1
|
Hantikainen E, Weichenberger CX, Dordevic N, Verri Hernandes V, Foco L, Gögele M, Melotti R, Pattaro C, Ralser M, Amari F, Farztdinov V, Mülleder M, Pramstaller PP, Rainer J, Domingues FS. Metabolite and protein associations with general health in the population-based CHRIS study. Sci Rep 2024; 14:26635. [PMID: 39496618 PMCID: PMC11535378 DOI: 10.1038/s41598-024-75627-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/07/2024] [Indexed: 11/06/2024] Open
Abstract
Identifying biomarkers able to discriminate individuals on different health trajectories is crucial to understand the molecular basis of age-related morbidity. We investigated multi-omics signatures of general health and organ-specific morbidity, as well as their interconnectivity. We examined cross-sectional metabolome and proteome data from 3,142 adults of the Cooperative Health Research in South Tyrol (CHRIS) study, an Alpine population study designed to investigate how human biology, environment, and lifestyle factors contribute to people's health over time. We had 174 metabolites and 148 proteins quantified from fasting serum and plasma samples. We used the Cumulative Illness Rating Scale (CIRS) Comorbidity Index (CMI), which considers morbidity in 14 organ systems, to assess health status (any morbidity vs. healthy). Omics-signatures for health status were identified using random forest (RF) classifiers. Linear regression models were fitted to assess directionality of omics markers and health status associations, as well as to identify omics markers related to organ-specific morbidity. Next to age, we identified 21 metabolites and 10 proteins as relevant predictors of health status and results confirmed associations for serotonin and glutamate to be age-independent. Considering organ-specific morbidity, several metabolites and proteins were jointly related to endocrine, cardiovascular, and renal morbidity. To conclude, circulating serotonin was identified as a potential novel predictor for overall morbidity.
Collapse
Affiliation(s)
| | | | | | | | - Luisa Foco
- Institute for Biomedicine, Eurac Research, Bolzano, Italy
| | - Martin Gögele
- Institute for Biomedicine, Eurac Research, Bolzano, Italy
| | | | | | - Markus Ralser
- Department of Biochemistry, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Fatma Amari
- Department of Biochemistry, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Vadim Farztdinov
- Core Facility, High-Throughput Mass Spectrometry, Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Michael Mülleder
- Core Facility, High-Throughput Mass Spectrometry, Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | | | | | | |
Collapse
|
2
|
Cai T, Shang K, Wang X, Qi X, Liu R, Wang X. Integration of Glutamate Dehydrogenase and Nanoporous Gold for Electrochemical Detection of Glutamate. BIOSENSORS 2023; 13:1023. [PMID: 38131783 PMCID: PMC10741451 DOI: 10.3390/bios13121023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/23/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023]
Abstract
Glutamate, a non-essential amino acid produced by fermentation, plays a significant role in disease diagnosis and food safety. It is important to enable the real-time monitoring of glutamate concentration for human health and nutrition. Due to the challenges in directly performing electrochemical oxidation-reduction reactions of glutamate, this study leverages the synergistic effect of glutamate dehydrogenase (GLDH) and nanoporous gold (NPG) to achieve the indirect and accurate detection of glutamate within the range of 50 to 700 μM by measuring the generated quantity of NADH during the enzymatic reaction. The proposed biosensor demonstrates remarkable performance characteristics, including a detection sensitivity of 1.95 μA mM-1 and a limit of detection (LOD) of 6.82 μM. The anti-interference tests indicate an average recognition error ranging from -3.85% to +2.60%, spiked sample recovery rates between 95% and 105%, and a relative standard deviation (RSD) of less than 4.97% for three replicate experiments. Therefore, the GLDH-NPG/GCE biosensor presented in this work exhibits excellent accuracy and repeatability, providing a novel alternative for rapid glutamate detection. This research contributes significantly to enhancing the precise monitoring of glutamate concentration, thereby offering more effective guidance and control for human health and nutrition.
Collapse
Affiliation(s)
| | | | | | | | | | - Xia Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; (T.C.); (K.S.); (X.W.); (X.Q.); (R.L.)
| |
Collapse
|
3
|
Li Y, Chitturi J, Yu B, Zhang Y, Wu J, Ti P, Hung W, Zhen M, Gao S. UBR-1 ubiquitin ligase regulates the balance between GABAergic and glutamatergic signaling. EMBO Rep 2023; 24:e57014. [PMID: 37811674 PMCID: PMC10626437 DOI: 10.15252/embr.202357014] [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: 02/16/2023] [Revised: 09/16/2023] [Accepted: 09/21/2023] [Indexed: 10/10/2023] Open
Abstract
Excitation/inhibition (E/I) balance is carefully maintained by the nervous system. The neurotransmitter GABA has been reported to be co-released with its sole precursor, the neurotransmitter glutamate. The genetic and circuitry mechanisms to establish the balance between GABAergic and glutamatergic signaling have not been fully elucidated. Caenorhabditis elegans DVB is an excitatory GABAergic motoneuron that drives the expulsion step in the defecation motor program. We show here that in addition to UNC-47, the vesicular GABA transporter, DVB also expresses EAT-4, a vesicular glutamate transporter. UBR-1, a conserved ubiquitin ligase, regulates DVB activity by suppressing a bidirectional inhibitory glutamate signaling. Loss of UBR-1 impairs DVB Ca2+ activity and expulsion frequency. These impairments are fully compensated by the knockdown of EAT-4 in DVB. Further, glutamate-gated chloride channels GLC-3 and GLC-2/4 receive DVB's glutamate signals to inhibit DVB and enteric muscle activity, respectively. These results implicate an intrinsic cellular mechanism that promotes the inherent asymmetric neural activity. We propose that elevated glutamate in ubr-1 mutants, being the cause of the E/I shift, potentially contributes to Johanson Blizzard syndrome.
Collapse
Affiliation(s)
- Yi Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
| | - Jyothsna Chitturi
- Lunenfeld‐Tanenbaum Research Institute, Mount Sinai HospitalUniversity of TorontoTorontoONCanada
| | - Bin Yu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
| | - Yongning Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
| | - Jing Wu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
| | - Panpan Ti
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
| | - Wesley Hung
- Lunenfeld‐Tanenbaum Research Institute, Mount Sinai HospitalUniversity of TorontoTorontoONCanada
| | - Mei Zhen
- Lunenfeld‐Tanenbaum Research Institute, Mount Sinai HospitalUniversity of TorontoTorontoONCanada
| | - Shangbang Gao
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Vascular Aging of the Ministry of Education, Tongji Hospital of Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
4
|
Mirveis Z, Howe O, Cahill P, Patil N, Byrne HJ. Monitoring and modelling the glutamine metabolic pathway: a review and future perspectives. Metabolomics 2023; 19:67. [PMID: 37482587 PMCID: PMC10363518 DOI: 10.1007/s11306-023-02031-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 07/03/2023] [Indexed: 07/25/2023]
Abstract
BACKGROUND Analysis of the glutamine metabolic pathway has taken a special place in metabolomics research in recent years, given its important role in cell biosynthesis and bioenergetics across several disorders, especially in cancer cell survival. The science of metabolomics addresses the intricate intracellular metabolic network by exploring and understanding how cells function and respond to external or internal perturbations to identify potential therapeutic targets. However, despite recent advances in metabolomics, monitoring the kinetics of a metabolic pathway in a living cell in situ, real-time and holistically remains a significant challenge. AIM This review paper explores the range of analytical approaches for monitoring metabolic pathways, as well as physicochemical modeling techniques, with a focus on glutamine metabolism. We discuss the advantages and disadvantages of each method and explore the potential of label-free Raman microspectroscopy, in conjunction with kinetic modeling, to enable real-time and in situ monitoring of the cellular kinetics of the glutamine metabolic pathway. KEY SCIENTIFIC CONCEPTS Given its important role in cell metabolism, the ability to monitor and model the glutamine metabolic pathways are highlighted. Novel, label free approaches have the potential to revolutionise metabolic biosensing, laying the foundation for a new paradigm in metabolomics research and addressing the challenges in monitoring metabolic pathways in living cells.
Collapse
Affiliation(s)
- Zohreh Mirveis
- FOCAS Research Institute, Technological University Dublin, City Campus, Camden Row, Dublin 8, Ireland.
- School of Physics and Optometric & Clinical Sciences, Technological University Dublin, City Campus, Grangegorman, Dublin 7, Ireland.
| | - Orla Howe
- School of Biological, Health and Sport Sciences, Technological University Dublin, City Campus, Grangegorman, Dublin 7, Ireland
| | - Paul Cahill
- School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Nitin Patil
- FOCAS Research Institute, Technological University Dublin, City Campus, Camden Row, Dublin 8, Ireland
- School of Physics and Optometric & Clinical Sciences, Technological University Dublin, City Campus, Grangegorman, Dublin 7, Ireland
| | - Hugh J Byrne
- FOCAS Research Institute, Technological University Dublin, City Campus, Camden Row, Dublin 8, Ireland
| |
Collapse
|
5
|
Sinclair GM, Di Giannantonio M, Jones OAH, Long SM. Is substrate choice an overlooked variable in ecotoxicology experiments? ENVIRONMENTAL MONITORING AND ASSESSMENT 2023; 195:344. [PMID: 36715783 PMCID: PMC9886613 DOI: 10.1007/s10661-023-10935-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 01/10/2023] [Indexed: 06/18/2023]
Abstract
It is crucial to understand the effects caused by experimental parameters such as temperature, light, and food type on lab and field-based ecotoxicology experiments, as these variables, and combinations thereof, can affect results. The type of substrate used in exposure experiments, however, is generally assumed to have no effect. This may not always be correct. The metabolic changes in the freshwater crustacean, Austrochiltonia subtenuis exposed to copper, using three common substrates, gauze; toilet paper; and cellulose were investigated. Substrate alone did not affect survival, but each substrate elicited a different metabolic response and adult and juvenile amphipods had different substrate preferences. Several classes of metabolites were shown to change in response to different substrates and toxicant. These included disaccharides, monosaccharides, fatty acids, and tricarboxylic acid cycle intermediates. The results illustrate that metabolomic responses can differ in response to experimental factors that were previously thought not to be significant. In fact, our data indicate that substrate should be viewed as an experimental factor as important to control for as more well-known confounders such as temperature or food, thus challenging the current paradigm. Assuming substrate type has no effect on the experiment could potentially lead to errors in contaminant toxicity assessments. We propose that ideal good practise would be that all experimental factors should be evaluated for their potential influence on metabolomic profiles prior to contaminant response experiments being undertaken.
Collapse
Affiliation(s)
- Georgia M Sinclair
- Australian Centre for Research on Separation Science (ACROSS), School of Science, RMIT University, PO Box 71, Bundoora West Campus, Bundoora, VIC, 3083, Australia.
| | - Michela Di Giannantonio
- National Research Council (CNR-IAS), Institute for the study of Anthropic Impacts and Sustainability in Marine Environment, Genoa, Italy
- Aquatic Environmental Stress (AQUEST) Research Group School of Science, RMIT University, Bundoora, VIC, 3083, Australia
| | - Oliver A H Jones
- Australian Centre for Research on Separation Science (ACROSS), School of Science, RMIT University, PO Box 71, Bundoora West Campus, Bundoora, VIC, 3083, Australia
| | - Sara M Long
- Aquatic Environmental Stress (AQUEST) Research Group School of Science, RMIT University, Bundoora, VIC, 3083, Australia
| |
Collapse
|
6
|
Daniyan MO, Fisusi FA, Adeoye OB. Neurotransmitters and molecular chaperones interactions in cerebral malaria: Is there a missing link? Front Mol Biosci 2022; 9:965569. [PMID: 36090033 PMCID: PMC9451049 DOI: 10.3389/fmolb.2022.965569] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 07/28/2022] [Indexed: 12/02/2022] Open
Abstract
Plasmodium falciparum is responsible for the most severe and deadliest human malaria infection. The most serious complication of this infection is cerebral malaria. Among the proposed hypotheses that seek to explain the manifestation of the neurological syndrome in cerebral malaria is the vascular occlusion/sequestration/mechanic hypothesis, the cytokine storm or inflammatory theory, or a combination of both. Unfortunately, despite the increasing volume of scientific information on cerebral malaria, our understanding of its pathophysiologic mechanism(s) is still very limited. In a bid to maintain its survival and development, P. falciparum exports a large number of proteins into the cytosol of the infected host red blood cell. Prominent among these are the P. falciparum erythrocytes membrane protein 1 (PfEMP1), P. falciparum histidine-rich protein II (PfHRP2), and P. falciparum heat shock proteins 70-x (PfHsp70-x). Functional activities and interaction of these proteins with one another and with recruited host resident proteins are critical factors in the pathology of malaria in general and cerebral malaria in particular. Furthermore, several neurological impairments, including cognitive, behavioral, and motor dysfunctions, are known to be associated with cerebral malaria. Also, the available evidence has implicated glutamate and glutamatergic pathways, coupled with a resultant alteration in serotonin, dopamine, norepinephrine, and histamine production. While seeking to improve our understanding of the pathophysiology of cerebral malaria, this article seeks to explore the possible links between host/parasite chaperones, and neurotransmitters, in relation to other molecular players in the pathology of cerebral malaria, to explore such links in antimalarial drug discovery.
Collapse
Affiliation(s)
- Michael Oluwatoyin Daniyan
- Department of Pharmacology, Faculty of Pharmacy, Obafemi Awolowo University, Ile-Ife, Osun State, Nigeria
| | - Funmilola Adesodun Fisusi
- Drug Research and Production Unit, Faculty of Pharmacy, Obafemi Awolowo University, Ile-Ife, Osun State, Nigeria
| | - Olufunso Bayo Adeoye
- Department of Biochemistry, Benjamin S. Carson (Snr.) College of Medicine, Babcock University, Ilishan-Remo, Ogun State, Nigeria
| |
Collapse
|
7
|
Germain A, Giloteaux L, Moore GE, Levine SM, Chia JK, Keller BA, Stevens J, Franconi CJ, Mao X, Shungu DC, Grimson A, Hanson MR. Plasma metabolomics reveals disrupted response and recovery following maximal exercise in myalgic encephalomyelitis/chronic fatigue syndrome. JCI Insight 2022; 7:e157621. [PMID: 35358096 PMCID: PMC9090259 DOI: 10.1172/jci.insight.157621] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/23/2022] [Indexed: 11/17/2022] Open
Abstract
Post-exertional malaise (PEM) is a hallmark symptom of myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). We monitored the evolution of 1157 plasma metabolites in 60 ME/CFS (45 female, 15 male) and 45 matched healthy control participants (30 female, 15 male) before and after 2 maximal cardiopulmonary exercise test (CPET) challenges separated by 24 hours, with the intent of provoking PEM in patients. Four time points allowed exploration of the metabolic response to maximal energy-producing capacity and the recovery pattern of participants with ME/CFS compared with the healthy control group. Baseline comparison identified several significantly different metabolites, along with an enriched percentage of yet-to-be identified compounds. Additionally, temporal measures demonstrated an increased metabolic disparity between cohorts, including unknown metabolites. The effects of exertion in the ME/CFS cohort predominantly highlighted lipid-related as well as energy-related pathways and chemical structure clusters, which were disparately affected by the first and second exercise sessions. The 24-hour recovery period was distinct in the ME/CFS cohort, with over a quarter of the identified pathways statistically different from the controls. The pathways that are uniquely different 24 hours after an exercise challenge provide clues to metabolic disruptions that lead to PEM. Numerous altered pathways were observed to depend on glutamate metabolism, a crucial component of the homeostasis of many organs in the body, including the brain.
Collapse
Affiliation(s)
- Arnaud Germain
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| | - Ludovic Giloteaux
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| | - Geoffrey E. Moore
- Department of Exercise Science and Athletic Training, Ithaca College, Ithaca, New York, USA
| | - Susan M. Levine
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| | | | - Betsy A. Keller
- Department of Exercise Science and Athletic Training, Ithaca College, Ithaca, New York, USA
| | | | - Carl J. Franconi
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| | - Xiangling Mao
- Department of Neuroradiology, Weill Cornell Medical College, New York, New York, USA
| | - Dikoma C. Shungu
- Department of Neuroradiology, Weill Cornell Medical College, New York, New York, USA
| | - Andrew Grimson
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| | - Maureen R. Hanson
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| |
Collapse
|
8
|
Afzaal A, Rehman K, Kamal S, Akash MSH. Versatile role of sirtuins in metabolic disorders: From modulation of mitochondrial function to therapeutic interventions. J Biochem Mol Toxicol 2022; 36:e23047. [PMID: 35297126 DOI: 10.1002/jbt.23047] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 01/11/2022] [Accepted: 03/02/2022] [Indexed: 12/17/2022]
Abstract
Sirtuins (SIRT1-7) are distinct histone deacetylases (HDACs) whose activity is determined by cellular metabolic status andnicotinamide adenine dinucleotide (NAD+ ) levels. HDACs of class III are the members of the SIRT's protein family. SIRTs are the enzymes that modulate mitochondrial activity and energy metabolism. SIRTs have been linked to a number of clinical and physiological operations, such as energy responses to low-calorie availability, aging, stress resistance, inflammation, and apoptosis. Mammalian SIRT2 orthologs have been identified as SIRT1-7 that are found in several subcellular sections, including the cytoplasm (SIRT1, 2), mitochondrial matrix (SIRT3, 4, 5), and the core (SIRT1, 2, 6, 7). For their deacetylase or ADP-ribosyl transferase action, all SIRTs require NAD+ and are linked to cellular energy levels. Evolutionarily, SIRT1 is related to yeast's SIRT2 as well as received primary attention in the circulatory system. An endogenous protein, SIRT1 is involved in the development of heart failure and plays a key role in cell death and survival. SIRT2 downregulation protects against ischemic-reperfusion damage. Increase in human longevity is caused by an increase in SIRT3 expression. Cardiomyocytes are also protected by SIRT3 from oxidative damage and aging, as well as suppressing cardiac hypertrophy. SIRT4 and SIRT5 perform their roles in the heart. SIRT6 has also been linked to a reduction in heart hypertrophy. SIRT7 is known to be involved in the regulation of stress responses and apoptosis in the heart.
Collapse
Affiliation(s)
- Ammara Afzaal
- Department of Pharmaceutical Chemistry, Government College University, Faisalabad, Pakistan
| | - Kanwal Rehman
- Department of Pharmacy, University of Agriculture, Faisalabad, Pakistan
| | - Shagufta Kamal
- Department of Biochemistry, Government College University, Faisalabad, Pakistan
| | | |
Collapse
|
9
|
The Interplay between Pathophysiological Pathways in Early-Onset Severe Preeclampsia Unveiled by Metabolomics. Life (Basel) 2022; 12:life12010086. [PMID: 35054479 PMCID: PMC8780941 DOI: 10.3390/life12010086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/02/2022] [Accepted: 01/04/2022] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Preeclampsia is a multi-system disorder unique to pregnancy responsible for a great part of maternal and perinatal morbidity and mortality. The precise pathogenesis of this complex disorder is still unrevealed. METHODS We examined the pathophysiological pathways involved in early-onset preeclampsia, a specific subgroup representing its most severe presentation, using LC-MS/MS metabolomic analysis based on multi-level extraction of lipids and small metabolites from maternal blood samples, collected at the time of diagnosis from 14 preeclamptic and six matched healthy pregnancies. Statistical analysis comprised multivariate and univariate approaches with the application of over representation analysis to identify differential pathways. RESULTS A clear difference between preeclamptic and control pregnancies was observed in principal component analysis. Supervised multivariate analysis using orthogonal partial least square discriminant analysis provided a robust model with goodness of fit (R2X = 0.91, p = 0.002) and predictive ability (Q2Y = 0.72, p < 0.001). Finally, univariate analysis followed by 5% false discovery rate correction indicated 82 metabolites significantly altered, corresponding to six overrepresented pathways: (1) aminoacyl-tRNA biosynthesis; (2) arginine biosynthesis; (3) alanine, aspartate and glutamate metabolism; (4) D-glutamine and D-glutamate metabolism; (5) arginine and proline metabolism; and (6) histidine metabolism. CONCLUSION Metabolomic analysis focusing specifically on the early-onset severe form of preeclampsia reveals the interplay between pathophysiological pathways involved in this form. Future studies are required to explore new therapeutic approaches targeting these altered metabolic pathways in early-onset preeclampsia.
Collapse
|
10
|
Kretzschmar T, Wu JMF, Schulze PC. Mitochondrial Homeostasis Mediates Lipotoxicity in the Failing Myocardium. Int J Mol Sci 2021; 22:1498. [PMID: 33540894 PMCID: PMC7867320 DOI: 10.3390/ijms22031498] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 01/17/2023] Open
Abstract
Heart failure remains the most common cause of death in the industrialized world. In spite of new therapeutic interventions that are constantly being developed, it is still not possible to completely protect against heart failure development and progression. This shows how much more research is necessary to understand the underlying mechanisms of this process. In this review, we give a detailed overview of the contribution of impaired mitochondrial dynamics and energy homeostasis during heart failure progression. In particular, we focus on the regulation of fatty acid metabolism and the effects of fatty acid accumulation on mitochondrial structural and functional homeostasis.
Collapse
Affiliation(s)
| | | | - P. Christian Schulze
- Department of Internal Medicine I, University Hospital Jena, 07747 Jena, Thüringen, Germany; (T.K.); (J.M.F.W.)
| |
Collapse
|
11
|
In Silico Prediction of the Mode of Action of Viola odorata in Diabetes. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2768403. [PMID: 33490239 PMCID: PMC7803256 DOI: 10.1155/2020/2768403] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/27/2020] [Accepted: 10/05/2020] [Indexed: 12/17/2022]
Abstract
Background The metabolic syndrome increases the risk of different diseases such as type 2 diabetes. The prevalence of metabolic syndrome has rapidly grown and affected more than 230 million people worldwide. Viola odorata is a traditionally used plant for the treatment of diabetes; however, its mechanism to manage diabetes is still unknown. Purpose This study was designed to systematically assess the mechanism of action of Viola odorata in diabetes. Methods An extensive literature search was made to establish an ingredient-target database of Viola odorata. Of these, targets related to diabetes were identified and used to develop a protein-protein interaction network (PPIN) by utilizing the STITCH database. The obtained PPIN was assessed through Gene Ontology (GO) enrichment analysis based on ClueGO plugin. Results According to the acquired data, there were about 143 chemical constituents present in Viola odorata having 119 protein targets. Of these, 31 targets were established to give the pharmacological effect against diabetes. The UniProt database was used for screening of 31 targets, out of which Homo sapiens contained 22 targets. Ultimately, 207 GO terms, grouped into 41 clusters, were found by gene analysis, and most of them were found to be linked with diabetes. According to findings, several proteins including TP53, BCL2, CDKN1A, 1L6, CCND1, CDKN2A, and RB1 have a significant role in the treatment of diabetes by Viola odorata. Conclusion The possible activity of Viola odorata in the management of diabetes may be mediated by several molecular mechanisms, including the glutamine metabolic process, IRE1-mediated unfolded protein response, and pentose metabolic process.
Collapse
|
12
|
da Silva I, da Costa Vieira R, Stella C, Loturco E, Carvalho AL, Veo C, Neto C, Silva SM, D'Amora P, Salzgeber M, Matos D, Silva CR, Oliveira JR, Rabelo I, Yamakawa P, Maciel R, Biscolla R, Chiamolera M, Fraietta R, Reis F, Mori M, Marchioni D, Carioca A, Maciel G, Tomioka R, Baracat E, Silva C, Granato C, Diaz R, Scarpellini B, Egle D, Fiegl H, Himmel I, Troi C, Nagourney R. Inborn-like errors of metabolism are determinants of breast cancer risk, clinical response and survival: a study of human biochemical individuality. Oncotarget 2018; 9:31664-31681. [PMID: 30167086 PMCID: PMC6114970 DOI: 10.18632/oncotarget.25839] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/12/2018] [Indexed: 01/16/2023] Open
Abstract
Breast cancer remains a leading cause of morbidity and mortality worldwide yet methods for early detection remain elusive. We describe the discovery and validation of biochemical signatures measured by mass spectrometry, performed upon blood samples from patients and controls that accurately identify (>95%) the presence of clinical breast cancer. Targeted quantitative MS/MS conducted upon 1225 individuals, including patients with breast and other cancers, normal controls as well as individuals with a variety of metabolic disorders provide a biochemical phenotype that accurately identifies the presence of breast cancer and predicts response and survival following the administration of neoadjuvant chemotherapy. The metabolic changes identified are consistent with inborn-like errors of metabolism and define a continuum from normal controls to elevated risk to invasive breast cancer. Similar results were observed in other adenocarcinomas but were not found in squamous cell cancers or hematologic neoplasms. The findings describe a new early detection platform for breast cancer and support a role for pre-existing, inborn-like errors of metabolism in the process of breast carcinogenesis that may also extend to other glandular malignancies. Statement of Significance: Findings provide a powerful tool for early detection and the assessment of prognosis in breast cancer and define a novel concept of breast carcinogenesis that characterizes malignant transformation as the clinical manifestation of underlying metabolic insufficiencies.
Collapse
Affiliation(s)
- Ismael da Silva
- Gynecology Department, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil.,Fleury Laboratories, São Paulo, Brazil.,Barretos Cancer Hospital (HCB), Barretos, Brazil
| | | | - Carolina Stella
- Gynecology Department, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Edson Loturco
- Department of Surgery, Urology Unit, Human Reproduction Division, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | | | - Carlos Veo
- Barretos Cancer Hospital (HCB), Barretos, Brazil
| | | | | | - Paulo D'Amora
- Gynecology Department, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Marcia Salzgeber
- Gynecology Department, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Delcio Matos
- Department of Surgery, Surgical Gastroenterology Division, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Celso R Silva
- Clinical and Experimental Oncology Department, Hematology and Hemotherapy Division, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Jose R Oliveira
- Clinical and Experimental Oncology Department, Hematology and Hemotherapy Division, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Iara Rabelo
- Clinical and Experimental Oncology Department, Hematology and Hemotherapy Division, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Patricia Yamakawa
- Clinical and Experimental Oncology Department, Hematology and Hemotherapy Division, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Rui Maciel
- Fleury Laboratories, São Paulo, Brazil.,Department of Medicine, Endocrinology Division, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Rosa Biscolla
- Department of Medicine, Endocrinology Division, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Maria Chiamolera
- Department of Medicine, Endocrinology Division, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Renato Fraietta
- Department of Surgery, Urology Unit, Human Reproduction Division, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Felipe Reis
- Biophysics Department, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Marcelo Mori
- Department of Biochemistry and Tissue Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Dirce Marchioni
- Nutrition Department, School of Public Health, University of São Paulo School of Medicine (FMUSP), São Paulo, Brazil
| | - Antonio Carioca
- Nutrition Department, School of Public Health, University of São Paulo School of Medicine (FMUSP), São Paulo, Brazil
| | - Gustavo Maciel
- Fleury Laboratories, São Paulo, Brazil.,Department of Obstetrics and Gynecology, University of São Paulo School of Medicine (HCFMUSP), São Paulo, Brazil
| | - Renato Tomioka
- Department of Obstetrics and Gynecology, University of São Paulo School of Medicine (HCFMUSP), São Paulo, Brazil
| | - Edmund Baracat
- Department of Obstetrics and Gynecology, University of São Paulo School of Medicine (HCFMUSP), São Paulo, Brazil
| | - Clovis Silva
- Department of Pediatrics, Children's Hospital, University of São Paulo School of Medicine (HCFMUSP), São Paulo, Brazil
| | - Celso Granato
- Fleury Laboratories, São Paulo, Brazil.,Retrovirology Laboratory, Infectious Diseases Unit, Medicine Department, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Ricardo Diaz
- Retrovirology Laboratory, Infectious Diseases Unit, Medicine Department, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Bruno Scarpellini
- Fleury Laboratories, São Paulo, Brazil.,Retrovirology Laboratory, Infectious Diseases Unit, Medicine Department, College of Medicine of the Federal University of São Paulo (EPM-UNIFESP), São Paulo, Brazil
| | - Daniel Egle
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, Innsbruck, Austria
| | - Heidi Fiegl
- Department of Gynecology, Meran Hospital, Meran, Italy
| | | | - Christina Troi
- Department of Gynecology, Brixen Hospital, Brixen, Italy
| | - Robert Nagourney
- Department of Obstetrics and Gynecology, Gynecological Oncology Unit, University of California Irvine (UCI), California, USA
| |
Collapse
|
13
|
Mathioudakis L, Bourbouli M, Daklada E, Kargatzi S, Michaelidou K, Zaganas I. Localization of Human Glutamate Dehydrogenases Provides Insights into Their Metabolic Role and Their Involvement in Disease Processes. Neurochem Res 2018; 44:170-187. [PMID: 29943084 DOI: 10.1007/s11064-018-2575-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 06/11/2018] [Accepted: 06/13/2018] [Indexed: 12/21/2022]
Abstract
Glutamate dehydrogenase (GDH) catalyzes the reversible deamination of L-glutamate to α-ketoglutarate and ammonia. In mammals, GDH contributes to important processes such as amino acid and carbohydrate metabolism, energy production, ammonia management, neurotransmitter recycling and insulin secretion. In humans, two isoforms of GDH are found, namely hGDH1 and hGDH2, with the former being ubiquitously expressed and the latter found mainly in brain, testis and kidney. These two iso-enzymes display highly divergent allosteric properties, especially concerning their basal activity, ADP activation and GTP inhibition. On the other hand, both enzymes are thought to predominantly localize in the mitochondrial matrix, even though alternative localizations have been proposed. To further study the subcellular localization of the two human iso-enzymes, we created HEK293 cell lines stably over-expressing hGDH1 and hGDH2. In these cell lines, immunofluorescence and enzymatic analyses verified the overexpression of both hGDH1 and hGDH2 iso-enzymes, whereas subcellular fractionation followed by immunoblotting showed their predominantly mitochondrial localization. Given that previous studies have only indirectly compared the subcellular localization of the two iso-enzymes, we co-expressed them tagged with different fluorescent dyes (green and red fluorescent protein for hGDH1 and hGDH2, respectively) and found them to co-localize. Despite the wealth of information related to the functional properties of hGDH1 and hGDH2 and the availability of the hGDH1 structure, there is still an ongoing debate concerning their metabolic role and their involvement in disease processes. Data on the localization of hGDHs, as the ones presented here, could contribute to better understanding of the function of these important human enzymes.
Collapse
Affiliation(s)
- Lambros Mathioudakis
- Neurology Laboratory, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Mara Bourbouli
- Neurology Laboratory, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Elisavet Daklada
- Neurology Laboratory, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Sofia Kargatzi
- Neurology Laboratory, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Kleita Michaelidou
- Neurology Laboratory, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Ioannis Zaganas
- Neurology Laboratory, Medical School, University of Crete, Heraklion, Crete, Greece. .,Department of Neurology, University Hospital of Heraklion, Heraklion, Crete, Greece.
| |
Collapse
|
14
|
Dong X, Zhou Z, Wang L, Saremi B, Helmbrecht A, Wang Z, Loor J. Increasing the availability of threonine, isoleucine, valine, and leucine relative to lysine while maintaining an ideal ratio of lysine:methionine alters mammary cellular metabolites, mammalian target of rapamycin signaling, and gene transcription. J Dairy Sci 2018; 101:5502-5514. [DOI: 10.3168/jds.2017-13707] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 02/05/2018] [Indexed: 12/21/2022]
|
15
|
Sarma SN, Saleem A, Lee JY, Tokumoto M, Hwang GW, Man Chan H, Satoh M. Effects of long-term cadmium exposure on urinary metabolite profiles in mice. J Toxicol Sci 2018; 43:89-100. [PMID: 29479038 DOI: 10.2131/jts.43.89] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Cadmium (Cd) is a common environmental pollutant with known toxic effects on the kidney. Urinary metabolomics is a promising approach to study mechanism by which Cd-induced nephrotoxicity. The aim of this study was to elucidate the mechanism of Cd toxicity and to develop specific biomarkers by identifying urinary metabolic changes after a long-term of Cd exposure and with the critical concentration of Cd in the kidney. Urine samples were collected from wild-type 129/Sv mice after 67 weeks of 300 ppm Cd exposure and analyzed by ultra performance liquid chromatography connected with quadrupole time of flight mass spectrometer (UPLC-QTOF-MS) based metabolomics approach. A total of 40 most differentiated metabolites (9 down-regulated and 31 up-regulated) between the control and Cd-exposed group were identified. The majority of the regulated metabolites are amino acids (glutamine, L-aspartic acid, phenylalanine, tryptophan, and D-proline) indicating that amino acid metabolism pathways are affected by long-term exposure of Cd. However, there are also some nucleotides (guanosine, guanosine monophosphate, cyclic AMP, uridine), amino acid derivatives (homoserine, N-acetyl-L-aspartate, N-acetylglutamine, acetyl-phenylalanine, carboxymethyllysine), and peptides. Results of pathway analysis showed that the arginine and proline metabolism, purine metabolism, alanine, aspartate and glutamate metabolism, and aminoacyl-tRNA biosynthesis were affected compared to the control. This study demonstrates that metabolomics is useful to elucidate the metabolic responses and biological effects induced by Cd-exposure.
Collapse
Affiliation(s)
| | - Ammar Saleem
- Department of Biology, University of Ottawa, Canada
| | - Jin-Yong Lee
- Laboratory of Pharmaceutical Health Sciences, School of Pharmacy, Aichi Gakuin University
| | - Maki Tokumoto
- Laboratory of Pharmaceutical Health Sciences, School of Pharmacy, Aichi Gakuin University
| | - Gi-Wook Hwang
- Laboratory of Molecular Biochemical Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University
| | | | - Masahiko Satoh
- Laboratory of Pharmaceutical Health Sciences, School of Pharmacy, Aichi Gakuin University
| |
Collapse
|
16
|
The UBR-1 ubiquitin ligase regulates glutamate metabolism to generate coordinated motor pattern in Caenorhabditis elegans. PLoS Genet 2018; 14:e1007303. [PMID: 29649217 PMCID: PMC5931689 DOI: 10.1371/journal.pgen.1007303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 05/02/2018] [Accepted: 03/08/2018] [Indexed: 12/14/2022] Open
Abstract
UBR1 is an E3 ubiquitin ligase best known for its ability to target protein degradation by the N-end rule. The physiological functions of UBR family proteins, however, remain not fully understood. We found that the functional loss of C. elegans UBR-1 leads to a specific motor deficit: when adult animals generate reversal movements, A-class motor neurons exhibit synchronized activation, preventing body bending. This motor deficit is rescued by removing GOT-1, a transaminase that converts aspartate to glutamate. Both UBR-1 and GOT-1 are expressed and critically required in premotor interneurons of the reversal motor circuit to regulate the motor pattern. ubr-1 and got-1 mutants exhibit elevated and decreased glutamate level, respectively. These results raise an intriguing possibility that UBR proteins regulate glutamate metabolism, which is critical for neuronal development and signaling. Ubiquitin-mediated protein degradation is central to diverse biological processes. The selection of substrates for degradation is carried out by the E3 ubiquitin ligases, which target specific groups of proteins for ubiquitination. The human genome encodes hundreds of E3 ligases; many exhibit sequence conservation across animal species, including one such ligase called UBR1. Patients carrying mutations in UBR1 exhibit severe systemic defects, but the biology behinds UBR1’s physiological function remains elusive. Here we found that the C. elegans UBR-1 regulates glutamate level. When UBR-1 is defective, C. elegans exhibits increased glutamate; this leads to synchronization of motor neuron activity, hence defective locomotion when animals reach adulthood. UBR1-mediated glutamate metabolism may contribute to the physiological defects of UBR1 mutations.
Collapse
|
17
|
Gar C, Rottenkolber M, Prehn C, Adamski J, Seissler J, Lechner A. Serum and plasma amino acids as markers of prediabetes, insulin resistance, and incident diabetes. Crit Rev Clin Lab Sci 2017; 55:21-32. [PMID: 29239245 DOI: 10.1080/10408363.2017.1414143] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Presently, routine screening misses many cases of prediabetes and early type 2 diabetes (T2D). Therefore, better biomarkers are needed for a simple and early detection of abnormalities of glucose metabolism and prediction of future T2D. Possible candidates for this include plasma or serum amino acids because glucose and amino acid metabolism are closely connected. This review presents the available evidence of this connectivity and discusses its clinical implications. First, we examine the underlying physiological, pre-analytical, and analytical issues. Then, we summarize results of human studies that evaluate amino acid levels as markers for insulin resistance, prediabetes, and future incident T2D. Finally, we illustrate the interconnection of amino acid levels and metabolic syndrome with our own data from a deeply phenotyped human cohort. We also discuss how amino acids may contribute to the pathophysiology of T2D. We conclude that elevated branched-chain amino acids and reduced glycine are currently the most robust and consistent amino acid markers for prediabetes, insulin resistance, and future T2D. Yet, we are cautious regarding the clinical potential even of these parameters because their discriminatory power is insufficient and their levels depend not only on glycemia, but also on other components of the metabolic syndrome. The identification of more precise intermediates of amino acid metabolism or combinations with other biomarkers will, therefore, be necessary to obtain in order to develop laboratory tests that can improve T2D screening.
Collapse
Affiliation(s)
- C Gar
- a Diabetes Research Group , Medizinische Klinik und Poliklinik IV, Klinikum der Universität München , Munich , Germany.,b Clinical Cooperation Group Type 2 Diabetes , Helmholtz Zentrum München , Neuherberg , Germany.,c Deutsches Zentrum für Diabetesforschung (DZD) , Neuherberg , Germany
| | - M Rottenkolber
- a Diabetes Research Group , Medizinische Klinik und Poliklinik IV, Klinikum der Universität München , Munich , Germany.,b Clinical Cooperation Group Type 2 Diabetes , Helmholtz Zentrum München , Neuherberg , Germany.,c Deutsches Zentrum für Diabetesforschung (DZD) , Neuherberg , Germany
| | - C Prehn
- d Institute of Experimental Genetics, Genome Analysis Center , Helmholtz Zentrum München, German Research Center for Environmental Health , Neuherberg , Germany
| | - J Adamski
- c Deutsches Zentrum für Diabetesforschung (DZD) , Neuherberg , Germany.,d Institute of Experimental Genetics, Genome Analysis Center , Helmholtz Zentrum München, German Research Center for Environmental Health , Neuherberg , Germany.,e Lehrstuhl fu¨r Experimentelle Genetik , Technische Universität München , Freising , Germany
| | - J Seissler
- a Diabetes Research Group , Medizinische Klinik und Poliklinik IV, Klinikum der Universität München , Munich , Germany.,b Clinical Cooperation Group Type 2 Diabetes , Helmholtz Zentrum München , Neuherberg , Germany.,c Deutsches Zentrum für Diabetesforschung (DZD) , Neuherberg , Germany
| | - A Lechner
- a Diabetes Research Group , Medizinische Klinik und Poliklinik IV, Klinikum der Universität München , Munich , Germany.,b Clinical Cooperation Group Type 2 Diabetes , Helmholtz Zentrum München , Neuherberg , Germany.,c Deutsches Zentrum für Diabetesforschung (DZD) , Neuherberg , Germany
| |
Collapse
|
18
|
Liao S, Ruiz Y, Gulzar H, Yelskaya Z, Ait Taouit L, Houssou M, Jaikaran T, Schvarts Y, Kozlitina K, Basu-Roy U, Mansukhani A, Mahajan SS. Osteosarcoma cell proliferation and survival requires mGluR5 receptor activity and is blocked by Riluzole. PLoS One 2017; 12:e0171256. [PMID: 28231291 PMCID: PMC5322947 DOI: 10.1371/journal.pone.0171256] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 01/17/2017] [Indexed: 11/18/2022] Open
Abstract
Osteosarcomas are malignant tumors of bone, most commonly seen in children and adolescents. Despite advances in modern medicine, the poor survival rate of metastatic osteosarcoma has not improved in two decades. In the present study we have investigated the effect of Riluzole on a human and mouse metastatic osteosarcoma cells. We show that LM7 cells secrete glutamate in the media and that mGluR5 receptors are required for the proliferation of LM7 cells. Riluzole, which is known to inhibit glutamate release, inhibits proliferation, induces apoptosis and prevents migration of LM7 cells. This is also seen with Fenobam, a specific blocker of mGluR5. We also show that Riluzole alters the phosphorylation status of AKT/P70 S6 kinase, ERK1/2 and JNK1/2. Thus Riluzole is an effective drug to inhibit proliferation and survival of osteosarcoma cells and has therapeutic potential for the treatment of osteosarcoma exhibiting autocrine glutamate signaling.
Collapse
Affiliation(s)
- Sally Liao
- Department of Medical Laboratory Sciences, Hunter College, City University of New York, New York, NY, United States of America
| | - Yuleisy Ruiz
- Department of Medical Laboratory Sciences, Hunter College, City University of New York, New York, NY, United States of America
| | - Hira Gulzar
- Department of Medical Laboratory Sciences, Hunter College, City University of New York, New York, NY, United States of America
| | - Zarina Yelskaya
- Department of Medical Laboratory Sciences, Hunter College, City University of New York, New York, NY, United States of America
| | - Lyes Ait Taouit
- Department of Medical Laboratory Sciences, Hunter College, City University of New York, New York, NY, United States of America
| | - Murielle Houssou
- Department of Medical Laboratory Sciences, Hunter College, City University of New York, New York, NY, United States of America
| | - Trisha Jaikaran
- Department of Medical Laboratory Sciences, Hunter College, City University of New York, New York, NY, United States of America
| | - Yuriy Schvarts
- Department of Medical Laboratory Sciences, Hunter College, City University of New York, New York, NY, United States of America
| | - Kristina Kozlitina
- Department of Medical Laboratory Sciences, Hunter College, City University of New York, New York, NY, United States of America
| | - Upal Basu-Roy
- Department of Microbiology & Perlmutter Cancer Center, NYU Langone Medical Center, New York, NY, United States of America
| | - Alka Mansukhani
- Department of Microbiology & Perlmutter Cancer Center, NYU Langone Medical Center, New York, NY, United States of America
| | - Shahana S. Mahajan
- Department of Medical Laboratory Sciences, Hunter College, City University of New York, New York, NY, United States of America
- Brain and Mind Research Institute, Weil Cornell Medical College, New York, NY, United States of America
| |
Collapse
|
19
|
Oyedeji O, Bakare MK, Adewale IO, Olutiola PO, Omoboye OO. Optimized production and characterization of thermostable invertase from Aspergillus niger IBK1, using pineapple peel as alternate substrate. BIOCATALYSIS AND AGRICULTURAL BIOTECHNOLOGY 2017. [DOI: 10.1016/j.bcab.2017.01.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
20
|
Bao Y, Liu X, Zhang W, Cao J, Li W, Li C, Lin Z. Identification of a regulation network in response to cadmium toxicity using blood clam Tegillarca granosa as model. Sci Rep 2016; 6:35704. [PMID: 27760991 PMCID: PMC5071765 DOI: 10.1038/srep35704] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 10/04/2016] [Indexed: 12/19/2022] Open
Abstract
Clam, a filter-feeding lamellibranch mollusk, is capable to accumulate high levels of trace metals and has therefore become a model for investigation the mechanism of heavy metal toxification. In this study, the effects of cadmium were characterized in the gills of Tegillarca granosa during a 96-hour exposure course using integrated metabolomic and proteomic approaches. Neurotoxicity and disturbances in energy metabolism were implicated according to the metabolic responses after Cd exposure, and eventually affected the osmotic function of gill tissue. Proteomic analysis showed that oxidative stress, calcium-binding and sulfur-compound metabolism proteins were key factors responding to Cd challenge. A knowledge-based network regulation model was constructed with both metabolic and proteomic data. The model suggests that Cd stimulation mainly inhibits a core regulation network that is associated with histone function, ribosome processing and tight junctions, with the hub proteins actin, gamma 1 and Calmodulin 1. Moreover, myosin complex inhibition causes abnormal tight junctions and is linked to the irregular synthesis of amino acids. For the first time, this study provides insight into the proteomic and metabolomic changes caused by Cd in the blood clam T. granosa and suggests a potential toxicological pathway for Cd.
Collapse
Affiliation(s)
- Yongbo Bao
- Zhejiang Key Laboratory of Aquatic Germplasm Resources, College of Biological & Environmental Sciences, Zhejiang Wanli University, Ningbo, Zhejiang, 315100, P.R. China
| | - Xiao Liu
- Department of Systems biology, GFK, Shanghai Biotech Inc., Shanghai, 201112, P.R. China
| | - Weiwei Zhang
- School of Marine Scienes, Ningbo University, Ningbo, Zhejiang, 315010, P.R. China
| | - Jianping Cao
- Ningbo Yinzhou Measurement and Test Center for Quality and Technique Supervising, Ningbo, Zhejiang, 315100, P.R. China
| | - Wei Li
- Zhejiang Key Laboratory of Aquatic Germplasm Resources, College of Biological & Environmental Sciences, Zhejiang Wanli University, Ningbo, Zhejiang, 315100, P.R. China
| | - Chenghua Li
- School of Marine Scienes, Ningbo University, Ningbo, Zhejiang, 315010, P.R. China
| | - Zhihua Lin
- Zhejiang Key Laboratory of Aquatic Germplasm Resources, College of Biological & Environmental Sciences, Zhejiang Wanli University, Ningbo, Zhejiang, 315100, P.R. China
| |
Collapse
|
21
|
Mutations in mitochondrial enzyme GPT2 cause metabolic dysfunction and neurological disease with developmental and progressive features. Proc Natl Acad Sci U S A 2016; 113:E5598-607. [PMID: 27601654 DOI: 10.1073/pnas.1609221113] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Mutations that cause neurological phenotypes are highly informative with regard to mechanisms governing human brain function and disease. We report autosomal recessive mutations in the enzyme glutamate pyruvate transaminase 2 (GPT2) in large kindreds initially ascertained for intellectual and developmental disability (IDD). GPT2 [also known as alanine transaminase 2 (ALT2)] is one of two related transaminases that catalyze the reversible addition of an amino group from glutamate to pyruvate, yielding alanine and α-ketoglutarate. In addition to IDD, all affected individuals show postnatal microcephaly and ∼80% of those followed over time show progressive motor symptoms, a spastic paraplegia. Homozygous nonsense p.Arg404* and missense p.Pro272Leu mutations are shown biochemically to be loss of function. The GPT2 gene demonstrates increasing expression in brain in the early postnatal period, and GPT2 protein localizes to mitochondria. Akin to the human phenotype, Gpt2-null mice exhibit reduced brain growth. Through metabolomics and direct isotope tracing experiments, we find a number of metabolic abnormalities associated with loss of Gpt2. These include defects in amino acid metabolism such as low alanine levels and elevated essential amino acids. Also, we find defects in anaplerosis, the metabolic process involved in replenishing TCA cycle intermediates. Finally, mutant brains demonstrate misregulated metabolites in pathways implicated in neuroprotective mechanisms previously associated with neurodegenerative disorders. Overall, our data reveal an important role for the GPT2 enzyme in mitochondrial metabolism with relevance to developmental as well as potentially to neurodegenerative mechanisms.
Collapse
|
22
|
Hughes G, Pemberton R, Fielden P, Hart J. A novel reagentless glutamate microband biosensor for real-time cell toxicity monitoring. Anal Chim Acta 2016; 933:82-8. [DOI: 10.1016/j.aca.2016.06.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 05/27/2016] [Accepted: 06/02/2016] [Indexed: 11/30/2022]
|
23
|
Changes of the plasma metabolome of newly born piglets subjected to postnatal hypoxia and resuscitation with air. Pediatr Res 2016; 80:284-92. [PMID: 27055187 DOI: 10.1038/pr.2016.66] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Accepted: 02/02/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND Perinatal hypoxic-ischemic brain damage is a major cause of mortality and morbidity in the neonatal period. Currently, limited ranges of biochemical tests assessing the intensity and duration of hypoxia are ready for clinical use. However, the need to initiate hypothermia therapy early after the clinical suspicion of hypoxic-ischemic encephalopathy requires the availability of early and reliable hypoxia markers. We have sought these biomarkers in an experimental model of hypoxia reoxygenation. METHODS Hypoxia and hypotension were induced in newborn piglets following a standardized model and reoxygenation was carried out using room air (RA). An untargeted liquid chromatography-time of flight mass spectrometry (LC-TOFMS) approach was used to assess changes in the metabolomic profile of plasma samples after intense hypoxia and upon reoxygenation. RESULTS At the end of hypoxia, the plasma metabolome showed an increased plasma concentration of analytes reflecting a metabolic adaptation to prolonged anaerobiosis. However, after resuscitation, metabolite levels returned to the starting values. CONCLUSION Severe hypoxia induces early, significant, and transient changes of specific metabolites in the plasma metabolome, which represent a snapshot of the biochemical adaptation of mammals to intense hypoxia. These metabolites could have applicability in predicting the severity of hypoxia in the clinical setting.
Collapse
|
24
|
Metabolomic Analysis of Biochemical Changes in the Plasma of High-Fat Diet and Streptozotocin-Induced Diabetic Rats after Treatment with Isoflavones Extract of Radix Puerariae. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:4701890. [PMID: 27042190 PMCID: PMC4794592 DOI: 10.1155/2016/4701890] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 01/10/2016] [Accepted: 01/31/2016] [Indexed: 12/14/2022]
Abstract
The main purpose of this study was to investigate the protective effects of total isoflavones from Radix Puerariae (PTIF) in diabetic rats. Diabetes was induced by a high-fat diet and intraperitoneal injection of low-dose streptozotocin (STZ; 40 mg/kg). At 26 weeks onwards, PTIF 421 mg/kg was administrated to the rats once daily consecutively for 10 weeks. Metabolic profiling changes were analyzed by Ultraperformance Liquid Chromatography-Quadrupole-Exactive Orbitrap-Mass Spectrometry (UPLC-Q-Exactive Orbitrap-MS). The principal component discriminant analysis (PCA-DA), partial least-squares discriminant analysis (PLS-DA), and orthogonal partial least-squares discriminant analysis (OPLS-DA) were used for multivariate analysis. Moreover, free amino acids in serum were determined by high-performance liquid chromatography with fluorescence detector (HPLC-FLD). Additionally, oxidative stress and inflammatory cytokines were evaluated. Eleven potential metabolite biomarkers, which are mainly related to the coagulation, lipid metabolism, and amino acid metabolism, have been identified. PCA-DA scores plots indicated that biochemical changes in diabetic rats were gradually restored to normal after administration of PTIF. Furthermore, the levels of BCAAs, glutamate, arginine, and tyrosine were significantly increased in diabetic rats. Treatment with PTIF could regulate the disturbed amino acid metabolism. Consequently, PTIF has great therapeutic potential in the treatment of DM by improving metabolism disorders and inhibiting oxidative damage.
Collapse
|
25
|
Yu LJ, Wall BA, Wangari-Talbot J, Chen S. Metabotropic glutamate receptors in cancer. Neuropharmacology 2016; 115:193-202. [PMID: 26896755 DOI: 10.1016/j.neuropharm.2016.02.011] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 02/07/2016] [Accepted: 02/09/2016] [Indexed: 01/31/2023]
Abstract
Metabotropic glutamate receptors (mGluRs) are widely known for their roles in synaptic signaling. However, accumulating evidence suggests roles of mGluRs in human malignancies in addition to synaptic transmission. Somatic cell homeostasis presents intriguing possibilities of mGluRs and glutamate signaling as novel targets for human cancers. More recently, aberrant glutamate signaling has been shown to participate in the transformation and maintenance of various cancer types, including glioma, melanoma skin cancer, breast cancer, and prostate cancer, indicating that genes encoding mGluRs, GRMs, can function as oncogenes. Here, we provide a review on the interactions of mGluRs and their ligand, glutamate, in processes that promote the growth of tumors of neuronal and non-neuronal origins. Further, we discuss the evolution of riluzole, a glutamate release inhibitor approved for amyotrophic lateral sclerosis (ALS), but now fashioned as an mGluR1 inhibitor for melanoma therapy and as a radio-sensitizer for tumors that have metastasized to the brain. With the success of riluzole, it is not far-fetched to believe that other drugs that may act directly or indirectly on other mGluRs can be beneficial for multiple applications. This article is part of the Special Issue entitled 'Metabotropic Glutamate Receptors, 5 years on'.
Collapse
Affiliation(s)
- Lumeng J Yu
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, NJ, USA
| | - Brian A Wall
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, NJ, USA; Global Product Safety, Colgate-Palmolive Company, Piscataway, NJ, USA
| | - Janet Wangari-Talbot
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, NJ, USA
| | - Suzie Chen
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers University, NJ, USA; The Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA.
| |
Collapse
|
26
|
Saran U, Humar B, Kolly P, Dufour JF. Hepatocellular carcinoma and lifestyles. J Hepatol 2016; 64:203-14. [PMID: 26341826 DOI: 10.1016/j.jhep.2015.08.028] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 08/19/2015] [Accepted: 08/24/2015] [Indexed: 12/31/2022]
Abstract
The majority of hepatocellular carcinoma occurs over pre-existing chronic liver diseases that share cirrhosis as an endpoint. In the last decade, a strong association between lifestyle and hepatocellular carcinoma has become evident. Abundance of energy-rich food and sedentary lifestyles have caused metabolic conditions such as obesity and diabetes mellitus to become global epidemics. Obesity and diabetes mellitus are both tightly linked to non-alcoholic fatty liver disease and also increase hepatocellular carcinoma risk independent of cirrhosis. Emerging data suggest that physical activity not only counteracts obesity, diabetes mellitus and non-alcoholic fatty liver disease, but also reduces cancer risk. Physical activity exerts significant anticancer effects in the absence of metabolic disorders. Here, we present a systematic review on lifestyles and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Uttara Saran
- Hepatology, Department of Clinical Research, University of Berne, Berne, Switzerland; University Clinic of Visceral Surgery and Medicine, Inselspital Berne, Berne, Switzerland
| | - Bostjan Humar
- Department of Visceral & Transplantation Surgery, University Hospital Zürich, Zürich, Switzerland
| | - Philippe Kolly
- Hepatology, Department of Clinical Research, University of Berne, Berne, Switzerland; University Clinic of Visceral Surgery and Medicine, Inselspital Berne, Berne, Switzerland
| | - Jean-François Dufour
- Hepatology, Department of Clinical Research, University of Berne, Berne, Switzerland; University Clinic of Visceral Surgery and Medicine, Inselspital Berne, Berne, Switzerland.
| |
Collapse
|
27
|
Perspectives in Intraoperative Diagnostics of Human Gliomas. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2015; 2015:479014. [PMID: 26543495 PMCID: PMC4620377 DOI: 10.1155/2015/479014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Accepted: 06/25/2015] [Indexed: 12/31/2022]
Abstract
Amongst large a variety of oncological diseases, malignant gliomas represent one of the most severe types of tumors. They are also the most common type of the brain tumors and account for over half of the astrocytic tumors. According to different sources, the average life expectancy of patients with various glioblastomas varies between 10 and 12 months and that of patients with anaplastic astrocytic tumors between 20 and 24 months. Therefore, studies of the physiology of transformed glial cells are critical for the development of treatment methods. Modern medical approaches offer complex procedures, including the microsurgical tumor removal, radiotherapy, and chemotherapy, supplemented with photodynamic therapy and immunotherapy. The most radical of them is surgical resection, which allows removing the largest part of the tumor, reduces the intracranial hypertension, and minimizes the degree of neurological deficit. However, complete removal of the tumor remains impossible. The main limitations are insufficient visualization of glioma boundaries, due to its infiltrative growth, and the necessity to preserve healthy tissue. This review is devoted to the description of advantages and disadvantages of modern intraoperative diagnostics of human gliomas and highlights potential perspectives for development of their treatment.
Collapse
|
28
|
Anderson SG, Dunn WB, Banerjee M, Brown M, Broadhurst DI, Goodacre R, Cooper GJS, Kell DB, Cruickshank JK. Evidence that multiple defects in lipid regulation occur before hyperglycemia during the prodrome of type-2 diabetes. PLoS One 2014; 9:e103217. [PMID: 25184286 PMCID: PMC4153569 DOI: 10.1371/journal.pone.0103217] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Accepted: 06/30/2014] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Blood-vessel dysfunction arises before overt hyperglycemia in type-2 diabetes (T2DM). We hypothesised that a metabolomic approach might identify metabolites/pathways perturbed in this pre-hyperglycemic phase. To test this hypothesis and for specific metabolite hypothesis generation, serum metabolic profiling was performed in young women at increased, intermediate and low risk of subsequent T2DM. METHODS Participants were stratified by glucose tolerance during a previous index pregnancy into three risk-groups: overt gestational diabetes (GDM; n = 18); those with glucose values in the upper quartile but below GDM levels (UQ group; n = 45); and controls (n = 43, below the median glucose values). Follow-up serum samples were collected at a mean 22 months postnatally. Samples were analysed in a random order using Ultra Performance Liquid Chromatography coupled to an electrospray hybrid LTQ-Orbitrap mass spectrometer. Statistical analysis included principal component (PCA) and multivariate methods. FINDINGS Significant between-group differences were observed at follow-up in waist circumference (86, 95%CI (79-91) vs 80 (76-84) cm for GDM vs controls, p<0.05), adiponectin (about 33% lower in GDM group, p = 0.004), fasting glucose, post-prandial glucose and HbA1c, but the latter 3 all remained within the 'normal' range. Substantial differences in metabolite profiles were apparent between the 2 'at-risk' groups and controls, particularly in concentrations of phospholipids (4 metabolites with p ≤ 0.01), acylcarnitines (3 with p ≤ 0.02), short- and long-chain fatty acids (3 with p< = 0.03), and diglycerides (4 with p ≤ 0.05). INTERPRETATION Defects in adipocyte function from excess energy storage as relatively hypoxic visceral and hepatic fat, and impaired mitochondrial fatty acid oxidation may initiate the observed perturbations in lipid metabolism. Together with evidence from the failure of glucose-directed treatments to improve cardiovascular outcomes, these data and those of others indicate that a new, quite different definition of type-2 diabetes is required. This definition would incorporate disturbed lipid metabolism prior to hyperglycemia.
Collapse
Affiliation(s)
- Simon G. Anderson
- Institute of Cardiovascular Sciences, Core Technology Facility, The University of Manchester, Manchester, United Kingdom
| | - Warwick B. Dunn
- Manchester Centre for Integrative Systems Biology, Manchester Institute of Biotechnology, The University of Manchester, Manchester, United Kingdom
- Centre for Advanced Discovery & Experimental Therapeutics (CADET), Central Manchester NHS Foundation Trust and School of Biomedicine, The University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Moulinath Banerjee
- Institute of Cardiovascular Sciences, Core Technology Facility, The University of Manchester, Manchester, United Kingdom
| | - Marie Brown
- Manchester Centre for Integrative Systems Biology, Manchester Institute of Biotechnology, The University of Manchester, Manchester, United Kingdom
| | - David I. Broadhurst
- Manchester Centre for Integrative Systems Biology, Manchester Institute of Biotechnology, The University of Manchester, Manchester, United Kingdom
- Division of General Internal Medicine, Department of Medicine, 4126A Katz Group Centre for Pharmacy & Health, University of Alberta, Edmonton, Alberta, Canada
| | - Royston Goodacre
- Manchester Centre for Integrative Systems Biology, Manchester Institute of Biotechnology, The University of Manchester, Manchester, United Kingdom
| | - Garth J. S. Cooper
- Centre for Advanced Discovery & Experimental Therapeutics (CADET), Central Manchester NHS Foundation Trust and School of Biomedicine, The University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom
- Maurice Wilkins Centre for Molecular Biodiscovery, Faculty of Science, University of Auckland, Auckland, New Zealand
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Douglas B. Kell
- Manchester Centre for Integrative Systems Biology, Manchester Institute of Biotechnology, The University of Manchester, Manchester, United Kingdom
| | - J. Kennedy Cruickshank
- Institute of Cardiovascular Sciences, Core Technology Facility, The University of Manchester, Manchester, United Kingdom
- Diabetes & Nutritional Sciences Division, King's College London, London, United Kingdom
| |
Collapse
|
29
|
Abstract
The objective of the article is to highlight various roles of glutamic acid like endogenic anticancer agent, conjugates to anticancer agents, and derivatives of glutamic acid as possible anticancer agents. Besides these emphases are given especially for two endogenous derivatives of glutamic acid such as glutamine and glutamate. Glutamine is a derivative of glutamic acid and is formed in the body from glutamic acid and ammonia in an energy requiring reaction catalyzed by glutamine synthase. It also possesses anticancer activity. So the transportation and metabolism of glutamine are also discussed for better understanding the role of glutamic acid. Glutamates are the carboxylate anions and salts of glutamic acid. Here the roles of various enzymes required for the metabolism of glutamates are also discussed.
Collapse
Affiliation(s)
- Satyajit Dutta
- Department of Pharmaceutical Chemistry, IIMT College of Medical Sciences, Meerut 250001, Uttar Pradesh, India
| | | | | |
Collapse
|
30
|
Grings M, Moura AP, Amaral AU, Parmeggiani B, Gasparotto J, Moreira JCF, Gelain DP, Wyse ATS, Wajner M, Leipnitz G. Sulfite disrupts brain mitochondrial energy homeostasis and induces mitochondrial permeability transition pore opening via thiol group modification. Biochim Biophys Acta Mol Basis Dis 2014; 1842:1413-22. [PMID: 24793416 DOI: 10.1016/j.bbadis.2014.04.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 04/23/2014] [Accepted: 04/25/2014] [Indexed: 11/16/2022]
Abstract
Sulfite oxidase (SO) deficiency is biochemically characterized by the accumulation of sulfite, thiosulfate and S-sulfocysteine in tissues and biological fluids of the affected patients. The main clinical symptoms include severe neurological dysfunction and brain abnormalities, whose pathophysiology is still unknown. The present study investigated the in vitro effects of sulfite and thiosulfate on mitochondrial homeostasis in rat brain mitochondria. It was verified that sulfite per se, but not thiosulfate, decreased state 3, CCCP-stimulated state and respiratory control ratio in mitochondria respiring with glutamate plus malate. In line with this, we found that sulfite inhibited the activities of glutamate and malate (MDH) dehydrogenases. In addition, sulfite decreased the activity of a commercial solution of MDH, that was prevented by antioxidants and dithiothreitol. Sulfite also induced mitochondrial swelling and reduced mitochondrial membrane potential, Ca(2+) retention capacity, NAD(P)H pool and cytochrome c immunocontent when Ca(2+) was present in the medium. These alterations were prevented by ruthenium red, cyclosporine A (CsA) and ADP, supporting the involvement of mitochondrial permeability transition (MPT) in these effects. We further observed that N-ethylmaleimide prevented the sulfite-elicited swelling and that sulfite decreased free thiol group content in brain mitochondria. These findings indicate that sulfite acts directly on MPT pore containing thiol groups. Finally, we verified that sulfite reduced cell viability in cerebral cortex slices and that this effect was prevented by CsA. Therefore, it may be presumed that disturbance of mitochondrial energy homeostasis and MPT induced by sulfite could be involved in the neuronal damage characteristic of SO deficiency.
Collapse
Affiliation(s)
- Mateus Grings
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil
| | - Alana P Moura
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil
| | - Alexandre U Amaral
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil
| | - Belisa Parmeggiani
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil
| | - Juciano Gasparotto
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil
| | - José C F Moreira
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil
| | - Daniel P Gelain
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil
| | - Angela T S Wyse
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil
| | - Moacir Wajner
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil; Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, CEP 90035-903 Porto Alegre, RS, Brazil
| | - Guilhian Leipnitz
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil.
| |
Collapse
|
31
|
Stark R, Kibbey RG. The mitochondrial isoform of phosphoenolpyruvate carboxykinase (PEPCK-M) and glucose homeostasis: has it been overlooked? BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1840:1313-30. [PMID: 24177027 PMCID: PMC3943549 DOI: 10.1016/j.bbagen.2013.10.033] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 10/13/2013] [Accepted: 10/18/2013] [Indexed: 01/03/2023]
Abstract
BACKGROUND Plasma glucose levels are tightly regulated within a narrow physiologic range. Insulin-mediated glucose uptake by tissues must be balanced by the appearance of glucose from nutritional sources, glycogen stores, or gluconeogenesis. In this regard, a common pathway regulating both glucose clearance and appearance has not been described. The metabolism of glucose to produce ATP is generally considered to be the primary stimulus for insulin release from beta-cells. Similarly, gluconeogenesis from phosphoenolpyruvate (PEP) is believed to be the primarily pathway via the cytosolic isoform of phosphoenolpyruvate carboxykinase (PEPCK-C). These models cannot adequately explain the regulation of insulin secretion or gluconeogenesis. SCOPE OF REVIEW A metabolic sensing pathway involving mitochondrial GTP (mtGTP) and PEP synthesis by the mitochondrial isoform of PEPCK (PEPCK-M) is associated with glucose-stimulated insulin secretion from pancreatic beta-cells. Here we examine whether there is evidence for a similar mtGTP-dependent pathway involved in gluconeogenesis. In both islets and the liver, mtGTP is produced at the substrate level by the enzyme succinyl CoA synthetase (SCS-GTP) with a rate proportional to the TCA cycle. In the beta-cell PEPCK-M then hydrolyzes mtGTP in the production of PEP that, unlike mtGTP, can escape the mitochondria to generate a signal for insulin release. Similarly, PEPCK-M and mtGTP might also provide a significant source of PEP in gluconeogenic tissues for the production of glucose. This review will focus on the possibility that PEPCK-M, as a sensor for TCA cycle flux, is a key mechanism to regulate both insulin secretion and gluconeogenesis suggesting conservation of this biochemical mechanism in regulating multiple aspects of glucose homeostasis. Moreover, we propose that this mechanism may be important for regulating insulin secretion and gluconeogenesis compared to canonical nutrient sensing pathways. MAJOR CONCLUSIONS PEPCK-M, initially believed to be absent in islets, carries a substantial metabolic flux in beta-cells. This flux is intimately involved with the coupling of glucose-stimulated insulin secretion. PEPCK-M activity may have been similarly underestimated in glucose producing tissues and could potentially be an unappreciated but important source of gluconeogenesis. GENERAL SIGNIFICANCE The generation of PEP via PEPCK-M may occur via a metabolic sensing pathway important for regulating both insulin secretion and gluconeogenesis. This article is part of a Special Issue entitled Frontiers of Mitochondrial Research.
Collapse
Affiliation(s)
- Romana Stark
- Department of Physiology, Monash University, Clayton, Victoria 3800, Australia.
| | - Richard G Kibbey
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06520-8020, USA.
| |
Collapse
|
32
|
The Odyssey of a Young Gene: Structure–Function Studies in Human Glutamate Dehydrogenases Reveal Evolutionary-Acquired Complex Allosteric Regulation Mechanisms. Neurochem Res 2014; 39:471-86. [DOI: 10.1007/s11064-014-1251-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Revised: 01/24/2014] [Accepted: 01/29/2014] [Indexed: 01/13/2023]
|
33
|
Effects of low-dose radiation on glutamate dehydrogenase activity in tissues of rats with transplanted Guerin's carcinoma. Bull Exp Biol Med 2013; 156:91-3. [PMID: 24319738 DOI: 10.1007/s10517-013-2285-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Glutamate dehydrogenase activity in mitochondrial fraction of the liver and muscle tissue of tumor-bearing rats was studied in the dynamics of Guerin's carcinoma growth and after preliminary low-dose irradiation. At the initial stages of Guerin's carcinoma growth, maximum activity of glutamate dehydrogenase was observed in the liver, while in the muscle tissue, catabolic transformations of amino acids was activated at the logarithmic phase of carcinogenesis and tended to inhibition at the terminal stages. Low-dose irradiation was followed by activation glutamate dehydrogenase in mitochondrial fraction of the liver and inhibition of this enzyme in mitochondrial fraction of the muscle tissue.
Collapse
|
34
|
Zhou Y, Qiu L, Xiao Q, Wang Y, Meng X, Xu R, Wang S, Na R. Obesity and diabetes related plasma amino acid alterations. Clin Biochem 2013; 46:1447-52. [DOI: 10.1016/j.clinbiochem.2013.05.045] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 05/04/2013] [Accepted: 05/06/2013] [Indexed: 01/19/2023]
|
35
|
Willard SS, Koochekpour S. Glutamate, glutamate receptors, and downstream signaling pathways. Int J Biol Sci 2013; 9:948-59. [PMID: 24155668 PMCID: PMC3805900 DOI: 10.7150/ijbs.6426] [Citation(s) in RCA: 204] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 07/11/2013] [Indexed: 12/23/2022] Open
Abstract
Glutamate is a nonessential amino acid, a major bioenergetic substrate for proliferating normal and neoplastic cells, and an excitatory neurotransmitter that is actively involved in biosynthetic, bioenergetic, metabolic, and oncogenic signaling pathways. Glutamate signaling activates a family of receptors consisting of metabotropic glutamate receptors (mGluRs) and ionotropic glutamate receptors (iGluRs), both of which have been implicated in chronic disabling brain disorders such as Schizophrenia and neurodegenerative diseases like Alzheimer's, Parkinson's, and multiple sclerosis. In this review, we discuss the structural and functional relationship of mGluRs and iGluRs and their downstream signaling pathways. The three groups of mGluRs, the associated second messenger systems, and subsequent activation of PI3K/Akt, MAPK, NFkB, PLC, and Ca/CaM signaling systems will be discussed in detail. The current state of human mGluR1a as one of the most important isoforms of Group I-mGluRs will be highlighted. The lack of studies on the human orthologues of mGluRs family will be outlined. We conclude that upon further study, human glutamate-initiated signaling pathways may provide novel therapeutic opportunities for a variety of non-malignant and malignant human diseases.
Collapse
Affiliation(s)
- Stacey S Willard
- Departments of Cancer Genetics and Urology, Center for Genetics and Pharmacology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY, USA
| | | |
Collapse
|
36
|
Willard SS, Koochekpour S. Glutamate signaling in benign and malignant disorders: current status, future perspectives, and therapeutic implications. Int J Biol Sci 2013; 9:728-42. [PMID: 23983606 PMCID: PMC3753409 DOI: 10.7150/ijbs.6475] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 06/15/2013] [Indexed: 11/05/2022] Open
Abstract
Glutamate, a nonessential amino acid, is the major excitatory neurotransmitter in the central nervous system. As such, glutamate has been shown to play a role in not only neural processes, such as learning and memory, but also in bioenergetics, biosynthetic and metabolic oncogenic pathways. Glutamate has been the target of intense investigation for its involvement not only in the pathogenesis of benign neurodegenerative diseases (NDDs) such as Parkinson's disease, Alzheimer's disease, schizophrenia, multiple sclerosis, and amyotropic lateral sclerosis (ALS), but also in carcinogenesis and progression of malignant diseases. In addition to its intracellular activities, glutamate in secreted form is a phylogenetically conserved cell signaling molecule. Glutamate binding activates multiple major receptor families including the metabotropic glutamate receptors (mGluRs) and ionotropic glutamate receptors (iGluRs), both of which have been implicated in various signaling pathways in cancer. Inhibition of extracellular glutamate release or glutamate receptor activation via competitive or non-competitive antagonists decreases growth, migration and invasion and induces apoptosis in breast cancer, melanoma, glioma and prostate cancer cells. In this review, we discuss the current state of glutamate signaling research as it relates to benign and malignant diseases. In addition, we provide a synopsis of clinical trials using glutamate antagonists for the treatment of NDD and malignant diseases. We conclude that in addition to its potential role as a metabolic biomarker, glutamate receptors and glutamate-initiated signaling pathways may provide novel therapeutic opportunities for cancer.
Collapse
Affiliation(s)
- Stacey S Willard
- Departments of Cancer Genetics and Urology, Center for Genetics and Pharmacology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | | |
Collapse
|
37
|
Goswami DB, Jernigan CS, Chandran A, Iyo AH, May WL, Austin MC, Stockmeier CA, Karolewicz B. Gene expression analysis of novel genes in the prefrontal cortex of major depressive disorder subjects. Prog Neuropsychopharmacol Biol Psychiatry 2013; 43:126-33. [PMID: 23261523 PMCID: PMC4089971 DOI: 10.1016/j.pnpbp.2012.12.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Revised: 12/11/2012] [Accepted: 12/11/2012] [Indexed: 12/21/2022]
Abstract
Dysregulation of the glutamatergic system has been implicated not only in the treatment of major depressive disorder (MDD), but also in the excitotoxic effects of stress and anxiety on the prefrontal cortex, which may precede the onset of a depressive episode. Our previous studies demonstrate marked deficits in prominent postsynaptic proteins involved in glutamate neurotransmission in the prefrontal cortex (PFC), Brodmann's area 10 (BA 10) from subjects diagnosed with major depressive disorder (MDD). In the same group of subjects we have identified deficits in expression and phosphorylation level of key components of the mammalian target of rapamycin (mTOR) signaling pathway, known to regulate translation initiation. Based on our previous findings, we have postulated that glutamate-dependent dysregulation of mTOR-initiated protein synthesis in the PFC may underlie the pathology of MDD. The aim of this study was to use the NanoString nCounter System to perform analysis of genes coding for glutamate transporters, glutamate metabolizing enzymes, neurotrophic factors and other intracellular signaling markers involved in glutamate signaling that were not previously investigated by our group in the PFC BA 10 from subjects with MDD. We have analyzed a total of 200 genes from 16 subjects with MDD and 16 healthy controls. These are part of the same cohort used in our previous studies. Setting our cutoff p-value≤0.01, marked upregulation of genes coding for mitochondrial glutamate carrier (GC1; p=0.0015), neuropilin 1 (NRP-1; p=0.0019), glutamate receptor ionotropic N-methyl-d-aspartate-associated protein 1 (GRINA; p=0.0060), and fibroblast growth factor receptor 1 (FGFR-1; p=0.010) was identified. No significant differences in expression of the remaining 196 genes were observed between MDD subjects and controls. While upregulation of FGFR-1 has been previously shown in MDD; abnormalities in GC-1, GRINA, and NRP-1 have not been reported. Therefore, this postmortem study identifies GC1, GRINA, and NRP-1 as novel factors associated with MDD; however, future studies will be needed to address the significance of these genes in the pathophysiology of depression and antidepressant activity.
Collapse
Affiliation(s)
- Dharmendra B. Goswami
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS 39216-4505, USA,New England Primate Research Center, Harvard Medical School, One Pine Hill Drive, Southborough, MA 01772, USA,Correspondence author at: Department of Psychiatry, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216-4505, USA. Tel.: +1 601-815-5614; fax: +1-601-984-5899.
| | - Courtney S. Jernigan
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS 39216-4505, USA
| | - Agata Chandran
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS 39216-4505, USA
| | - Abiye H. Iyo
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS 39216-4505, USA
| | - Warren L. May
- Center of Biostatistics and Bioinformatic, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS 39216-4505, USA
| | - Mark C. Austin
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS 39216-4505, USA
| | - Craig A. Stockmeier
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS 39216-4505, USA,Department of Psychiatry, Case Western Reserve University, Cleveland, OH 44106
| | - Beata Karolewicz
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS 39216-4505, USA
| |
Collapse
|
38
|
Del Rizzo M, Burlina AP, Sass JO, Beermann F, Zanco C, Cazzorla C, Bordugo A, Giordano L, Manara R, Burlina AB. Metabolic stroke in a late-onset form of isolated sulfite oxidase deficiency. Mol Genet Metab 2013; 108:263-6. [PMID: 23414711 DOI: 10.1016/j.ymgme.2013.01.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 01/21/2013] [Accepted: 01/21/2013] [Indexed: 11/26/2022]
Abstract
We report the first case of late-onset isolated sulfite oxidase deficiency (ISOD) presenting with a stroke-like episode. Clinical, biochemical and neuroradiological features at diagnosis and during follow-up after dietary treatment intervention are described. Furthermore, pathogenic mechanisms possibly leading to stroke in ISOD are discussed.
Collapse
Affiliation(s)
- Monica Del Rizzo
- Division of Metabolic Diseases, Department of Woman and Child Health, University Hospital of Padua, Via Giustiniani 3 35128 Padua, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Intertissue Differences for the Role of Glutamate Dehydrogenase in Metabolism. Neurochem Res 2013; 39:516-26. [DOI: 10.1007/s11064-013-0998-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 01/24/2013] [Accepted: 02/01/2013] [Indexed: 11/26/2022]
|
40
|
Komlos D, Mann KD, Zhuo Y, Ricupero CL, Hart RP, Liu AYC, Firestein BL. Glutamate dehydrogenase 1 and SIRT4 regulate glial development. Glia 2012; 61:394-408. [PMID: 23281078 DOI: 10.1002/glia.22442] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Accepted: 10/22/2012] [Indexed: 01/10/2023]
Abstract
Congenital hyperinsulinism/hyperammonemia (HI/HA) syndrome is caused by an activation mutation of glutamate dehydrogenase 1 (GDH1), a mitochondrial enzyme responsible for the reversible interconversion between glutamate and α-ketoglutarate. The syndrome presents clinically with hyperammonemia, significant episodic hypoglycemia, seizures, and frequent incidences of developmental and learning defects. Clinical research has implicated that although some of the developmental and neurological defects may be attributed to hypoglycemia, some characteristics cannot be ascribed to low glucose and as hyperammonemia is generally mild and asymptomatic, there exists the possibility that altered GDH1 activity within the brain leads to some clinical changes. GDH1 is allosterically regulated by many factors, and has been shown to be inhibited by the ADP-ribosyltransferase sirtuin 4 (SIRT4), a mitochondrially localized sirtuin. Here we show that SIRT4 is localized to mitochondria within the brain. SIRT4 is highly expressed in glial cells, specifically astrocytes, in the postnatal brain and in radial glia during embryogenesis. Furthermore, SIRT4 protein decreases in expression during development. We show that factors known to allosterically regulate GDH1 alter gliogenesis in CTX8 cells, a novel radial glial cell line. We find that SIRT4 and GDH1 overexpression play antagonistic roles in regulating gliogenesis and that a mutant variant of GDH1 found in HI/HA patients accelerates the development of glia from cultured radial glia cells.
Collapse
Affiliation(s)
- Daniel Komlos
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Sookoian S, Pirola CJ. Alanine and aspartate aminotransferase and glutamine-cycling pathway: Their roles in pathogenesis of metabolic syndrome. World J Gastroenterol 2012; 18:3775-81. [PMID: 22876026 PMCID: PMC3413046 DOI: 10.3748/wjg.v18.i29.3775] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Revised: 06/15/2012] [Accepted: 06/28/2012] [Indexed: 02/06/2023] Open
Abstract
Although new research technologies are constantly used to look either for genes or biomarkers in the prediction of metabolic syndrome (MS), the pathogenesis and pathophysiology of this complex disease remains a major challenge. Interestingly, Cheng et al recently investigated possible pathways underlying MS by high-throughput metabolite profiling in two large and well characterized community-based cohorts. The authors explored by liquid chromatography and mass spectrometry the plasma concentrations of 45 distinct metabolites and examined their relation to cardiometabolic risk, and observed that metabolic risk factors such as obesity, insulin resistance (IR), high blood pressure, and dyslipidemia were associated with several metabolites, including branched-chain amino acids, other hydrophobic amino acids, tryptophan breakdown products, and nucleotide metabolites. In addition, the authors found a significant association of IR traits with glutamine, glutamate and the glutamine-to-glutamate ratio. These data provide new insight into the pathogenesis of MS-associated phenotypes and introduce a crucial role of glutamine-cycling pathway as prominently involved in the development of metabolic risk. We consider that the hypothesis about the role of abnormal glutamate metabolism in the pathogenesis of the MS is certainly challenging and suggests the critical role of the liver in the global metabolic modulation as glutamate metabolism is linked with aminotransferase reactions. We discuss here the critical role of the “liver metabolism” in the pathogenesis of the MS and IR, and postulate that before fatty liver develops, abnormal levels of liver enzymes, such as alanine and aspartate aminotransferases might reflect high levels of hepatic transamination of amino acids in the liver.
Collapse
|
42
|
Shi X, Wahlang B, Wei X, Yin X, Falkner KC, Prough RA, Kim SH, Mueller EG, McClain CJ, Cave M, Zhang X. Metabolomic analysis of the effects of polychlorinated biphenyls in nonalcoholic fatty liver disease. J Proteome Res 2012; 11:3805-15. [PMID: 22686559 DOI: 10.1021/pr300297z] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Polychlorinated biphenyls (PCBs) are persistent organic pollutants and have been associated with abnormal liver enzymes and suspected nonalcoholic fatty liver disease (NAFLD), obesity, and the metabolic syndrome in epidemiological studies. In epidemiological surveys of human PCB exposure, PCB 153 has the highest serum levels among PCB congeners. To determine the hepatic effects of PCB 153 in mice, C57BL/6J mice were fed either a control diet (CD) or a high fat diet (HFD) for 12 weeks, with or without PCB 153 coexposure. The metabolite extracts from mouse livers were analyzed using linear trap quadrupole-Fourier transform ion cyclotron resonance mass spectrometer (LTQ-FTICR MS) via direct infusion nanoelectrospray ionization (DI-nESI) mass spectrometry. The metabolomics analysis indicated no difference in the metabolic profile between mice fed the control diet with PCB 153 exposure (CD+PCB 153) and mice fed the control diet (CD) without PCB 153 exposure. However, compared with CD group, levels of 10 metabolites were increased and 15 metabolites were reduced in mice fed HFD. Moreover, compared to CD+PCB 153 group, the abundances of 6 metabolites were increased and 18 metabolites were decreased in the mice fed high fat diet with PCB 153 exposure (HFD+PCB 153). Compared with HFD group, the abundances of 2 metabolites were increased and of 12 metabolites were reduced in HFD+PCB 153 group. These observations agree with the histological results and indicate that the metabolic effects of PCB 153 were highly dependent on macronutrient interactions with HFD. Antioxidant depletion is likely to be an important consequence of this interaction, as this metabolic disturbance has previously been implicated in obesity and NAFLD.
Collapse
Affiliation(s)
- Xue Shi
- Department of Chemistry, University of Louisville, Louisville, Kentucky 40292, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
da Silva PMR, Batista TM, Ribeiro RA, Zoppi CC, Boschero AC, Carneiro EM. Decreased insulin secretion in islets from protein malnourished rats is associated with impaired glutamate dehydrogenase function: effect of leucine supplementation. Metabolism 2012; 61:721-32. [PMID: 22078937 DOI: 10.1016/j.metabol.2011.09.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Revised: 08/27/2011] [Accepted: 09/27/2011] [Indexed: 02/06/2023]
Abstract
We herein studied the role of glutamate dehydrogenase (GDH), in response to leucine (LEU) supplementation, upon insulin secretion of malnourished rats. Weaned male Wistar rats were fed normal-protein (17%) or low-protein diet (6%, LP) for 8 weeks. Half of the rats of each group were supplemented with LEU (1.5%) in the drinking water for the following 4 weeks. Gene and protein expressions, static insulin secretion, and cytoplasmic Ca(2+) oscillations were measured. Glutamate dehydrogenase messenger RNA was 58% lower in LP islets, and LEU supplementation augmented it in 28%. The LP islets secreted less insulin when exposed to 20 mmol/L LEU, 20 mmol/L LEU + 2 mmol/L glutamine (with or without 5 mmol/L aminooxyacetic acid, a branched chain aminotransferase inhibitor, or 20 μmol/L epigallocatechin gallate, a GDH inhibitor), 20 mmol/L α-ketoisocaproate, glutamine + 20 mmol/L β-2-aminobicyclo[2.2.1]heptane-2-carboxylic acid (a GDH activator), and 22.2 mmol/L glucose. Leucine supplementation augmented insulin secretion to levels found in normal-protein islets in all the above conditions, an effect that was blunted when islets were incubated with epigallocatechin gallate. The glutamine + β-2-aminobicyclo[2.2.1]heptane-2-carboxylic acid-induced increased [Ca(2+)](i) and oscillations were higher than those for LP islets. Leucine supplementation normalized these parameters in LP islets. Impaired GDH function was associated with lower insulin release in LP islets, and LEU supplementation normalized insulin secretion via restoration of GDH function. In addition, GDH may contribute to insulin secretion through ameliorations of Ca(2+) handling in LP islets.
Collapse
Affiliation(s)
- Priscilla Muniz Ribeiro da Silva
- Department of Anatomy, Cellular Biology and Physiology and Biophysics, Institute of Biology, University of Campinas,PO Box 6109, CEP 13083-970 Campinas, SP, Brazil
| | | | | | | | | | | |
Collapse
|
44
|
Structural basis for the allosteric inhibitory mechanism of human kidney-type glutaminase (KGA) and its regulation by Raf-Mek-Erk signaling in cancer cell metabolism. Proc Natl Acad Sci U S A 2012; 109:7705-10. [PMID: 22538822 DOI: 10.1073/pnas.1116573109] [Citation(s) in RCA: 153] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Besides thriving on altered glucose metabolism, cancer cells undergo glutaminolysis to meet their energy demands. As the first enzyme in catalyzing glutaminolysis, human kidney-type glutaminase isoform (KGA) is becoming an attractive target for small molecules such as BPTES [bis-2-(5 phenylacetamido-1, 2, 4-thiadiazol-2-yl) ethyl sulfide], although the regulatory mechanism of KGA remains unknown. On the basis of crystal structures, we reveal that BPTES binds to an allosteric pocket at the dimer interface of KGA, triggering a dramatic conformational change of the key loop (Glu312-Pro329) near the catalytic site and rendering it inactive. The binding mode of BPTES on the hydrophobic pocket explains its specificity to KGA. Interestingly, KGA activity in cells is stimulated by EGF, and KGA associates with all three kinase components of the Raf-1/Mek2/Erk signaling module. However, the enhanced activity is abrogated by kinase-dead, dominant negative mutants of Raf-1 (Raf-1-K375M) and Mek2 (Mek2-K101A), protein phosphatase PP2A, and Mek-inhibitor U0126, indicative of phosphorylation-dependent regulation. Furthermore, treating cells that coexpressed Mek2-K101A and KGA with suboptimal level of BPTES leads to synergistic inhibition on cell proliferation. Consequently, mutating the crucial hydrophobic residues at this key loop abrogates KGA activity and cell proliferation, despite the binding of constitutive active Mek2-S222/226D. These studies therefore offer insights into (i) allosteric inhibition of KGA by BPTES, revealing the dynamic nature of KGA's active and inhibitory sites, and (ii) cross-talk and regulation of KGA activities by EGF-mediated Raf-Mek-Erk signaling. These findings will help in the design of better inhibitors and strategies for the treatment of cancers addicted with glutamine metabolism.
Collapse
|
45
|
Plaitakis A, Latsoudis H, Spanaki C. The human GLUD2 glutamate dehydrogenase and its regulation in health and disease. Neurochem Int 2011; 59:495-509. [DOI: 10.1016/j.neuint.2011.03.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2010] [Revised: 03/04/2011] [Accepted: 03/06/2011] [Indexed: 01/12/2023]
|
46
|
Quan Y, Barszczyk A, Feng ZP, Sun HS. Current understanding of K ATP channels in neonatal diseases: focus on insulin secretion disorders. Acta Pharmacol Sin 2011; 32:765-80. [PMID: 21602835 PMCID: PMC4009965 DOI: 10.1038/aps.2011.57] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Accepted: 04/13/2011] [Indexed: 12/25/2022] Open
Abstract
ATP-sensitive potassium (K(ATP)) channels are cell metabolic sensors that couple cell metabolic status to electric activity, thus regulating many cellular functions. In pancreatic beta cells, K(ATP) channels modulate insulin secretion in response to fluctuations in plasma glucose level, and play an important role in glucose homeostasis. Recent studies show that gain-of-function and loss-of-function mutations in K(ATP) channel subunits cause neonatal diabetes mellitus and congenital hyperinsulinism respectively. These findings lead to significant changes in the diagnosis and treatment for neonatal insulin secretion disorders. This review describes the physiological and pathophysiological functions of K(ATP) channels in glucose homeostasis, their specific roles in neonatal diabetes mellitus and congenital hyperinsulinism, as well as future perspectives of K(ATP) channels in neonatal diseases.
Collapse
Affiliation(s)
- Yi Quan
- Departments of Physiology, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada, M5S 1A8
| | - Andrew Barszczyk
- Departments of Physiology, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada, M5S 1A8
| | - Zhong-ping Feng
- Departments of Physiology, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada, M5S 1A8
| | - Hong-shuo Sun
- Departments of Physiology, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada, M5S 1A8
- Departments of Surgery, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada, M5S 1A8
- Departments of Pharmacology, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada, M5S 1A8
- Institute of Medical Science, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada, M5S 1A8
| |
Collapse
|
47
|
Tranah GJ. Mitochondrial-nuclear epistasis: implications for human aging and longevity. Ageing Res Rev 2011; 10:238-52. [PMID: 20601194 DOI: 10.1016/j.arr.2010.06.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2010] [Revised: 06/17/2010] [Accepted: 06/17/2010] [Indexed: 12/22/2022]
Abstract
There is substantial evidence that mitochondria are involved in the aging process. Mitochondrial function requires the coordinated expression of hundreds of nuclear genes and a few dozen mitochondrial genes, many of which have been associated with either extended or shortened life span. Impaired mitochondrial function resulting from mtDNA and nuclear DNA variation is likely to contribute to an imbalance in cellular energy homeostasis, increased vulnerability to oxidative stress, and an increased rate of cellular senescence and aging. The complex genetic architecture of mitochondria suggests that there may be an equally complex set of gene interactions (epistases) involving genetic variation in the nuclear and mitochondrial genomes. Results from Drosophila suggest that the effects of mtDNA haplotypes on longevity vary among different nuclear allelic backgrounds, which could account for the inconsistent associations that have been observed between mitochondrial DNA (mtDNA) haplogroups and survival in humans. A diversity of pathways may influence the way mitochondria and nuclear-mitochondrial interactions modulate longevity, including: oxidative phosphorylation; mitochondrial uncoupling; antioxidant defenses; mitochondrial fission and fusion; and sirtuin regulation of mitochondrial genes. We hypothesize that aging and longevity, as complex traits having a significant genetic component, are likely to be controlled by nuclear gene variants interacting with both inherited and somatic mtDNA variability.
Collapse
|
48
|
Magni DV, Souza MA, Oliveira APF, Furian AF, Oliveira MS, Ferreira J, Santos ARS, Mello CF, Royes LFF, Fighera MR. Lipopolysaccharide enhances glutaric acid-induced seizure susceptibility in rat pups: Behavioral and electroencephalographic approach. Epilepsy Res 2011; 93:138-48. [DOI: 10.1016/j.eplepsyres.2010.11.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2010] [Revised: 11/23/2010] [Accepted: 11/25/2010] [Indexed: 12/31/2022]
|
49
|
Treberg JR, Clow KA, Greene KA, Brosnan ME, Brosnan JT. Systemic activation of glutamate dehydrogenase increases renal ammoniagenesis: implications for the hyperinsulinism/hyperammonemia syndrome. Am J Physiol Endocrinol Metab 2010; 298:E1219-25. [PMID: 20332361 DOI: 10.1152/ajpendo.00028.2010] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The hyperinsulism/hyperammonemia (HI/HA) syndrome is caused by glutamate dehydrogenase (GDH) gain-of-function mutations that reduce the inhibition by GTP, consequently increasing the activity of GDH in vivo. The source of the hyperammonemia in the HI/HA syndrome remains unclear. We examined the effect of systemic activation of GDH on ammonia metabolism in the rat. 2-Aminobicyclo[2,2,1]heptane-2-carboxylic acid (BCH) is a nonmetabolizable analog of the natural GDH allosteric activator leucine. A dose of 100 mumol BCH/100 g rat resulted in a mild systemic hyperammonemia. Using arterial-venous (A-V) differences, we exclude the liver, intestine, and skeletal muscle as major contributors to this BCH-induced hyperammonemia. However, renal ammonia output increased, as demonstrated by an increase in A-V difference for ammonia across the kidney in BCH-treated animals. Isolated renal cortical tubules incubated with BCH increased the rate of ammoniagenesis from glutamine by 40%. The flux through GDH increased more than twofold when BCH was added to renal mitochondria respiring on glutamine. The flux through glutaminase was not affected by BCH, whereas glutamate-oxaloacetate transaminase flux decreased when normalized to glutaminase flux. These data show that increased renal ammoniagenesis due to activation of GDH can explain the BCH-induced hyperammonemia. These results are discussed in relation to the organ source of the ammonia in the HI/HA syndrome as well as the role of GDH in regulating renal ammoniagenesis.
Collapse
Affiliation(s)
- Jason R Treberg
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada.
| | | | | | | | | |
Collapse
|
50
|
Emesh S, Rapson TD, Rajapakshe A, Kappler U, Bernhardt PV, Tollin G, Enemark JH. Intramolecular electron transfer in sulfite-oxidizing enzymes: elucidating the role of a conserved active site arginine. Biochemistry 2009; 48:2156-63. [PMID: 19226119 DOI: 10.1021/bi801553q] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
All reported sulfite-oxidizing enzymes have a conserved arginine in their active site which hydrogen bonds to the equatorial oxygen ligand on the Mo atom. Previous studies on the pathogenic R160Q mutant of human sulfite oxidase (HSO) have shown that Mo-heme intramolecular electron transfer (IET) is dramatically slowed when positive charge is lost at this position. To improve our understanding of the function that this conserved positively charged residue plays in IET, we have studied the equivalent uncharged substitutions R55Q and R55M as well as the positively charged substitution R55K in bacterial sulfite dehydrogenase (SDH). The heme and molybdenum cofactor (Moco) subunits are tightly associated in SDH, which makes it an ideal system for improving our understanding of residue function in IET without the added complexity of the interdomain movement that occurs in HSO. Unexpectedly, the uncharged SDH variants (R55Q and R55M) exhibited increased IET rate constants relative to that of the wild type (3-4-fold) when studied by laser flash photolysis. The gain in function observed in SDH(R55Q) and SDH(R55M) suggests that the reduction in the level of IET seen in HSO(R160Q) is not due to a required role of this residue in the IET pathway itself, but to the fact that it plays an important role in heme orientation during the interdomain movement necessary for IET in HSO (as seen in viscosity experiments). The pH profiles of SDH(WT), SDH(R55M), and SDH(R55Q) show that the arginine substitution also alters the behavior of the Mo-heme IET equilibrium (K(eq)) and rate constants (k(et)) of both variants with respect to the SDH(WT) enzyme. SDH(WT) has a k(et) that is independent of pH and a K(eq) that increases as pH decreases; on the other hand, both SDH(R55M) and SDH(R55Q) have a k(et) that increases as pH decreases, and SDH(R55M) has a K(eq) that is pH-independent. IET in the SDH(R55Q) variant is inhibited by sulfate in laser flash photolysis experiments, a behavior that differs from that of SDH(WT), but which also occurs in HSO. IET in SDH(R55K) is slower than in SDH(WT). A new analysis of the possible mechanistic pathways for sulfite-oxidizing enzymes is presented and related to available kinetic and EPR results for these enzymes.
Collapse
Affiliation(s)
- Safia Emesh
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona 85721, USA
| | | | | | | | | | | | | |
Collapse
|