1
|
Qiu Y, Stamatatos OT, Hu Q, Ruiter Swain J, Russo S, Sann A, Costa ASH, Violante S, Spector DL, Cross JR, Lukey MJ. The unique catalytic properties of PSAT1 mediate metabolic adaptation to glutamine blockade. Nat Metab 2024; 6:1529-1548. [PMID: 39192144 PMCID: PMC11490312 DOI: 10.1038/s42255-024-01104-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 07/09/2024] [Indexed: 08/29/2024]
Abstract
Cultured cancer cells frequently rely on the consumption of glutamine and its subsequent hydrolysis by glutaminase (GLS). However, this metabolic addiction can be lost in the tumour microenvironment, rendering GLS inhibitors ineffective in the clinic. Here we show that glutamine-addicted breast cancer cells adapt to chronic glutamine starvation, or GLS inhibition, via AMPK-mediated upregulation of the serine synthesis pathway (SSP). In this context, the key product of the SSP is not serine, but α-ketoglutarate (α-KG). Mechanistically, we find that phosphoserine aminotransferase 1 (PSAT1) has a unique capacity for sustained α-KG production when glutamate is depleted. Breast cancer cells with resistance to glutamine starvation or GLS inhibition are highly dependent on SSP-supplied α-KG. Accordingly, inhibition of the SSP prevents adaptation to glutamine blockade, resulting in a potent drug synergism that suppresses breast tumour growth. These findings highlight how metabolic redundancy can be context dependent, with the catalytic properties of different metabolic enzymes that act on the same substrate determining which pathways can support tumour growth in a particular nutrient environment. This, in turn, has practical consequences for therapies targeting cancer metabolism.
Collapse
Affiliation(s)
- Yijian Qiu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | | | - Qingting Hu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- Graduate Program in Genetics, Stony Brook University, Stony Brook, NY, USA
| | - Jed Ruiter Swain
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- CSHL School of Biological Sciences, Cold Spring Harbor, NY, USA
| | - Suzanne Russo
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Ava Sann
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Ana S H Costa
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- Matterworks Inc., Somerville, MA, USA
| | - Sara Violante
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Justin R Cross
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | |
Collapse
|
2
|
Cui Z, Ayva CE, Liew YJ, Guo Z, Mutschler R, Dreier B, Fiorito MM, Walden P, Howard CB, Ely F, Plückthun A, Pretorius C, Ungerer JPJ, Buckle AM, Alexandrov K. mRNA Display Pipeline for Protein Biosensor Construction. ACS Sens 2024; 9:2846-2857. [PMID: 38807313 PMCID: PMC11218749 DOI: 10.1021/acssensors.3c02471] [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: 11/20/2023] [Revised: 05/01/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024]
Abstract
Despite the significant potential of protein biosensors, their construction remains a trial-and-error process. The most obvious approach for addressing this is to utilize modular biosensor architectures where specificity-conferring modalities can be readily generated to recognize new targets. Toward this goal, we established a workflow that uses mRNA display-based selection of hyper-stable monobody domains for the target of choice or ribosome display to select equally stable DARPins. These binders were integrated into a two-component allosteric biosensor architecture based on a calmodulin-reporter chimera. This workflow was tested by developing biosensors for liver toxicity markers such as cytosolic aspartate aminotransferase, mitochondrial aspartate aminotransferase, and alanine aminotransferase 1. We demonstrate that our pipeline consistently produced >103 unique binders for each target within a week. Our analysis revealed that the affinity of the binders for their targets was not a direct predictor of the binder's performance in a biosensor context. The interactions between the binding domains and the reporter module affect the biosensor activity and the dynamic range. We conclude that following binding domain selection, the multiplexed biosensor assembly and prototyping appear to be the most promising approach for identifying biosensors with the desired properties.
Collapse
Affiliation(s)
- Zhenling Cui
- ARC
Centre of Excellence in Synthetic Biology, Brisbane, Queensland 4001, Australia
- Centre
for Agriculture and the Bioeconomy, Queensland
University of Technology, Brisbane, Queensland 4001, Australia
- School
of Biology and Environmental Science, Queensland
University of Technology, Brisbane, Queensland 4001, Australia
| | - Cagla Ergun Ayva
- Centre
for Agriculture and the Bioeconomy, Queensland
University of Technology, Brisbane, Queensland 4001, Australia
- School
of Biology and Environmental Science, Queensland
University of Technology, Brisbane, Queensland 4001, Australia
| | - Yi Jin Liew
- CSIRO
Health & Biosecurity, Westmead, New South Wales 2145,Australia
| | - Zhong Guo
- ARC
Centre of Excellence in Synthetic Biology, Brisbane, Queensland 4001, Australia
- Centre
for Agriculture and the Bioeconomy, Queensland
University of Technology, Brisbane, Queensland 4001, Australia
- School
of Biology and Environmental Science, Queensland
University of Technology, Brisbane, Queensland 4001, Australia
| | - Roxane Mutschler
- Centre
for Agriculture and the Bioeconomy, Queensland
University of Technology, Brisbane, Queensland 4001, Australia
- School
of Biology and Environmental Science, Queensland
University of Technology, Brisbane, Queensland 4001, Australia
| | - Birgit Dreier
- Department
of Biochemistry, University of Zurich, Winterthurerstrasse 190, Zurich CH-8057, Switzerland
| | - Maria M Fiorito
- Centre
for Agriculture and the Bioeconomy, Queensland
University of Technology, Brisbane, Queensland 4001, Australia
- School
of Biology and Environmental Science, Queensland
University of Technology, Brisbane, Queensland 4001, Australia
| | - Patricia Walden
- Centre
for Agriculture and the Bioeconomy, Queensland
University of Technology, Brisbane, Queensland 4001, Australia
- School
of Biology and Environmental Science, Queensland
University of Technology, Brisbane, Queensland 4001, Australia
| | - Christopher B Howard
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | | | - Andreas Plückthun
- Department
of Biochemistry, University of Zurich, Winterthurerstrasse 190, Zurich CH-8057, Switzerland
| | - Carel Pretorius
- Department
of Chemical Pathology, Pathology Queensland, Brisbane, Queensland 4006, Australia
- Faculty
of Health and Behavioural Sciences, The
University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jacobus PJ Ungerer
- Department
of Chemical Pathology, Pathology Queensland, Brisbane, Queensland 4006, Australia
- Faculty
of Health and Behavioural Sciences, The
University of Queensland, Brisbane, Queensland 4072, Australia
| | | | - Kirill Alexandrov
- ARC
Centre of Excellence in Synthetic Biology, Brisbane, Queensland 4001, Australia
- Centre
for Agriculture and the Bioeconomy, Queensland
University of Technology, Brisbane, Queensland 4001, Australia
- School
of Biology and Environmental Science, Queensland
University of Technology, Brisbane, Queensland 4001, Australia
| |
Collapse
|
3
|
Improvement of Fusel Alcohol Production by Engineering of the Yeast Branched-Chain Amino Acid Aminotransaminase. Appl Environ Microbiol 2022; 88:e0055722. [PMID: 35699439 PMCID: PMC9275217 DOI: 10.1128/aem.00557-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Branched-chain higher alcohols (BCHAs), or fusel alcohols, including isobutanol, isoamyl alcohol, and active amyl alcohol, are useful compounds in several industries. The yeast Saccharomyces cerevisiae can synthesize these compounds via the metabolic pathways of branched-chain amino acids (BCAAs). Branched-chain amino acid aminotransaminases (BCATs) are the key enzymes for BCHA production via the Ehrlich pathway of BCAAs. BCATs catalyze a bidirectional transamination reaction between branched-chain α-keto acids (BCKAs) and BCAAs. In S. cerevisiae, there are two BCAT isoforms, Bat1 and Bat2, which are encoded by the genes BAT1 and BAT2. Although many studies have shown the effects of deletion or overexpression of BAT1 and BAT2 on BCHA production, there have been no reports on the enhancement of BCHA production by functional variants of BCATs. Here, to improve BCHA productivity, we designed variants of Bat1 and Bat2 with altered enzyme activity by using in silico computational analysis: the Gly333Ser and Gly333Trp Bat1 and corresponding Gly316Ser and Gly316Trp Bat2 variants, respectively. When expressed in S. cerevisiae cells, most of these variants caused a growth defect in minimal medium. Interestingly, the Gly333Trp Bat1 and Gly316Ser Bat2 variants achieved 18.7-fold and 17.4-fold increases in isobutanol above that for the wild-type enzyme, respectively. The enzyme assay revealed that the catalytic activities of all four BCAT variants were lower than that of the wild-type enzyme. Our results indicate that the decreased BCAT activity enhanced BCHA production by reducing BCAA biosynthesis, which occurs via a pathway that directly competes with BCHA production. IMPORTANCE Recently, several studies have attempted to increase the production of branched-chain higher alcohols (BCHAs) in the yeast Saccharomyces cerevisiae. The key enzymes for BCHA biosynthesis in S. cerevisiae are the branched-chain amino acid aminotransaminases (BCATs) Bat1 and Bat2. Deletion or overexpression of the genes encoding BCATs has an impact on the production of BCHAs; however, amino acid substitution variants of Bat1 and Bat2 that could affect enzymatic properties—and ultimately BCHA productivity—have not been fully studied. By using in silico analysis, we designed variants of Bat1 and Bat2 and expressed them in yeast cells. We found that the engineered BCATs decreased catalytic activities and increased BCHA production. Our approach provides new insight into the functions of BCATs and will be useful in the future construction of enzymes optimized for high-level production of BCHAs.
Collapse
|
4
|
The BCAT1 CXXC Motif Provides Protection against ROS in Acute Myeloid Leukaemia Cells. Antioxidants (Basel) 2022; 11:antiox11040683. [PMID: 35453368 PMCID: PMC9030579 DOI: 10.3390/antiox11040683] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/23/2022] [Accepted: 03/28/2022] [Indexed: 01/15/2023] Open
Abstract
The cytosolic branched-chain aminotransferase (BCAT1) has received attention for its role in myeloid leukaemia development, where studies indicate metabolic adaptations due to BCAT1 up-regulation. BCAT1, like the mitochondria isoform (BCAT2), shares a conserved CXXC motif ~10 Å from the active site. This CXXC motif has been shown to act as a ‘redox-switch’ in the enzymatic regulation of the BCAT proteins, however the response to reactive oxygen species (ROS) differs between BCAT isoforms. Studies indicate that the BCAT1 CXXC motif is several orders of magnitude less sensitive to the effects of ROS compared with BCAT2. Moreover, estimation of the reduction mid-point potential of BCAT1, indicates that BCAT1 is more reductive in nature and may possess antioxidant properties. Therefore, the aim of this study was to further characterise the BCAT1 CXXC motif and evaluate its role in acute myeloid leukaemia. Our biochemical analyses show that purified wild-type (WT) BCAT1 protein could metabolise H2O2 in vitro, whereas CXXC motif mutant or WT BCAT2 could not, demonstrating for the first time a novel antioxidant role for the BCAT1 CXXC motif. Transformed U937 AML cells over-expressing WT BCAT1, showed lower levels of intracellular ROS compared with cells over-expressing the CXXC motif mutant (CXXS) or Vector Controls, indicating that the BCAT1 CXXC motif may buffer intracellular ROS, impacting on cell proliferation. U937 AML cells over-expressing WT BCAT1 displayed less cellular differentiation, as observed by a reduction of the myeloid markers; CD11b, CD14, CD68, and CD36. This finding suggests a role for the BCAT1 CXXC motif in cell development, which is an important pathological feature of myeloid leukaemia, a disease characterised by a block in myeloid differentiation. Furthermore, WT BCAT1 cells were more resistant to apoptosis compared with CXXS BCAT1 cells, an important observation given the role of ROS in apoptotic signalling and myeloid leukaemia development. Since CD36 has been shown to be Nrf2 regulated, we investigated the expression of the Nrf2 regulated gene, TrxRD1. Our data show that the expression of TrxRD1 was downregulated in transformed U937 AML cells overexpressing WT BCAT1, which taken with the reduction in CD36 implicates less Nrf2 activation. Therefore, this finding may implicate the BCAT1 CXXC motif in wider cellular redox-mediated processes. Altogether, this study provides the first evidence to suggest that the BCAT1 CXXC motif may contribute to the buffering of ROS levels inside AML cells, which may impact ROS-mediated processes in the development of myeloid leukaemia.
Collapse
|
5
|
Sonnabend R, Seiler L, Gressler M. Regulation of the Leucine Metabolism in Mortierella alpina. J Fungi (Basel) 2022; 8:196. [PMID: 35205950 PMCID: PMC8880518 DOI: 10.3390/jof8020196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 12/20/2022] Open
Abstract
The oleaginous fungus Mortierella alpina is a safe source of polyunsaturated fatty acids (PUFA) in industrial food and feed production. Besides PUFA production, pharmaceutically relevant surface-active and antimicrobial oligopeptides were isolated from this basal fungus. Both production of fatty acids and oligopeptides rely on the biosynthesis and high turnover of branched-chain-amino acids (BCAA), especially l-leucine. However, the regulation of BCAA biosynthesis in basal fungi is largely unknown. Here, we report on the regulation of the leucine, isoleucine, and valine metabolism in M. alpina. In contrast to higher fungi, the biosynthetic genes for BCAA are hardly transcriptionally regulated, as shown by qRT-PCR analysis, which suggests a constant production of BCAAs. However, the enzymes of the leucine metabolism are tightly metabolically regulated. Three enzymes of the leucine metabolism were heterologously produced in Escherichia coli, one of which is inhibited by allosteric feedback loops: The key regulator is the α-isopropylmalate synthase LeuA1, which is strongly disabled by l-leucine, α-ketoisocaproate, and propionyl-CoA, the precursor of the odd-chain fatty acid catabolism. Its gene is not related to homologs from higher fungi, but it has been inherited from a phototrophic ancestor by horizontal gene transfer.
Collapse
Affiliation(s)
| | | | - Markus Gressler
- Pharmaceutical Microbiology, Friedrich-Schiller-University Jena, Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute, Winzerlaer Strasse 2, 07745 Jena, Germany; (R.S.); (L.S.)
| |
Collapse
|
6
|
Effects of Chronic Arginase Inhibition with Norvaline on Tau Pathology and Brain Glucose Metabolism in Alzheimer's Disease Mice. Neurochem Res 2022; 47:1255-1268. [DOI: 10.1007/s11064-021-03519-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 12/11/2021] [Accepted: 12/27/2021] [Indexed: 12/23/2022]
|
7
|
Ma QX, Zhu WY, Lu XC, Jiang D, Xu F, Li JT, Zhang L, Wu YL, Chen ZJ, Yin M, Huang HY, Lei QY. BCAA-BCKA axis regulates WAT browning through acetylation of PRDM16. Nat Metab 2022; 4:106-122. [PMID: 35075301 DOI: 10.1038/s42255-021-00520-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 12/09/2021] [Indexed: 12/11/2022]
Abstract
The link between branched-chain amino acids (BCAAs) and obesity has been known for decades but the functional role of BCAA metabolism in white adipose tissue (WAT) of obese individuals remains vague. Here, we show that mice with adipose tissue knockout of Bcat2, which converts BCAAs to branched-chain keto acids (BCKAs), are resistant to high-fat diet-induced obesity due to increased inguinal WAT browning and thermogenesis. Mechanistically, acetyl-CoA derived from BCKA suppresses WAT browning by acetylation of PR domain-containing protein 16 (PRDM16) at K915, disrupting the interaction between PRDM16 and peroxisome proliferator-activated receptor-γ (PPARγ) to maintain WAT characteristics. Depletion of BCKA-derived acetyl-CoA robustly prompts WAT browning and energy expenditure. In contrast, BCKA supplementation re-establishes high-fat diet-induced obesity in Bcat2 knockout mice. Moreover, telmisartan, an anti-hypertension drug, significantly represses Bcat2 activity via direct binding, resulting in enhanced WAT browning and reduced adiposity. Strikingly, BCKA supplementation reverses the lean phenotype conferred by telmisartan. Thus, we uncover the critical role of the BCAA-BCKA axis in WAT browning.
Collapse
Affiliation(s)
- Qi-Xiang Ma
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; Shanghai Key Laboratory of Radiation Oncology; The Shanghai Key Laboratory of Medical Epigenetics; Department of Oncology; State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wen-Ying Zhu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; Shanghai Key Laboratory of Radiation Oncology; The Shanghai Key Laboratory of Medical Epigenetics; Department of Oncology; State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiao-Chen Lu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; Shanghai Key Laboratory of Radiation Oncology; The Shanghai Key Laboratory of Medical Epigenetics; Department of Oncology; State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Duo Jiang
- Key Laboratory of Metabolism and Molecular Medicine of Chinese Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Feng Xu
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Jin-Tao Li
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; Shanghai Key Laboratory of Radiation Oncology; The Shanghai Key Laboratory of Medical Epigenetics; Department of Oncology; State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lei Zhang
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Ying-Li Wu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng-Jun Chen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Miao Yin
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; Shanghai Key Laboratory of Radiation Oncology; The Shanghai Key Laboratory of Medical Epigenetics; Department of Oncology; State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Hai-Yan Huang
- Key Laboratory of Metabolism and Molecular Medicine of Chinese Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Qun-Ying Lei
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; Shanghai Key Laboratory of Radiation Oncology; The Shanghai Key Laboratory of Medical Epigenetics; Department of Oncology; State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
8
|
Toyokawa Y, Koonthongkaew J, Takagi H. An overview of branched-chain amino acid aminotransferases: functional differences between mitochondrial and cytosolic isozymes in yeast and human. Appl Microbiol Biotechnol 2021; 105:8059-8072. [PMID: 34622336 DOI: 10.1007/s00253-021-11612-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 09/20/2021] [Accepted: 09/20/2021] [Indexed: 01/07/2023]
Abstract
Branched-chain amino acid aminotransferase (BCAT) catalyzes bidirectional transamination in the cell between branched-chain amino acids (BCAAs; valine, leucine, and isoleucine) and branched-chain α-keto acids (BCKAs; α-ketoisovalerate, α-ketoisocaproate, and α-keto-β-methylvalerate). Eukaryotic cells contain two types of paralogous BCATs: mitochondrial BCAT (BCATm) and cytosolic BCAT (BCATc). Both isozymes have identical enzymatic functions, so they have long been considered to perform similar physiological functions in the cells. However, many studies have gradually revealed the differences in physiological functions and regulatory mechanisms between them. In this article, we present overviews of BCATm and BCATc in both yeast and human. We also introduce BCAT variants found natively or constructed artificially, which could have significant implications for research into the relationship between the primary structures and protein functions of BCATs. KEY POINTS: • BCAT catalyzes bidirectional transamination in the cell between BCAAs and BCKAs. • BCATm and BCATc are different in the metabolic roles and regulatory mechanisms. • BCAT variants offer insight into a relationship between the structure and function.
Collapse
Affiliation(s)
- Yoichi Toyokawa
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara, 630-0192, Japan
| | - Jirasin Koonthongkaew
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara, 630-0192, Japan
| | - Hiroshi Takagi
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara, 630-0192, Japan.
| |
Collapse
|
9
|
Mann G, Mora S, Madu G, Adegoke OAJ. Branched-chain Amino Acids: Catabolism in Skeletal Muscle and Implications for Muscle and Whole-body Metabolism. Front Physiol 2021; 12:702826. [PMID: 34354601 PMCID: PMC8329528 DOI: 10.3389/fphys.2021.702826] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/24/2021] [Indexed: 12/20/2022] Open
Abstract
Branched-chain amino acids (BCAAs) are critical for skeletal muscle and whole-body anabolism and energy homeostasis. They also serve as signaling molecules, for example, being able to activate mammalian/mechanistic target of rapamycin complex 1 (mTORC1). This has implication for macronutrient metabolism. However, elevated circulating levels of BCAAs and of their ketoacids as well as impaired catabolism of these amino acids (AAs) are implicated in the development of insulin resistance and its sequelae, including type 2 diabetes, cardiovascular disease, and of some cancers, although other studies indicate supplements of these AAs may help in the management of some chronic diseases. Here, we first reviewed the catabolism of these AAs especially in skeletal muscle as this tissue contributes the most to whole body disposal of the BCAA. We then reviewed emerging mechanisms of control of enzymes involved in regulating BCAA catabolism. Such mechanisms include regulation of their abundance by microRNA and by post translational modifications such as phosphorylation, acetylation, and ubiquitination. We also reviewed implications of impaired metabolism of BCAA for muscle and whole-body metabolism. We comment on outstanding questions in the regulation of catabolism of these AAs, including regulation of the abundance and post-transcriptional/post-translational modification of enzymes that regulate BCAA catabolism, as well the impact of circadian rhythm, age and mTORC1 on these enzymes. Answers to such questions may facilitate emergence of treatment/management options that can help patients suffering from chronic diseases linked to impaired metabolism of the BCAAs.
Collapse
Affiliation(s)
| | | | | | - Olasunkanmi A. J. Adegoke
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| |
Collapse
|
10
|
Abstract
Significance: Unique to the branched-chain aminotransferase (BCAT) proteins is their redox-active CXXC motif. Subjected to post-translational modification by reactive oxygen species and reactive nitrogen species, these proteins have the potential to adopt numerous cellular roles, which may be fundamental to their role in oncogenesis and neurodegenerative diseases. An understanding of the interplay of the redox regulation of BCAT with important cell signaling mechanisms will identify new targets for future therapeutics. Recent Advances: The BCAT proteins have been assigned novel thiol oxidoreductase activity that can accelerate the refolding of proteins, in particular when S-glutathionylated, supporting a chaperone role for BCAT in protein folding. Other metabolic proteins were also shown to have peroxide-mediated redox associations with BCAT, indicating that the cellular function of BCAT is more diverse. Critical Issues: While the role of branched-chain amino acid metabolism and its metabolites has dominated aspects of cancer research, less is known about the role of BCAT. The importance of the CXXC motif in regulating the BCAT activity under hypoxic conditions, a characteristic of tumors, has not been addressed. Understanding how these proteins operate under various cellular redox conditions will become important, in particular with respect to their moonlighting roles. Future Directions: Advances in the quantification of thiols, their measurement, and the manipulation of metabolons that rely on redox-based interactions should accelerate the investigation of the cellular role of moonlighting proteins such as BCAT. Given the importance of cross talk between signaling pathways, research should focus more on these "housekeeping" proteins paying attention to their wider application. Antioxid. Redox Signal. 34, 1048-1067.
Collapse
Affiliation(s)
- Myra Elizabeth Conway
- Department of Applied Science, University of the West of England, Bristol, United Kingdom
| |
Collapse
|
11
|
Baier J, Gänsbauer M, Giessler C, Arnold H, Muske M, Schleicher U, Lukassen S, Ekici A, Rauh M, Daniel C, Hartmann A, Schmid B, Tripal P, Dettmer K, Oefner PJ, Atreya R, Wirtz S, Bogdan C, Mattner J. Arginase impedes the resolution of colitis by altering the microbiome and metabolome. J Clin Invest 2020; 130:5703-5720. [PMID: 32721946 PMCID: PMC7598089 DOI: 10.1172/jci126923] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 07/16/2020] [Indexed: 02/06/2023] Open
Abstract
Arginase 1 (Arg1), which converts l-arginine into ornithine and urea, exerts pleiotropic immunoregulatory effects. However, the function of Arg1 in inflammatory bowel disease (IBD) remains poorly characterized. Here, we found that Arg1 expression correlated with the degree of inflammation in intestinal tissues from IBD patients. In mice, Arg1 was upregulated in an IL-4/IL-13- and intestinal microbiota-dependent manner. Tie2-Cre Arg1fl/fl mice lacking Arg1 in hematopoietic and endothelial cells recovered faster from colitis than Arg1-expressing (Arg1fl/fl) littermates. This correlated with decreased vessel density, compositional changes in intestinal microbiota, diminished infiltration by myeloid cells, and an accumulation of intraluminal polyamines that promote epithelial healing. The proresolving effect of Arg1 deletion was reduced by an l-arginine-free diet, but rescued by simultaneous deletion of other l-arginine-metabolizing enzymes, such as Arg2 or Nos2, demonstrating that protection from colitis requires l-arginine. Fecal microbiota transfers from Tie2-Cre Arg1fl/fl mice into WT recipients ameliorated intestinal inflammation, while transfers from WT littermates into Arg1-deficient mice prevented an advanced recovery from colitis. Thus, an increased availability of l-arginine as well as altered intestinal microbiota and metabolic products accounts for the accelerated resolution from colitis in the absence of Arg1. Consequently, l-arginine metabolism may serve as a target for clinical intervention in IBD patients.
Collapse
Affiliation(s)
- Julia Baier
- Mikrobiologisches Institut, Klinische Mikrobiologie, Immunologie und Hygiene
| | | | - Claudia Giessler
- Mikrobiologisches Institut, Klinische Mikrobiologie, Immunologie und Hygiene
| | - Harald Arnold
- Mikrobiologisches Institut, Klinische Mikrobiologie, Immunologie und Hygiene
| | - Mercedes Muske
- Mikrobiologisches Institut, Klinische Mikrobiologie, Immunologie und Hygiene
| | - Ulrike Schleicher
- Mikrobiologisches Institut, Klinische Mikrobiologie, Immunologie und Hygiene
| | | | | | | | | | - Arndt Hartmann
- Pathologisches Institut, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Benjamin Schmid
- Optical Imaging Centre Erlangen (OICE), FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Philipp Tripal
- Optical Imaging Centre Erlangen (OICE), FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Katja Dettmer
- Institut für Funktionelle Genomik, Universität Regensburg, Regensburg, Germany
| | - Peter J. Oefner
- Institut für Funktionelle Genomik, Universität Regensburg, Regensburg, Germany
| | - Raja Atreya
- Medizinische Klinik 1–Gastroenterologie, Pneumologie and Endokrinologie, Universitätsklinikum Erlangen and FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Stefan Wirtz
- Medizinische Klinik 1–Gastroenterologie, Pneumologie and Endokrinologie, Universitätsklinikum Erlangen and FAU Erlangen-Nürnberg, Erlangen, Germany
- Medical Immunology Campus Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Bogdan
- Mikrobiologisches Institut, Klinische Mikrobiologie, Immunologie und Hygiene
- Medical Immunology Campus Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Jochen Mattner
- Mikrobiologisches Institut, Klinische Mikrobiologie, Immunologie und Hygiene
- Medical Immunology Campus Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
12
|
Hindy MEL, Conway ME. Redox-Regulated, Targeted Affinity Isolation of NADH-Dependent Protein Interactions with the Branched Chain Aminotransferase Proteins. Methods Mol Biol 2020; 1990:151-163. [PMID: 31148070 DOI: 10.1007/978-1-4939-9463-2_13] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Isolation and identification of protein targets for redox-active proteins is challenging. The human branched chain aminotransferase (hBCAT) proteins are redox active transaminases that can be regulated through oxidation, S-nitrosation and S-glutathionylation. This metabolic protein was shown to associate with the E1 decarboxylase component of the branched-chain α-keto acid dehydrogenase complex in a NADH-dependent manner, where mutation of the CXXC center was shown to prevent complex formation. To determine if the redox state of the CXXC motif can influence other NADH-dependent protein-protein interactions, proteins were extracted from neuronal cells treated under reduced and oxidized conditions and then isolated using targeted affinity chromatography, resolved using 2D electrophoresis. Select proteins spots were excised and identified using a quadrupole time of flight mass spectrometer (Thermo) with a precursor tolerance of 10 ppm and subsequently analyzed using Proteome Discoverer 2.1 with Swiss-Prot human DB. Mass tolerances for precursor/product were set to 10 ppm/0.6 Da and data were filtered by peptide confidence with PD2.1. It was determined that the protein profile considerably altered in both number and abundance dependent on the redox state of the cell and also on the availability of the redox active thiol groups. The biological relevance of the newly identified partners was determined using DAVID, the bioinformatics database, which indicated that proteins important to cytoskeletal function, protein transport, protein synthesis, chaperone activity, and cell signaling.
Collapse
Affiliation(s)
- Maya E L Hindy
- Faculty of Health and Applied Sciences, University of the West of England, Bristol, UK
| | - Myra E Conway
- Department of Applied Sciences, University of the West of England, Bristol, UK.
| |
Collapse
|
13
|
Herbert D, Gibbs S, Riddick A, Conway M, Dong M. Crystal structure of an oxidized mutant of human mitochondrial branched-chain aminotransferase. Acta Crystallogr F Struct Biol Commun 2020; 76:14-19. [PMID: 31929181 PMCID: PMC6957111 DOI: 10.1107/s2053230x19016480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 12/06/2019] [Indexed: 12/02/2022] Open
Abstract
This study presents the crystal structure of a thiol variant of the human mitochondrial branched-chain aminotransferase protein. Human branched-chain aminotransferase (hBCAT) catalyzes the transamination of the branched-chain amino acids leucine, valine and isoleucine and α-ketoglutarate to their respective α-keto acids and glutamate. hBCAT activity is regulated by a CXXC center located approximately 10 Å from the active site. This redox-active center facilitates recycling between the reduced and oxidized states, representing hBCAT in its active and inactive forms, respectively. Site-directed mutagenesis of the redox sensor (Cys315) results in a significant loss of activity, with no loss of activity reported on the mutation of the resolving cysteine (Cys318), which allows the reversible formation of a disulfide bond between Cys315 and Cys318. The crystal structure of the oxidized form of the C318A variant was used to better understand the contributions of the individual cysteines and their oxidation states. The structure reveals the modified CXXC center in a conformation similar to that in the oxidized wild type, supporting the notion that its regulatory mechanism depends on switching the Cys315 side chain between active and inactive conformations. Moreover, the structure reveals conformational differences in the N-terminal and inter-domain region that may correlate with the inactivated state of the CXXC center.
Collapse
Affiliation(s)
- Darius Herbert
- Department of Chemistry, North Carolina Agricultural and Technical State University, USA
| | - Synphane Gibbs
- Department of Biology, North Carolina Agricultural and Technical State University, USA
| | - Alexys Riddick
- Department of Chemistry, North Carolina Agricultural and Technical State University, USA
| | - Myra Conway
- Department of Applied Science, University of the West of England, England
| | - Ming Dong
- Department of Chemistry, North Carolina Agricultural and Technical State University, USA
| |
Collapse
|
14
|
Polis B, Samson AO. Role of the metabolism of branched-chain amino acids in the development of Alzheimer's disease and other metabolic disorders. Neural Regen Res 2020; 15:1460-1470. [PMID: 31997805 PMCID: PMC7059578 DOI: 10.4103/1673-5374.274328] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Alzheimer’s disease is an incurable chronic neurodegenerative disorder and the leading cause of dementia, imposing a growing economic burden upon society. The disease progression is associated with gradual deposition of amyloid plaques and the formation of neurofibrillary tangles within the brain parenchyma, yet severe dementia is the culminating phase of the enduring pathology. Converging evidence suggests that Alzheimer’s disease-related cognitive decline is the outcome of an extremely complex and persistent pathophysiological process. The disease is characterized by distinctive abnormalities apparent at systemic, histological, macromolecular, and biochemical levels. Moreover, besides the well-defined and self-evident characteristic profuse neurofibrillary tangles, dystrophic neurites, and amyloid-beta deposits, the Alzheimer’s disease-associated pathology includes neuroinflammation, substantial neuronal loss, apoptosis, extensive DNA damage, considerable mitochondrial malfunction, compromised energy metabolism, and chronic oxidative stress. Likewise, distinctive metabolic dysfunction has been named a leading cause and a hallmark of Alzheimer’s disease that is apparent decades prior to disease manifestation. State-of-the-art metabolomics studies demonstrate that altered branched-chain amino acids (BCAAs) metabolism accompanies Alzheimer’s disease development. Lower plasma valine levels are correlated with accelerated cognitive decline, and, conversely, an increase in valine concentration is associated with reduced risk of Alzheimer’s disease. Additionally, a clear BCAAs-related metabolic signature has been identified in subjects with obesity, diabetes, and atherosclerosis. Also, arginine metabolism is dramatically altered in Alzheimer’s disease human brains and animal models. Accordingly, a potential role of the urea cycle in the Alzheimer’s disease development has been hypothesized, and preclinical studies utilizing intervention in the urea cycle and/or BCAAs metabolism have demonstrated clinical potential. Continual failures to offer a competent treatment strategy directed against amyloid-beta or Tau proteins-related lesions, which could face all challenges of the multifaceted Alzheimer’s disease pathology, led to the hypothesis that hyperphosphorylated Tau and deposited amyloid-beta proteins are just hallmarks or epiphenomena, but not the ultimate causes of Alzheimer’s disease. Therefore, approaches targeting amyloid-beta or Tau are not adequate to cure the disease. Accordingly, the modern scientific vision of Alzheimer’s disease etiology and pathogenesis must reach beyond the hallmarks, and look for alternative strategies and areas of research.
Collapse
Affiliation(s)
- Baruh Polis
- Drug Discovery Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Abraham O Samson
- Drug Discovery Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| |
Collapse
|
15
|
Polis B, Gilinsky MA, Samson AO. Reports of L-Norvaline Toxicity in Humans May Be Greatly Overstated. Brain Sci 2019; 9:brainsci9120382. [PMID: 31861122 PMCID: PMC6955955 DOI: 10.3390/brainsci9120382] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 12/16/2019] [Indexed: 11/30/2022] Open
Abstract
Recently, a study published in “Toxicology In Vitro” (Kate Samardzic and Kenneth J. Rodgers) was entitled: “Cytotoxicity and Mitochondrial Dysfunction Caused by the Dietary Supplement L-Norvaline”. The title may be greatly overstated, and here we provide several arguments showing that norvaline is not as toxic as reported.
Collapse
Affiliation(s)
- Baruh Polis
- Drug Discovery Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel;
- Correspondence: ; Tel.: +972-525-654-451
| | - Michael A. Gilinsky
- Scientific Research Institute of Physiology and Basic Medicine, 4 Timakova St., Novosibirsk 630117, Russia;
| | - Abraham O. Samson
- Drug Discovery Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel;
| |
Collapse
|
16
|
New tools for an old question: dependence of ATP and bicarbonate for branched-chain keto acids oxidation. Biochem J 2019; 476:2235-2237. [PMID: 31416902 DOI: 10.1042/bcj20190368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/28/2019] [Accepted: 07/29/2019] [Indexed: 11/17/2022]
Abstract
Branched-chain keto acids (BCKA) metabolism involves several well-regulated steps within mitochondria, requires cofactors, and is modulated according to the metabolic status of the cells. This regulation has made it challenging to utilize in vitro approaches to determine the contribution of branched-chain amino acid oxidation to energy production. These methodological issues were elegantly addressed in a recent publication within the Biochemical Journal. In this issue, Goldberg et al. [Biochem. J. (2019) 476, 1521-1537] demonstrated in a well-designed system the dependence of ATP and bicarbonate for BCKA full oxidation. In addition, the utilized system allowed the authors to characterize specific biochemical routes within mitochondria for each BCKA. Among them, a quantitative analysis of the participation of BCKA on mitochondrial flux was estimated between tissues. These findings are milestones with meaningful impact in several fields of metabolism.
Collapse
|
17
|
Forshaw TE, Conway ME. Detection of S-Nitrosation and S-Glutathionylation of the Human Branched-Chain Aminotransferase Proteins. Methods Mol Biol 2019; 1990:71-84. [PMID: 31148063 DOI: 10.1007/978-1-4939-9463-2_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The human branched-chain aminotransferase (hBCAT) enzymes play an integral role in brain glutamate and branched-chain amino acid (BCAA) metabolism. Optimal hBCAT activity is dependent on the oxidation state of their redox reactive thiols, where post-translational modification by nitric oxide (NO) and glutathione results in reversible inhibition. Incubation of the cytosolic isoform (hBCATc) with S-nitrosating agents was found to inhibit in both a time and dose dependent manner through formation of a mixture of products including cysteine-nitric oxide (SNO) and S-glutathionylation. Mechanistic details of these redox interactions were studied using labeling with fluorescein-5-maleimide and confirmed via mass spectrometry and Western blot analysis. Though the mitochondrial isoform (hBCATm) was inhibited by nitrosating agents adduct formation could only be observed by DTNB titration as neither SNO, S-glutathionylation or disulfide bond formation could be detected. These studies revealed that the two isoforms of hBCAT, namely hBCATc and hBCATm, were differently regulated by S-nitrosation or S-glutathionylation pointing to distinct functional/mechanistic responses to GSNO modification. Detection of these adducts is essential for studies into the effect of NO on cells and the redox proteome which can offer insight into several pathological states and normal functioning of the cell.
Collapse
Affiliation(s)
- Thomas E Forshaw
- Faculty of Health and Applied Sciences, University of the West of England, Bristol, UK
| | - Myra E Conway
- Department of Applied Sciences, University of the West of England, Bristol, UK.
| |
Collapse
|
18
|
Caldwell RW, Rodriguez PC, Toque HA, Narayanan SP, Caldwell RB. Arginase: A Multifaceted Enzyme Important in Health and Disease. Physiol Rev 2018; 98:641-665. [PMID: 29412048 PMCID: PMC5966718 DOI: 10.1152/physrev.00037.2016] [Citation(s) in RCA: 290] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 08/14/2017] [Accepted: 08/17/2017] [Indexed: 12/15/2022] Open
Abstract
The arginase enzyme developed in early life forms and was maintained during evolution. As the last step in the urea cycle, arginase cleaves l-arginine to form urea and l-ornithine. The urea cycle provides protection against excess ammonia, while l-ornithine is needed for cell proliferation, collagen formation, and other physiological functions. In mammals, increases in arginase activity have been linked to dysfunction and pathologies of the cardiovascular system, kidney, and central nervous system and also to dysfunction of the immune system and cancer. Two important aspects of the excessive activity of arginase may be involved in diseases. First, overly active arginase can reduce the supply of l-arginine needed for the production of nitric oxide (NO) by NO synthase. Second, too much l-ornithine can lead to structural problems in the vasculature, neuronal toxicity, and abnormal growth of tumor cells. Seminal studies have demonstrated that increased formation of reactive oxygen species and key inflammatory mediators promote this pathological elevation of arginase activity. Here, we review the involvement of arginase in diseases affecting the cardiovascular, renal, and central nervous system and cancer and discuss the value of therapies targeting the elevated activity of arginase.
Collapse
Affiliation(s)
- R William Caldwell
- Department of Pharmacology & Toxicology, Vision Discovery Institute, Department of Medicine-Hematology and Oncology, Department of Occupational Therapy, School of Allied Health Sciences, and Vascular Biology Center, Medical College of Georgia, Augusta University , Augusta, Georgia ; and VA Medical Center, Augusta, Georgia
| | - Paulo C Rodriguez
- Department of Pharmacology & Toxicology, Vision Discovery Institute, Department of Medicine-Hematology and Oncology, Department of Occupational Therapy, School of Allied Health Sciences, and Vascular Biology Center, Medical College of Georgia, Augusta University , Augusta, Georgia ; and VA Medical Center, Augusta, Georgia
| | - Haroldo A Toque
- Department of Pharmacology & Toxicology, Vision Discovery Institute, Department of Medicine-Hematology and Oncology, Department of Occupational Therapy, School of Allied Health Sciences, and Vascular Biology Center, Medical College of Georgia, Augusta University , Augusta, Georgia ; and VA Medical Center, Augusta, Georgia
| | - S Priya Narayanan
- Department of Pharmacology & Toxicology, Vision Discovery Institute, Department of Medicine-Hematology and Oncology, Department of Occupational Therapy, School of Allied Health Sciences, and Vascular Biology Center, Medical College of Georgia, Augusta University , Augusta, Georgia ; and VA Medical Center, Augusta, Georgia
| | - Ruth B Caldwell
- Department of Pharmacology & Toxicology, Vision Discovery Institute, Department of Medicine-Hematology and Oncology, Department of Occupational Therapy, School of Allied Health Sciences, and Vascular Biology Center, Medical College of Georgia, Augusta University , Augusta, Georgia ; and VA Medical Center, Augusta, Georgia
| |
Collapse
|
19
|
Polis B, Samson AO. Arginase as a Potential Target in the Treatment of Alzheimer’s Disease. ACTA ACUST UNITED AC 2018. [DOI: 10.4236/aad.2018.74009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
20
|
Yudkoff M. Interactions in the Metabolism of Glutamate and the Branched-Chain Amino Acids and Ketoacids in the CNS. Neurochem Res 2017; 42:10-18. [PMID: 27696119 PMCID: PMC5285401 DOI: 10.1007/s11064-016-2057-z] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 08/29/2016] [Accepted: 09/01/2016] [Indexed: 01/17/2023]
Abstract
Glutamatergic neurotransmission entails a tonic loss of glutamate from nerve endings into the synapse. Replacement of neuronal glutamate is essential in order to avoid depletion of the internal pool. In brain this occurs primarily via the glutamate-glutamine cycle, which invokes astrocytic synthesis of glutamine and hydrolysis of this amino acid via neuronal phosphate-dependent glutaminase. This cycle maintains constancy of internal pools, but it does not provide a mechanism for inevitable losses of glutamate N from brain. Import of glutamine or glutamate from blood does not occur to any appreciable extent. However, the branched-chain amino acids (BCAA) cross the blood-brain barrier swiftly. The brain possesses abundant branched-chain amino acid transaminase activity which replenishes brain glutamate and also generates branched-chain ketoacids. It seems probable that the branched-chain amino acids and ketoacids participate in a "glutamate-BCAA cycle" which involves shuttling of branched-chain amino acids and ketoacids between astrocytes and neurons. This mechanism not only supports the synthesis of glutamate, it also may constitute a mechanism by which high (and potentially toxic) concentrations of glutamate can be avoided by the re-amination of branched-chain ketoacids.
Collapse
Affiliation(s)
- Marc Yudkoff
- Department of Pediatrics, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
21
|
Ashby EL, Kierzkowska M, Hull J, Kehoe PG, Hutson SM, Conway ME. Altered Expression of Human Mitochondrial Branched Chain Aminotransferase in Dementia with Lewy Bodies and Vascular Dementia. Neurochem Res 2016; 42:306-319. [PMID: 26980008 PMCID: PMC5283609 DOI: 10.1007/s11064-016-1855-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 01/27/2016] [Accepted: 01/28/2016] [Indexed: 11/22/2022]
Abstract
Cytosolic and mitochondrial human branched chain aminotransferase (hBCATc and hBCATm, respectively) play an integral role in brain glutamate metabolism. Regional increased levels of hBCATc in the CA1 and CA4 region of Alzheimer’s disease (AD) brain together with increased levels of hBCATm in frontal and temporal cortex of AD brains, suggest a role for these proteins in glutamate excitotoxicity. Glutamate toxicity is a key pathogenic feature of several neurological disorders including epilepsy associated dementia, AD, vascular dementia (VaD) and dementia with Lewy bodies (DLB). To further understand if these increases are specific to AD, the expression profiles of hBCATc and hBCATm were examined in other forms of dementia including DLB and VaD. Similar to AD, levels of hBCATm were significantly increased in the frontal and temporal cortex of VaD cases and in frontal cortex of DLB cases compared to controls, however there were no observed differences in hBCATc between groups in these areas. Moreover, multiple forms of hBCATm were observed that were particular to the disease state relative to matched controls. Real-time PCR revealed similar expression of hBCATm mRNA in frontal and temporal cortex for all cohort comparisons, whereas hBCATc mRNA expression was significantly increased in VaD cases compared to controls. Collectively our results suggest that hBCATm protein expression is significantly increased in the brains of DLB and VaD cases, similar to those reported in AD brain. These findings indicate a more global response to altered glutamate metabolism and suggest common metabolic responses that might reflect shared neurodegenerative mechanisms across several forms of dementia.
Collapse
Affiliation(s)
- Emma L Ashby
- Department of Applied Science, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK
| | - Marta Kierzkowska
- Department of Applied Science, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK
| | - Jonathon Hull
- Department of Applied Science, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK
| | - Patrick G Kehoe
- Dementia Research Group, Faculty of Medicine and Dentistry, University of Bristol, Bristol, BS16 1LE, UK
| | - Susan M Hutson
- Human Nutrition, Foods, and Exercise, Virginia Tech, 1981 Kraft Drive, 1008 ILSB, Blacksburg, VA, 24060, USA
| | - Myra E Conway
- Department of Applied Science, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK.
| |
Collapse
|
22
|
Conway ME, Hutson SM. BCAA Metabolism and NH3 Homeostasis. ADVANCES IN NEUROBIOLOGY 2016; 13:99-132. [DOI: 10.1007/978-3-319-45096-4_5] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
23
|
Wang XL, Li CJ, Xing Y, Yang YH, Jia JP. Hypervalinemia and hyperleucine-isoleucinemia caused by mutations in the branched-chain-amino-acid aminotransferase gene. J Inherit Metab Dis 2015; 38:855-61. [PMID: 25653144 DOI: 10.1007/s10545-015-9814-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 01/09/2015] [Accepted: 01/12/2015] [Indexed: 10/24/2022]
Abstract
Valine, leucine, and isoleucine are essential branched chain amino acids (BCAAs). When BCAA metabolism is genetically impaired in human, serum levels of BCAA and/or their metabolites rise considerably, causing severe neurological dysfunction. The first step in BCAA catabolism is catalyzed by branched chain aminotransferase (BCAT). Hypervalinemia and hyperleucine-isoleucinemia caused by BCAT gene mutation in human have not been reported previously. A 25-year-old man presented with headache complaints and mild memory impairment for about six years. Brain MRI showed symmetric white matter abnormal signals. Metabolic studies revealed remarkably elevated plasma valine and leucine concentrations. Maple syrup urine disease (MSUD) diagnosis was not supported since all genes for the branched-chain α-keto acid dehydrogenase complex (BCKD) gene were normal. Interestingly, two heterogeneous BCAT2 gene mutations were found in the patient, including c.509G > A (p.Arg170Gln) and c.790G > A (p.Glu264Lys). In addition, c.509G > A (p.Arg170Gln) and c.790G > A (p.Glu264Lys) were found in his father and mother, respectively, suggesting an autosomal recessive disorder. BCAT2 functional studies demonstrated that the two BCAT2 gene mutations resulted in decreased BCAT2 enzyme activity. After treatment with vitamin B6, the levels of BCAA, especially valine were remarkably decreased and brain MRI lesions were improved. These findings suggest a new type of branched chain amino acid metabolism disorder. This rare case provides great insight into the further understanding of BCAA metabolism and its defect in human. BCAT2 gene mutations can cause hypervalinemia and hyperleucine-isoleucinemia, which are associated with brain white matter lesions.
Collapse
Affiliation(s)
- X L Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, People's Republic of China
| | | | | | | | | |
Collapse
|
24
|
Caldwell RB, Toque HA, Narayanan SP, Caldwell RW. Arginase: an old enzyme with new tricks. Trends Pharmacol Sci 2015; 36:395-405. [PMID: 25930708 PMCID: PMC4461463 DOI: 10.1016/j.tips.2015.03.006] [Citation(s) in RCA: 221] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 03/23/2015] [Accepted: 03/30/2015] [Indexed: 01/05/2023]
Abstract
Arginase has roots in early life-forms. It converts L-arginine to urea and ornithine. The former provides protection against NH3; the latter serves to stimulate cell growth and other physiological functions. Excessive arginase activity in mammals has been associated with cardiovascular and nervous system dysfunction and disease. Two relevant aspects of this elevated activity may be involved in these disease states. First, excessive arginase activity reduces the supply of L-arginine needed by nitric oxide (NO) synthase to produce NO. Second, excessive production of ornithine leads to vascular structural problems and neural toxicity. Recent research has identified inflammatory agents and reactive oxygen species (ROS) as drivers of this pathologic elevation of arginase activity and expression. We review the involvement of arginase in cardiovascular and nervous system dysfunction, and discuss potential therapeutic interventions targeting excess arginase.
Collapse
Affiliation(s)
- Ruth B. Caldwell
- VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA
- Vision Discovery Institute, School of Allied Health Sciences, Medical College of Georgia, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
- Vascular Biology Center, School of Allied Health Sciences, Medical College of Georgia, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
| | - Haroldo A. Toque
- Department of Pharmacology & Toxicology, School of Allied Health Sciences, Medical College of Georgia, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
| | - S. Priya Narayanan
- Vision Discovery Institute, School of Allied Health Sciences, Medical College of Georgia, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
- Vascular Biology Center, School of Allied Health Sciences, Medical College of Georgia, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
- Department of Occupational Therapy, School of Allied Health Sciences, Medical College of Georgia, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
| | - R. William Caldwell
- Vision Discovery Institute, School of Allied Health Sciences, Medical College of Georgia, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
- Department of Pharmacology & Toxicology, School of Allied Health Sciences, Medical College of Georgia, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
| |
Collapse
|
25
|
Chronic activation of mTOR complex 1 by branched chain amino acids and organ hypertrophy. Amino Acids 2015; 47:1167-82. [PMID: 25721400 DOI: 10.1007/s00726-015-1944-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 02/14/2015] [Indexed: 12/11/2022]
Abstract
The mitochondrial branched chain aminotransferase-deficient mouse model (BCATm KO), which exhibits elevated plasma and tissue branched chain amino acids (BCAAs), was used to study the effect of BCAAs on mammalian target of rapamycin complex 1 (mTORC1) regulation of organ size. BCATm is the first enzyme in the BCAA catabolic pathway. BCATm KO mouse exhibited hypertrophy of heart, kidneys, and spleen. On the other hand, the mass of the gastrocnemius was reduced relative to body mass. Feeding the mice with a diet supplemented with rapamycin prevented the enlargement of the heart and spleen, suggesting that mTORC1 is the mediator of these effects. Consistently, enlargement of these organs was accompanied by the activation of mTORC1 complex as evidenced by enhanced levels of S6 and 4E-BP1 phosphorylation. HSP20, HSP27 and GAPDH were also increased in the heart but not gastrocnemius, consistent with mTORC1 activation. Liver, however, displayed no weight difference between the KO and the wild-type mice despite the highest activation level of mTORC1 complex. These observations suggest that the anabolic effect of mTORC1 activation at the organ level by BCAAs and inhibition by rapamycin are complex phenomenon and tissue-specific. In addition, it suggests that rapamycin can be used to counter hypertrophy of the organs when activation of mTORC1 is the underlying cause.
Collapse
|
26
|
Hull J, Patel VB, Hutson SM, Conway ME. New insights into the role of the branched-chain aminotransferase proteins in the human brain. J Neurosci Res 2015; 93:987-98. [PMID: 25639459 DOI: 10.1002/jnr.23558] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 11/26/2014] [Accepted: 12/21/2014] [Indexed: 11/10/2022]
Abstract
The human cytosolic branched-chain aminotransferase (hBCATc) enzyme is strategically located in glutamatergic neurons, where it is thought to provide approximately 30% of de novo nitrogen for brain glutamate synthesis. In health, glutamate plays a dominant role in facilitating learning and memory. However, in patients with Alzheimer's disease (AD), synaptic levels of glutamate become toxic, resulting in a direct increase in postsynaptic neuronal calcium, causing a cascade of events that contributes to the destruction of neuronal integrity and cell death, pathological features of AD. Our group is the first to map the hBCAT proteins to the human brain, where cell-specific compartmentation indicates key roles for these proteins in regulating glutamate homeostasis. Moreover, increased expression of hBCAT was observed in the brains of patients with AD relative to matched controls. We reflect on the importance of the redox-active CXXC motif, which confers novel roles for the hBCAT proteins, particularly with respect to substrate channeling and protein folding. This implies that, in addition to their role in glutamate metabolism, these proteins have additional functional roles that might impact redox cell signaling. This review discusses how these proteins behave as potential neuroprotectors during periods of oxidative stress. These findings are particularly important because an increase in misfolded proteins, linked to increased oxidative stress, occurs in several neurodegenerative conditions. Together, these studies give an overview of the diverse role that these proteins play in brain metabolism, in which a dysregulation of their expression may contribute to neurodegenerative conditions such as AD.
Collapse
Affiliation(s)
- Jonathon Hull
- Department of Applied Science, University of the West of England, Bristol, United Kingdom
| | - Vinood B Patel
- Department of Applied Science, University of Westminster, London, United Kingdom
| | - Susan M Hutson
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, Virginia, 24061
| | - Myra E Conway
- Department of Applied Science, University of the West of England, Bristol, United Kingdom
| |
Collapse
|
27
|
El Hindy M, Hezwani M, Corry D, Hull J, El Amraoui F, Harris M, Lee C, Forshaw T, Wilson A, Mansbridge A, Hassler M, Patel VB, Kehoe PG, Love S, Conway ME. The branched-chain aminotransferase proteins: novel redox chaperones for protein disulfide isomerase--implications in Alzheimer's disease. Antioxid Redox Signal 2014; 20:2497-513. [PMID: 24094038 PMCID: PMC4026213 DOI: 10.1089/ars.2012.4869] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
AIMS The human branched-chain aminotransferase proteins (hBCATm and hBCATc) are regulated through oxidation and S-nitrosation. However, it remains unknown whether they share common redox characteristics to enzymes such as protein disulfide isomerase (PDI) in terms of regulating cellular repair and protein misfolding. RESULTS Here, similar to PDI, the hBCAT proteins showed dithiol-disulfide isomerase activity that was mediated through an S-glutathionylated mechanism. Site-directed mutagenesis of the active thiols of the CXXC motif demonstrates that they are fundamental to optimal protein folding. Far Western analysis indicated that both hBCAT proteins can associate with PDI. Co-immunoprecipitation studies demonstrated that hBCATm directly binds to PDI in IMR-32 cells and the human brain. Electron and confocal microscopy validated the expression of PDI in mitochondria (using Mia40 as a mitochondrial control), where both PDI and Mia40 were found to be co-localized with hBCATm. Under conditions of oxidative stress, this interaction is decreased, suggesting that the proposed chaperone role for hBCATm may be perturbed. Moreover, immunohistochemistry studies show that PDI and hBCAT are expressed in the same neuronal and endothelial cells of the vasculature of the human brain, supporting a physiological role for this binding. INNOVATION This study identifies a novel redox role for hBCAT and confirms that hBCATm differentially binds to PDI under cellular stress. CONCLUSION These studies indicate that hBCAT may play a role in the stress response of the cell as a novel redox chaperone, which, if compromised, may result in protein misfolding, creating aggregates as a key feature in neurodegenerative conditions such as Alzheimer's disease.
Collapse
Affiliation(s)
- Maya El Hindy
- 1 Faculty of Health and Life Sciences, University of the West of England , Coldharbor Lane, Bristol, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Zohir N, Afifi R, Ahmed A, Aly Z, Elsobekey M, Kareem H, Helmy R. Role of CYP2C9, VKORC1 and Calumenin Genotypes in Monitoring Warfarin Therapy: An Egyptian Study. Open Access Maced J Med Sci 2013. [DOI: 10.3889/oamjms.2013.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background: Oral anticoagulant therapy is conditioned by environmental and genetic factors.Objectives: To verify the effect of the calumenin, cytochrome P-450 variants and VKORC1 genetic polymorphisms on the response to warfarin therapy and warfarin dose adjustment.Patients and Methods: We selected fifty warfarin treated patients with dose adjusted at INR value between 2 and 3. PCR-RFLP is used for of calumenin gene polymorphism. Insitu Hybridization was used for identification of VKORC1 promoter and CYP2C9 variants polymorphisms.Results: The warfarin dose in the patients with Calumenin and CYP2C9 genetic polymorphism was lower than the wild type gene. The warfarin dose in the patients with VKORC1 variants was statistically lower compared to that of the wild-type. The presence of combined CYP2C9 genetic variants and VKORC1 polymorphism was associated with lower warfarin dose than that the wild types.Conclusion: Calumenin (CALU) might be a new genetic factor involved in the pharmacogenetics of anticoagulant therapy.
Collapse
|
29
|
Narayanan SP, Rojas M, Suwanpradid J, Toque HA, Caldwell RW, Caldwell RB. Arginase in retinopathy. Prog Retin Eye Res 2013; 36:260-80. [PMID: 23830845 PMCID: PMC3759622 DOI: 10.1016/j.preteyeres.2013.06.002] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 06/14/2013] [Accepted: 06/25/2013] [Indexed: 12/12/2022]
Abstract
Ischemic retinopathies, such as diabetic retinopathy (DR), retinopathy of prematurity and retinal vein occlusion are a major cause of blindness in developed nations worldwide. Each of these conditions is associated with early neurovascular dysfunction. However, conventional therapies target clinically significant macula edema or neovascularization, which occur much later. Intra-ocular injections of anti-VEGF show promise in reducing retinal edema, but the effects are usually transient and the need for repeated injections increases the risk of intraocular infection. Laser photocoagulation can control pathological neovascularization, but may impair vision and in some patients the retinopathy continues to progress. Moreover, neither treatment targets early stage disease or promotes repair. This review examines the potential role of the ureahydrolase enzyme arginase as a therapeutic target for the treatment of ischemic retinopathy. Arginase metabolizes l-arginine to form proline, polyamines and glutamate. Excessive arginase activity reduces the l-arginine supply for nitric oxide synthase (NOS), causing it to become uncoupled and produce superoxide and less NO. Superoxide and NO react and form the toxic oxidant peroxynitrite. The catabolic products of polyamine oxidation and glutamate can induce more oxidative stress and DNA damage, both of which can cause cellular injury. Studies indicate that neurovascular injury during retinopathy is associated with increased arginase expression/activity, decreased NO, polyamine oxidation, formation of superoxide and peroxynitrite and dysfunction and injury of both vascular and neural cells. Furthermore, data indicate that the cytosolic isoform arginase I (AI) is involved in hyperglycemia-induced dysfunction and injury of vascular endothelial cells whereas the mitochondrial isoform arginase II (AII) is involved in neurovascular dysfunction and death following hyperoxia exposure. Thus, we postulate that activation of the arginase pathway causes neurovascular injury by uncoupling NOS and inducing polyamine oxidation and glutamate formation, thereby reducing NO and increasing oxidative stress, all of which contribute to the retinopathic process.
Collapse
Affiliation(s)
- S. Priya Narayanan
- Vision Discovery Institute, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
- Vascular Biology Center, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
| | - Modesto Rojas
- Vision Discovery Institute, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
- Vascular Biology Center, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
| | - Jutamas Suwanpradid
- Vision Discovery Institute, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
- Vascular Biology Center, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
| | - Haroldo A. Toque
- Department of Pharmacology & Toxicology, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
| | - R. William Caldwell
- Vision Discovery Institute, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
- Department of Pharmacology & Toxicology, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
| | - Ruth B. Caldwell
- Vision Discovery Institute, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
- Vascular Biology Center, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
- VA Medical Center, One Freedom Way, Augusta, GA, USA
| |
Collapse
|
30
|
Tönjes M, Barbus S, Park YJ, Wang W, Schlotter M, Lindroth AM, Pleier SV, Bai AHC, Karra D, Piro RM, Felsberg J, Addington A, Lemke D, Weibrecht I, Hovestadt V, Rolli CG, Campos B, Turcan S, Sturm D, Witt H, Chan TA, Herold-Mende C, Kemkemer R, König R, Schmidt K, Hull WE, Pfister SM, Jugold M, Hutson SM, Plass C, Okun JG, Reifenberger G, Lichter P, Radlwimmer B. BCAT1 promotes cell proliferation through amino acid catabolism in gliomas carrying wild-type IDH1. Nat Med 2013; 19:901-908. [PMID: 23793099 DOI: 10.1038/nm.3217] [Citation(s) in RCA: 369] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 05/01/2013] [Indexed: 12/11/2022]
Abstract
Here we show that glioblastoma express high levels of branched-chain amino acid transaminase 1 (BCAT1), the enzyme that initiates the catabolism of branched-chain amino acids (BCAAs). Expression of BCAT1 was exclusive to tumors carrying wild-type isocitrate dehydrogenase 1 (IDH1) and IDH2 genes and was highly correlated with methylation patterns in the BCAT1 promoter region. BCAT1 expression was dependent on the concentration of α-ketoglutarate substrate in glioma cell lines and could be suppressed by ectopic overexpression of mutant IDH1 in immortalized human astrocytes, providing a link between IDH1 function and BCAT1 expression. Suppression of BCAT1 in glioma cell lines blocked the excretion of glutamate and led to reduced proliferation and invasiveness in vitro, as well as significant decreases in tumor growth in a glioblastoma xenograft model. These findings suggest a central role for BCAT1 in glioma pathogenesis, making BCAT1 and BCAA metabolism attractive targets for the development of targeted therapeutic approaches to treat patients with glioblastoma.
Collapse
Affiliation(s)
- Martje Tönjes
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sebastian Barbus
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Yoon Jung Park
- Division of Epigenomics and Cancer Risk Factors, DKFZ, Heidelberg, Germany.,Department of Nutritional Science and Food Management, College of Health Science, Ewha Womans University, Seoul, South Korea
| | - Wei Wang
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Magdalena Schlotter
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anders M Lindroth
- Division of Epigenomics and Cancer Risk Factors, DKFZ, Heidelberg, Germany
| | - Sabrina V Pleier
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Division of Pediatric Neurooncology, DKFZ, Heidelberg, Germany
| | - Alfa H C Bai
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniela Karra
- Department of Neuropathology, Heinrich Heine University, Düsseldorf, Germany
| | - Rosario M Piro
- Department of Bioinformatics and Functional Genomics, Institute of Pharmacy and Molecular Biotechnology and Bioquant, University of Heidelberg, Heidelberg, Germany.,Division of Theoretical Bioinformatics, DKFZ, Heidelberg, Germany
| | - Jörg Felsberg
- Department of Neuropathology, Heinrich Heine University, Düsseldorf, Germany
| | - Adele Addington
- Department of Human Nutrition, Foods and Exercise, Virginia Polytechnic Institute, Blacksburg, Virginia, USA
| | - Dieter Lemke
- Clinical Cooperation Unit Neurooncology, DKFZ, Heidelberg, Germany
| | - Irene Weibrecht
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Volker Hovestadt
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Claudio G Rolli
- Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, Stuttgart, Germany
| | - Benito Campos
- Division of Experimental Neurosurgery, University of Heidelberg, Heidelberg, Germany.,Department of Neurosurgery, University of Heidelberg, Heidelberg, Germany
| | - Sevin Turcan
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | - Dominik Sturm
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Division of Pediatric Neurooncology, DKFZ, Heidelberg, Germany.,Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Hendrik Witt
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Division of Pediatric Neurooncology, DKFZ, Heidelberg, Germany.,Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Timothy A Chan
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | - Christel Herold-Mende
- Division of Experimental Neurosurgery, University of Heidelberg, Heidelberg, Germany.,Department of Neurosurgery, University of Heidelberg, Heidelberg, Germany
| | - Ralf Kemkemer
- Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, Stuttgart, Germany.,Reutlingen University of Applied Science, Reutlingen, Germany
| | - Rainer König
- Department of Bioinformatics and Functional Genomics, Institute of Pharmacy and Molecular Biotechnology and Bioquant, University of Heidelberg, Heidelberg, Germany.,Division of Theoretical Bioinformatics, DKFZ, Heidelberg, Germany
| | - Kathrin Schmidt
- Department of General Pediatrics, Division of Inborn Metabolic Diseases, University Children's Hospital, Heidelberg, Germany
| | | | - Stefan M Pfister
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Division of Pediatric Neurooncology, DKFZ, Heidelberg, Germany.,Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Manfred Jugold
- Core Facility, Small Animal Imaging Center, DKFZ, Heidelberg, Germany
| | - Susan M Hutson
- Department of Human Nutrition, Foods and Exercise, Virginia Polytechnic Institute, Blacksburg, Virginia, USA
| | - Christoph Plass
- Division of Epigenomics and Cancer Risk Factors, DKFZ, Heidelberg, Germany
| | - Jürgen G Okun
- Department of General Pediatrics, Division of Inborn Metabolic Diseases, University Children's Hospital, Heidelberg, Germany
| | - Guido Reifenberger
- Department of Neuropathology, Heinrich Heine University, Düsseldorf, Germany.,German Cancer Consortium (DKTK), DKFZ, Heidelberg, Germany
| | - Peter Lichter
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Bernhard Radlwimmer
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
31
|
Hull J, Hindy ME, Kehoe PG, Chalmers K, Love S, Conway ME. Distribution of the branched chain aminotransferase proteins in the human brain and their role in glutamate regulation. J Neurochem 2012; 123:997-1009. [PMID: 23043456 DOI: 10.1111/jnc.12044] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 08/15/2012] [Accepted: 10/02/2012] [Indexed: 12/16/2022]
Abstract
The branched chain aminotransferase enzymes (BCAT) serve as nitrogen donors for the production of 30% of de novo glutamate synthesis in rat brain. Despite the importance of this major metabolite and excitatory neurotransmitter, the distribution of BCAT proteins in the human brain (hBCAT) remains unreported. We have studied this and report, for the first time, that the mitochondrial isoform, hBCATm is largely confined to vascular endothelial cells, whereas the cytosolic hBCATc is restricted to neurons. The majority of hBCATc-labelled neurons were either GABA-ergic or glutamatergic showing both cell body and axonal staining indicating a role for hBCATc in both glutamate production and glutamate release during excitation. Strong staining in hormone secreting cells suggests a further role for the transaminases in hormone regulation potentially similar to that proposed for insulin secretion. Expression of hBCATm in the endothelial cells of the vasculature demonstrates for the first time that glutamate could be metabolized by aminotranferases in these cells. This has important implications given that the dysregulation of glutamate metabolism, leading to glutamate excitotoxicity, is an important contributor to the pathogenesis of several neurodegenerative conditions, where the role of hBCATm in metabolizing excess glutamate may factor more prominently.
Collapse
Affiliation(s)
- Jonathon Hull
- Faculty of Health and Life Sciences, University of the West of England, Bristol, UK
| | | | | | | | | | | |
Collapse
|
32
|
Crystal structures of complexes of the branched-chain aminotransferase from Deinococcus radiodurans with α-ketoisocaproate and L-glutamate suggest the radiation resistance of this enzyme for catalysis. J Bacteriol 2012; 194:6206-16. [PMID: 22984263 DOI: 10.1128/jb.01659-12] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Branched-chain aminotransferases (BCAT), which utilize pyridoxal 5'-phosphate (PLP) as a cofactor, reversibly catalyze the transfer of the α-amino groups of three of the most hydrophobic branched-chain amino acids (BCAA), leucine, isoleucine, and valine, to α-ketoglutarate to form the respective branched-chain α-keto acids and glutamate. The BCAT from Deinococcus radiodurans (DrBCAT), an extremophile, was cloned and expressed in Escherichia coli for structure and functional studies. The crystal structures of the native DrBCAT with PLP and its complexes with L-glutamate and α-ketoisocaproate (KIC), respectively, have been determined. The DrBCAT monomer, comprising 358 amino acids, contains large and small domains connected with an interdomain loop. The cofactor PLP is located at the bottom of the active site pocket between two domains and near the dimer interface. The substrate (L-glutamate or KIC) is bound with key residues through interactions of the hydrogen bond and the salt bridge near PLP inside the active site pocket. Mutations of some interaction residues, such as Tyr71, Arg145, and Lys202, result in loss of the specific activity of the enzymes. In the interdomain loop, a dynamic loop (Gly173 to Gly179) clearly exhibits open and close conformations in structures of DrBCAT without and with substrates, respectively. DrBCAT shows the highest specific activity both in nature and under ionizing radiation, but with lower thermal stability above 60 °C, than either BCAT from Escherichia coli (eBCAT) or from Thermus thermophilus (HB8BCAT). The dimeric molecular packing and the distribution of cysteine residues at the active site and the molecular surface might explain the resistance to radiation but small thermal stability of DrBCAT.
Collapse
|
33
|
Coles SJ, Hancock JT, Conway ME. Differential redox potential between the human cytosolic and mitochondrial branched-chain aminotransferase. Acta Biochim Biophys Sin (Shanghai) 2012; 44:172-6. [PMID: 22107788 DOI: 10.1093/abbs/gmr103] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The human branched-chain aminotransferase (hBCAT) isoenzymes are CXXC motif redox sensitive homodimers central to glutamate metabolism in the central nervous system. These proteins respond differently to oxidation by H(2)O(2), NO, and S-glutathionylation, suggesting that the redox potential is distinct between isoenzymes. Using various reduced to oxidized glutathione ratios (GSH:GSSG) to alter the redox environment, we demonstrate that hBCATc (cytosolic) has an overall redox potential that is 30 mV lower than hBCATm (mitochondrial). Furthermore, the CXXC motif of hBCATc was estimated to be 80 mV lower, suggesting that hBCATm is more oxidizing in nature. Western blot analysis revealed close correlations between hBCAT S-glutathionylation and the redox status of the assay environment, offering the hBCAT isoenzymes as novel biomarkers for cytosolic and mitochondrial oxidative stress.
Collapse
Affiliation(s)
- Steven J Coles
- Department of Medical Genetics, Haematology and Pathology, School of Medicine, Cardiff University, South Wales, UK
| | | | | |
Collapse
|
34
|
Hutson SM, Islam MM, Zaganas I. Interaction between glutamate dehydrogenase (GDH) and l-leucine catabolic enzymes: Intersecting metabolic pathways. Neurochem Int 2011; 59:518-24. [DOI: 10.1016/j.neuint.2011.05.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 03/28/2011] [Accepted: 05/03/2011] [Indexed: 10/18/2022]
|
35
|
Abstract
The perception of wine flavor and aroma is the result of a multitude of interactions between a large number of chemical compounds and sensory receptors. Compounds interact and combine and show synergistic (i.e., the presence of one compound enhances the perception of another) and antagonistic (a compound suppresses the perception of another) interactions. The chemical profile of a wine is derived from the grape, the fermentation microflora (in particular the yeast Saccharomyces cerevisiae), secondary microbial fermentations that may occur, and the aging and storage conditions. Grape composition depends on the varietal and clonal genotype of the vine and on the interaction of the genotype and its phenotype with many environmental factors which, in wine terms, are usually grouped under the concept of "terroir" (macro, meso and microclimate, soil, topography). The microflora, and in particular the yeast responsible for fermentation, contributes to wine aroma by several mechanisms: firstly by utilizing grape juice constituents and biotransforming them into aroma- or flavor-impacting components, secondly by producing enzymes that transform neutral grape compounds into flavor-active compounds, and lastly by the de novo synthesis of many flavor-active primary (e.g., ethanol, glycerol, acetic acid, and acetaldehyde) and secondary metabolites (e.g., esters, higher alcohols, fatty acids). This review aims to present an overview of the formation of wine flavor and aroma-active components, including the varietal precursor molecules present in grapes and the chemical compounds produced during alcoholic fermentation by yeast, including compounds directly related to ethanol production or secondary metabolites. The contribution of malolactic fermentation, ageing, and maturation on the aroma and flavor of wine is also discussed.
Collapse
|
36
|
Mitręga K, Zorniak M, Varghese B, Lange D, Nożynski J, Porc M, Białka S, Krzemiński TF. Beneficial effects of l-leucine and l-valine on arrhythmias, hemodynamics and myocardial morphology in rats. Pharmacol Res 2011; 64:218-25. [PMID: 21605982 DOI: 10.1016/j.phrs.2011.04.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Revised: 04/14/2011] [Accepted: 04/26/2011] [Indexed: 01/19/2023]
Abstract
Branched chain amino acids (BCAA) have been shown to have a general protective effect on the heart in different animal models as well as in humans. However, so far no attempt has been made to specifically elucidate their influence on arrhythmias. Our study was performed to evaluate whether an infusion of either l-leucine or l-valine in a dose of 1mgkg(-1)h(-1) 10min before a 7-min period of left anterior descending artery occlusion followed by 15min of reperfusion, had an effect on arrhythmias measured during the reperfusion phase in the ischemia- and reperfusion-induced arrhythmias model in rats in vivo. The effect of the infusion of these substances on mean arterial blood pressure was monitored throughout the experiment. Both of the tested amino acids exhibited significant antiarrhythmic properties. l-Leucine reduced the duration of ventricular fibrillation (P<0.05) and l-valine decreased the duration of ventricular fibrillation (P<0.001) and ventricular tachycardia (P<0.05). The two amino acids were generally hypotensive. l-Valine lowered blood pressure in all phases of the experiment (P<0.05) while l-leucine lowered this parameter mainly towards the end of occlusion and reperfusion (P<0.05). In addition, 30min infusion of the amino acids in the used dose did not produce any apparent adverse histological changes that were remarkably different from control. In summary, the results of our study suggest that l-leucine and l-valine in the dose that was used attenuates arrhythmias and are hypotensive in their influence. Our findings lend support to the many ongoing investigations into the benefit of the application of l-leucine and l-valine in cardiology like their addition to cardioplegic solutions.
Collapse
Affiliation(s)
- Katarzyna Mitręga
- Chair and Department of Pharmacology, Medical University of Silesia, ul. Jordana 19, 41-808 Zabrze, Poland.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Karimpour S, Davoodi J, Ghahremani MH. Integrity of ATP binding site is essential for effective inhibition of the intrinsic apoptosis pathway by NAIP. Biochem Biophys Res Commun 2011; 407:158-62. [PMID: 21371431 DOI: 10.1016/j.bbrc.2011.02.130] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2011] [Accepted: 02/25/2011] [Indexed: 11/30/2022]
Abstract
The importance of the ATP binding site of human Neuronal Apoptosis Inhibitory Protein (NAIP) on its ability in prevention of intrinsic apoptotic pathway was investigated. Thus, ATP binding lysine 476 of NAIP, which is located at the Nucleotide Binding Oligomerization Domain (NOD) was mutated to threonine and the effect of this mutation on autoproteolysis of procaspase-9 and the cleavage of procaspase-3 by apoptosome was investigated. Formation of apoptosome was induced by the addition of cytochrome c and dATP to lysates of HeLa cells transfected with pcDNA-NAIP or pcDNA-NAIP (K476T). Full length wild type NAIP prevented the cleavage of both procaspase-9 to caspase-9 and procaspase-3 to caspase-3. However, K476T variant of NAIP did not block autocleavage of procaspase-9 efficiently. Furthermore, cleavage pattern of procaspase-9 was altered in the presence of mutant NAIP. Interestingly no effect on the procaspase-3 cleavage by apoptosome was observed. The presence of NOD domain by itself had no effect on autocleavage of procaspase-9 yet slightly reduced the cleavage of procaspase-3 by apoptosome. Pull down experiment showed direct interaction of the NOD domain of NAIP with the CARD-NOD domain of Apoptotic Protease Activating Factor 1 (APAF-1). The physical association of these domains was confirmed by pull-down assays. These observations taken with previous findings indicate that the integrity of the NOD domain is essential for effective inhibition of procaspase-9 and procaspase-3 cleavage by the NAIP protein.
Collapse
Affiliation(s)
- Sarvenaz Karimpour
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417653761, Iran
| | | | | |
Collapse
|
38
|
Wrenger C, Müller IB, Schifferdecker AJ, Jain R, Jordanova R, Groves MR. Specific inhibition of the aspartate aminotransferase of Plasmodium falciparum. J Mol Biol 2010; 405:956-71. [PMID: 21087616 DOI: 10.1016/j.jmb.2010.11.018] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Revised: 11/05/2010] [Accepted: 11/09/2010] [Indexed: 10/18/2022]
Abstract
Aspartate aminotransferases (AspATs; EC 2.6.1.1) catalyze the conversion of aspartate and α-ketoglutarate into oxaloacetate and glutamate and are key enzymes in the nitrogen metabolism of all organisms. Recent findings suggest that the plasmodial enzyme [Plasmodium falciparum aspartate aminotransferase (PfAspAT)] may also play a pivotal role in energy metabolism and in the de novo biosynthesis of pyrimidines. However, while PfAspAT is a potential drug target, the high homology between the active sites of currently available AspAT structures hinders the development of specific inhibitors of these enzymes. In this article, we report the X-ray structure of the PfAspAT homodimer at a resolution of 2.8 Å. While the overall fold is similar to the currently available structures of other AspATs, the structure presented shows a significant divergence in the conformation of the N-terminal residues. Deletion of these divergent PfAspAT N-terminal residues results in a loss of activity for the recombinant protein, and addition of a peptide containing these 13 N-terminal residues results in inhibition both in vitro and in a lysate isolated from cultured parasites, while the activity of human cytosolic AspAT is unaffected. The finding that the divergent N-terminal amino acids of PfAspAT play a role in catalytic activity indicates that specific inhibition of the enzyme may provide a lead for the development of novel compounds in the treatment of malaria. We also report on the localization of PfAspAT to the parasite cytosol and discuss the implications of the role of PfAspAT in the supply of malate to the parasite mitochondria.
Collapse
Affiliation(s)
- Carsten Wrenger
- Department of Biochemistry, Bernhard Nocht Institute for Tropical Medicine, Bernhard Nocht Strasse 74, D-20359 Hamburg, Germany
| | | | | | | | | | | |
Collapse
|
39
|
Jain R, Jordanova R, Müller IB, Wrenger C, Groves MR. Purification, crystallization and preliminary X-ray analysis of the aspartate aminotransferase of Plasmodium falciparum. Acta Crystallogr Sect F Struct Biol Cryst Commun 2010; 66:409-12. [PMID: 20383010 PMCID: PMC2852332 DOI: 10.1107/s1744309110003933] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Accepted: 02/01/2010] [Indexed: 11/10/2022]
Abstract
Aspartate aminotransferases (EC 2.6.1.1) catalyse the conversion of aspartate and alpha-ketoglutarate to oxaloacetate and glutamate in a reversible manner. Thus, the aspartate aminotransferase of Plasmodium falciparum (PfAspAT) plays a central role in the transamination of amino acids. Recent findings suggest that PfAspAT may also play a pivotal role in energy metabolism and the de novo biosynthesis of pyrimidines. While therapeutics based upon the inhibition of other proteins in these pathways are already used in the treatment of malaria, the advent of multidrug-resistant strains has limited their efficacy. The presence of PfAspAT in these pathways may offer additional opportunities for the development of novel therapeutics. In order to gain a deeper understanding of the function and role of PfAspAT, it has been expressed and purified to homogeneity. The successful crystallization of PfAspAT, the collection of a 2.8 A diffraction data set and initial attempts to solve the structure using molecular replacement are reported.
Collapse
Affiliation(s)
- Rishabh Jain
- European Molecular Biology Laboratory Hamburg Outstation, Notkestrasse 85, D-22603 Hamburg, Germany
| | - Rositsa Jordanova
- European Molecular Biology Laboratory Hamburg Outstation, Notkestrasse 85, D-22603 Hamburg, Germany
| | - Ingrid B. Müller
- Bernhard Nocht Institute for Tropical Medicine, Department of Biochemistry, Bernhard Nocht Strasse 74, D-20359 Hamburg, Germany
| | - Carsten Wrenger
- Bernhard Nocht Institute for Tropical Medicine, Department of Biochemistry, Bernhard Nocht Strasse 74, D-20359 Hamburg, Germany
| | - Matthew R. Groves
- European Molecular Biology Laboratory Hamburg Outstation, Notkestrasse 85, D-22603 Hamburg, Germany
| |
Collapse
|
40
|
Gonda I, Bar E, Portnoy V, Lev S, Burger J, Schaffer AA, Tadmor Y, Gepstein S, Giovannoni JJ, Katzir N, Lewinsohn E. Branched-chain and aromatic amino acid catabolism into aroma volatiles in Cucumis melo L. fruit. JOURNAL OF EXPERIMENTAL BOTANY 2010; 61:1111-23. [PMID: 20065117 PMCID: PMC2826658 DOI: 10.1093/jxb/erp390] [Citation(s) in RCA: 181] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2009] [Revised: 10/29/2009] [Accepted: 12/02/2009] [Indexed: 05/18/2023]
Abstract
The unique aroma of melons (Cucumis melo L., Cucurbitaceae) is composed of many volatile compounds biosynthetically derived from fatty acids, carotenoids, amino acids, and terpenes. Although amino acids are known precursors of aroma compounds in the plant kingdom, the initial steps in the catabolism of amino acids into aroma volatiles have received little attention. Incubation of melon fruit cubes with amino acids and alpha-keto acids led to the enhanced formation of aroma compounds bearing the side chain of the exogenous amino or keto acid supplied. Moreover, L-[(13)C(6)]phenylalanine was also incorporated into aromatic volatile compounds. Amino acid transaminase activities extracted from the flesh of mature melon fruits converted L-isoleucine, L-leucine, L-valine, L-methionine, or L-phenylalanine into their respective alpha-keto acids, utilizing alpha-ketoglutarate as the amine acceptor. Two novel genes were isolated and characterized (CmArAT1 and CmBCAT1) encoding 45.6 kDa and 42.7 kDa proteins, respectively, that displayed aromatic and branched-chain amino acid transaminase activities, respectively, when expressed in Escherichia coli. The expression of CmBCAT1 and CmArAT1 was low in vegetative tissues, but increased in flesh and rind tissues during fruit ripening. In addition, ripe fruits of climacteric aromatic cultivars generally showed high expression of CmBCAT1 and CmArAT1 in contrast to non-climacteric non-aromatic fruits. The results presented here indicate that in melon fruit tissues, the catabolism of amino acids into aroma volatiles can initiate through a transamination mechanism, rather than decarboxylation or direct aldehyde synthesis, as has been demonstrated in other plants.
Collapse
Affiliation(s)
- Itay Gonda
- Institute of Plant Sciences, Newe Ya'ar Research Center, Agricultural Research Organization, PO Box 1021, Ramat Yishay 30095, Israel
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Einat Bar
- Institute of Plant Sciences, Newe Ya'ar Research Center, Agricultural Research Organization, PO Box 1021, Ramat Yishay 30095, Israel
| | - Vitaly Portnoy
- Institute of Plant Sciences, Newe Ya'ar Research Center, Agricultural Research Organization, PO Box 1021, Ramat Yishay 30095, Israel
| | - Shery Lev
- Institute of Plant Sciences, Newe Ya'ar Research Center, Agricultural Research Organization, PO Box 1021, Ramat Yishay 30095, Israel
| | - Joseph Burger
- Institute of Plant Sciences, Newe Ya'ar Research Center, Agricultural Research Organization, PO Box 1021, Ramat Yishay 30095, Israel
| | - Arthur A. Schaffer
- Institute of Plant Sciences, Newe Ya'ar Research Center, Agricultural Research Organization, PO Box 1021, Ramat Yishay 30095, Israel
| | - Ya'akov Tadmor
- Institute of Plant Sciences, Newe Ya'ar Research Center, Agricultural Research Organization, PO Box 1021, Ramat Yishay 30095, Israel
| | - Shimon Gepstein
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - James J. Giovannoni
- United States Department of Agriculture and Boyce Thompson Institute for Plant Research, Cornell University, Tower Road, Ithaca, NY 14853, USA
| | - Nurit Katzir
- Institute of Plant Sciences, Newe Ya'ar Research Center, Agricultural Research Organization, PO Box 1021, Ramat Yishay 30095, Israel
| | - Efraim Lewinsohn
- Institute of Plant Sciences, Newe Ya'ar Research Center, Agricultural Research Organization, PO Box 1021, Ramat Yishay 30095, Israel
| |
Collapse
|
41
|
Islam MM, Nautiyal M, Wynn RM, Mobley JA, Chuang DT, Hutson SM. Branched-chain amino acid metabolon: interaction of glutamate dehydrogenase with the mitochondrial branched-chain aminotransferase (BCATm). J Biol Chem 2010; 285:265-76. [PMID: 19858196 PMCID: PMC2804173 DOI: 10.1074/jbc.m109.048777] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Revised: 10/08/2009] [Indexed: 01/23/2023] Open
Abstract
The catabolic pathway for branched-chain amino acids includes deamination followed by oxidative decarboxylation of the deaminated product branched-chain alpha-keto acids, catalyzed by the mitochondrial branched-chain aminotransferase (BCATm) and branched-chain alpha-keto acid dehydrogenase enzyme complex (BCKDC). We found that BCATm binds to the E1 decarboxylase of BCKDC, forming a metabolon that allows channeling of branched-chain alpha-keto acids from BCATm to E1. The protein complex also contains glutamate dehydrogenase (GDH1), 4-nitrophenylphosphatase domain and non-neuronal SNAP25-like protein homolog 1, pyruvate carboxylase, and BCKDC kinase. GDH1 binds to the pyridoxamine 5'-phosphate (PMP) form of BCATm (PMP-BCATm) but not to the pyridoxal 5'-phosphate-BCATm and other metabolon proteins. Leucine activates GDH1, and oxidative deamination of glutamate is increased further by addition of PMP-BCATm. Isoleucine and valine are not allosteric activators of GDH1, but in the presence of 5'-phosphate-BCATm, they convert BCATm to PMP-BCATm, stimulating GDH1 activity. Sensitivity to ADP activation of GDH1 was unaffected by PMP-BCATm; however, addition of a 3 or higher molar ratio of PMP-BCATm to GDH1 protected GDH1 from GTP inhibition by 50%. Kinetic results suggest that GDH1 facilitates regeneration of the form of BCATm that binds to E1 decarboxylase of the BCKDC, promotes metabolon formation, branched-chain amino acid oxidation, and cycling of nitrogen through glutamate.
Collapse
Affiliation(s)
- Mohammad Mainul Islam
- From the Department of Human Nutrition, Foods and Exercise, Virginia Polytechnic Institute, Blacksburg, Virginia 24061
| | - Manisha Nautiyal
- the Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157
| | - R. Max Wynn
- the Departments of Biochemistry and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, and
| | - James A. Mobley
- the Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - David T. Chuang
- the Departments of Biochemistry and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, and
| | - Susan M. Hutson
- From the Department of Human Nutrition, Foods and Exercise, Virginia Polytechnic Institute, Blacksburg, Virginia 24061
| |
Collapse
|
42
|
Recombinant production of eight human cytosolic aminotransferases and assessment of their potential involvement in glyoxylate metabolism. Biochem J 2009; 422:265-72. [PMID: 19545238 DOI: 10.1042/bj20090748] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PH1 (primary hyperoxaluria type 1) is a severe inborn disorder of glyoxylate metabolism caused by a functional deficiency of the peroxisomal enzyme AGXT (alanine-glyoxylate aminotransferase), which converts glyoxylate into glycine using L-alanine as the amino-group donor. Even though pre-genomic studies indicate that other human transaminases can convert glyoxylate into glycine, in PH1 patients these enzymes are apparently unable to compensate for the lack of AGXT, perhaps due to their limited levels of expression, their localization in an inappropriate cell compartment or the scarcity of the required amino-group donor. In the present paper, we describe the cloning of eight human cytosolic aminotransferases, their recombinant expression as His6-tagged proteins and a comparative study on their ability to transaminate glyoxylate, using any standard amino acid as an amino-group donor. To selectively quantify the glycine formed, we have developed and validated an assay based on bacterial GO (glycine oxidase); this assay allows the detection of enzymes that produce glycine by transamination in the presence of mixtures of potential amino-group donors and without separation of the product from the substrates. We show that among the eight enzymes tested, only GPT (alanine transaminase) and PSAT1 (phosphoserine aminotransferase 1) can transaminate glyoxylate with good efficiency, using L-glutamate (and, for GPT, also L-alanine) as the best amino-group donor. These findings confirm that glyoxylate transamination can occur in the cytosol, in direct competition with the conversion of glyoxylate into oxalate. The potential implications for the treatment of primary hyperoxaluria are discussed.
Collapse
|
43
|
Morris SM. Recent advances in arginine metabolism: roles and regulation of the arginases. Br J Pharmacol 2009; 157:922-30. [PMID: 19508396 DOI: 10.1111/j.1476-5381.2009.00278.x] [Citation(s) in RCA: 344] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
As arginine can serve as precursor to a wide range of compounds, including nitric oxide, creatine, urea, polyamines, proline, glutamate and agmatine, there is considerable interest in elucidating mechanisms underlying regulation of its metabolism. It is now becoming apparent that the two isoforms of arginase in mammals play key roles in regulation of most aspects of arginine metabolism in health and disease. In particular, work over the past several years has focused on the roles and regulation of the arginases in vascular disease, pulmonary disease, infectious disease, immune cell function and cancer. As most of these topics have been considered in recent review articles, this review will focus more closely on results of recent studies on expression of the arginases in endothelial and vascular smooth muscle cells, post-translational modulation of arginase activity and applications of arginase inhibitors in vivo.
Collapse
Affiliation(s)
- Sidney M Morris
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
44
|
Hutson SM, Poole LB, Coles S, Conway ME. Redox regulation and trapping sulfenic acid in the peroxide-sensitive human mitochondrial branched chain aminotransferase. Methods Mol Biol 2009; 476:139-52. [PMID: 19157014 DOI: 10.1007/978-1-59745-129-1_10] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
Abstract
The human branched chain aminotransferase enzymes are key regulators of glutamate metabolism in the brain and are among a growing number of redox-sensitive proteins. Studies that use thiol-specific reagents and electrospray ionization mass spectrometry demonstrate that the mitochondrial BCAT enzyme has a redox-active CXXC center, which on oxidation forms a disulfide bond (RSSR), via a cysteine sulfenic acid intermediate. Mechanistic details of this redox regulation were revealed by the use of mass spectrometry and dimedone modification. We discovered that the thiol group at position C315 of the CXXC motif acts a redox sensor, whereas the thiol group at position C318 permits reversible regulation by forming an intrasubunit disulphide bond. Because of their roles in redox regulation and catalysis, there is a growing interest in cysteine sulphenic acids. Therefore, development of chemical tags/methods to trap these transient intermediates is of immense importance.
Collapse
Affiliation(s)
- Susan M Hutson
- Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | | | | | | |
Collapse
|
45
|
Abstract
Methionine adenosyltransferases (MATs) are the family of enzymes that synthesize the main biological methyl donor, S-adenosylmethionine. The high sequence conservation among catalytic subunits from bacteria and eukarya preserves key residues that control activity and oligomerization, which is reflected in the protein structure. However, structural differences among complexes with substrates and products have led to proposals of several reaction mechanisms. In parallel, folding studies begin to explain how the three intertwined domains of the catalytic subunit are produced, and to highlight the importance of certain intermediates in attaining the active final conformation. This review analyzes the available structural data and proposes a consensus interpretation that facilitates an understanding of the pathological problems derived from impairment of MAT function. In addition, new research opportunities directed toward clarification of aspects that remain obscure are also identified.
Collapse
Affiliation(s)
- G. D. Markham
- Institute for Cancer Research, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111 USA
| | - M. A. Pajares
- Instituto de Investigaciones Biomédicas “Alberto Sols” (CSIC-UAM), Arturo Duperier 4, 28029 Madrid, Spain
| |
Collapse
|
46
|
Coles SJ, Easton P, Sharrod H, Hutson SM, Hancock J, Patel VB, Conway ME. S-Nitrosoglutathione Inactivation of the Mitochondrial and Cytosolic BCAT Proteins: S-Nitrosation and S-Thiolation. Biochemistry 2009; 48:645-56. [PMID: 19119849 DOI: 10.1021/bi801805h] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Steven J. Coles
- Faculty of Health and Life Sciences, University of the West of England, Coldharbor Lane, Bristol BS16 1QY, U.K., Department of Biochemistry, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157, and Department of Biomedical Sciences, School of Biosciences, University of Westminster, London W1W 6UW, U.K
| | - Peter Easton
- Faculty of Health and Life Sciences, University of the West of England, Coldharbor Lane, Bristol BS16 1QY, U.K., Department of Biochemistry, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157, and Department of Biomedical Sciences, School of Biosciences, University of Westminster, London W1W 6UW, U.K
| | - Hayley Sharrod
- Faculty of Health and Life Sciences, University of the West of England, Coldharbor Lane, Bristol BS16 1QY, U.K., Department of Biochemistry, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157, and Department of Biomedical Sciences, School of Biosciences, University of Westminster, London W1W 6UW, U.K
| | - Susan M. Hutson
- Faculty of Health and Life Sciences, University of the West of England, Coldharbor Lane, Bristol BS16 1QY, U.K., Department of Biochemistry, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157, and Department of Biomedical Sciences, School of Biosciences, University of Westminster, London W1W 6UW, U.K
| | - John Hancock
- Faculty of Health and Life Sciences, University of the West of England, Coldharbor Lane, Bristol BS16 1QY, U.K., Department of Biochemistry, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157, and Department of Biomedical Sciences, School of Biosciences, University of Westminster, London W1W 6UW, U.K
| | - Vinood B. Patel
- Faculty of Health and Life Sciences, University of the West of England, Coldharbor Lane, Bristol BS16 1QY, U.K., Department of Biochemistry, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157, and Department of Biomedical Sciences, School of Biosciences, University of Westminster, London W1W 6UW, U.K
| | - Myra E. Conway
- Faculty of Health and Life Sciences, University of the West of England, Coldharbor Lane, Bristol BS16 1QY, U.K., Department of Biochemistry, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157, and Department of Biomedical Sciences, School of Biosciences, University of Westminster, London W1W 6UW, U.K
| |
Collapse
|
47
|
Mugford P, Wagner UG, Jiang Y, Faber K, Kazlauskas R. Enantiocomplementary Enzymes: Classification, Molecular Basis for Their Enantiopreference, and Prospects for Mirror-Image Biotransformations. Angew Chem Int Ed Engl 2008; 47:8782-93. [DOI: 10.1002/anie.200705159] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
48
|
Mugford P, Wagner U, Jiang Y, Faber K, Kazlauskas R. Enantiokomplementäre Enzyme: Klassifizierung, molekulare Grundlage der Enantiopräferenz und Prognosen für spiegelbildliche Biotransformationen. Angew Chem Int Ed Engl 2008. [DOI: 10.1002/ange.200705159] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
49
|
Conway ME, Coles SJ, Islam MM, Hutson SM. Regulatory Control of Human Cytosolic Branched-Chain Aminotransferase by Oxidation and S-Glutathionylation and Its Interactions with Redox Sensitive Neuronal Proteins. Biochemistry 2008; 47:5465-79. [DOI: 10.1021/bi800303h] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Myra E. Conway
- Faculty of Health and Life Sciences, University of the West of England, Coldharbor Lane, Bristol, BS16 1QY, U.K., and Department of Biochemistry, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| | - Steven J. Coles
- Faculty of Health and Life Sciences, University of the West of England, Coldharbor Lane, Bristol, BS16 1QY, U.K., and Department of Biochemistry, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| | - Mohammad M. Islam
- Faculty of Health and Life Sciences, University of the West of England, Coldharbor Lane, Bristol, BS16 1QY, U.K., and Department of Biochemistry, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| | - Susan M. Hutson
- Faculty of Health and Life Sciences, University of the West of England, Coldharbor Lane, Bristol, BS16 1QY, U.K., and Department of Biochemistry, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| |
Collapse
|
50
|
Islam MM, Wallin R, Wynn RM, Conway M, Fujii H, Mobley JA, Chuang DT, Hutson SM. A novel branched-chain amino acid metabolon. Protein-protein interactions in a supramolecular complex. J Biol Chem 2007; 282:11893-903. [PMID: 17314104 DOI: 10.1074/jbc.m700198200] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The catabolic pathways of branched-chain amino acids have two common steps. The first step is deamination catalyzed by the vitamin B(6)-dependent branched-chain aminotransferase isozymes (BCATs) to produce branched-chain alpha-keto acids (BCKAs). The second step is oxidative decarboxylation of the BCKAs mediated by the branched-chain alpha-keto acid dehydrogenase enzyme complex (BCKD complex). The BCKD complex is organized around a cubic core consisting of 24 lipoate-bearing dihydrolipoyl transacylase (E2) subunits, associated with the branched-chain alpha-keto acid decarboxylase/dehydrogenase (E1), dihydrolipoamide dehydrogenase (E3), BCKD kinase, and BCKD phosphatase. In this study, we provide evidence that human mitochondrial BCAT (hBCATm) associates with the E1 decarboxylase component of the rat or human BCKD complex with a K(D) of 2.8 microM. NADH dissociates the complex. The E2 and E3 components do not interact with hBCATm. In the presence of hBCATm, k(cat) values for E1-catalyzed decarboxylation of the BCKAs are enhanced 12-fold. Mutations of hBCATm proteins in the catalytically important CXXC center or E1 proteins in the phosphorylation loop residues prevent complex formation, indicating that these regions are important for the interaction between hBCATm and E1. Our results provide evidence for substrate channeling between hBCATm and BCKD complex and formation of a metabolic unit (termed branched-chain amino acid metabolon) that can be influenced by the redox state in mitochondria.
Collapse
Affiliation(s)
- Mohammad Mainul Islam
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | | | | | | | | | | | | | | |
Collapse
|