent in the guts in the underwater environment. The total number of cultured gut bacteria decreased significantly in the semi-aquatic environment. In semi-aquatic mudskippers, we also detected reductions and increases in the length and width of gut villi, respectively, whereas the width of gut villi declined and the number of goblet cells increased significantly in mudskippers maintained underwater. The mRNA expression of multiple gut transporters for glucose, long-chain fatty acids, and amino acids was found to increase markedly in both underwater and semi-aquatic environments, with the expression of most transporters being significantly higher in those mudskippers exposed to an underwater environment. Furthermore, we detected significant increases in the mRNA expression of pro-inflammatory cytokine transcripts in the guts of both underwater and semi-aquatic mudskippers on days 2, 4, and 6 of exposure, whereas the expression of IL-10 and TGFβ mRNA was more pronounced on days 4 and 8, respectively. Comparatively, we found that expression levels of cytokines in the guts of underwater mudskipper were substantially higher than those in the guts of semi-aquatic mudskippers. Collectively, our findings revealed notable differences in the gut microbiota and energy metabolism requirements of mudskippers exposed to underwater and semi-aquatic conditions, thereby providing a theoretical basis explaining the maintenance of a homeostatic state in mudskippers that constantly transition between these contrasting amphibious habitats.
Collapse
|
27
|
Tripathi R, Aggarwal T, Fredriksson R. SLC38A10 Transporter Plays a Role in Cell Survival Under Oxidative Stress and Glutamate Toxicity. Front Mol Biosci 2021; 8:671865. [PMID: 34026845 PMCID: PMC8133219 DOI: 10.3389/fmolb.2021.671865] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/09/2021] [Indexed: 11/13/2022] Open
Abstract
Solute carrier (SLC) transporters regulate amino acids, glucose, ions, and metabolites that flow across cell membranes. In the brain, SLCs are the key regulators of neurotransmission, in particular, the glutamate/GABA-glutamine (GGG) cycle. Genetic mutations in SLCs are associated with various neurodevelopmental and neurodegenerative diseases. In this study, we have investigated the role of SLC38A10 under acute oxidative and glutamate stress in mouse primary cortical cells from SLC38A10 knockout (KO) mice. The ER/golgi localized transporter, SLC38A10, transports glutamate, glutamine, and alanine in brain cells, and the aim of this study was to determine the possible effects of removal of SLC38A10 in primary cortical cells under glutamate and oxidative challenges. Primary cortical neuronal cultures of wild-type (WT) cell and SLC38A10 KO mice were subjected to different concentrations of glutamate and hydrogen peroxide. There was no morphological change observed between KO and WT cortical neurons in culture. Interestingly, KO cells showed significantly lower cell viability and higher cell death compared to WT cells under both glutamate and hydrogen peroxide exposure. Further, we evaluated the possible role of p53 in neuronal cell apoptosis in KO cells. We found decreased intracellular p53 protein levels under glutamate and hydrogen peroxide treatment in KO cortical cells. In contrast, caspase 3/7 activity remains unaltered under all conditions. These results demonstrate an indirect relationship between the expression of SLC38A10 and p53 and a role in the cell defense mechanism against neurotoxicity.
Collapse
Affiliation(s)
- Rekha Tripathi
- Molecular Neuropharmacology, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Tanya Aggarwal
- Molecular Neuropharmacology, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Robert Fredriksson
- Molecular Neuropharmacology, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
28
|
Maezawa T, Ishikawa M, Sekii K, Nagamatsu G, Furukawa R, Kobayashi K. D-Tryptophan enhances the reproductive organ-specific expression of the amino acid transporter homolog Dr-SLC38A9 involved in the sexual induction of planarian Dugesia ryukyuensis. ZOOLOGICAL LETTERS 2021; 7:4. [PMID: 33743841 PMCID: PMC7981857 DOI: 10.1186/s40851-021-00173-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 03/10/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Many animals switch between asexual and sexual reproduction in nature. We previously established a system for the sexual induction of planarian Dugesia ryukyuensis by feeding asexual planarians with minced sexual planarians. We identified DL-tryptophan (Trp) as one of the sex-inducing substances. DL-Trp can induce ovarian development, the first and essential step of sexual induction. D-Trp must act as a principal bioactive compound in terms of ovarian development, because the ovary-inducing activity of D-Trp was 500 times more potent than that of L-Trp. However, how Trp controls sexual induction is still unknown. RESULTS In this study, qRT-PCR analyses suggested that the putative amino acid transporter gene Dr-SLC38A9 is highly expressed in sexual worms, especially in the yolk glands. In situ hybridization analyses showed that Dr-SLC38A9 is expressed in the ovarian primordia of asexual worms and in the mature ovaries, testes, and yolk glands of sexual worms. In addition, Dr-SLC38A9 RNA interference during sexual induction resulted in the suppression of the development of reproductive organs. These results suggest that Dr-SLC38A9 is involved in the development of these organs. Moreover, we demonstrated that the reproductive organ-specific expression of Dr-SLC38A9 is enhanced by the addition of D-Trp. CONCLUSION We propose that D-Trp activates the expression of Dr-SLC38A9 to promote sexual induction in the planarian D. ryukyuensis.
Collapse
Affiliation(s)
- Takanobu Maezawa
- Advanced Science Course, Department of Integrated Science and Technology, National Institute of Technology, Tsuyama College, 624-1 Numa, Tsuyama, Okayama, 708-8509, Japan.
| | - Masaki Ishikawa
- Faculty of Agriculture and Life Science, Hirosaki University, 3 Bunkyo-cho, Hirosaki, Aomori, 036-8561, Japan
| | - Kiyono Sekii
- Faculty of Agriculture and Life Science, Hirosaki University, 3 Bunkyo-cho, Hirosaki, Aomori, 036-8561, Japan
| | - Go Nagamatsu
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Ryohei Furukawa
- Department of Biology, Research and Education Center for Natural Sciences, Keio University, 4-1-1 Hiyoshi, Kohoku-ku, Yokohama, Kanagawa, 223-8521, Japan
| | - Kazuya Kobayashi
- Faculty of Agriculture and Life Science, Hirosaki University, 3 Bunkyo-cho, Hirosaki, Aomori, 036-8561, Japan.
| |
Collapse
|
29
|
Saric A, Freeman SA. Endomembrane Tension and Trafficking. Front Cell Dev Biol 2021; 8:611326. [PMID: 33490077 PMCID: PMC7820182 DOI: 10.3389/fcell.2020.611326] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/09/2020] [Indexed: 12/11/2022] Open
Abstract
Eukaryotic cells employ diverse uptake mechanisms depending on their specialized functions. While such mechanisms vary widely in their defining criteria: scale, molecular machinery utilized, cargo selection, and cargo destination, to name a few, they all result in the internalization of extracellular solutes and fluid into membrane-bound endosomes. Upon scission from the plasma membrane, this compartment is immediately subjected to extensive remodeling which involves tubulation and vesiculation/budding of the limiting endomembrane. This is followed by a maturation process involving concomitant retrograde transport by microtubule-based motors and graded fusion with late endosomes and lysosomes, organelles that support the degradation of the internalized content. Here we review an important determinant for sorting and trafficking in early endosomes and in lysosomes; the control of tension on the endomembrane. Remodeling of endomembranes is opposed by high tension (caused by high hydrostatic pressure) and supported by the relief of tension. We describe how the timely and coordinated efflux of major solutes along the endocytic pathway affords the cell control over such tension. The channels and transporters that expel the smallest components of the ingested medium from the early endocytic fluid are described in detail as these systems are thought to enable endomembrane deformation by curvature-sensing/generating coat proteins. We also review similar considerations for the lysosome where resident hydrolases liberate building blocks from luminal macromolecules and transporters flux these organic solutes to orchestrate trafficking events. How the cell directs organellar trafficking based on the luminal contents of organelles of the endocytic pathway is not well-understood, however, we propose that the control over membrane tension by solute transport constitutes one means for this to ensue.
Collapse
Affiliation(s)
- Amra Saric
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Spencer A Freeman
- Program in Cell Biology, Peter Gilgan Center for Research and Learning, Hospital for Sick Children, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
30
|
Transport of L-Arginine Related Cardiovascular Risk Markers. J Clin Med 2020; 9:jcm9123975. [PMID: 33302555 PMCID: PMC7764698 DOI: 10.3390/jcm9123975] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 12/15/2022] Open
Abstract
L-arginine and its derivatives, asymmetric and symmetric dimethylarginine (ADMA and SDMA) and L-homoarginine, have emerged as cardiovascular biomarkers linked to cardiovascular outcomes and various metabolic and functional pathways such as NO-mediated endothelial function. Cellular uptake and efflux of L-arginine and its derivatives are facilitated by transport proteins. In this respect the cationic amino acid transporters CAT1 and CAT2 (SLC7A1 and SLC7A2) and the system y+L amino acid transporters (SLC7A6 and SLC7A7) have been most extensively investigated, so far, but the number of transporters shown to mediate the transport of L-arginine and its derivatives is constantly increasing. In the present review we assess the growing body of evidence regarding the function, expression, and clinical relevance of these transporters and their possible relation to cardiovascular diseases.
Collapse
|
31
|
Zhou Y, Eid T, Hassel B, Danbolt NC. Novel aspects of glutamine synthetase in ammonia homeostasis. Neurochem Int 2020; 140:104809. [DOI: 10.1016/j.neuint.2020.104809] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 02/07/2023]
|
32
|
Hamuro J, Deguchi H, Fujita T, Ueda K, Tokuda Y, Hiramoto N, Numa K, Nakano M, Bush J, Ueno M, Sotozono C, Kinoshita S. Polarized Expression of Ion Channels and Solute Carrier Family Transporters on Heterogeneous Cultured Human Corneal Endothelial Cells. Invest Ophthalmol Vis Sci 2020; 61:47. [PMID: 32455435 PMCID: PMC7405722 DOI: 10.1167/iovs.61.5.47] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Purpose To clarify the expression profiles of ion channels and transporters of metabolic substrates among heterogeneous cultured human corneal endothelial cells (cHCECs) distinct in their effectiveness in reconstituting the corneal endothelium. Methods Integrated proteomics for cell lysates by liquid chromatography–tandem mass spectrometry was carried out from three aliquots of cHCECs enriched in either cluster of definition (CD)44−/+ (mature) cHCECs or CD44++/+++ cell-state transition (CST) cHCECs. The expression profiles of cations/anions, monocarboxylic acid transporters (MCTs), and solute carrier (SLC) family proteins, as well as carbonic anhydrases (CAs), were investigated. Results The polarized expression of cations/anions, MCTs, and SLC family proteins, as well as CAs, was clarified for mature and CST cHCECs. Most SLC4 family members, including SLC4A11 and SLC4A4 (NBCe1), were upregulated in the CST cHCECs, whereas SLC9A1 (Na+/H+ exchanger isoform one [NHE1]) and CA5B were detected only in the mature cHCECs. In addition, SLC25A42, catalyzing the entry of coenzyme A into the mitochondria, and SLC25A18, functioning as a mitochondrial glutamate carrier 2 (both relevant for providing the substrates for mitochondrial bioenergetics), were selectively expressed in the mature cHCECs. Conclusions Our findings may suggest the relevance of qualifying the polarized expression of these ion channels and transporter-like proteins to ensure not only the suitability but also the in vivo biological functionality of cHCECs selected for use in a cell-injection therapy.
Collapse
|
33
|
Hong JK, Lee JB, Ramayo-Caldas Y, Kim SD, Cho ES, Kim YS, Cho KH, Lee DH, Park HB. Single-step genome-wide association study for social genetic effects and direct genetic effects on growth in Landrace pigs. Sci Rep 2020; 10:14958. [PMID: 32917921 PMCID: PMC7486944 DOI: 10.1038/s41598-020-71647-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 08/03/2020] [Indexed: 02/08/2023] Open
Abstract
In livestock social interactions, social genetic effects (SGE) represent associations between phenotype of one individual and genotype of another. Such associations occur when the trait of interest is affected by transmissible phenotypes of social partners. The aim of this study was to estimate SGE and direct genetic effects (DGE, genetic effects of an individual on its own phenotype) on average daily gain (ADG) in Landrace pigs, and to conduct single-step genome-wide association study using SGE and DGE as dependent variables to identify quantitative trait loci (QTLs) and their positional candidate genes. A total of 1,041 Landrace pigs were genotyped using the Porcine SNP 60K BeadChip. Estimates of the two effects were obtained using an extended animal model. The SGE contributed 16% of the total heritable variation of ADG. The total heritability estimated by the extended animal model including both SGE and DGE was 0.52. The single-step genome-wide association study identified a total of 23 QTL windows for the SGE on ADG distributed across three chromosomes (i.e., SSC1, SSC2, and SSC6). Positional candidate genes within these QTL regions included PRDM13, MAP3K7, CNR1, HTR1E, IL4, IL5, IL13, KIF3A, EFHD2, SLC38A7, mTOR, CNOT1, PLCB2, GABRR1, and GABRR2, which have biological roles in neuropsychiatric processes. The results of biological pathway and gene network analyses also support the association of the neuropsychiatric processes with SGE on ADG in pigs. Additionally, a total of 11 QTL windows for DGE on ADG in SSC2, 3, 6, 9, 10, 12, 14, 16, and 17 were detected with positional candidate genes such as ARL15. We found a putative pleotropic QTL for both SGE and DGE on ADG on SSC6. Our results in this study provide important insights that can help facilitate a better understanding of the molecular basis of SGE for socially affected traits.
Collapse
Affiliation(s)
- Joon-Ki Hong
- National Institute of Animal Science, Rural Development Administration, Cheonan, 31000, Republic of Korea
| | - Jae-Bong Lee
- Korea Zoonosis Research Institute, Chonbuk National University, 54531, Iksan, Republic of Korea
| | - Yuliaxis Ramayo-Caldas
- Animal Breeding and Genetics Program, Institute for Research and Technology in Food and Agriculture (IRTA), Torre Marimon, 08140, Caldes de Montbui, Spain
| | - Si-Dong Kim
- National Institute of Animal Science, Rural Development Administration, Cheonan, 31000, Republic of Korea
| | - Eun-Seok Cho
- National Institute of Animal Science, Rural Development Administration, Cheonan, 31000, Republic of Korea
| | - Young-Sin Kim
- National Institute of Animal Science, Rural Development Administration, Cheonan, 31000, Republic of Korea
| | - Kyu-Ho Cho
- National Institute of Animal Science, Rural Development Administration, Cheonan, 31000, Republic of Korea
| | - Deuk-Hwan Lee
- Department of Animal Life Resources, Hankyong National University, Anseong, 17579, Republic of Korea
| | - Hee-Bok Park
- Department of Animal Resources Science, Kongju National University, Yesan, 32439, Republic of Korea.
| |
Collapse
|
34
|
D-Serine Signaling and NMDAR-Mediated Synaptic Plasticity Are Regulated by System A-Type of Glutamine/D-Serine Dual Transporters. J Neurosci 2020; 40:6489-6502. [PMID: 32661027 DOI: 10.1523/jneurosci.0801-20.2020] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 05/22/2020] [Accepted: 06/25/2020] [Indexed: 02/03/2023] Open
Abstract
D-serine is a physiologic coagonist of NMDA receptors (NMDARs) required for synaptic plasticity, but mechanisms that terminate D-serine signaling are unclear. In particular, the identity of unidirectional plasma membrane transporters that mediate D-serine reuptake has remained elusive. We report that D-serine and glutamine share the same neuronal transport system, consisting of the classic system A transporters Slc38a1 and Slc38a2. We show that these transporters are not saturated with glutamine in vivo and regulate the extracellular levels of D-serine and NMDAR activity. Glutamine increased the NMDAR-dependent long-term potentiation and the isolated NMDAR potentials at the Schaffer collateral-CA1 synapses, but without affecting basal neurotransmission in male mice. Glutamine did not increase the NMDAR potentials in slices from serine racemase knock-out mice, which are devoid of D-serine, indicating that the effect of glutamine is caused by outcompeting D-serine for a dual glutamine-D-serine transport system. Inhibition of the system A reduced the uptake of D-serine in synaptosomes and neuronal cultures of mice of either sex, while increasing the extracellular D-serine concentration in slices and in vivo by microdialysis. When compared with Slc38a2, the Slc38a1 transporter displayed more favorable kinetics toward the D-enantiomer. Biochemical experiments with synaptosomes from Slc38a1 knock-down mice of either sex further support its role as a D-serine reuptake system. Our study identifies the first concentrative and electrogenic transporters mediating D-serine reuptake in vivo In addition to their classical role in the glutamine-glutamate cycle, system A transporters regulate the synaptic turnover of D-serine and its effects on NMDAR synaptic plasticity.SIGNIFICANCE STATEMENT Despite the plethora of roles attributed to D-serine, the regulation of its synaptic turnover is poorly understood. We identified the system A transporters Slc38a1 and Slc38a2 as the main pathway for neuronal reuptake of D-serine. These transporters are not saturated with glutamine in vivo and provide an unexpected link between the serine shuttle pathway, responsible for regulating D-serine synaptic turnover, and the glutamine-glutamate cycle. Our observations suggest that Slc38a1 and Slc38a2 have a dual role in regulating neurotransmission. In addition to their classical role as the glutamine providers, the system A transporters regulate extracellular D-serine and therefore affect NMDAR-dependent synaptic plasticity. Higher glutamine export from astrocytes would increase extracellular D-serine, providing a feedforward mechanism to increase synaptic NMDAR activation.
Collapse
|
35
|
Zhang H, McClatchie T, Baltz JM. l-Serine transport in growing and maturing mouse oocytes. J Cell Physiol 2020; 235:8585-8600. [PMID: 32329057 DOI: 10.1002/jcp.29702] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/27/2020] [Accepted: 03/28/2020] [Indexed: 01/24/2023]
Abstract
Serine has roles in cell metabolism besides protein synthesis including providing one-carbon units to the folate cycle. Since growing mouse oocytes undergo a burst of folate accumulation as they near full size, we have investigated whether oocytes transport serine. Substantial serine transport appeared in oocytes near the end of their growth. Serine transport continued when oocytes resumed meiosis but ceased partway through first meiotic metaphase, remaining quiescent in mature eggs in second meiotic metaphase. The serine transporter was sodium dependent and inhibited by alanine, cysteine, leucine, or histidine, and had a Michaelis-Menten constant (Km ) for serine of 200 µM. Unexpectedly, exposing cumulus cell-enclosed oocytes to the physiological mediator of meiotic arrest, natriuretic peptide precursor Type C, substantially stimulated serine transport by the enclosed oocyte. Finally, in addition to transport by the oocyte itself, cumulus cells also supply serine to the enclosed oocyte via gap junctions within intact cumulus-oocyte complexes.
Collapse
Affiliation(s)
- Han Zhang
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Division of Reproductive Medicine, Department of Obstetrics and Gynecology, University of Ottawa Faculty of Medicine, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa Faculty of Medicine, Ottawa, Ontario, Canada
| | - Taylor McClatchie
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Division of Reproductive Medicine, Department of Obstetrics and Gynecology, University of Ottawa Faculty of Medicine, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa Faculty of Medicine, Ottawa, Ontario, Canada
| | - Jay M Baltz
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Division of Reproductive Medicine, Department of Obstetrics and Gynecology, University of Ottawa Faculty of Medicine, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa Faculty of Medicine, Ottawa, Ontario, Canada
| |
Collapse
|
36
|
Bagchi S, Perland E, Hosseini K, Lundgren J, Al-Walai N, Kheder S, Fredriksson R. Probable role for major facilitator superfamily domain containing 6 (MFSD6) in the brain during variable energy consumption. Int J Neurosci 2020; 130:476-489. [PMID: 31906755 DOI: 10.1080/00207454.2019.1694020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Purpose: The major facilitator superfamily (MFS) is known as the largest and most diverse superfamily containing human transporters, and these transporters are essential as they sustain the homeostasis within cellular compartments by moving substances over lipid membranes.Methods: We have identified a novel MFS protein, named Major facilitator superfamily domain containing 6 (MFSD6), and confirmed that it is phylogenetically related to the human Solute Carrier (SLC) transporter family. A homology model of MFSD6 revealed 12 predicted transmembrane segments (TMS) with the classical MFS fold between TMS 6 and 7.Results: Immunohistological analyses showed specific MFSD6 staining in neurons of wildtype mouse brain tissue, but no expression in astrocytes. Furthermore, we explored expression and probable function(s) of MFSD6 in relation to its phylogenetically related proteins, major facilitator superfamily domain containing 8 (MFSD8) and 10 (MFSD10), which is of interest as both these proteins are involved in diseases.Conclusions: We showed that expression levels of Mfsd6 and Mfsd10 were decreased with elevated or depleted energy consumption, while that of Mfsd8 remained unaffected.
Collapse
Affiliation(s)
- Sonchita Bagchi
- Department of Pharmaceutical Biosciences, Unit of Molecular Neuropharmacology, Uppsala University, Uppsala, Sweden
| | - Emelie Perland
- Department of Pharmaceutical Biosciences, Unit of Molecular Neuropharmacology, Uppsala University, Uppsala, Sweden.,Department of Neuroscience, Unit of Functional Pharmacology, Uppsala University, Uppsala, Sweden
| | - Kimia Hosseini
- Department of Pharmaceutical Biosciences, Unit of Molecular Neuropharmacology, Uppsala University, Uppsala, Sweden
| | - Johanna Lundgren
- Department of Neuroscience, Unit of Functional Pharmacology, Uppsala University, Uppsala, Sweden
| | - Noura Al-Walai
- Department of Pharmaceutical Biosciences, Unit of Molecular Neuropharmacology, Uppsala University, Uppsala, Sweden
| | - Sania Kheder
- Department of Neuroscience, Unit of Functional Pharmacology, Uppsala University, Uppsala, Sweden
| | - Robert Fredriksson
- Department of Pharmaceutical Biosciences, Unit of Molecular Neuropharmacology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
37
|
Fagundes NS, Milfort MC, Williams SM, Da Costa MJ, Fuller AL, Menten JF, Rekaya R, Aggrey SE. Dietary methionine level alters growth, digestibility, and gene expression of amino acid transporters in meat-type chickens. Poult Sci 2020; 99:67-75. [PMID: 32416854 PMCID: PMC7587823 DOI: 10.3382/ps/pez588] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 10/01/2019] [Indexed: 01/16/2023] Open
Abstract
Imbalance in nutrients can affect digestibility of amino acids by altering gene expression of amino acid transporters. We investigated digestibility and molecular transporters of essential amino acids in chickens fed a methionine-deficient diet. A total of 40 chicks (23 D old) were randomly assigned to either a control (0.49% methionine) or a deficient (0.28%) diet until 41 D when they were sampled for Pectoralis (P.) major, kidney, ileum, and hypothalamus for mRNA expression analysis. The ileal content was collected for apparent ileal digestibility (AID) analysis. Birds fed the deficient diet had reduced growth and worse feed efficiency compared to control. The AID of methionine was similar between both groups. The AID of other essential amino acids was higher in the deficient group than control. mRNA expression of b0,+ AT and LAT4 were upregulated in the ileum and kidney but LAT1 was downregulated only in kidney of the deficient group compared to control. In the P. major, SNAT1, SNAT2, and CAT1 were upregulated in the deficient group compared to control. A diet deficiency in methionine affects digestibility of essential amino acids and cysteine, but not the digestibility of methionine. The change in digestibility is reflected in the mRNA expression of amino acid transporters across different tissues.
Collapse
Affiliation(s)
- Naiara S Fagundes
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602; Department of Animal Science, University of Sao Paulo, Piracicaba, Sao Paulo, Brazil
| | - Marie C Milfort
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602
| | - Susan M Williams
- Department of Population Health, University of Georgia, Athens, GA 30602
| | - Manuel J Da Costa
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602
| | - Alberta L Fuller
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602
| | - José F Menten
- Department of Animal Science, University of Sao Paulo, Piracicaba, Sao Paulo, Brazil
| | - Romdhane Rekaya
- Department of Animal and Dairy Science, University of Georgia, Athens, GA 30602
| | - Samuel E Aggrey
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602.
| |
Collapse
|
38
|
Tripathi R, Hosseini K, Arapi V, Fredriksson R, Bagchi S. SLC38A10 (SNAT10) is Located in ER and Golgi Compartments and Has a Role in Regulating Nascent Protein Synthesis. Int J Mol Sci 2019; 20:ijms20246265. [PMID: 31842320 PMCID: PMC6940841 DOI: 10.3390/ijms20246265] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/03/2019] [Accepted: 12/09/2019] [Indexed: 01/27/2023] Open
Abstract
The solute carrier (SLC) family-38 of transporters has eleven members known to transport amino acids, with glutamine being a common substrate for ten of them, with SLC38A9 being the exception. In this study, we examine the subcellular localization of SNAT10 in several independent immortalized cell lines and stem cell-derived neurons. Co-localization studies confirmed the SNAT10 was specifically localized to secretory organelles. SNAT10 is expressed in both excitatory and inhibitory neurons in the mouse brain, predominantly in the endoplasmic reticulum, and in the Golgi apparatus. Knock-down experiments of SNAT10, using Slc38a10-specific siRNA in PC12 cells reduced nascent protein synthesis by more than 40%, suggesting that SNAT10 might play a role in signaling pathways that regulate protein synthesis, and may act as a transceptor in a similar fashion to what has been shown previously for SLC38A2 (SNAT2) and SNAT9(SLC38A9).
Collapse
|
39
|
Yamada D, Kawabe K, Tosa I, Tsukamoto S, Nakazato R, Kou M, Fujikawa K, Nakamura S, Ono M, Oohashi T, Kaneko M, Go S, Hinoi E, Yoneda Y, Takarada T. Inhibition of the glutamine transporter SNAT1 confers neuroprotection in mice by modulating the mTOR-autophagy system. Commun Biol 2019; 2:346. [PMID: 31552299 PMCID: PMC6751179 DOI: 10.1038/s42003-019-0582-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 08/19/2019] [Indexed: 01/31/2023] Open
Abstract
The pathophysiological role of mammalian target of rapamycin complex 1 (mTORC1) in neurodegenerative diseases is established, but possible therapeutic targets responsible for its activation in neurons must be explored. Here we identified solute carrier family 38a member 1 (SNAT1, Slc38a1) as a positive regulator of mTORC1 in neurons. Slc38a1flox/flox and Synapsin I-Cre mice were crossed to generate mutant mice in which Slc38a1 was selectively deleted in neurons. Measurement of 2,3,5-triphenyltetrazolium chloride (TTC) or the MAP2-negative area in a mouse model of middle cerebral artery occlusion (MCAO) revealed that Slc38a1 deficiency decreased infarct size. We found a transient increase in the phosphorylation of p70S6k1 (pp70S6k1) and a suppressive effect of rapamycin on infarct size in MCAO mice. Autophagy inhibitors completely mitigated the suppressive effect of SNAT1 deficiency on neuronal cell death under in vitro stroke culture conditions. These results demonstrate that SNAT1 promoted ischemic brain damage via mTOR-autophagy system.
Collapse
Affiliation(s)
- Daisuke Yamada
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558 Japan
| | - Kenji Kawabe
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558 Japan
| | - Ikue Tosa
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558 Japan
| | - Shunpei Tsukamoto
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558 Japan
| | - Ryota Nakazato
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School, Kanazawa, Ishikawa 920-1192 Japan
| | - Miki Kou
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School, Kanazawa, Ishikawa 920-1192 Japan
| | - Koichi Fujikawa
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School, Kanazawa, Ishikawa 920-1192 Japan
| | - Saki Nakamura
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School, Kanazawa, Ishikawa 920-1192 Japan
| | - Mitsuaki Ono
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558 Japan
| | - Toshitaka Oohashi
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558 Japan
| | - Mari Kaneko
- Laboratory for Animal Resource Development Unit and Genetic Engineering Team, RIKEN Center for Life Science Technologies, 2-2-3 Minatojima Minami, Chuou-ku, Kobe, Hyogo 650-0047 Japan
| | - Shioi Go
- Laboratory for Animal Resource Development Unit and Genetic Engineering Team, RIKEN Center for Life Science Technologies, 2-2-3 Minatojima Minami, Chuou-ku, Kobe, Hyogo 650-0047 Japan
| | - Eiichi Hinoi
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School, Kanazawa, Ishikawa 920-1192 Japan
| | - Yukio Yoneda
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School, Kanazawa, Ishikawa 920-1192 Japan
| | - Takeshi Takarada
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558 Japan
| |
Collapse
|
40
|
Scalise M, Galluccio M, Pochini L, Cosco J, Trotta M, Rebsamen M, Superti-Furga G, Indiveri C. Insights into the transport side of the human SLC38A9 transceptor. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1861:1558-1567. [PMID: 31295473 DOI: 10.1016/j.bbamem.2019.07.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 05/17/2019] [Accepted: 07/03/2019] [Indexed: 01/06/2023]
Abstract
The lysosomal amino acid transporter SLC38A9 is referred to as transceptor, i.e. a transporter with a receptor function. The protein is responsible for coupling amino acid transport across the lysosomal membrane according to the substrate availability to mTORC1 signal transduction. This process allows cells to sense amino acid level responding to growth stimuli in physiological and pathological conditions triggering mTOR regulation. The main substrates underlying this function are glutamine and arginine. The functional and kinetic characterization of glutamine and arginine transport was performed using human SLC38A9 produced in E. coli, purified by affinity chromatography and reconstituted in liposomes. A cooperative behaviour for the wild type protein was revealed for both the substrates. A novel Na+ binding site, namely T453, was described by combined approaches of bioinformatics, site-directed mutagenesis and transport assay. Stimulation by cholesterol of glutamine and arginine transport was observed. The biological function of SLC38A9 relies on the interaction between its N-terminus and components of the mTOR complex; a deletion mutant of the N-terminus tail was produced and transport of glutamine was assayed revealing that this portion does not play any role in the intrinsic transport function of the human SLC38A9. Different features for glutamine and arginine transport were revealed: human SLC38A9 is competent for glutamine efflux, while that of arginine is negligible. In line with these results, imposed ∆pH stimulated glutamine, not arginine transport. Arginine plays, on the contrary, a modulatory function and is able to stimulate glutamine efflux. Interestingly, reciprocal inhibition experiments also supported by bioinformatics, suggested that glutamine and arginine may bind to different sites in the human SLC38A9 transporter.
Collapse
Affiliation(s)
- Mariafrancesca Scalise
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, 87036 Arcavacata di Rende, Italy
| | - Michele Galluccio
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, 87036 Arcavacata di Rende, Italy
| | - Lorena Pochini
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, 87036 Arcavacata di Rende, Italy
| | - Jessica Cosco
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, 87036 Arcavacata di Rende, Italy
| | - Miriam Trotta
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, 87036 Arcavacata di Rende, Italy
| | - Manuele Rebsamen
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Giulio Superti-Furga
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria; Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Cesare Indiveri
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, 87036 Arcavacata di Rende, Italy; CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnology, via Amendola 165/A, 70126 Bari, Italy.
| |
Collapse
|
41
|
Crouse MS, Greseth NP, McLean KJ, Crosswhite MR, Pereira NN, Ward AK, Reynolds LP, Dahlen CR, Neville BW, Borowicz PP, Caton JS. Maternal nutrition and stage of early pregnancy in beef heifers: impacts on hexose and AA concentrations in maternal and fetal fluids1. J Anim Sci 2019; 97:1296-1316. [PMID: 30649334 DOI: 10.1093/jas/skz013] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 01/08/2019] [Indexed: 01/12/2023] Open
Abstract
We examined the hypothesis that maternal nutrition and day of gestation would affect the concentrations of AAs and hexoses in bovine utero-placental fluids and maternal serum from days 16 to 50 of gestation. Forty-nine cross-bred Angus heifers were bred via artificial insemination and fed a control diet (CON = 100% of requirements for growth) or a restricted diet (RES = 60% of CON) and ovariohysterectomized on days 16, 34, or 50 of gestation; nonpregnant controls were not bred and ovariohysterectomized on day 16 of the synchronized estrous cycle. The resulting design was a completely randomized design with a 2 × 3 factorial + 1 arrangement of treatments. Maternal serum, histotroph, allantoic fluid, and amniotic fluid were collected at time of ovariohysterectomy. Samples were then analyzed for concentrations of AAs and intermediary metabolites: alanine (Ala), arginine, asparagine (Asn), aspartate (Asp), citrulline, cysteine, glutamine, glutamate (Glu), glycine (Gly), histidine, isoleucine, leucine (Leu), lysine, methionine (Met), ornithine, phenylalanine (Phe), proline (Pro), serine (Ser), threonine (Thr), tryptophan, tyrosine (Tyr), and valine (Val). The concentrations of Gly, Ser, and Thr in maternal serum were greater (P ≤ 0.05) in CON compared with RES. Furthermore, day of gestation affected (P ≤ 0.05) concentrations of Asn, Glu, Phe, Thr, and Tyr in maternal serum. Status of maternal nutrition affected the Asp concentration of histotroph where RES was greater (P = 0.02) than CON. In histotroph, Ala, Leu, Met, and Val concentrations were greater (P ≤ 0.05) on day 50 compared with day 16. Additionally, Glu and Pro concentrations in histotroph were greater (P < 0.01) on days 34 and 50 compared with day 16. A day × treatment interaction was observed for the concentration of Val in allantoic fluid where day 34 CON was greater (P = 0.05) than all other days and nutritional treatments. In addition, the concentration of Gln in amniotic fluid experienced a day × treatment interaction where day 34 RES was greater (P ≤ 0.05) than day 34 CON, which was greater (P ≤ 0.05) than day 50 CON and RES. These data support our hypothesis that day of gestation and maternal nutrition affect the concentrations of various neutral and acidic AA in beef heifer utero-placental fluids and maternal serum from days 16 to 50 of gestation.
Collapse
Affiliation(s)
- Matthew S Crouse
- Center for Nutrition and Pregnancy, Department of Animal Sciences, North Dakota State University, Fargo, ND
| | | | - Kyle J McLean
- Department of Animal Science, University of Tennessee, Knoxville, TN
| | | | - Nicolas Negrin Pereira
- Center for Nutrition and Pregnancy, Department of Animal Sciences, North Dakota State University, Fargo, ND
| | - Alison K Ward
- Center for Nutrition and Pregnancy, Department of Animal Sciences, North Dakota State University, Fargo, ND
| | - Lawrence P Reynolds
- Center for Nutrition and Pregnancy, Department of Animal Sciences, North Dakota State University, Fargo, ND
| | - Carl R Dahlen
- Center for Nutrition and Pregnancy, Department of Animal Sciences, North Dakota State University, Fargo, ND
| | | | - Pawel P Borowicz
- Center for Nutrition and Pregnancy, Department of Animal Sciences, North Dakota State University, Fargo, ND
| | - Joel S Caton
- Center for Nutrition and Pregnancy, Department of Animal Sciences, North Dakota State University, Fargo, ND
| |
Collapse
|
42
|
Dudenkov TM, Liu D, Cairns J, Devarajan S, Zhuang Y, Ingle JN, Buzdar AU, Robson ME, Kubo M, Batzler A, Barman P, Jenkins GD, Carlson EE, Goetz MP, Northfelt DW, Moreno-Aspitia A, Desta Z, Reid JM, Kalari KR, Wang L, Weinshilboum RM. Anastrozole Aromatase Inhibitor Plasma Drug Concentration Genome-Wide Association Study: Functional Epistatic Interaction Between SLC38A7 and ALPPL2. Clin Pharmacol Ther 2019; 106:219-227. [PMID: 30648747 PMCID: PMC6612579 DOI: 10.1002/cpt.1359] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 12/13/2018] [Indexed: 01/13/2023]
Abstract
Anastrozole is a widely prescribed aromatase inhibitor for the therapy of estrogen receptor positive (ER+) breast cancer. We performed a genome‐wide association study (GWAS) for plasma anastrozole concentrations in 687 postmenopausal women with ER+ breast cancer. The top single‐nucleotide polymorphism (SNP) signal mapped across SLC38A7 (rs11648166, P = 2.3E‐08), which we showed to encode an anastrozole influx transporter. The second most significant signal (rs28845026, P = 5.4E‐08) mapped near ALPPL2 and displayed epistasis with the SLC38A7 signal. Both of these SNPs were cis expression quantitative trait loci (eQTL)s for these genes, and patients homozygous for variant genotypes for both SNPs had the highest drug concentrations, the highest SLC38A7 expression, and the lowest ALPPL2 expression. In summary, our GWAS identified a novel gene encoding an anastrozole transporter, SLC38A7, as well as epistatic interaction between SNPs in that gene and SNPs near ALPPL2 that influenced both the expression of the transporter and anastrozole plasma concentrations.
Collapse
Affiliation(s)
- Tanda M Dudenkov
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Duan Liu
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Junmei Cairns
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Sandhya Devarajan
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Yongxian Zhuang
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - James N Ingle
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Aman U Buzdar
- Department of Breast Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Mark E Robson
- Breast Medicine Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Michiaki Kubo
- RIKEN Center for Integrative Medical Sciences, Yokohama City, Japan
| | - Anthony Batzler
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Poulami Barman
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Gregory D Jenkins
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Erin E Carlson
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Matthew P Goetz
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Donald W Northfelt
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona, USA
| | - Alvaro Moreno-Aspitia
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | - Zeruesenay Desta
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Joel M Reid
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Krishna R Kalari
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Liewei Wang
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Richard M Weinshilboum
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
43
|
Exchange-mode glutamine transport across CNS cell membranes. Neuropharmacology 2019; 161:107560. [PMID: 30853601 DOI: 10.1016/j.neuropharm.2019.03.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 02/28/2019] [Accepted: 03/02/2019] [Indexed: 12/18/2022]
Abstract
CNS cell membranes possess four transporters capable of exchanging Lglutamine (Gln) for other amino acids: the large neutral amino acid (LNAA) transporters LAT1 and LAT2, the hybrid basic amino acid (L-arginine (Arg), L-leucine (Leu)/LNAA transporter y+LAT2, and the L-alanine/L-serine/L-cysteine transporter 2 (ASCT2). LAT1/LAT2 and y+LAT2 are present in astrocytes, neurons and the blood brain barrier (BBB) - forming cerebral vascular endothelial cells (CVEC), while the location of ASCT2 in the individual cell types is a matter of debate. In the healthy brain, contribution of the exchangers to Gln shuttling from astrocytes to neurons and thus their role in controlling the conversion of Gln to the amino acid neurotransmitters l-glutamate (Glu) and γ-aminobutyric acid (GABA) and Gln flux across the BBB appears negligible as compared to the system A and system N uniporters. Insofar, except for the contribution of LAT1 to the maintenance of Gln homeostasis in the interstitial fluid (ISF), no well-defined CNS-specific function has been established for either of the three transporters in the healthy brain. The Gln-accepting amino acid exchangers appear to gain significance under conditions of excessive brain Gln load (glutaminosis). Excess Gln efflux across the BBB enhances influx into the brain of L-tryptophan (Trp). Excess of Trp is responsible for overloading the brain with neuroactive compounds: serotonin, kynurenic acid, quinolinic acid and/or oxindole, which contribute to neurotransmission imbalance accompanying hyperammonemia. In turn, alterations of y+LAT2-mediated Gln/Arg exchange and Arg uptake in astrocyte, modulate astrocytic nitric oxide synthesis and oxidative/nitrosative stress in ammonia-overexposed brain. This article is part of the issue entitled 'Special Issue on Neurotransmitter Transporters'.
Collapse
|
44
|
Freeman SA, Grinstein S. Resolution of macropinosomes, phagosomes and autolysosomes: Osmotically driven shrinkage enables tubulation and vesiculation. Traffic 2018; 19:965-974. [DOI: 10.1111/tra.12614] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 08/15/2018] [Accepted: 08/16/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Spencer A. Freeman
- Program in Cell Biology; Peter Gilgan Centre for Research and Learning, Hospital for Sick Children; Toronto Ontario Canada
| | - Sergio Grinstein
- Program in Cell Biology; Peter Gilgan Centre for Research and Learning, Hospital for Sick Children; Toronto Ontario Canada
- Keenan Research Centre of the Li Ka Shing Knowledge Institute, St. Michael's Hospital; Toronto Ontario Canada
- Department of Biochemistry; University of Toronto; Toronto Ontario Canada
| |
Collapse
|
45
|
Hellsten SV, Tripathi R, Ceder MM, Fredriksson R. Nutritional Stress Induced by Amino Acid Starvation Results in Changes for Slc38 Transporters in Immortalized Hypothalamic Neuronal Cells and Primary Cortex Cells. Front Mol Biosci 2018; 5:45. [PMID: 29868606 PMCID: PMC5952004 DOI: 10.3389/fmolb.2018.00045] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/18/2018] [Indexed: 01/01/2023] Open
Abstract
Amino acid sensing and signaling is vital for cells, and both gene expression and protein levels of amino acid transporters are regulated in response to amino acid availability. Here, the aim was to study the regulation of all members of the SLC38 amino acid transporter family, Slc38a1-11, in mouse brain cells following amino acid starvation. We reanalyzed microarray data for the immortalized hypothalamic cell line N25/2 subjected to complete amino acid starvation for 1, 2, 3, 5, or 16 h, focusing specifically on the SLC38 family. All 11 Slc38 genes were expressed in the cell line, and Slc38a1, Slc38a2, and Slc38a7 were significantly upregulated at 5 h and most strongly at 16 h. Here, protein level changes were measured for SLC38A7 and the orphan family member SLC38A11 which has not been studied under different amino acid starvation condition at protein level. At 5 h, no significant alteration on protein level for either SLC38A7 or SLC38A11 could be detected. In addition, primary embryonic cortex cells were deprived of nine amino acids, the most common amino acids transported by the SLC38 family members, for 3 h, 7 h or 12 h, and the gene expression was measured using qPCR. Slc38a1, Slc38a2, Slc38a5, Slc38a6, Slc38a9, and Slc38a10 were upregulated, while Slc38a3 and Slc38a7 were downregulated. Slc38a8 was upregulated at 5 h and downregulated at 12 h. In conclusion, several members from the SLC38 family are regulated depending on amino acid levels and are likely to be involved in amino acid sensing and signaling in brain.
Collapse
Affiliation(s)
- Sofie V Hellsten
- Molecular Neuropharmacology, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Rekha Tripathi
- Molecular Neuropharmacology, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Mikaela M Ceder
- Molecular Neuropharmacology, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Robert Fredriksson
- Molecular Neuropharmacology, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
46
|
Greseth NP, Crouse MS, McLean KJ, Crosswhite MR, Pereira NN, Dahlen CR, Borowicz PP, Reynolds LP, Ward AK, Neville BW, Caton JS. The effects of maternal nutrition on the messenger ribonucleic acid expression of neutral and acidic amino acid transporters in bovine uteroplacental tissues from day sixteen to fifty of gestation. J Anim Sci 2018; 95:4668-4676. [PMID: 29108050 DOI: 10.2527/jas2017.1713] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We hypothesized that both day of gestation and maternal nutrition would alter the relative mRNA expression of neutral and acid AA transporters , , , , and . Crossbred Angus heifers ( = 49) were synchronized, bred via AI, assigned to nutritional treatment (100% of NRC requirements for 0.45 kg/d gain [control heifers {CON}] and 60% of CON [restricted heifers {RES}]), and ovariohysterectomized on d 16, 34, or 50 of gestation ( = 6 to 9/d). Nonbred, nonpregnant (NB-NP) controls were ovariohysterectomized on d 16 of the estrous cycle ( = 6) after synchronization. The resulting arrangement was a 2 × 3 factorial + 1 (CON vs. RES × d 16, 34, or 50 + NB-NP controls). Tissues collected included caruncular endometrium (CAR), intercaruncular endometrium (ICAR), fetal membranes (FM; chorioallantois; d 16 and 34), cotyledonary placenta (COT; d 50 only), intercotyledonary placenta (ICOT; d 50 only), and amnion (AMN; d 50 only]). Relative expression of , , , , and was determined for each tissue using NB-NP CAR and NB-NP ICAR tissues for the baseline; for FM, endometrium from NB-NP controls served as the baseline. In CAR, no day × treatment interaction was observed ( > 0.05). However, day of gestation affected relative expression of , where expression on d 16 was greater ( < 0.01) than expression on d 34 and 50. Additionally, relative expression of and was greater ( ≤ 0.05) in pregnant heifers compared with NB-NP heifers. For ICAR, was influenced by a day × treatment interaction ( < 0.01), where expression in d 16 RES was greater ( ≤ 0.05) than that of any other day or nutritional treatment. Furthermore, expression in d 16 CON was greater ( ≤ 0.05) than that in d 50 RES, with those in d 34 CON and RES and d 50 CON being intermediate. In addition, was affected by day of gestation, where expression on d 16 was greater ( < 0.01) than that on d 34 and 50. A day × treatment interaction was not observed ( > 0.05) in FM; however, expression on d 34 was greater ( = 0.02) than on d 50, with that on d 16 being intermediate. Day of gestation also affected expression of , where expression on d 34 and 50 was greater ( < 0.01) than that on d 16. These data support our hypothesis in that both day of gestation and maternal nutrition affected the relative mRNA expression of AA transporter in ICAR, whereas day of gestation has a greater effect on the relative mRNA expression of other neutral and acidic AA transporters in the various tissues studied.
Collapse
|
47
|
Ekizoglu S, Seven D, Ulutin T, Guliyev J, Buyru N. Investigation of the SLC22A23 gene in laryngeal squamous cell carcinoma. BMC Cancer 2018; 18:477. [PMID: 29703252 PMCID: PMC5921549 DOI: 10.1186/s12885-018-4381-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 04/17/2018] [Indexed: 01/10/2023] Open
Abstract
Background Laryngeal squamous cell carcinoma (LSCC) is the second most common cancer of the head and neck. In order to identify differentially expressed genes which may have a role in LSCC carcinogenesis, we performed GeneFishing Assay. One of the differentially expressed genes was the SLC22A23 (solute carrier family 22, member 23) gene. SLC22A23 belongs to a family of organic ion transporters that are responsible for the absorption or excretion of many drugs, xenobiotics and endogenous compounds in a variety of tissues. SLC22A23 is expressed in a various tissues but no substrates or functions have been identified for it. Although the exact function is unknown, single nucleotide polymorphisms (SNPs) which are located in SLC22A23 gene were associated with inflammatory bowel disease (IBD), endometriosis-related infertility and the clearance of antipsychotic drugs. On the other hand SLC22A23 is identified as a prognostic gene to predict the recurrence of triple-negative breast cancer. Methods To understand the role of the SLC22A23 gene in laryngeal carcinogenesis, we investigated its mRNA expression level in laryngeal tumor tissue and adjacent non-cancerous tissue samples obtained from 83 patients by quantitative real-time PCR. To understand the association between SNPs in SLC22A23 and LSCC, selected genetic variations (rs4959235, rs6923667, rs9503518) were genotyped. Results We found that SLC22A23 expression was increased in 46 of 83 tumor tissues (55.4%) and was decreased in 30 of 83 (36.1%) tumor tissues compared to normal tissues. 77.2% of patients were homozygote for genotype rs9503518-AA and they most frequently had histological grade 2 and 3 tumors. We also found that rs9503518-AA genotype is associated with increased SLC22A23 expression. Conclusions Our results indicate that SLC22A23 may play a role in the development of laryngeal cancer.
Collapse
Affiliation(s)
- Seda Ekizoglu
- Cerrahpasa Medical Faculty, Department of Medical Biology, Istanbul University, Kocamustafapasa, 34098, Istanbul, Turkey
| | - Didem Seven
- Cerrahpasa Medical Faculty, Department of Medical Biology, Istanbul University, Kocamustafapasa, 34098, Istanbul, Turkey
| | - Turgut Ulutin
- Cerrahpasa Medical Faculty, Department of Medical Biology, Istanbul University, Kocamustafapasa, 34098, Istanbul, Turkey
| | - Jalal Guliyev
- Cerrahpasa Medical Faculty, Department of Otorhinolaryngology, Istanbul University, Istanbul, Turkey
| | - Nur Buyru
- Cerrahpasa Medical Faculty, Department of Medical Biology, Istanbul University, Kocamustafapasa, 34098, Istanbul, Turkey.
| |
Collapse
|
48
|
Lister A, Bourgeois S, Imenez Silva PH, Rubio-Aliaga I, Marbet P, Walsh J, Shelton LM, Keller B, Verrey F, Devuyst O, Giesbertz P, Daniel H, Goldring CE, Copple IM, Wagner CA, Odermatt A. NRF2 regulates the glutamine transporter Slc38a3 (SNAT3) in kidney in response to metabolic acidosis. Sci Rep 2018; 8:5629. [PMID: 29618784 PMCID: PMC5884861 DOI: 10.1038/s41598-018-24000-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 03/19/2018] [Indexed: 12/28/2022] Open
Abstract
Expression of the glutamine transporter SNAT3 increases in kidney during metabolic acidosis, suggesting a role during ammoniagenesis. Microarray analysis of Nrf2 knock-out (KO) mouse kidney identified Snat3 as the most significantly down-regulated transcript compared to wild-type (WT). We hypothesized that in the absence of NRF2 the kidney would be unable to induce SNAT3 under conditions of metabolic acidosis and therefore reduce the availability of glutamine for ammoniagenesis. Metabolic acidosis was induced for 7 days in WT and Nrf2 KO mice. Nrf2 KO mice failed to induce Snat3 mRNA and protein expression during metabolic acidosis. However, there were no differences in blood pH, bicarbonate, pCO2, chloride and calcium or urinary pH, ammonium and phosphate levels. Normal induction of ammoniagenic enzymes was observed whereas several amino acid transporters showed differential regulation. Moreover, Nrf2 KO mice during acidosis showed increased expression of renal markers of oxidative stress and injury and NRF2 activity was increased during metabolic acidosis in WT kidney. We conclude that NRF2 is required to adapt the levels of SNAT3 in response to metabolic acidosis. In the absence of NRF2 and SNAT3, the kidney does not have any major acid handling defect; however, increased oxidative stress and renal injury may occur.
Collapse
Affiliation(s)
- Adam Lister
- Department of Pharmaceutical Sciences, Division of Molecular and Systems Toxicology, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland.,National Center for Competence in Research Kidney.CH, Zürich, Switzerland
| | - Soline Bourgeois
- Institute of Physiology, Zürich Centre for Integrative Human Physiology, University of Zürich, Winterthurerstrasse 190, 8057, Zürich, Switzerland.,National Center for Competence in Research Kidney.CH, Zürich, Switzerland
| | - Pedro H Imenez Silva
- Institute of Physiology, Zürich Centre for Integrative Human Physiology, University of Zürich, Winterthurerstrasse 190, 8057, Zürich, Switzerland.,National Center for Competence in Research Kidney.CH, Zürich, Switzerland
| | - Isabel Rubio-Aliaga
- Institute of Physiology, Zürich Centre for Integrative Human Physiology, University of Zürich, Winterthurerstrasse 190, 8057, Zürich, Switzerland.,National Center for Competence in Research Kidney.CH, Zürich, Switzerland
| | - Philippe Marbet
- Department of Pharmaceutical Sciences, Division of Molecular and Systems Toxicology, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland.,National Center for Competence in Research Kidney.CH, Zürich, Switzerland
| | - Joanne Walsh
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Liverpool, L69 3GE, UK
| | - Luke M Shelton
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Liverpool, L69 3GE, UK
| | - Bettina Keller
- Institute of Physiology, Zürich Centre for Integrative Human Physiology, University of Zürich, Winterthurerstrasse 190, 8057, Zürich, Switzerland
| | - Francois Verrey
- Institute of Physiology, Zürich Centre for Integrative Human Physiology, University of Zürich, Winterthurerstrasse 190, 8057, Zürich, Switzerland.,National Center for Competence in Research Kidney.CH, Zürich, Switzerland
| | - Olivier Devuyst
- Institute of Physiology, Zürich Centre for Integrative Human Physiology, University of Zürich, Winterthurerstrasse 190, 8057, Zürich, Switzerland.,National Center for Competence in Research Kidney.CH, Zürich, Switzerland
| | - Pieter Giesbertz
- Department of Biochemistry, ZIEL Research Center of Nutrition and Food Sciences, Technische Universität München, Freising, Germany
| | - Hannelore Daniel
- Department of Biochemistry, ZIEL Research Center of Nutrition and Food Sciences, Technische Universität München, Freising, Germany
| | - Christopher E Goldring
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Liverpool, L69 3GE, UK
| | - Ian M Copple
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Liverpool, L69 3GE, UK
| | - Carsten A Wagner
- Institute of Physiology, Zürich Centre for Integrative Human Physiology, University of Zürich, Winterthurerstrasse 190, 8057, Zürich, Switzerland. .,National Center for Competence in Research Kidney.CH, Zürich, Switzerland.
| | - Alex Odermatt
- Department of Pharmaceutical Sciences, Division of Molecular and Systems Toxicology, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland. .,National Center for Competence in Research Kidney.CH, Zürich, Switzerland.
| |
Collapse
|
49
|
Wang XX, Li YB, Feng MR, Smith DE. Semi-Mechanistic Population Pharmacokinetic Modeling of L-Histidine Disposition and Brain Uptake in Wildtype and Pht1 Null Mice. Pharm Res 2018; 35:19. [PMID: 29305823 DOI: 10.1007/s11095-017-2322-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 11/22/2017] [Indexed: 01/06/2023]
Abstract
PURPOSE To develop a semi-mechanistic population pharmacokinetic (PK) model to quantitate the disposition kinetics of L-histidine, a peptide-histidine transporter 1 (PHT1) substrate, in the plasma, cerebrospinal fluid and brain parenchyma of wildtype (WT) and Pht1 knockout (KO) mice. METHODS L-[14C]Hisidine (L-His) was administrated to WT and KO mice via tail vein injection, after which plasma, cerebrospinal fluid (CSF) and brain parenchyma samples were collected. A PK model was developed using non-linear mixed effects modeling (NONMEM). The disposition of L-His between the plasma, brain, and CSF was described by a combination of PHT1-mediated uptake, CSF bulk flow and first-order micro-rate constants. RESULTS The PK profile of L-His was best described by a four-compartment model. A more rapid uptake of L-His in brain parenchyma was observed in WT mice due to PHT1-mediated uptake, a process characterized by a Michaelis-Menten component (Vmax = 0.051 nmoL/min and Km = 34.94 μM). CONCLUSIONS A semi-mechanistic population PK model was successfully developed, for the first time, to quantitatively characterize the disposition kinetics of L-His in brain under in vivo conditions. This model may prove a useful tool in predicting the uptake of L-His, and possibly other PHT1 peptide/mimetic substrates, for drug delivery to the brain.
Collapse
Affiliation(s)
- Xiao-Xing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, Michigan, 48109-1065, USA
| | - Yang-Bing Li
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Meihua R Feng
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, Michigan, 48109-1065, USA
| | - David E Smith
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, Michigan, 48109-1065, USA.
| |
Collapse
|
50
|
Bustillo JR, Patel V, Jones T, Jung R, Payaknait N, Qualls C, Canive JM, Liu J, Perrone-Bizzozero NI, Calhoun VD, Turner JA, Gasparovic C. Risk-Conferring Glutamatergic Genes and Brain Glutamate Plus Glutamine in Schizophrenia. Front Psychiatry 2017; 8:79. [PMID: 28659829 PMCID: PMC5466972 DOI: 10.3389/fpsyt.2017.00079] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 04/24/2017] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND The proton magnetic resonance spectroscopy (1H-MRS) signals from glutamate (or the combined glutamate and glutamine signal-Glx) have been found to be greater in various brain regions in people with schizophrenia. Recently, the Psychiatric Genetics Consortium reported that several common single-nucleotide polymorphisms (SNPs) in glutamate-related genes confer increased risk of schizophrenia. Here, we examined the relationship between presence of these risk polymorphisms and brain Glx levels in schizophrenia. METHODS 1H-MRS imaging data from an axial, supraventricular tissue slab were acquired in 56 schizophrenia patients and 67 healthy subjects. Glx was measured in gray matter (GM) and white matter (WM) regions. The genetic data included six polymorphisms genotyped across an Illumina 5M SNP array. Only three of six glutamate as well as calcium-related SNPs were available for examination. These included three glutamate-related polymorphisms (rs10520163 in CLCN3, rs12704290 in GRM3, and rs12325245 in SLC38A7), and three calcium signaling polymorphisms (rs1339227 in RIMS1, rs7893279 in CACNB2, and rs2007044 in CACNA1C). Summary risk scores for the three glutamate and the three calcium polymorphisms were calculated. RESULTS Glx levels in GM positively correlated with glutamate-related genetic risk score but only in younger (≤36 years) schizophrenia patients (p = 0.01). Glx levels did not correlate with calcium risk scores. Glx was higher in the schizophrenia group compared to levels in controls in GM and WM regardless of age (p < 0.001). CONCLUSION Elevations in brain Glx are in part, related to common allelic variants of glutamate-related genes known to increase the risk for schizophrenia. Since the glutamate risk scores did not differ between groups, some other genetic or environmental factors likely interact with the variability in glutamate-related risk SNPs to contribute to an increase in brain Glx early in the illness.
Collapse
Affiliation(s)
- Juan R. Bustillo
- Department of Psychiatry, University of New Mexico, Albuquerque, NM, United States
- Department of Behavioral Sciences, University of New Mexico, Albuquerque, NM, United States
- Department of Neurosciences, University of New Mexico, Albuquerque, NM, United States
| | - Veena Patel
- Mind Research Network, Albuquerque, NM, United States
| | - Thomas Jones
- Department of Psychiatry, University of New Mexico, Albuquerque, NM, United States
- Department of Behavioral Sciences, University of New Mexico, Albuquerque, NM, United States
| | - Rex Jung
- Department of Neurosurgery, University of New Mexico, Albuquerque, NM, United States
| | - Nattida Payaknait
- Department of Psychiatry, University of New Mexico, Albuquerque, NM, United States
- Department of Behavioral Sciences, University of New Mexico, Albuquerque, NM, United States
| | - Clifford Qualls
- Department of Mathematics and Statistics, University of New Mexico, Albuquerque, NM, United States
| | - Jose M. Canive
- Department of Psychiatry, University of New Mexico, Albuquerque, NM, United States
- Department of Behavioral Sciences, University of New Mexico, Albuquerque, NM, United States
- Department of Neurosciences, University of New Mexico, Albuquerque, NM, United States
- The New Mexico VA Health Care System, Albuquerque, NM, United States
| | - Jingyu Liu
- Mind Research Network, Albuquerque, NM, United States
- Department of Electrical Engineering, University of New Mexico, Albuquerque, NM, United States
| | - Nora Irma Perrone-Bizzozero
- Department of Psychiatry, University of New Mexico, Albuquerque, NM, United States
- Department of Behavioral Sciences, University of New Mexico, Albuquerque, NM, United States
- Department of Neurosciences, University of New Mexico, Albuquerque, NM, United States
| | - Vince D. Calhoun
- Department of Psychiatry, University of New Mexico, Albuquerque, NM, United States
- Department of Behavioral Sciences, University of New Mexico, Albuquerque, NM, United States
- Department of Neurosciences, University of New Mexico, Albuquerque, NM, United States
- Mind Research Network, Albuquerque, NM, United States
- Department of Electrical Engineering, University of New Mexico, Albuquerque, NM, United States
| | - Jessica A. Turner
- Mind Research Network, Albuquerque, NM, United States
- Department of Psychology, Georgia State University, Atlanta, GA, United States
| | | |
Collapse
|