1
|
Su J, Pan Y, Zhong F, Zhong Y, Huang J, Liu S, Wang K, Lin K, Gu X, Li D, Wu Q, Geng H, Guan Y, Xu G. Mitochondrial SLC3A1 regulates sexual dimorphism in cystinuria. Genes Dis 2025; 12:101472. [PMID: 40110490 PMCID: PMC11919626 DOI: 10.1016/j.gendis.2024.101472] [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: 04/16/2024] [Revised: 09/19/2024] [Accepted: 11/16/2024] [Indexed: 03/22/2025] Open
Abstract
Cystinuria is the most common inheritable cause of kidney stone disease, with males exhibiting a higher susceptibility than females. However, the cellular origin and underlying mechanisms of sex differences in cystinuria remain elusive. This study aims to investigate the mechanism using Slc3a1 knockout mice. We found that male mice lacking the Slc3a1 gene exhibited more severe stone formation and renal injuries, unaffected by double knockout of another sex-dependent-expressed cystine transporter Slc7a13 or orchidectomy procedure. Further investigations revealed aberrant mitochondrial functions as the primary factor contributing to the severity of cystinuria in Slc3a1 knockout male mice. Mechanistically, higher SLC3A1 levels in male kidneys could enhance mitochondrial functions through modulation of mitochondrial NAD+ uptake primarily in proximal tubule cells. Supplementation with an NAD+ precursor rescued the sex differences caused by Slc3a1 knockout. Our studies uncover the crucial role of Slc3a1 in mitochondrial functions and provide novel insights into potential interventions for sexual dimorphism of cystinuria.
Collapse
Affiliation(s)
- Jingyi Su
- Department of Pediatric Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yongdong Pan
- Department of Pediatric Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Fengbo Zhong
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yi Zhong
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jiaxin Huang
- Department of Pediatric Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Shengnan Liu
- Department of Pediatric Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Kaiyuan Wang
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Kai Lin
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xiangchen Gu
- Department of Nephrology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Dali Li
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Qihui Wu
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Hongquan Geng
- Department of Urology, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Yuting Guan
- Department of Pediatric Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
- Chongqing Key Laboratory of Precision Optics, Chongqing Institute of East China Normal University, Chongqing 401120, China
| | - Guofeng Xu
- Department of Pediatric Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| |
Collapse
|
2
|
Kasper C. Animal board invited review: Heritability of nitrogen use efficiency in fattening pigs: Current state and possible directions. Animal 2024; 18:101225. [PMID: 39013333 DOI: 10.1016/j.animal.2024.101225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 07/18/2024] Open
Abstract
Pork, an important component of human nutrition worldwide, contributes considerably to anthropogenic nitrogen and greenhouse gas emissions. Reducing the environmental impact of pig production is therefore essential. This can be achieved through system-level strategies, such as optimising resource use, improving manure management and recycling leftovers from human food production, and at the individual animal level by maintaining pig health and fine-tuning dietary protein levels to individual requirements. Breeding, coupled with nutritional strategies, offers a lasting solution to improve nitrogen use efficiency (NUE) - the ratio of nitrogen retained in the body to nitrogen ingested. With a heritability as high as 0.54, incorporating NUE into breeding programmes appears promising. Nitrogen use efficiency involves multiple tissues and metabolic processes, and is influenced by the environment and individual animal characteristics, including its genetic background. Heritable genetic variation in NUE may therefore occur in many different processes, including the central nervous regulation of feed intake, the endocrine system, the gastrointestinal tract where digestion and absorption take place, and the composition of the gut microbiome. An animal's postabsorptive protein metabolism might also harbour important genetic variation, especially in the maintenance requirements of tissues and organs. Precise phenotyping, although challenging and costly, is essential for successful breeding. Various measurement techniques, such as imaging techniques and mechanistic models, are being explored for their potential in genetic analysis. Despite the difficulties in phenotyping, some studies have estimated the heritability and genetic correlations of NUE. These studies suggest that direct selection for NUE is more effective than indirect methods through feed efficiency. The complexity of NUE indicates a polygenic trait architecture, which has been confirmed by genome-wide association studies that have been unable to identify significant quantitative trait loci. Building sufficiently large reference populations to train genomic prediction models is an important next step. However, this will require the development of truly high-throughput phenotyping methods. In conclusion, breeding pigs with higher NUE is both feasible and necessary but will require increased efforts in high-throughput phenotyping and improved genome annotation.
Collapse
Affiliation(s)
- C Kasper
- Animal GenoPhenomics, Agroscope, Posieux, Switzerland.
| |
Collapse
|
3
|
Huang M, Teng Q, Cao F, Huang J, Pang J. Ferroptosis and ferroptosis-inducing nanomedicine as a promising weapon in combination therapy of prostate cancer. Biomater Sci 2024; 12:1617-1629. [PMID: 38379396 DOI: 10.1039/d3bm01894f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Incidence and mortality of prostate cancer (PCa) rank in the top five among male tumors. However, single treatment modalities are often restricted due to biochemical recurrence and drug resistance, necessitating the development of new approaches for the combination treatment of castration-resistant and neuroendocrine PCa. Ferroptosis is characterized by the accumulation of iron-overload-mediated lipid peroxidation and has shown promising outcomes in anticancer treatment, prompting us to present a review reporting the application of ferroptosis in the treatment of PCa. First, the process and mechanism of ferroptosis are briefly reviewed. Second, research advances combining ferroptosis-inducing agents and clinical treatment regimens, which exhibit a "two-pronged approach" effect, are further summarized. Finally, the recent progress on ferroptosis-inducing nanomaterials for combination anticancer therapy is presented. This review is expected to provide novel insights into ferroptosis-based combination treatment in drug-resistant PCa.
Collapse
Affiliation(s)
- Mengjun Huang
- Department of Urology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.
| | - Qiliang Teng
- Department of Urology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.
| | - Fei Cao
- Department of Urology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.
| | - Jinsheng Huang
- Department of Urology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.
| | - Jun Pang
- Department of Urology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.
| |
Collapse
|
4
|
Liu D, Zhao Y, Xue X, Hou X, Xu H, Zhao X, Tian Y, Tang W, Guo J, Xu C. Novel compound heterozygous pathogenic variants in the SLC3A1 gene in a Chinese family with cystinuria. BMC Med Genomics 2023; 16:333. [PMID: 38114997 PMCID: PMC10731833 DOI: 10.1186/s12920-023-01767-6] [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: 10/08/2023] [Accepted: 12/05/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Cystinuria is an autosomal recessive disorder characterized by a cystine transport deficiency in the renal tubules due to mutations in two genes: SLC3A1 and SLC7A9. Cystinuria can be classified into three forms based on the genotype: type A, due to mutations in the SLC3A1 gene; type B, due to mutations in the SLC7A9 gene; and type AB, due to mutations in both genes. METHODS We report a 12-year-old boy from central China with cystine stones. He was from a non-consanguineous family that had no known history of genetic disease. A physical examination showed normal development and neurological behaviors. Whole-exome and Sanger sequencing were used to identify and verify the suspected pathogenic variants. RESULTS The compound heterozygous variants c.898_905del (p.Arg301AlafsTer6) is located in exon5 and c.1898_1899insAT (p.Asp634LeufsTer46) is located in exon10 of SLC3A1 (NM_000341.4) were deemed responsible for type A cystinuria family. The variant c.898_905del was reported in a Japanese patient in 2000, and the variant c.1898_1899insAT is novel. CONCLUSION A novel pathogenic heterozygous variant pair of the SLC3A1 gene was identified in a Chinese boy with type A cystinuria, enriching the mutational spectrum of the SLC3A1 gene. We attempted to find a pattern for the association between the genotype of SLC3A1 variants and the manifestations of cystinuria in patients with different onset ages. Our findings have important implications for genetic counseling and the early clinical diagnosis of cystinuria.
Collapse
Affiliation(s)
- Danhua Liu
- The Research and Application Center of Precision Medicine, The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, 450000, China
| | - Yongli Zhao
- Department of Urology, the Second Affiliated Hospital of Zhengzhou University, NO. 2 Jingba Road, Zhengzhou, 450014, China
| | - Xia Xue
- Henan Key Laboratory of Helicobacter Pylori & Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450002, China
| | - Xinyue Hou
- Precision Medicine Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Hongen Xu
- Precision Medicine Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Xinghua Zhao
- Department of Urology, the Second Affiliated Hospital of Zhengzhou University, NO. 2 Jingba Road, Zhengzhou, 450014, China
| | - Yongan Tian
- Precision Medicine Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Wenxue Tang
- The Research and Application Center of Precision Medicine, The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, 450000, China
- Precision Medicine Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
- Henan Institute of Medical and Pharmaceutical Sciences, BGI College, Zhengzhou University, Zhengzhou, 450052, China
| | - Jiancheng Guo
- The Research and Application Center of Precision Medicine, The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, 450000, China
- Precision Medicine Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
- Henan Institute of Medical and Pharmaceutical Sciences, BGI College, Zhengzhou University, Zhengzhou, 450052, China
| | - Changbao Xu
- Department of Urology, the Second Affiliated Hospital of Zhengzhou University, NO. 2 Jingba Road, Zhengzhou, 450014, China.
| |
Collapse
|
5
|
Kahlhofer J, Teis D. The human LAT1-4F2hc (SLC7A5-SLC3A2) transporter complex: Physiological and pathophysiological implications. Basic Clin Pharmacol Toxicol 2023; 133:459-472. [PMID: 36460306 PMCID: PMC11497297 DOI: 10.1111/bcpt.13821] [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: 09/19/2022] [Revised: 11/24/2022] [Accepted: 11/28/2022] [Indexed: 12/04/2022]
Abstract
LAT1 and 4F2hc form a heterodimeric membrane protein complex, which functions as one of the best characterized amino acid transporters. Since LAT1-4F2hc is required for the efficient uptake of essential amino acids and hormones, it promotes cellular growth, in part, by stimulating mTORC1 (mechanistic target of rapamycin complex 1) signalling and by repressing the integrated stress response (ISR). Gain or loss of LAT1-4F2hc function is associated with cancer, diabetes, and immunological and neurological diseases. Hence, LAT1-4F2hc represents an attractive drug target for disease treatment. Specific targeting of LAT1-4F2hc will be facilitated by the increasingly detailed understanding of its molecular architecture, which provides important concepts for its function and regulation. Here, we summarize (i) structural insights that help to explain how LAT1 and 4F2hc assemble to transport amino acids across membranes, (ii) the role of LAT1-4F2hc in key metabolic signalling pathways, and (iii) how derailing these processes could contribute to diseases.
Collapse
Affiliation(s)
- Jennifer Kahlhofer
- Institute for Cell Biology, BiocenterMedical University InnsbruckInnsbruckAustria
| | - David Teis
- Institute for Cell Biology, BiocenterMedical University InnsbruckInnsbruckAustria
| |
Collapse
|
6
|
Chen W, Yan Q, Zhong R, Tan Z. Amino acid profiles, amino acid sensors and transporters expression and intestinal microbiota are differentially altered in goats infected with Haemonchus contortus. Amino Acids 2023; 55:371-384. [PMID: 36648537 DOI: 10.1007/s00726-023-03235-y] [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: 08/23/2022] [Accepted: 01/06/2023] [Indexed: 01/18/2023]
Abstract
Infection with the nematode Haemonchus contortus causes host malnutrition and gastrointestinal injuries. The objective of this study was to investigate the effects of H. contortus infection on gastrointestinal contents of free amino acids (AA), the expression of AA transporters and microbiota with a focus on amino acid metabolism. Twenty-four Xiangdong black goats (13 ± 1.5 kg, 6 months old) were randomly assigned into the control group (n = 8) and the infected group (n = 16). The results showed that H. contortus infection increased (P < 0.05) the free AA contents in jejunum and ileum digesta. The concentrations of blood threonine, phenylalanine and tyrosine were lower (P < 0.05) in the infected group as compared to the control group. In the jejunum and ileum epithelium, H. contortus infection significantly (P < 0.05) down-regulated the expression of AA transporter b0,+AT/rBAT and B0AT1, but up-regulated (P < 0.05) the expression of transporter CAT2 and xCT. Furthermore, microbiota in both jejunum (Bifidobacteriaceae, Lachnospiraceae, Bacteroidaceae, Enterobacteriaceae, and Micrococcaceae) and ileum (Acidaminococcaceae, Desulfovibrionaceae, Bacteroidaceae, and Peptostreptococcaceae) were also altered at the family level by H. contortus infection. The commensal bacteria of jejunum showed a close correlation with amino acids, AA transporters, and amino acid metabolism, especially cystine. In conclusion, H. contortus infection affected the intestinal AA contents and the expression of intestinal AA transporters, suggesting altered AA metabolism and absorption, which were accompanied by changes in the relative abundances of gut bacteria that mediate amino acid metabolism.
Collapse
Affiliation(s)
- Wenxun Chen
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, 410125, Hunan, People's Republic of China
- University of Chinese Academy of Science, Beijing, 100049, People's Republic of China
| | - Qiongxian Yan
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, 410125, Hunan, People's Republic of China.
| | - Rongzhen Zhong
- Jilin Provincial Key Laboratory of Grassland Farming, Northeast Institute of Geography and Agroecology, Chinese Academy of Sciences, Changchun, 130102, Jilin, People's Republic of China
| | - Zhiliang Tan
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, 410125, Hunan, People's Republic of China
| |
Collapse
|
7
|
Yamasaki A, Maruyama-Takahashi K, Nishida K, Okazaki S, Okita K, Akiyama Y, Suzuki H, Endo Y, Masuko K, Masuko T, Tomioka Y. CD98 regulates the phosphorylation of HER2 and a bispecific anti-HER2/CD98 antibody inhibits the growth signal of human breast cancer cells. Genes Cells 2023; 28:374-382. [PMID: 36811310 DOI: 10.1111/gtc.13016] [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: 01/09/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023]
Abstract
Human epidermal growth factor receptor (HER) family proteins are currently major targets of therapeutic monoclonal antibodies against various epithelial cancers. However, the resistance of cancer cells to HER family-targeted therapies, which may be caused by cancer heterogeneity and persistent HER phosphorylation, often reduces overall therapeutic effects. We herein showed that a newly discovered molecular complex between CD98 and HER2 affected HER function and cancer cell growth. The immunoprecipitation of the HER2 or HER3 protein from lysates of SKBR3 breast cancer (BrCa) cells revealed the HER2-CD98 or HER3-CD98 complex. The knockdown of CD98 by small interfering RNAs inhibited the phosphorylation of HER2 in SKBR3 cells. A bispecific antibody (BsAb) that recognized the HER2 and CD98 proteins was constructed from a humanized anti-HER2 (SER4) IgG and an anti-CD98 (HBJ127) single chain variable fragment, and this BsAb significantly inhibited the cell growth of SKBR3 cells. Prior to the inhibition of AKT phosphorylation, BsAb inhibited the phosphorylation of HER2, however, significant inhibition of HER2 phosphorylation was not observed in anti-HER2 pertuzumab, trastuzumab, SER4 or anti-CD98 HBJ127 in SKBR3 cells. The dual targeting of HER2 and CD98 has potential as a new therapeutic strategy for BrCa.
Collapse
Affiliation(s)
- Akitaka Yamasaki
- Laboratory of Oncology Pharmacy Practice and Science, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai-shi, Japan.,Cell Biology Laboratory, Faculty of Pharmacy, Kindai University, Higashiosaka-shi, Japan
| | - Kumiko Maruyama-Takahashi
- Department of Hygienic Chemistry (April 1975-March 1994), Pharmaceutical Institute, Tohoku University, Sendai-shi, Japan
| | - Kento Nishida
- Laboratory of Oncology Pharmacy Practice and Science, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai-shi, Japan
| | - Shogo Okazaki
- Cell Biology Laboratory, Faculty of Pharmacy, Kindai University, Higashiosaka-shi, Japan.,Department of Microbiology, Division of Immunology and Pathobiology, Nihon University School of Density, Chiyoda-ku, Japan
| | - Kouki Okita
- Cell Biology Laboratory, Faculty of Pharmacy, Kindai University, Higashiosaka-shi, Japan.,Production and Manufacturing, Carna Biosciences Inc., Chuo-ku, Japan
| | - Yasutoshi Akiyama
- Laboratory of Oncology Pharmacy Practice and Science, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai-shi, Japan
| | - Hideaki Suzuki
- Department of Hygienic Chemistry (April 1975-March 1994), Pharmaceutical Institute, Tohoku University, Sendai-shi, Japan
| | - Yuichi Endo
- Laboratory of Natural Drug Resources, Faculty of Pharmacy, Kindai University, Higashiosaka-shi, Japan
| | - Kazue Masuko
- Cell Biology Laboratory, Faculty of Pharmacy, Kindai University, Higashiosaka-shi, Japan.,Department of Hygienic Chemistry (April 1975-March 1994), Pharmaceutical Institute, Tohoku University, Sendai-shi, Japan
| | - Takashi Masuko
- Cell Biology Laboratory, Faculty of Pharmacy, Kindai University, Higashiosaka-shi, Japan.,Department of Hygienic Chemistry (April 1975-March 1994), Pharmaceutical Institute, Tohoku University, Sendai-shi, Japan.,Laboratory of Natural Drug Resources, Faculty of Pharmacy, Kindai University, Higashiosaka-shi, Japan
| | - Yoshihisa Tomioka
- Laboratory of Oncology Pharmacy Practice and Science, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai-shi, Japan.,Department of Hygienic Chemistry (April 1975-March 1994), Pharmaceutical Institute, Tohoku University, Sendai-shi, Japan
| |
Collapse
|
8
|
Insights into the Transport Cycle of LAT1 and Interaction with the Inhibitor JPH203. Int J Mol Sci 2023; 24:ijms24044042. [PMID: 36835453 PMCID: PMC9965313 DOI: 10.3390/ijms24044042] [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: 12/30/2022] [Revised: 02/08/2023] [Accepted: 02/15/2023] [Indexed: 02/19/2023] Open
Abstract
The large Amino Acid Transporter 1 (LAT1) is an interesting target in drug discovery since this transporter is overexpressed in several human cancers. Furthermore, due to its location in the blood-brain barrier (BBB), LAT1 is interesting for delivering pro-drugs to the brain. In this work, we focused on defining the transport cycle of LAT1 using an in silico approach. So far, studies of the interaction of LAT1 with substrates and inhibitors have not considered that the transporter must undergo at least four different conformations to complete the transport cycle. We built outward-open and inward-occluded conformations of LAT1 using an optimized homology modelling procedure. We used these 3D models and the cryo-EM structures in outward-occluded and inward-open conformations to define the substrate/protein interaction during the transport cycle. We found that the binding scores for the substrate depend on the conformation, with the occluded states as the crucial steps affecting the substrate affinity. Finally, we analyzed the interaction of JPH203, a high-affinity inhibitor of LAT1. The results indicate that conformational states must be considered for in silico analyses and early-stage drug discovery. The two built models, together with the available cryo-EM 3D structures, provide important information on the LAT1 transport cycle, which could be used to speed up the identification of potential inhibitors through in silico screening.
Collapse
|
9
|
Gyimesi G, Hediger MA. Transporter-Mediated Drug Delivery. Molecules 2023; 28:molecules28031151. [PMID: 36770817 PMCID: PMC9919865 DOI: 10.3390/molecules28031151] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/12/2023] [Accepted: 01/18/2023] [Indexed: 01/27/2023] Open
Abstract
Transmembrane transport of small organic and inorganic molecules is one of the cornerstones of cellular metabolism. Among transmembrane transporters, solute carrier (SLC) proteins form the largest, albeit very diverse, superfamily with over 400 members. It was recognized early on that xenobiotics can directly interact with SLCs and that this interaction can fundamentally determine their efficacy, including bioavailability and intertissue distribution. Apart from the well-established prodrug strategy, the chemical ligation of transporter substrates to nanoparticles of various chemical compositions has recently been used as a means to enhance their targeting and absorption. In this review, we summarize efforts in drug design exploiting interactions with specific SLC transporters to optimize their therapeutic effects. Furthermore, we describe current and future challenges as well as new directions for the advanced development of therapeutics that target SLC transporters.
Collapse
|
10
|
Hang TD, Hung HM, Beckers P, Desmet N, Lamrani M, Massie A, Hermans E, Vanommeslaeghe K. Structural investigation of human cystine/glutamate antiporter system xc− (Sxc−) using homology modeling and molecular dynamics. Front Mol Biosci 2022; 9:1064199. [DOI: 10.3389/fmolb.2022.1064199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 11/15/2022] [Indexed: 12/04/2022] Open
Abstract
The cystine/glutamate antiporter system xc− (Sxc−) belongs to the SLC7 family of plasma membrane transporters. It exports intracellular glutamate along the latter’s concentration gradient as a driving force for cellular uptake of cystine. Once imported, cystine is mainly used for the production of glutathione, a tripeptide thiol crucial in maintenance of redox homeostasis and protection of cells against oxidative stress. Overexpression of Sxc− has been found in several cancer cells, where it is thought to counteract the increased oxidative stress. In addition, Sxc− is important in the central nervous system, playing a complex role in regulating glutamatergic neurotransmission and glutamate toxicity. Accordingly, this transporter is considered a potential target for the treatment of cancer as well as neurodegenerative diseases. Till now, no specific inhibitors are available. We herein present four conformations of Sxc− along its transport pathway, obtained using multi-template homology modeling and refined by means of Molecular Dynamics. Comparison with a very recently released cryo-EM structure revealed an excellent agreement with our inward-open conformation. Intriguingly, our models contain a structured N-terminal domain that is unresolved in the experimental structures and is thought to play a gating role in the transport mechanism of other SLC7 family members. In contrast to the inward-open model, there is no direct experimental counterpart for the other three conformations we obtained, although they are in fair agreement with the other stages of the transport mechanism seen in other SLC7 transporters. Therefore, our models open the prospect for targeting alternative Sxc− conformations in structure-based drug design efforts.
Collapse
|
11
|
Şeker E, Süt H, Özışık S, Koç A. Can prenatal renal pelvicalyceal echogenic foci support the diagnosis of cystinuria? J Turk Ger Gynecol Assoc 2022; 23:327-329. [DOI: 10.4274/jtgga.galenos.2022.2021-11-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
12
|
Console L, Scalise M, Salerno S, Scanga R, Giudice D, De Bartolo L, Tonazzi A, Indiveri C. N-glycosylation is crucial for trafficking and stability of SLC3A2 (CD98). Sci Rep 2022; 12:14570. [PMID: 36028562 PMCID: PMC9418156 DOI: 10.1038/s41598-022-18779-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/18/2022] [Indexed: 11/30/2022] Open
Abstract
The type II glycoprotein CD98 (SLC3A2) is a membrane protein with pleiotropic roles in cells, ranging from modulation of inflammatory processes, host–pathogen interactions to association with membrane transporters of the SLC7 family. The recent resolution of CD98 structure in complex with LAT1 showed that four Asn residues, N365, N381, N424, N506, harbour N-glycosylation moieties. Then, the role of N-glycosylation on CD98 trafficking and stability was investigated by combining bioinformatics, site-directed mutagenesis and cell biology approach. Single, double, triple and quadruple mutants of the four Asn exhibited altered electrophoretic mobility, with apparent molecular masses from 95 to 70 kDa. The quadruple mutant displayed a single band of 70 kDa corresponding to the unglycosylated protein. The presence in the membrane and the trafficking of CD98 were evaluated by a biotinylation assay and a brefeldin assay, respectively. Taken together, the results highlighted that the quadruple mutation severely impaired both the stability and the trafficking of CD98 to the plasma membrane. The decreased presence of CD98 at the plasma membrane, correlated with a lower presence of LAT1 (SLC7A5) and its transport activity. This finding opens new perspectives for human therapy. Indeed, the inhibition of CD98 trafficking would act synergistically with LAT1 inhibitors that are under clinical trial for anticancer therapy.
Collapse
Affiliation(s)
- Lara Console
- Department DiBEST (Biologia, Ecologia, Scienze Della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via Bucci 4C, 87036, Arcavacata di Rende, Italy
| | - Mariafrancesca Scalise
- Department DiBEST (Biologia, Ecologia, Scienze Della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via Bucci 4C, 87036, Arcavacata di Rende, Italy
| | - Simona Salerno
- CNR Institute on Membrane Technology, National Research Council of Italy (CNR-ITM), Via P. Bucci, cubo 17/C, 87036, Rende, Italy
| | - Raffaella Scanga
- Department DiBEST (Biologia, Ecologia, Scienze Della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via Bucci 4C, 87036, Arcavacata di Rende, Italy
| | - Deborah Giudice
- Department DiBEST (Biologia, Ecologia, Scienze Della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via Bucci 4C, 87036, Arcavacata di Rende, Italy
| | - Loredana De Bartolo
- CNR Institute on Membrane Technology, National Research Council of Italy (CNR-ITM), Via P. Bucci, cubo 17/C, 87036, Rende, Italy
| | - Annamaria Tonazzi
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), Via Amendola 122/O, 70126, Bari, Italy
| | - Cesare Indiveri
- Department DiBEST (Biologia, Ecologia, Scienze Della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via Bucci 4C, 87036, Arcavacata di Rende, Italy. .,CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), Via Amendola 122/O, 70126, Bari, Italy.
| |
Collapse
|
13
|
D'Ambrosio V, Capolongo G, Goldfarb D, Gambaro G, Ferraro PM. Cystinuria: an update on pathophysiology, genetics, and clinical management. Pediatr Nephrol 2022; 37:1705-1711. [PMID: 34812923 DOI: 10.1007/s00467-021-05342-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 09/22/2021] [Accepted: 10/12/2021] [Indexed: 11/25/2022]
Abstract
Cystinuria is the most common genetic cause of nephrolithiasis in children. It is considered a heritable aminoaciduria as the genetic defect affects the reabsorption of cystine and three other amino acids (ornithine, lysine, and arginine) in the renal proximal tubule. Patients affected by this condition have elevated excretion of cystine in the urine, and because of this amino acid's low solubility at normal urine pH, patients tend to form cystine calculi. To date, two genes have been identified as disease-causative: SLC3A1 and SLC7A9, encoding for the two subunits of the heterodimeric transporter. The clinical features of this condition are solely related to nephrolithiasis. The diagnosis is usually made during infancy or adolescence, but cases of late diagnosis are common. The goal of therapy is to reduce excretion and increase the solubility of cystine, through both modifications of dietary habits and pharmacological treatment. However, therapeutic interventions are not always sufficient, and patients often have to undergo several surgical procedures during their lives to treat recurrent nephrolithiasis. The goal of this literature review is to synthesize the available evidence on diagnosis and management of patients affected by cystinuria in order to provide physicians with a practical tool that can be used in daily clinical practice. This review also aims to shed some light on new therapy directions with the aim of ameliorating kidney outcomes while improving adherence to treatment and quality of life of cystinuric patients.
Collapse
Affiliation(s)
- Viola D'Ambrosio
- Dipartimento Di Scienze Mediche E Chirurgiche, U.O.S. Terapia Conservativa Della Malattia Renale Cronica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, U.O.C. Nefrologia, 00168, Rome, Italy
- Dipartimento Universitario Di Medicina E Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giovanna Capolongo
- Department of Translational Medical Sciences, Unit of Nephrology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - David Goldfarb
- Nephrology Section, VA, New York Harbor Healthcare System, New York, NY, USA
- Division of Nephrology, New York University Langone Medical Center, New York, NY, USA
| | - Giovanni Gambaro
- Department of Medicine, Division of Nephrology and Dialysis, Renal Unit, University of Verona, Verona, Italy
| | - Pietro Manuel Ferraro
- Dipartimento Di Scienze Mediche E Chirurgiche, U.O.S. Terapia Conservativa Della Malattia Renale Cronica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, U.O.C. Nefrologia, 00168, Rome, Italy.
- Dipartimento Universitario Di Medicina E Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy.
| |
Collapse
|
14
|
Shan K, Feng N, Zhu D, Qu H, Fu G, Li J, Cui J, Chen H, Wang R, Qi Y, Chen YQ. Free docosahexaenoic acid promotes ferroptotic cell death via lipoxygenase dependent and independent pathways in cancer cells. Eur J Nutr 2022; 61:4059-4075. [PMID: 35804267 DOI: 10.1007/s00394-022-02940-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 06/15/2022] [Indexed: 02/07/2023]
Abstract
PURPOSE Ferroptosis is a form of regulated cell death that has the potential to be targeted as a cancer therapeutic strategy. But cancer cells have a wide range of sensitivities to ferroptosis, which limits its therapeutic potential. Accumulation of lipid peroxides determines the occurrence of ferroptosis. However, the type of lipid involved in peroxidation and the mechanism of lipid peroxide accumulation are less studied. METHODS The effects of fatty acids (10 μM) with different carbon chain length and unsaturation on ferroptosis were evaluated by MTT and LDH release assay in cell lines derived from prostate cancer (PC3, 22RV1, DU145 and LNCaP), colorectal cancer (HT-29), cervical cancer (HeLa) and liver cancer (HepG2). Inhibitors of apoptosis, necroptosis, autophagy and ferroptosis were used to determine the type of cell death. Then the regulation of reactive oxygen species (ROS) and lipid peroxidation by docosahexaenoic acid (DHA) was measured by HPLC-MS and flow cytometry. The avtive form of DHA was determined by siRNA mediated gene silencing. The role of lipoxygenases was checked by inhibitors and gene silencing. Finally, the effect of DHA on ferroptosis-mediated tumor killing was verified in xenografts. RESULTS The sensitivity of ferroptosis was positively correlated with the unsaturation of exogenously added fatty acid. DHA (22:6 n-3) sensitized cancer cells to ferroptosis-inducing reagents (FINs) at the highest level in vitro and in vivo. In this process, DHA increased ROS accumulation, lipid peroxidation and protein oxidation independent of its membrane receptor, GPR120. Inhibition of long chain fatty acid-CoA ligases and lysophosphatidylcholine acyltransferases didn't affect the role of DHA. DHA-involved ferroptosis can be induced in both arachidonate lipoxygenase 5 (ALOX5) negative and positive cells. Down regulation of ALOX5 inhibited ferroptosis, while overexpression of ALOX5 promoted ferroptosis. CONCLUSION DHA can effectively promote ferroptosis-mediated tumor killing by increasing intracellular lipid peroxidation. Both ALOX5 dependent and independent pathways are involved in DHA-FIN induced ferroptosis. And during this process, free DHA plays an important role.
Collapse
Affiliation(s)
- Kai Shan
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
| | - Ninghan Feng
- Department of Urology, Wuxi No. 2 People's Hospital, Wuxi, 214000, Jiangsu Province, China
| | - Doudou Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
| | - Hongyan Qu
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
| | - Guoling Fu
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
| | - Jiaqi Li
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
| | - Jing Cui
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
| | - Heyan Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
| | - Rong Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
| | - Yumin Qi
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
| | - Yong Q Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu Province, China.
| |
Collapse
|
15
|
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
|
16
|
Fort J, Nicolàs-Aragó A, Palacín M. The Ectodomains of rBAT and 4F2hc Are Fake or Orphan α-Glucosidases. Molecules 2021; 26:6231. [PMID: 34684812 PMCID: PMC8537225 DOI: 10.3390/molecules26206231] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 11/22/2022] Open
Abstract
It is known that 4F2hc and rBAT are the heavy subunits of the heteromeric amino acid transporters (HATs). These heavy subunits are N-glycosylated proteins, with an N-terminal domain, one transmembrane domain and a bulky extracellular domain (ectodomain) that belongs to the α-amylase family. The heavy subunits are covalently linked to a light subunit from the SLC7 family, which is responsible for the amino acid transport activity, forming a heterodimer. The functions of 4F2hc and rBAT are related mainly to the stability and trafficking of the HATs in the plasma membrane of vertebrates, where they exert the transport activity. Moreover, 4F2hc is a modulator of integrin signaling, has a role in cell fusion and it is overexpressed in some types of cancers. On the other hand, some mutations in rBAT are found to cause the malfunctioning of the b0,+ transport system, leading to cystinuria. The ectodomains of 4F2hc and rBAT share both sequence and structure homology with α-amylase family members. Very recently, cryo-EM has revealed the structure of several HATs, including the ectodomains of rBAT and 4F2hc. Here, we analyze available data on the ectodomains of rBAT and 4Fhc and their relationship with the α-amylase family. The physiological relevance of this relationship remains largely unknown.
Collapse
Affiliation(s)
- Joana Fort
- Laboratory of Amino Acid Transporters and Disease, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028 Barcelona, Spain; (A.N.-A.); (M.P.)
- CIBERER (Centro Español en Red de Biomedicina de Enfermedades Raras), 08028 Barcelona, Spain
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Adrià Nicolàs-Aragó
- Laboratory of Amino Acid Transporters and Disease, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028 Barcelona, Spain; (A.N.-A.); (M.P.)
| | - Manuel Palacín
- Laboratory of Amino Acid Transporters and Disease, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028 Barcelona, Spain; (A.N.-A.); (M.P.)
- CIBERER (Centro Español en Red de Biomedicina de Enfermedades Raras), 08028 Barcelona, Spain
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, 08028 Barcelona, Spain
| |
Collapse
|
17
|
Kovaříková S, Maršálek P, Vrbová K. Cystinuria in Dogs and Cats: What Do We Know after Almost 200 Years? Animals (Basel) 2021; 11:2437. [PMID: 34438894 PMCID: PMC8388795 DOI: 10.3390/ani11082437] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/08/2021] [Accepted: 08/13/2021] [Indexed: 11/18/2022] Open
Abstract
The purpose of this review is to summarize current knowledge on canine and feline cystinuria from available scientific reports. Cystinuria is an inherited metabolic defect characterized by abnormal intestinal and renal amino acid transport in which cystine and the dibasic amino acids ornithine, lysine, and arginine are involved (COLA). At a normal urine pH, ornithine, lysine, and arginine are soluble, but cysteine forms a dimer, cystine, which is relatively insoluble, resulting in crystal precipitation. Mutations in genes coding COLA transporter and the mode of inheritance were identified only in some canine breeds. Cystinuric dogs may form uroliths (mostly in lower urinary tract) which are associated with typical clinical symptoms. The prevalence of cystine urolithiasis is much higher in European countries (up to 14% according to the recent reports) when compared to North America (United States and Canada) where it is approximately 1-3%. Cystinuria may be diagnosed by the detection of cystine urolithiasis, cystine crystalluria, assessment of amino aciduria, or using genetic tests. The management of cystinuria is aimed at urolith removal or dissolution which may be reached by dietary changes or medical treatment. In dogs with androgen-dependent cystinuria, castration will help. In cats, cystinuria occurs less frequently in comparison with dogs.
Collapse
Affiliation(s)
- Simona Kovaříková
- Department of Animal Protection and Welfare and Veterinary Public Health, Faculty of Veterinary Hygiene and Ecology, University of Veterinary Sciences, 612 42 Brno, Czech Republic;
| | - Petr Maršálek
- Department of Animal Protection and Welfare and Veterinary Public Health, Faculty of Veterinary Hygiene and Ecology, University of Veterinary Sciences, 612 42 Brno, Czech Republic;
| | - Kateřina Vrbová
- Faculty of Veterinary Medicine, University of Veterinary Sciences, 612 42 Brno, Czech Republic;
| |
Collapse
|
18
|
Citrulline, Biomarker of Enterocyte Functional Mass and Dietary Supplement. Metabolism, Transport, and Current Evidence for Clinical Use. Nutrients 2021; 13:nu13082794. [PMID: 34444954 PMCID: PMC8398474 DOI: 10.3390/nu13082794] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/28/2021] [Accepted: 07/28/2021] [Indexed: 12/25/2022] Open
Abstract
L-Citrulline is a non-essential but still important amino acid that is released from enterocytes. Because plasma levels are reduced in case of impaired intestinal function, it has become a biomarker to monitor intestinal integrity. Moreover, oxidative stress induces protein citrullination, and antibodies against anti-citrullinated proteins are useful to monitor rheumatoid diseases. Citrullinated histones, however, may even predict a worse outcome in cancer patients. Supplementation of citrulline is better tolerated compared to arginine and might be useful to slightly improve muscle strength or protein balance. The following article shall provide an overview of L-citrulline properties and functions, as well as the current evidence for its use as a biomarker or as a therapeutic supplement.
Collapse
|
19
|
Fairweather SJ, Shah N, Brӧer S. Heteromeric Solute Carriers: Function, Structure, Pathology and Pharmacology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 21:13-127. [PMID: 33052588 DOI: 10.1007/5584_2020_584] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Solute carriers form one of three major superfamilies of membrane transporters in humans, and include uniporters, exchangers and symporters. Following several decades of molecular characterisation, multiple solute carriers that form obligatory heteromers with unrelated subunits are emerging as a distinctive principle of membrane transporter assembly. Here we comprehensively review experimentally established heteromeric solute carriers: SLC3-SLC7 amino acid exchangers, SLC16 monocarboxylate/H+ symporters and basigin/embigin, SLC4A1 (AE1) and glycophorin A exchanger, SLC51 heteromer Ost α-Ost β uniporter, and SLC6 heteromeric symporters. The review covers the history of the heteromer discovery, transporter physiology, structure, disease associations and pharmacology - all with a focus on the heteromeric assembly. The cellular locations, requirements for complex formation, and the functional role of dimerization are extensively detailed, including analysis of the first complete heteromer structures, the SLC7-SLC3 family transporters LAT1-4F2hc, b0,+AT-rBAT and the SLC6 family heteromer B0AT1-ACE2. We present a systematic analysis of the structural and functional aspects of heteromeric solute carriers and conclude with common principles of their functional roles and structural architecture.
Collapse
Affiliation(s)
- Stephen J Fairweather
- Research School of Biology, Australian National University, Canberra, ACT, Australia. .,Resarch School of Chemistry, Australian National University, Canberra, ACT, Australia.
| | - Nishank Shah
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Stefan Brӧer
- Research School of Biology, Australian National University, Canberra, ACT, Australia.
| |
Collapse
|
20
|
Galagovsky D, Depetris-Chauvin A, Manière G, Geillon F, Berthelot-Grosjean M, Noirot E, Alves G, Grosjean Y. Sobremesa L-type Amino Acid Transporter Expressed in Glia Is Essential for Proper Timing of Development and Brain Growth. Cell Rep 2019; 24:3156-3166.e4. [PMID: 30231999 PMCID: PMC6167638 DOI: 10.1016/j.celrep.2018.08.067] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 07/13/2018] [Accepted: 08/22/2018] [Indexed: 02/07/2023] Open
Abstract
In Drosophila, ecdysone hormone levels determine the timing of larval development. Its production is regulated by the stereotypical rise in prothoracicotropic hormone (PTTH) levels. Additionally, ecdysone levels can also be modulated by nutrition (specifically by amino acids) through their action on Drosophila insulin-like peptides (Dilps). Moreover, in glia, amino-acid-sensitive production of Dilps regulates brain development. In this work, we describe the function of an SLC7 amino acid transporter, Sobremesa (Sbm). Larvae with reduced Sbm levels in glia remain in third instar for an additional 24 hr. These larvae show reduced brain growth with increased body size but do not show reduction in insulin signaling or production. Interestingly, Sbm downregulation in glia leads to reduced Ecdysone production and a surprising delay in the rise of PTTH levels. Our work highlights Sbm as a modulator of both brain development and the timing of larval development via an amino-acid-sensitive and Dilp-independent function of glia. Glia express the SLC7 amino acid transporter Sobremesa, which controls development Sobremesa downregulation in glia leads to contrasting effects: small brain and big body size Sobremesa downregulation results in reduced ecdysone production Sobremesa downregulation causes a delayed rise in PTTH
Collapse
Affiliation(s)
- Diego Galagovsky
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, 21000 Dijon, France
| | - Ana Depetris-Chauvin
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, 21000 Dijon, France; Department of Evolutionary Neuroethology, Max Planck Institute for Chemical Ecology, 07745 Jena, Germany
| | - Gérard Manière
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, 21000 Dijon, France
| | - Flore Geillon
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, 21000 Dijon, France
| | - Martine Berthelot-Grosjean
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, 21000 Dijon, France
| | - Elodie Noirot
- Plateforme DImaCell, INRA, Université Bourgogne Franche-Comté, 21000 Dijon, France
| | - Georges Alves
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, 21000 Dijon, France
| | - Yael Grosjean
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, 21000 Dijon, France.
| |
Collapse
|
21
|
Inducible Slc7a7 Knockout Mouse Model Recapitulates Lysinuric Protein Intolerance Disease. Int J Mol Sci 2019; 20:ijms20215294. [PMID: 31653080 PMCID: PMC6862226 DOI: 10.3390/ijms20215294] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 12/30/2022] Open
Abstract
Lysinuric protein intolerance (LPI) is a rare autosomal disease caused by defective cationic amino acid (CAA) transport due to mutations in SLC7A7, which encodes for the y+LAT1 transporter. LPI patients suffer from a wide variety of symptoms, which range from failure to thrive, hyperammonemia, and nephropathy to pulmonar alveolar proteinosis (PAP), a potentially life-threatening complication. Hyperammonemia is currently prevented by citrulline supplementation. However, the full impact of this treatment is not completely understood. In contrast, there is no defined therapy for the multiple reported complications of LPI, including PAP, for which bronchoalveolar lavages do not prevent progression of the disease. The lack of a viable LPI model prompted us to generate a tamoxifen-inducible Slc7a7 knockout mouse (Slc7a7-/-). The Slc7a7-/- model resembles the human LPI phenotype, including malabsorption and impaired reabsorption of CAA, hypoargininemia and hyperammonemia. Interestingly, the Slc7a7-/- mice also develops PAP and neurological impairment. We observed that citrulline treatment improves the metabolic derangement and survival. On the basis of our findings, the Slc7a7-/- model emerges as a promising tool to further study the complexity of LPI, including its immune-like complications, and to design evidence-based therapies to halt its progression.
Collapse
|
22
|
Abstract
The small intestine mediates the absorption of amino acids after ingestion of protein and sustains the supply of amino acids to all tissues. The small intestine is an important contributor to plasma amino acid homeostasis, while amino acid transport in the large intestine is more relevant for bacterial metabolites and fluid secretion. A number of rare inherited disorders have contributed to the identification of amino acid transporters in epithelial cells of the small intestine, in particular cystinuria, lysinuric protein intolerance, Hartnup disorder, iminoglycinuria, and dicarboxylic aminoaciduria. These are most readily detected by analysis of urine amino acids, but typically also affect intestinal transport. The genes underlying these disorders have all been identified. The remaining transporters were identified through molecular cloning techniques to the extent that a comprehensive portrait of functional cooperation among transporters of intestinal epithelial cells is now available for both the basolateral and apical membranes. Mouse models of most intestinal transporters illustrate their contribution to amino acid homeostasis and systemic physiology. Intestinal amino acid transport activities can vary between species, but these can now be explained as differences of amino acid transporter distribution along the intestine. © 2019 American Physiological Society. Compr Physiol 9:343-373, 2019.
Collapse
Affiliation(s)
- Stefan Bröer
- Research School of Biology, The Australian National University, Canberra, ACT 2601, Australia
| | - Stephen J Fairweather
- Research School of Biology, The Australian National University, Canberra, ACT 2601, Australia
| |
Collapse
|
23
|
In silico analysis of SLC3A1 and SLC7A9 mutations in Iranian patients with Cystinuria. Mol Biol Rep 2018; 45:1165-1173. [PMID: 30069816 DOI: 10.1007/s11033-018-4269-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 07/16/2018] [Indexed: 01/02/2023]
Abstract
Cystinuria is an autosomal recessive defect in reabsorptive transport of cystine and the dibasic amino acids ornithine, arginine, and lysine from renal tubule and small intestine. Mutations in two genes: SLC3A1, encoding the heavy chain rbAT of the renal cystine transport system and SLC7A9, the gene of its light chain b0, + AT have a crucial role in the diseases. In our previous studies from Iranian populations with Cystinuria totally six and eleven novel mutations respectively identified in SLC3A1 and SLC7A9 genes. In this study, we conducted an in silico functional analysis to explore the possible association between these genetic mutations and Cystinuria. MutationTaster, PolyPhen-2, PANTHER, FATHMM. PhDSNP and MutPred was applied to predict the degree of pathogenicity for the missense mutations. Furthermore, Residue Interaction Network (RIN) and Intron variant analyses was performed using Cytoscape and Human Slicing Finder softwares. These genetic variants can provide a better understanding of genotype-phenotype relationships in patients with Cystinuria. In the future, the findings may also facilitate the development of new molecular diagnostic markers for the diseases.
Collapse
|
24
|
Scalise M, Galluccio M, Console L, Pochini L, Indiveri C. The Human SLC7A5 (LAT1): The Intriguing Histidine/Large Neutral Amino Acid Transporter and Its Relevance to Human Health. Front Chem 2018; 6:243. [PMID: 29988369 PMCID: PMC6023973 DOI: 10.3389/fchem.2018.00243] [Citation(s) in RCA: 195] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 06/06/2018] [Indexed: 12/22/2022] Open
Abstract
SLC7A5, known as LAT1, belongs to the APC superfamily and forms a heterodimeric amino acid transporter interacting with the glycoprotein CD98 (SLC3A2) through a conserved disulfide. The complex is responsible for uptake of essential amino acids in crucial body districts such as placenta and blood brain barrier. LAT1/CD98 heterodimer has been studied over the years to unravel the transport mechanism and the role of each subunit. Studies conducted in intact cells demonstrated that LAT1/CD98 mediates a Na+ and pH independent antiport of amino acids. Some novel insights into the function of LAT1 derived from studies conducted in proteoliposomes reconstituted with the recombinant human LAT1. Using this experimental tool, it has been demonstrated that the preferred substrate is histidine and that CD98 is not required for transport being, plausibly, involved in routing LAT1 to the plasma membrane. Since a 3D structure of LAT1 is not available, homology models have been built on the basis of the AdiC transporter from E.coli. Crucial residues for substrate recognition and gating have been identified using a combined approach of bioinformatics and site-directed mutagenesis coupled to functional assays. Over the years, the interest around LAT1 increased because this transporter is involved in important human diseases such as neurological disorders and cancer. Therefore, LAT1 became an important pharmacological target together with other nutrient membrane transporters. Moving from knowledge on structure/function relationships, two cysteine residues, lying on the substrate binding site, have been exploited for designing thiol reacting covalent inhibitors. Some lead compounds have been characterized whose efficacy has been tested in a cancer cell line.
Collapse
Affiliation(s)
- Mariafrancesca Scalise
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Rende, Italy
| | - Michele Galluccio
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Rende, Italy
| | - Lara Console
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Rende, Italy
| | - Lorena Pochini
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Rende, Italy
| | - Cesare Indiveri
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Rende, Italy.,CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnology, Bari, Italy
| |
Collapse
|
25
|
Rotoli BM, Barilli A, Visigalli R, Ingoglia F, Milioli M, Di Lascia M, Riccardi B, Puccini P, Dall'Asta V. Downregulation of SLC7A7 Triggers an Inflammatory Phenotype in Human Macrophages and Airway Epithelial Cells. Front Immunol 2018; 9:508. [PMID: 29616026 PMCID: PMC5868322 DOI: 10.3389/fimmu.2018.00508] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 02/26/2018] [Indexed: 11/29/2022] Open
Abstract
Lysinuric protein intolerance (LPI) is a recessively inherited aminoaciduria caused by mutations of SLC7A7, the gene encoding y+LAT1 light chain of system y+L for cationic amino acid transport. The pathogenesis of LPI is still unknown. In this study, we have utilized a gene silencing approach in macrophages and airway epithelial cells to investigate whether complications affecting lung and immune system are directly ascribable to the lack of SLC7A7 or, rather, mediated by an abnormal accumulation of arginine in mutated cells. When SLC7A7/y+LAT1 was silenced in human THP-1 macrophages and A549 airway epithelial cells by means of short interference RNA (siRNA), a significant induction of the expression and release of the inflammatory mediators IL1β and TNFα was observed, no matter the intracellular arginine availability. This effect was mainly regulated at transcriptional level through the activation of NFκB signaling pathway. Moreover, since respiratory epithelial cells are the important sources of chemokines in response to pro-inflammatory stimuli, the effect of IL1β has been addressed on SLC7A7 silenced A549 cells. Results obtained indicated that the downregulation of SLC7A7/y+LAT1 markedly strengthened the stimulatory effect of the cytokine on CCL5/RANTES expression and release without affecting the levels of CXCL8/IL8. Consistently, also the conditioned medium of silenced THP-1 macrophages activated airway epithelial cells in terms of CCL5/RANTES expression due to the presence of elevated amount of proinflammatory cytokines. In conclusion, our results point to a novel thus far unknown function of SLC7A7/y+LAT1, that, under physiological conditions, besides transporting arginine, may act as a brake to restrain inflammation.
Collapse
Affiliation(s)
- Bianca Maria Rotoli
- Department of Medicine and Surgery (DiMeC), University of Parma, Parma, Italy
| | - Amelia Barilli
- Department of Medicine and Surgery (DiMeC), University of Parma, Parma, Italy
| | - Rossana Visigalli
- Department of Medicine and Surgery (DiMeC), University of Parma, Parma, Italy
| | - Filippo Ingoglia
- Department of Medicine and Surgery (DiMeC), University of Parma, Parma, Italy
| | - Marco Milioli
- Preclinical Pharmacokinetics, Biochemistry and Metabolism Department, Chiesi Farmaceutici, Parma, Italy
| | - Maria Di Lascia
- Preclinical Pharmacokinetics, Biochemistry and Metabolism Department, Chiesi Farmaceutici, Parma, Italy
| | - Benedetta Riccardi
- Preclinical Pharmacokinetics, Biochemistry and Metabolism Department, Chiesi Farmaceutici, Parma, Italy
| | - Paola Puccini
- Preclinical Pharmacokinetics, Biochemistry and Metabolism Department, Chiesi Farmaceutici, Parma, Italy
| | - Valeria Dall'Asta
- Department of Medicine and Surgery (DiMeC), University of Parma, Parma, Italy
| |
Collapse
|
26
|
Gaildrat P, Lebbah S, Tebani A, Sudrié-Arnaud B, Tostivint I, Bollee G, Tubeuf H, Charles T, Bertholet-Thomas A, Goldenberg A, Barbey F, Martins A, Saugier-Veber P, Frébourg T, Knebelmann B, Bekri S. Clinical and molecular characterization of cystinuria in a French cohort: relevance of assessing large-scale rearrangements and splicing variants. Mol Genet Genomic Med 2017; 5:373-389. [PMID: 28717662 PMCID: PMC5511796 DOI: 10.1002/mgg3.294] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Revised: 03/27/2017] [Accepted: 04/06/2017] [Indexed: 02/04/2023] Open
Abstract
Background Cystinuria is an autosomal recessive disorder of dibasic amino acid transport in the kidney and the intestine leading to increased urinary cystine excretion and nephrolithiasis. Two genes, SLC3A1 and SLC7A9, coding respectively for rBAT and b0,+AT, account for the genetic basis of cystinuria. Methods This study reports the clinical and molecular characterization of a French cohort including 112 cystinuria patients and 25 relatives from 99 families. Molecular screening was performed using sequencing and Quantitative Multiplex PCR of Short Fluorescent Fragments analyses. Functional minigene‐based assays have been used to characterize splicing variants. Results Eighty‐eight pathogenic nucleotide changes were identified in SLC3A1 (63) and SLC7A9 (25) genes, of which 42 were novel. Interestingly, 17% (15/88) and 11% (10/88) of the total number of variants correspond, respectively, to large‐scale rearrangements and splicing mutations. Functional minigene‐based assays were performed for six variants located outside the most conserved sequences of the splice sites; three variants affect splice sites, while three others modify exonic splicing regulatory elements (ESR), in good agreement with a new in silico prediction based on ΔtESRseq values. Conclusion This report expands the spectrum of SLC3A1 and SLC7A9 variants and supports that digenic inheritance is unlikely. Furthermore, it highlights the relevance of assessing large‐scale rearrangements and splicing mutations to fully characterize cystinuria patients at the molecular level.
Collapse
Affiliation(s)
- Pascaline Gaildrat
- Inserm U1245UNIROUENNormandie UnivNormandy Centre for Genomic and Personalized MedicineRouenFrance
| | - Said Lebbah
- Department of NephrologyNecker HospitalParisFrance
| | - Abdellah Tebani
- Inserm U1245UNIROUENNormandie UnivNormandy Centre for Genomic and Personalized MedicineRouenFrance.,Department of Metabolic BiochemistryRouen University HospitalRouenFrance
| | | | | | | | - Hélène Tubeuf
- Inserm U1245UNIROUENNormandie UnivNormandy Centre for Genomic and Personalized MedicineRouenFrance.,Interactive BiosoftwareRouenFrance
| | | | | | | | - Frederic Barbey
- Department of TransplantationCHUV Department of PediatricsLausanne University HospitalLausanneSwitzerland
| | - Alexandra Martins
- Inserm U1245UNIROUENNormandie UnivNormandy Centre for Genomic and Personalized MedicineRouenFrance
| | - Pascale Saugier-Veber
- Inserm U1245UNIROUENNormandie UnivNormandy Centre for Genomic and Personalized MedicineRouenFrance.,Department of GeneticsRouen University HospitalRouenFrance
| | - Thierry Frébourg
- Inserm U1245UNIROUENNormandie UnivNormandy Centre for Genomic and Personalized MedicineRouenFrance.,Department of GeneticsRouen University HospitalRouenFrance
| | | | - Soumeya Bekri
- Inserm U1245UNIROUENNormandie UnivNormandy Centre for Genomic and Personalized MedicineRouenFrance.,Department of Metabolic BiochemistryRouen University HospitalRouenFrance
| |
Collapse
|
27
|
Markazi S, Kheirollahi M, Doosti A, Mohammadi M. Report of SLC3A1/rBAT gene mutations in Iranian cystinuria patients: A direct sequencing study. JOURNAL OF RESEARCH IN MEDICAL SCIENCES 2017; 22:33. [PMID: 28461819 PMCID: PMC5390536 DOI: 10.4103/1735-1995.202149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/10/2016] [Accepted: 12/21/2016] [Indexed: 11/04/2022]
Abstract
Background: Considering a few studies on the genetic basis of the cystinuria in the Middle East and the population-specific distribution of mutations in the SLC3A1, we tried to find genetic variants in three exons (1, 3, and 8) of SLC3A1. Materials and Methods: In this study, exons 1, 3, and 8 of SLC3A1 gene of 25 unrelated cystinuria patients searched for genetic variations by polymerase chain reaction and sequencing. Results: There were five different variations in our studied population. We found one mutation in the SLC3A1 gene including missense variant M467K and identified three polymorphisms: nonsynonymous variant G38G, c. 610 + 169C>T and c. 610 + 147C>G within the SLC3A1 gene, and one new variant. Conclusion: Our results confirm that cystinuria is a heterogeneous disorder at the molecular level and more studies are needed to identify the distribution and frequency of mutations causing cystinuria in the Iranian population.
Collapse
Affiliation(s)
- Samaneh Markazi
- Department of Molecular Genetics, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Sahrekord, Iran
| | - Majid Kheirollahi
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Noncommunicable Disease, Isfahan, Iran.,Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Abbas Doosti
- Department of Molecular Genetics, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Sahrekord, Iran
| | - Mehrdad Mohammadi
- Department of Urology, Urology and Kidney Transplantation Research Center, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
28
|
Shin CS, Mishra P, Watrous JD, Carelli V, D'Aurelio M, Jain M, Chan DC. The glutamate/cystine xCT antiporter antagonizes glutamine metabolism and reduces nutrient flexibility. Nat Commun 2017; 8:15074. [PMID: 28429737 PMCID: PMC5413954 DOI: 10.1038/ncomms15074] [Citation(s) in RCA: 218] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 02/24/2017] [Indexed: 01/05/2023] Open
Abstract
As noted by Warburg, many cancer cells depend on the consumption of glucose. We performed a genetic screen to identify factors responsible for glucose addiction and recovered the two subunits of the xCT antiporter (system xc−), which plays an antioxidant role by exporting glutamate for cystine. Disruption of the xCT antiporter greatly improves cell viability after glucose withdrawal, because conservation of glutamate enables cells to maintain mitochondrial respiration. In some breast cancer cells, xCT antiporter expression is upregulated through the antioxidant transcription factor Nrf2 and contributes to their requirement for glucose as a carbon source. In cells carrying patient-derived mitochondrial DNA mutations, the xCT antiporter is upregulated and its inhibition improves mitochondrial function and cell viability. Therefore, although upregulation of the xCT antiporter promotes antioxidant defence, it antagonizes glutamine metabolism and restricts nutrient flexibility. In cells with mitochondrial dysfunction, the potential utility of xCT antiporter inhibition should be further tested. The factors that limit the nutrient flexibility of cells remain largely unknown. Here, the authors identify the glutamate/cysteine antiporter xCT in a genetic screen for glucose dependency and show it determines the ability of cells to survive under conditions of low glucose by limiting the utilization of glutamine.
Collapse
Affiliation(s)
- Chun-Shik Shin
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA
| | - Prashant Mishra
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA.,Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-8502, USA
| | - Jeramie D Watrous
- Department of Medicine and Pharmacology, University of California, La Jolla, San Diego, California 92093, USA
| | - Valerio Carelli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Via Altura 3, 40139 Bologna, Italy.,Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Via Altura 3, 40139 Bologna, Italy
| | - Marilena D'Aurelio
- Department of Neurology and Neuroscience, Weill Medical College of Cornell University, 1300 York Avenue, A501, New York, New York 10065, USA
| | - Mohit Jain
- Department of Medicine and Pharmacology, University of California, La Jolla, San Diego, California 92093, USA
| | - David C Chan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA
| |
Collapse
|
29
|
Janeček Š, Gabriško M. Remarkable evolutionary relatedness among the enzymes and proteins from the α-amylase family. Cell Mol Life Sci 2016; 73:2707-25. [PMID: 27154042 PMCID: PMC11108405 DOI: 10.1007/s00018-016-2246-6] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 04/22/2016] [Indexed: 12/17/2022]
Abstract
The α-amylase is a ubiquitous starch hydrolase catalyzing the cleavage of the α-1,4-glucosidic bonds in an endo-fashion. Various α-amylases originating from different taxonomic sources may differ from each other significantly in their exact substrate preference and product profile. Moreover, it also seems to be clear that at least two different amino acid sequences utilizing two different catalytic machineries have evolved to execute the same α-amylolytic specificity. The two have been classified in the Cabohydrate-Active enZyme database, the CAZy, in the glycoside hydrolase (GH) families GH13 and GH57. While the former and the larger α-amylase family GH13 evidently forms the clan GH-H with the families GH70 and GH77, the latter and the smaller α-amylase family GH57 has only been predicted to maybe define a future clan with the family GH119. Sequences and several tens of enzyme specificities found throughout all three kingdoms in many taxa provide an interesting material for evolutionarily oriented studies that have demonstrated remarkable observations. This review emphasizes just the three of them: (1) a close relatedness between the plant and archaeal α-amylases from the family GH13; (2) a common ancestry in the family GH13 of animal heavy chains of heteromeric amino acid transporter rBAT and 4F2 with the microbial α-glucosidases; and (3) the unique sequence features in the primary structures of amylomaltases from the genus Borrelia from the family GH77. Although the three examples cannot represent an exhaustive list of exceptional topics worth to be interested in, they may demonstrate the importance these enzymes possess in the overall scientific context.
Collapse
Affiliation(s)
- Štefan Janeček
- Laboratory of Protein Evolution, Institute of Molecular Biology, Slovak Academy of Sciences, Dúbravská cesta 21, 84551, Bratislava, Slovakia.
- Department of Biology, Faculty of Natural Sciences, University of SS. Cyril and Methodius in Trnava, Nám. J. Herdu 2, 91701, Trnava, Slovakia.
| | - Marek Gabriško
- Laboratory of Protein Evolution, Institute of Molecular Biology, Slovak Academy of Sciences, Dúbravská cesta 21, 84551, Bratislava, Slovakia
| |
Collapse
|
30
|
Rodríguez-Banqueri A, Errasti-Murugarren E, Bartoccioni P, Kowalczyk L, Perálvarez-Marín A, Palacín M, Vázquez-Ibar JL. Stabilization of a prokaryotic LAT transporter by random mutagenesis. ACTA ACUST UNITED AC 2016; 147:353-68. [PMID: 26976827 PMCID: PMC4810068 DOI: 10.1085/jgp.201511510] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 02/22/2016] [Indexed: 12/31/2022]
Abstract
The knowledge of three-dimensional structures at atomic resolution of membrane transport proteins has improved considerably our understanding of their physiological roles and pathological implications. However, most structural biology techniques require an optimal candidate within a protein family for structural determination with (a) reasonable production in heterologous hosts and (b) good stability in detergent micelles. SteT, the Bacillus subtilis L-serine/L-threonine exchanger is the best-known prokaryotic paradigm of the mammalian L-amino acid transporter (LAT) family. Unfortunately, SteT's lousy stability after extracting from the membrane prevents its structural characterization. Here, we have used an approach based on random mutagenesis to engineer stability in SteT. Using a split GFP complementation assay as reporter of protein expression and membrane insertion, we created a library of 70 SteT mutants each containing random replacements of one or two residues situated in the transmembrane domains. Analysis of expression and monodispersity in detergent of this library permitted the identification of evolved versions of SteT with a significant increase in both expression yield and stability in detergent with respect to wild type. In addition, these experiments revealed a correlation between the yield of expression and the stability in detergent micelles. Finally, and based on protein delipidation and relipidation assays together with transport experiments, possible mechanisms of SteT stabilization are discussed. Besides optimizing a member of the LAT family for structural determination, our work proposes a new approach that can be used to optimize any membrane protein of interest.
Collapse
Affiliation(s)
- Arturo Rodríguez-Banqueri
- Institute for Research in Biomedicine (IRB), Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Ekaitz Errasti-Murugarren
- Institute for Research in Biomedicine (IRB), Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Paola Bartoccioni
- Institute for Research in Biomedicine (IRB), Barcelona Institute of Science and Technology, 08028 Barcelona, Spain Spanish Biomedical Research Center in Rare Diseases (CIBERER), 08028 Barcelona, Spain
| | - Lukasz Kowalczyk
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Alex Perálvarez-Marín
- Biophysics Unit, Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona, 08193 Cerdanyola del Vallés, Spain
| | - Manuel Palacín
- Institute for Research in Biomedicine (IRB), Barcelona Institute of Science and Technology, 08028 Barcelona, Spain Spanish Biomedical Research Center in Rare Diseases (CIBERER), 08028 Barcelona, Spain Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, 08028 Barcelona, Spain
| | - José Luis Vázquez-Ibar
- Institute for Integrative Biology of the Cell (I2BC), CEA, French National Centre for Scientific Research (CNRS) UMR 9198, University Paris-Sud, University Paris-Saclay, F-91198 Gif-sur-Yvette, France
| |
Collapse
|
31
|
Antonissen G, Van Immerseel F, Pasmans F, Ducatelle R, Janssens GPJ, De Baere S, Mountzouris KC, Su S, Wong EA, De Meulenaer B, Verlinden M, Devreese M, Haesebrouck F, Novak B, Dohnal I, Martel A, Croubels S. Mycotoxins Deoxynivalenol and Fumonisins Alter the Extrinsic Component of Intestinal Barrier in Broiler Chickens. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:10846-10855. [PMID: 26632976 DOI: 10.1021/acs.jafc.5b04119] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Deoxynivalenol (DON) and fumonisins (FBs) are secondary metabolites produced by Fusarium fungi that frequently contaminate broiler feed. The aim of this study was to investigate the impact of DON and/or FBs on the intestinal barrier in broiler chickens, more specifically on the mucus layer and antioxidative response to oxidative stress. One-day-old broiler chicks were divided into four groups, each consisting of eight pens of seven birds each, and were fed for 15 days either a control diet, a DON-contaminated diet (4.6 mg DON/kg feed), a FBs-contaminated diet (25.4 mg FB1 + FB2/kg feed), or a DON+FBs-contaminated diet (4.3 mg DON and 22.9 mg FB1 + FB2/kg feed). DON and FBs affected the duodenal mucus layer by suppressing intestinal mucin (MUC) 2 gene expression and altering the mucin monosaccharide composition. Both mycotoxins decreased gene expression of the intestinal zinc transporter (ZnT)-1 and regulated intracellular methionine homeostasis, which are both important for preserving the cell's critical antioxidant activity. Feeding a DON- and/or FBs-contaminated diet, at concentrations close to the European Union maximum guidance levels (5 mg DON and 20 mg FB1 + FB2/kg feed) changes the intestinal mucus layer and several intestinal epithelial antioxidative mechanisms.
Collapse
Affiliation(s)
- Gunther Antonissen
- Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University , Salisburylaan 133, 9820 Merelbeke, Belgium
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University , Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Filip Van Immerseel
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University , Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Frank Pasmans
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University , Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Richard Ducatelle
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University , Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Geert P J Janssens
- Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University , Heidestraat 19, 9820 Merelbeke, Belgium
| | - Siegrid De Baere
- Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University , Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Konstantinos C Mountzouris
- Department of Nutritional Physiology and Feeding, Agricultural University of Athens , Iera Odos 75, 11855 Athens, Greece
| | - Shengchen Su
- Department of Animal and Poultry Sciences, Virginia Tech , Blacksburg, Virginia 24061, United States
| | - Eric A Wong
- Department of Animal and Poultry Sciences, Virginia Tech , Blacksburg, Virginia 24061, United States
| | - Bruno De Meulenaer
- Department of Food Safety and Food Quality (Partner in Food2Know), Faculty of Bioscience Engineering, Ghent University , Coupure Links 653, 9000 Gent, Belgium
| | - Marc Verlinden
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University , Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Mathias Devreese
- Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University , Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Freddy Haesebrouck
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University , Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Barbara Novak
- Biomin Research Center , Technopark 1, 3430 Tulln, Austria
| | - Ilse Dohnal
- Biomin Research Center , Technopark 1, 3430 Tulln, Austria
| | - An Martel
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University , Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Siska Croubels
- Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University , Salisburylaan 133, 9820 Merelbeke, Belgium
| |
Collapse
|
32
|
Abstract
Most extracellular glutamate in the brain is released by xCT, a glial antiporter that exports glutamate and imports cystine. The function of xCT, and extracellular glutamate in general, remains unclear. Several lines of evidence suggest that glutamate from xCT could act in a paracrine fashion to suppress glutamatergic synapse strength by triggering removal of postsynaptic glutamate receptors. To test this idea, we used whole-cell patch-clamp electrophysiology and immunohistochemistry to quantify receptor number and synapse function in xCT knock-out mouse hippocampal CA3-CA1 synapses. Consistent with the hypothesis that xCT suppresses glutamate receptor number and synapse strength, xCT knock-out synapses showed increased AMPA receptor abundance with concomitant large enhancements of spontaneous and evoked synaptic transmission. We saw no evidence for changes in GABA receptor abundance or the overall number of glutamatergic synapses. The xCT knock-out phenotype was replicated by incubating slices in the xCT inhibitor (S)-4-carboxyphenylglycine, and consistent with the idea that xCT works by regulating extracellular glutamate, the xCT knock-out phenotype could be reproduced in controls by incubating the slices in glutamate-free aCSF. We conclude that glutamate secreted via xCT suppresses glutamatergic synapse strength by triggering removal of postsynaptic AMPA receptors.
Collapse
|
33
|
Vuille-dit-Bille RN, Camargo SM, Emmenegger L, Sasse T, Kummer E, Jando J, Hamie QM, Meier CF, Hunziker S, Forras-Kaufmann Z, Kuyumcu S, Fox M, Schwizer W, Fried M, Lindenmeyer M, Götze O, Verrey F. Human intestine luminal ACE2 and amino acid transporter expression increased by ACE-inhibitors. Amino Acids 2014; 47:693-705. [DOI: 10.1007/s00726-014-1889-6] [Citation(s) in RCA: 229] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 12/03/2014] [Indexed: 02/06/2023]
|
34
|
Featherstone DE, Yanoga F, Grosjean Y. Accelerated bang recovery in Drosophila genderblind mutants. Commun Integr Biol 2014; 1:14-17. [PMID: 19430543 DOI: 10.4161/cib.1.1.6437] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Cystine-glutamate transporters import cystine into cells for glutathione synthesis and protection from oxidative stress, but also export significant amounts of glutamate. Increasing evidence suggests that 'ambient extracellular glutamate' secreted by cystine-glutamate transporters in the nervous system modulates glutamatergic synapse strength and behavior. To date, the only cystine-glutamate transporter mutants examined behaviorally are Drosophila genderblind mutants. These animals contain loss-of-function mutations in the 'genderblind' gene, which encodes an xCT subunit essential for cystine-glutamate transporter function. Genderblind was named based on a mutant courtship phenotype: male genderblind mutants are attracted to normally aversive male pheromones and thus court and attempt to copulate with both male and female partners equally. However, genderblind protein is expressed in many parts of the fly brain and thus might be expected to also regulate other behaviors, including behaviors not related to male courtship or chemosensation. Here, we show that genderblind mutants display faster recovery and increased negative geotaxis after strong mechanical stimuli (e.g., they climb faster and farther after vial banging). This phenotype is displayed by both males and females, consistent with strong genderblind expression in both sexes.
Collapse
Affiliation(s)
- David E Featherstone
- Department of Biological Sciences; University of Illinois at Chicago; Chicago, Illinois, USA
| | | | | |
Collapse
|
35
|
Camargo SMR, Vuille-dit-Bille RN, Mariotta L, Ramadan T, Huggel K, Singer D, Götze O, Verrey F. The molecular mechanism of intestinal levodopa absorption and its possible implications for the treatment of Parkinson's disease. J Pharmacol Exp Ther 2014; 351:114-23. [PMID: 25073474 DOI: 10.1124/jpet.114.216317] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025] Open
Abstract
Levodopa (L-DOPA) is the naturally occurring precursor amino acid for dopamine and the main therapeutic agent for neurologic disorders due to dopamine depletion, such as Parkinson's disease. l-DOPA absorption in small intestine has been suggested to be mediated by the large neutral amino acids transport machinery, but the identity of the involved transporters is unknown. Clinically, coadministration of l-DOPA and dietary amino acids is avoided to decrease competition for transport in intestine and at the blood-brain barrier. l-DOPA is routinely coadministered with levodopa metabolism inhibitors (dopa-decarboxylase and cathechol-O-methyl transferase inhibitors) that share structural similarity with levodopa. In this systematic study involving Xenopus laevis oocytes and Madin-Darby canine kidney epithelia expression systems and ex vivo preparations from wild-type and knockout mice, we identified the neutral and dibasic amino acids exchanger (antiporter) b(0,+)AT-rBAT (SLC7A9-SLC3A1) as the luminal intestinal l-DOPA transporter. The major luminal cotransporter (symporter) B(0)AT1 (SLC6A19) was not involved in levodopa transport. L-Leucine and L-arginine competed with levodopa across the luminal enterocyte membrane as expected for b(0,+)AT-rBAT substrates, whereas dopa-decarboxylase and cathechol-O-methyl transferase inhibitors had no effect. The presence of amino acids in the basolateral compartment mimicking the postprandial phase increased transepithelial levodopa transport by stimulating basolateral efflux via the antiporter LAT2-4F2 (SLC7A8-SLC3A2). Additionally, the aromatic amino acid uniporter TAT1 (SLC16A10) was shown to play a major role in l-DOPA efflux from intestinal enterocytes. These results identify the molecular mechanisms mediating small intestinal levodopa absorption and suggest strategies for optimization of delivery and absorption of this important prodrug.
Collapse
Affiliation(s)
- Simone M R Camargo
- Institute of Physiology and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland (S.M.R.C., R.N.V.-d.-B., L.M., T.R., K.H., D.S., F.V.); and Division of Gastroenterology and Hepatology, University Hospital of Zurich, Zurich, Switzerland (O.G.)
| | - Raphael N Vuille-dit-Bille
- Institute of Physiology and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland (S.M.R.C., R.N.V.-d.-B., L.M., T.R., K.H., D.S., F.V.); and Division of Gastroenterology and Hepatology, University Hospital of Zurich, Zurich, Switzerland (O.G.)
| | - Luca Mariotta
- Institute of Physiology and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland (S.M.R.C., R.N.V.-d.-B., L.M., T.R., K.H., D.S., F.V.); and Division of Gastroenterology and Hepatology, University Hospital of Zurich, Zurich, Switzerland (O.G.)
| | - Tamara Ramadan
- Institute of Physiology and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland (S.M.R.C., R.N.V.-d.-B., L.M., T.R., K.H., D.S., F.V.); and Division of Gastroenterology and Hepatology, University Hospital of Zurich, Zurich, Switzerland (O.G.)
| | - Katja Huggel
- Institute of Physiology and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland (S.M.R.C., R.N.V.-d.-B., L.M., T.R., K.H., D.S., F.V.); and Division of Gastroenterology and Hepatology, University Hospital of Zurich, Zurich, Switzerland (O.G.)
| | - Dustin Singer
- Institute of Physiology and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland (S.M.R.C., R.N.V.-d.-B., L.M., T.R., K.H., D.S., F.V.); and Division of Gastroenterology and Hepatology, University Hospital of Zurich, Zurich, Switzerland (O.G.)
| | - Oliver Götze
- Institute of Physiology and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland (S.M.R.C., R.N.V.-d.-B., L.M., T.R., K.H., D.S., F.V.); and Division of Gastroenterology and Hepatology, University Hospital of Zurich, Zurich, Switzerland (O.G.)
| | - François Verrey
- Institute of Physiology and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland (S.M.R.C., R.N.V.-d.-B., L.M., T.R., K.H., D.S., F.V.); and Division of Gastroenterology and Hepatology, University Hospital of Zurich, Zurich, Switzerland (O.G.)
| |
Collapse
|
36
|
[Nephrolithiasis: metabolic defects and terapeutic implications]. Urologia 2014; 81:1-11. [PMID: 24744215 DOI: 10.5301/uro.5000058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2014] [Indexed: 12/31/2022]
Abstract
Over the past 10 years, major progress has been made in the knowledge of urinary lithogenesis, including the potential pathogenetic role of Randall's plaques and renal tubular crystal retention. Urine supersaturation is the driving force of this process and can be induced by some risk factors, including low urine volume, high urinary excretion of calcium oxalate and uric acid and low urinary excretion of citrate. Primary hypercalciuria can be due to intestinal overabsorption renal leak and bone reabsorption of calcium. Prophilaxis is mainly conducted with thiazides and low calcium diet which is indicated only in the intestinal form. Primary hyperoxaluria is treated with pyridoxine and may require in the severe forms simultaneous renal and liver transplantation. Enteric hyperoxaluria is secondary to fatty acids malabsorption and requires diet, oral calcium and cholestiramine. Hyperuricosuria is caused by diet endogenous overproduction, mainly due to enzymatic defects or high renal excretion of uric acid. Urine alkalinization with K or K and Mg citrate can prevent stone formation even in idiopathic uric acid nephrolithiasis, in which a defect of urine acidification is supposed to be the main abnormality, and in hypocitraturic patients. Cystinuria is a rare inherited defect with an intense clinical impact. It can be classified in three forms and urinary stone formation is the role. Increased solubility and conversion of cystine in a more soluble form are the main goals of the prophylaxis which includes K citrate and thiol agents administration. Tiopronin is preferred to D-penicillamine due to its lower side effects.
Collapse
|
37
|
Sahoo S, Aurich MK, Jonsson JJ, Thiele I. Membrane transporters in a human genome-scale metabolic knowledgebase and their implications for disease. Front Physiol 2014; 5:91. [PMID: 24653705 PMCID: PMC3949408 DOI: 10.3389/fphys.2014.00091] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 02/17/2014] [Indexed: 01/18/2023] Open
Abstract
Membrane transporters enable efficient cellular metabolism, aid in nutrient sensing, and have been associated with various diseases, such as obesity and cancer. Genome-scale metabolic network reconstructions capture genomic, physiological, and biochemical knowledge of a target organism, along with a detailed representation of the cellular metabolite transport mechanisms. Since the first reconstruction of human metabolism, Recon 1, published in 2007, progress has been made in the field of metabolite transport. Recently, we published an updated reconstruction, Recon 2, which significantly improved the metabolic coverage and functionality. Human metabolic reconstructions have been used to investigate the role of metabolism in disease and to predict biomarkers and drug targets. Given the importance of cellular transport systems in understanding human metabolism in health and disease, we analyzed the coverage of transport systems for various metabolite classes in Recon 2. We will review the current knowledge on transporters (i.e., their preferred substrates, transport mechanisms, metabolic relevance, and disease association for each metabolite class). We will assess missing coverage and propose modifications and additions through a transport module that is functional when combined with Recon 2. This information will be valuable for further refinements. These data will also provide starting points for further experiments by highlighting areas of incomplete knowledge. This review represents the first comprehensive overview of the transporters involved in central metabolism and their transport mechanisms, thus serving as a compendium of metabolite transporters specific for human metabolic reconstructions.
Collapse
Affiliation(s)
- Swagatika Sahoo
- Center for Systems Biology, University of Iceland Reykjavik, Iceland ; Molecular Systems Physiology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg Belval, Luxembourg
| | - Maike K Aurich
- Center for Systems Biology, University of Iceland Reykjavik, Iceland ; Molecular Systems Physiology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg Belval, Luxembourg
| | - Jon J Jonsson
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Iceland Reykjavik, Iceland ; Department of Genetics and Molecular Medicine, Landspitali, National University Hospital of Iceland Reykjavik, Iceland
| | - Ines Thiele
- Center for Systems Biology, University of Iceland Reykjavik, Iceland ; Molecular Systems Physiology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg Belval, Luxembourg
| |
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW Epithelial neutral amino acid transporters have been identified at the molecular level in recent years. Mouse models have now established the crucial role of these transporters for systemic amino acid homeostasis. This review summarizes recent progress in this field. RECENT FINDINGS Epithelial neutral amino acid transporters play an important role in the homeostasis of neutral amino acid levels in the body. They are important for the maintenance of body weight and muscle mass and serve as fuels. They also serve a role in providing nutrients to epithelial cells. Changes of plasma amino acid levels are not necessarily correlated to the amino acids appearing in the urine; changes in organ amino acid metabolism need to be taken into account. SUMMARY Genetic deletion of neutral amino acid transporters provides insight into their role in protein nutrition and homeostasis.
Collapse
|
39
|
Galluccio M, Pingitore P, Scalise M, Indiveri C. Cloning, large scale over-expression in E. coli and purification of the components of the human LAT 1 (SLC7A5) amino acid transporter. Protein J 2014; 32:442-8. [PMID: 23912240 DOI: 10.1007/s10930-013-9503-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The high yield expression of the human LAT1 transporter has been obtained for the first time using E. coli. The hLAT1 cDNA was amplified from HEK293 cells and cloned in pH6EX3 vector. The construct pH6EX3-6His-hLAT1 was used to express the 6His-hLAT1 protein in the Rosetta(DE3)pLysS strain of E. coli. The highest level of expression was detected 8 h after induction by IPTG at 28 °C. The expressed protein was collected in the insoluble fraction of cell lysate. On SDS-PAGE the apparent molecular mass of the polypeptide was 40 kDa. After solubilization with sarkosyl and denaturation with urea the protein carrying a 6His N-terminal tag was purified by Ni(2+)-chelating affinity chromatography and identified by anti-His antibody. The yield of the over-expressed protein after purification was 3.5 mg/L (cell culture). The human CD98 cDNA amplified from Imagene plasmid was cloned in pGEX-4T1. The construct pGEX-4T1-hCD98 was used to express the GST-hCD98 protein in the Rosetta(DE3)pLysS strain of E. coli. The highest level of expression was detected in this case 4 h after induction by IPTG at 28 °C. The expressed protein was accumulated in the soluble fraction of cell lysate. The molecular mass was determined on the basis of marker proteins on SDS-PAGE; it was about 110 kDa. GST was cleaved from the protein construct by incubation with thrombin for 12 h and the hCD98 was separated by Sephadex G-200 chromatography (size exclusion). hCD98 showed a 62 kDa apparent molecular mass, as determined on the basis of molecular mass markers using SDS-PAGE. The yield of CD98 was 2 mg/L of cell culture.
Collapse
Affiliation(s)
- Michele Galluccio
- Unit of Biochemistry and Molecular Biotechnology, Department BEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Via P. Bucci 4c, 87036, Arcavacata di Rende, Italy
| | | | | | | |
Collapse
|
40
|
|
41
|
Maruo T, Kanemaki N, Onda K, Sato R, Ichihara N, Ochiai H. Canine amino acid transport system Xc(-): cDNA sequence, distribution and cystine transport activity in lens epithelial cells. J Vet Med Sci 2013; 76:523-30. [PMID: 24366150 PMCID: PMC4064136 DOI: 10.1292/jvms.13-0170] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The cystine transport activity of a lens epithelial cell line originated from a
canine mature cataract was investigated. The distinct cystine transport activity was
observed, which was inhibited to 28% by extracellular 1 mM glutamate. The cDNA sequences
of canine cysteine/glutamate exchanger (xCT) and 4F2hc were determined. The predicted
amino acid sequences were 527 and 533 amino acid polypeptides, respectively. The amino
acid sequences of canine xCT and 4F2hc showed high similarities (>80%) to those of
humans. The expression of xCT in lens epithelial cell line was confirmed by western blot
analysis. RT-PCR analysis revealed high level expression only in the brain, and it was
below the detectable level in other tissues.
Collapse
Affiliation(s)
- Takuya Maruo
- Veterinary Teaching Hospital, School of Veterinary Medicine, Azabu University, 1-17-71 Fuchinobe, Chuou-ku, Sagamihara, Kanagawa 252-5201, Japan
| | | | | | | | | | | |
Collapse
|
42
|
Giacopo AD, Rubio-Aliaga I, Cantone A, Artunc F, Rexhepaj R, Frey-Wagner I, Font-Llitjós M, Gehring N, Stange G, Jaenecke I, Mohebbi N, Closs EI, Palacín M, Nunes V, Daniel H, Lang F, Capasso G, Wagner CA. Differential cystine and dibasic amino acid handling after loss of function of the amino acid transporter b0,+AT (Slc7a9) in mice. Am J Physiol Renal Physiol 2013; 305:F1645-55. [DOI: 10.1152/ajprenal.00221.2013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cystinuria is an autosomal recessive disease caused by mutations in SLC3A1 ( rBAT) and SLC7A9 ( b 0,+ AT). Gene targeting of the catalytic subunit ( Slc7a9) in mice leads to excessive excretion of cystine, lysine, arginine, and ornithine. Here, we studied this non-type I cystinuria mouse model using gene expression analysis, Western blotting, clearance, and brush-border membrane vesicle (BBMV) uptake experiments to further characterize the renal and intestinal consequences of losing Slc7a9 function. The electrogenic and BBMV flux studies in the intestine suggested that arginine and ornithine are transported via other routes apart from system b0,+. No remarkable gene expression changes were observed in other amino acid transporters and the peptide transporters in the intestine and kidney. Furthermore, the glomerular filtration rate (GFR) was reduced by 30% in knockout animals compared with wild-type animals. The fractional excretion of arginine was increased as expected (∼100%), but fractional excretions of lysine (∼35%), ornithine (∼16%), and cystine (∼11%) were less affected. Loss of function of b0,+AT reduced transport of cystine and arginine in renal BBMVs and completely abolished the exchanger activity of dibasic amino acids with neutral amino acids. In conclusion, loss of Slc7a9 function decreases the GFR and increases the excretion of several amino acids to a lesser extent than expected with no clear regulation at the mRNA and protein level of alternative transporters and no increased renal epithelial uptake. These observations indicate that transporters located in distal segments of the kidney and/or metabolic pathways may partially compensate for Slc7a9 loss of function.
Collapse
Affiliation(s)
- Andrea Di Giacopo
- Institute of Physiology-Zürich Center for Integrative Human Physiology (ZIHP), University of Zürich, Zürich, Switzerland
| | - Isabel Rubio-Aliaga
- Institute of Physiology-Zürich Center for Integrative Human Physiology (ZIHP), University of Zürich, Zürich, Switzerland
| | - Alessandra Cantone
- Department of Internal Medicine, Chair of Nephrology, Second University of Naples, Naples, Italy
| | - Ferruh Artunc
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Rexhep Rexhepaj
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | | | - Mariona Font-Llitjós
- Medical and Molecular Genetics Center, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Spain
- U730 CIBERER, Barcelona, Spain
| | - Nicole Gehring
- Institute of Physiology-Zürich Center for Integrative Human Physiology (ZIHP), University of Zürich, Zürich, Switzerland
| | - Gerti Stange
- Institute of Physiology-Zürich Center for Integrative Human Physiology (ZIHP), University of Zürich, Zürich, Switzerland
| | - Isabel Jaenecke
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Nilufar Mohebbi
- Institute of Physiology-Zürich Center for Integrative Human Physiology (ZIHP), University of Zürich, Zürich, Switzerland
| | - Ellen I. Closs
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Manuel Palacín
- IRB Barcelona, Department of Biochemistry and Molecular Biology, University of Barcelona and U731 CIBERER, Barcelona, Spain
| | - Virginia Nunes
- Medical and Molecular Genetics Center, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Spain
- Department of Physiological Sciences II, University of Barcelona, Spain; and
- U730 CIBERER, Barcelona, Spain
| | - Hannelore Daniel
- Molecular Nutrition Unit, Technical University of Munich, Freising, Germany
| | - Florian Lang
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Giovambattista Capasso
- Department of Internal Medicine, Chair of Nephrology, Second University of Naples, Naples, Italy
| | - Carsten A. Wagner
- Institute of Physiology-Zürich Center for Integrative Human Physiology (ZIHP), University of Zürich, Zürich, Switzerland
| |
Collapse
|
43
|
Newell JL, Keyari CM, McDaniel SW, Diaz PJ, Natale NR, Patel SA, Bridges RJ. Novel di-aryl-substituted isoxazoles act as noncompetitive inhibitors of the system Xc(-) cystine/glutamate exchanger. Neurochem Int 2013; 73:132-8. [PMID: 24333322 DOI: 10.1016/j.neuint.2013.11.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 11/26/2013] [Accepted: 11/29/2013] [Indexed: 01/18/2023]
Abstract
The system xc(-) antiporter is a plasma membrane transporter that mediates the exchange of extracellular l-cystine with intracellular l-glutamate. This exchange is significant within the context of the CNS because the import of l-cystine is required for the synthesis of the antioxidant glutathione, while the efflux of l-glutamate has the potential to contribute to either excitatory signaling or excitotoxic pathology. Changes in the activity of the transport system have been linked to the underlying pathological mechanisms of a variety of CNS disorders, one of the most prominent of which is its highly enriched expression in glial brain tumors. In an effort to produce more potent system xc(-) blockers, we have been using amino-3-carboxy-5-methylisoxazole propionic acid (ACPA) as a scaffold for inhibitor development. We previously demonstrated that the addition of lipophilic aryl groups to either the #4 or #5 position on the isoxazole ring markedly increased the inhibitory activity at system xc(-). In the present work a novel series of analogues has been prepared in which aryl groups have been introduced at both the #4 and #5 positions. In contrast to the competitive action of the mono-substituted analogues, kinetic analyses indicate that the di-substituted isoxazoles block system xc(-)-mediated uptake of (3)H-l-glutamate into SNB-19 cells by a noncompetitive mechanism. These new analogues appear to be the first noncompetitive inhibitors identified for this transport system, as well as being among the most potent blockers identified to date. These diaryl-isoxazoles should be of value in assessing the physiological roles and molecular pharmacology of system xc(-).
Collapse
Affiliation(s)
- J L Newell
- Center for Structural & Functional Neuroscience, Department of Biomedical & Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Montana, Missoula, MT 59812, United States
| | - C M Keyari
- Center for Structural & Functional Neuroscience, Department of Biomedical & Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Montana, Missoula, MT 59812, United States
| | - S W McDaniel
- Center for Structural & Functional Neuroscience, Department of Biomedical & Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Montana, Missoula, MT 59812, United States
| | - P J Diaz
- Center for Structural & Functional Neuroscience, Department of Biomedical & Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Montana, Missoula, MT 59812, United States
| | - N R Natale
- Center for Structural & Functional Neuroscience, Department of Biomedical & Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Montana, Missoula, MT 59812, United States
| | - S A Patel
- Center for Structural & Functional Neuroscience, Department of Biomedical & Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Montana, Missoula, MT 59812, United States
| | - R J Bridges
- Center for Structural & Functional Neuroscience, Department of Biomedical & Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Montana, Missoula, MT 59812, United States.
| |
Collapse
|
44
|
Zhou Y, Waanders LF, Holmseth S, Guo C, Berger UV, Li Y, Lehre AC, Lehre KP, Danbolt NC. Proteome analysis and conditional deletion of the EAAT2 glutamate transporter provide evidence against a role of EAAT2 in pancreatic insulin secretion in mice. J Biol Chem 2013; 289:1329-44. [PMID: 24280215 DOI: 10.1074/jbc.m113.529065] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Islet function is incompletely understood in part because key steps in glutamate handling remain undetermined. The glutamate (excitatory amino acid) transporter 2 (EAAT2; Slc1a2) has been hypothesized to (a) provide islet cells with glutamate, (b) protect islet cells against high extracellular glutamate concentrations, (c) mediate glutamate release, or (d) control the pH inside insulin secretory granules. Here we floxed the EAAT2 gene to produce the first conditional EAAT2 knock-out mice. Crossing with Nestin-cyclization recombinase (Cre) eliminated EAAT2 from the brain, resulting in epilepsy and premature death, confirming the importance of EAAT2 for brain function and validating the genetic construction. Crossing with insulin-Cre lines (RIP-Cre and IPF1-Cre) to obtain pancreas-selective deletion did not appear to affect survival, growth, glucose tolerance, or β-cell number. We found (using TaqMan RT-PCR, immunoblotting, immunocytochemistry, and proteome analysis) that the EAAT2 levels were too low to support any of the four hypothesized functions. The proteome analysis detected more than 7,000 islet proteins of which more than 100 were transporters. Although mitochondrial glutamate transporters and transporters for neutral amino acids were present at high levels, all other transporters with known ability to transport glutamate were strikingly absent. Glutamate-metabolizing enzymes were abundant. The level of glutamine synthetase was 2 orders of magnitude higher than that of glutaminase. Taken together this suggests that the uptake of glutamate by islets from the extracellular fluid is insignificant and that glutamate is intracellularly produced. Glutamine synthetase may be more important for islets than assumed previously.
Collapse
Affiliation(s)
- Yun Zhou
- From The Neurotransporter Group, Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, 0317 Oslo, Norway
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Brons AK, Henthorn PS, Raj K, Fitzgerald CA, Liu J, Sewell AC, Giger U. SLC3A1 and SLC7A9 mutations in autosomal recessive or dominant canine cystinuria: a new classification system. J Vet Intern Med 2013; 27:1400-8. [PMID: 24001348 DOI: 10.1111/jvim.12176] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 07/11/2013] [Accepted: 07/24/2013] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Cystinuria, one of the first recognized inborn errors of metabolism, has been reported in many dog breeds. HYPOTHESIS/OBJECTIVES To determine urinary cystine concentrations, inheritance, and mutations in the SLC3A1 and SLC7A9 genes associated with cystinuria in 3 breeds. ANIMALS Mixed and purebred Labrador Retrievers (n = 6), Australian Cattle Dogs (6), Miniature Pinschers (4), and 1 mixed breed dog with cystine urolithiasis, relatives and control dogs. METHODS Urinary cystinuria and aminoaciduria was assessed and exons of the SLC3A1 and SLC7A9 genes were sequenced from genomic DNA. RESULTS In each breed, male and female dogs, independent of neuter status, were found to form calculi. A frameshift mutation in SLC3A1 (c.350delG) resulting in a premature stop codon was identified in autosomal-recessive (AR) cystinuria in Labrador Retrievers and mixed breed dogs. A 6 bp deletion (c.1095_1100del) removing 2 threonines in SLC3A1 was found in autosomal-dominant (AD) cystinuria with a more severe phenotype in homozygous than in heterozygous Australian Cattle Dogs. A missense mutation in SLC7A9 (c.964G>A) was discovered in AD cystinuria in Miniature Pinschers with only heterozygous affected dogs observed to date. Breed-specific DNA tests were developed, but the prevalence of each mutation remains unknown. CONCLUSIONS AND CLINICAL IMPORTANCE These studies describe the first AD inheritance and the first putative SLC7A9 mutation to cause cystinuria in dogs and expand our understanding of this phenotypically and genetically heterogeneous disease, leading to a new classification system for canine cystinuria and better therapeutic management and genetic control in these breeds.
Collapse
Affiliation(s)
- A-K Brons
- Section of Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | | | | | | | | | | | | |
Collapse
|
46
|
le Roex N, Koets AP, van Helden PD, Hoal EG. Gene polymorphisms in African buffalo associated with susceptibility to bovine tuberculosis infection. PLoS One 2013; 8:e64494. [PMID: 23691232 PMCID: PMC3654904 DOI: 10.1371/journal.pone.0064494] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 04/15/2013] [Indexed: 12/19/2022] Open
Abstract
Bovine tuberculosis (BTB) is a chronic, highly infectious disease that affects humans, cattle and numerous species of wildlife. In developing countries such as South Africa, the existence of extensive wildlife-human-livestock interfaces poses a significant risk of Mycobacterium bovis transmission between these groups, and has far-reaching ecological, economic and public health impacts. The African buffalo (Syncerus caffer), acts as a maintenance host for Mycobacterium bovis, and maintains and transmits the disease within the buffalo and to other species. In this study we aimed to investigate genetic susceptibility of buffalo for Mycobacterium bovis infection. Samples from 868 African buffalo of the Cape buffalo subspecies were used in this study. SNPs (n = 69), with predicted functional consequences in genes related to the immune system, were genotyped in this buffalo population by competitive allele-specific SNP genotyping. Case-control association testing and statistical analyses identified three SNPs associated with BTB status in buffalo. These SNPs, SNP41, SNP137 and SNP144, are located in the SLC7A13, DMBT1 and IL1α genes, respectively. SNP137 remained significantly associated after permutation testing. The three genetic polymorphisms identified are located in promising candidate genes for further exploration into genetic susceptibility to BTB in buffalo and other bovids, such as the domestic cow. These polymorphisms/genes may also hold potential for marker-assisted breeding programmes, with the aim of breeding more BTB-resistant animals and herds within both the national parks and the private sector.
Collapse
Affiliation(s)
- Nikki le Roex
- Division of Molecular Biology and Human Genetics, DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, Faculty of Health Sciences, Stellenbosch University, Cape Town, South Africa.
| | | | | | | |
Collapse
|
47
|
Schweikhard ES, Ziegler CM. Amino acid secondary transporters: toward a common transport mechanism. CURRENT TOPICS IN MEMBRANES 2013. [PMID: 23177982 DOI: 10.1016/b978-0-12-394316-3.00001-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Solute carriers (SLC) that transport amino acids are key players in health and diseases in humans. Their prokaryotic relatives are often involved in essential physiological processes in microorganisms, e.g. in homeostasis and acidic/osmotic stress response. High-resolution X-ray structures of the sequence-unrelated amino acid transporters unraveled a striking structural similarity between carriers, which were formerly assigned to different families. The highly conserved fold is characterized by two inverted structural repeats of five transmembrane helices each and indicates common mechanistic transport concepts if not an evolutionary link among a large number of amino acid transporters. Therefore, these transporters are classified now into the structural amino acid-polyamine-organocation superfamily (APCS). The APCS includes among others the mammalian SLC6 transporters and the heterodimeric SLC7/SLC3 transporters. However, it has to be noted that the APCS is not limited entirely to amino acid transporters but contains also transporters for, e.g. amino acid derivatives and sugars. For instance, the betaine-choline-carnitine transporter family of bacterial activity-regulated Na(+)- and H(+)-coupled symporters for glycine betaine and choline is also part of this second largest structural superfamily. The APCS fold provides different possibilities to transport the same amino acid. Arginine can be transported by an H(+)-coupled symport or by antiport mechanism in exchange against agmatine for example. The convergence of the mechanistic concept of transport under comparable physiological conditions allows speculating if structurally unexplored amino acid transporters, e.g. the members of the SLC36 and SLC38 family, belong to the APCS, too. In the kidney, which is an organ that depends critically on the regulated amino acid transport, these different SLC transporters have to work together to account for proper function. Here, we will summarize the basic concepts of Na(+)- and H(+)-coupled amino acid symport and amino acid-product antiport in the light of the respective physiological requirements.
Collapse
Affiliation(s)
- Eva S Schweikhard
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt, Germany
| | | |
Collapse
|
48
|
Popovska-Jankovic K, Tasic V, Bogdanovic R, Miljkovic P, Golubovic E, Soylu A, Saraga M, Pavicevic S, Baskin E, Akil I, Gregoric A, Lilova M, Topaloglu R, Sukarova Stefanovska E, Plaseska-Karanfilska D. Molecular characterization of cystinuria in south-eastern European countries. Urolithiasis 2012; 41:21-30. [PMID: 23532419 DOI: 10.1007/s00240-012-0531-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 12/08/2012] [Indexed: 10/27/2022]
Abstract
Cystinuria is an autosomal recessive disorder caused by defective transport of cystine and dibasic amino acids in the proximal renal tubules and small intestine. So far, more than 128 mutations in SLC3A1 gene, and 93 in SLC7A9 gene have been described as a cause of cystinuria. We present a molecular characterization of the cystinuria in 47 unrelated south-east European families. The molecular methodology included direct sequencing, single strand conformational polymorphism, and restriction fragment length polymorphism. A total of 93 (94.9 %) out of 98 unrelated cystinuric chromosomes have been characterized. Mutations in SLC3A1 gene account for 64.3 % and in SLC7A9 gene for 30.6 % of the cystinuric chromosomes. Ten different mutations in SLC3A1 gene were found, and two of them were novel (C242R and L573X), while in SLC7A9 gene seven mutations were found, of which three were novel (G73R, V375I and c.1048_1051delACTC). The most common mutations in this study were T216M (24.5 %), M467T (16.3 %) and R365L (11.2 %) in SLC3A1 and G105R (21.4 %) in SLC7A9 gene. A population specificity of cystinuria mutations was observed; T216M mutation was the only mutation present among Gypsies, G105R was the most common mutation among Albanians and Macedonians, and R365L among Serbs. The results of this study allowed introduction of rapid, simple and cost-effective genetic diagnosis of cystinuria that enables an early preventive care of affected patients and a prenatal diagnosis in affected families.
Collapse
Affiliation(s)
- Katerina Popovska-Jankovic
- Macedonian Academy of Science and Arts, Research Centre for Genetic Engineering and Biotechnology "Georgi D. Efremov", 1000, Skopje, Republic of Macedonia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Yang Y, Toy W, Choong LY, Hou P, Ashktorab H, Smoot DT, Yeoh KG, Lim YP. Discovery of SLC3A2 cell membrane protein as a potential gastric cancer biomarker: implications in molecular imaging. J Proteome Res 2012; 11:5736-47. [PMID: 23116296 DOI: 10.1021/pr300555y] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Despite decreasing incidence and mortality, gastric cancer remains the second leading cause of cancer-related deaths in the world. Successful management of gastric cancer is hampered by lack of highly sensitive and specific biomarkers especially for early cancer detection. Cell surface proteins that are aberrantly expressed between normal and cancer cells are potentially useful for cancer imaging and therapy due to easy accessibility of these targets. Combining two-phase partition and isobaric tags for relative and absolute quantification methods, we compared the relative expression levels of membrane proteins between noncancer and gastric cancer cells. About 33% of the data set was found to be plasma membrane and associated proteins using this approach (compared to only 11% in whole cell analysis), several of which have never been previously implicated in gastric cancer. Upregulation of SLC3A2 in gastric cancer cells was validated by immunoblotting of a panel of 13 gastric cancer cell lines and immunohistochemistry on tissue microarrays comprising 85 matched pairs of normal and tumor tissues. Immunofluorescence and immunohistochemistry both confirmed the plasma membrane localization of SLC3A2 in gastric cancer cells. The data supported the notion that SLC3A2 is a potential biomarker that could be exploited for molecular imaging-based detection of gastric cancer.
Collapse
Affiliation(s)
- Yixuan Yang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore 117599
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Kimura T, Yoshida K, Yamamoto C, Suzuki M, Uno T, Isobe M, Naka H, Yasuike S, Satoh M, Kaji T, Uchiyama M. Bis(L-cysteinato)zincate(lI) as a coordination compound that induces metallothionein gene transcription without inducing cell-stress-related gene transcription. J Inorg Biochem 2012; 117:140-6. [PMID: 23085594 DOI: 10.1016/j.jinorgbio.2012.07.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 07/25/2012] [Accepted: 07/31/2012] [Indexed: 11/16/2022]
Abstract
Zinc is an essential micronutrient, deficiency of which results in growth retardation, immunodeficiency, and neurological diseases such as dysgeusia. Several zinc coordination compounds are used for zinc supplementation; however, supplemented zinc ions have no specificity and interact with various groups of molecules. Here, we found that, from a library of 30 zinc coordination compounds, bis(L-cysteinato)zincate(II), designated Z01, functioned as a metallothionein (MT) inducer. Z01 induced MT expression mediated by the transcription factor MTF-1, without inducing cell-stress-related heme oxygenase-1 gene expression at specific concentration. The zinc ion was necessary for the MT induction. (65)Zn incorporation following treatment with (65)Zn-labeled Z01 suggested that Z01 did not act as zinc ionophore despite its hydrophilicity. Electrophoretic mobility shift assays revealed that Z01 facilitates MTF-1-MRE complex formation, and, by inference, transfer of zinc from Z01 to MTF-1. Phosphorylated ERK levels were increased by ZnSO(4) treatment but not by Z01. Although our data do not definitely prove that Z01 is an MTF-1-specific activator, our observations suggest that zinc coordination compounds can regulate zinc distribution and act as zinc donors for specific molecules.
Collapse
Affiliation(s)
- Tomoki Kimura
- Department of Toxicology, Faculty of Pharmaceutical Sciences, Setsunan University, 45-1, Nagaotoge-cho, Hirakata, Osaka 573-0101, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|