1
|
Walters TH, Wiah S, Shekarabi A, Milton M, Reddy S, Zhao P, Mokkarala PS, Potula R, Rawls SM. Clavulanic acid inhibits methamphetamine locomotor sensitization in mice and normalizes methamphetamine-induced changes in glutaminase mRNA levels in the nucleus accumbens. Physiol Behav 2024; 284:114646. [PMID: 39053627 PMCID: PMC11347075 DOI: 10.1016/j.physbeh.2024.114646] [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: 03/19/2024] [Revised: 06/14/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
Clavulanic acid (CLAV) is a component of Augmentin® that preserves antibiotic efficacy by inhibiting β-lactamase activity. It also enhances cellular glutamate uptake and is a potential CNS therapeutic. Because increased glutamate transmission in brain reward circuits facilitates methamphetamine (METH) locomotor activation and sensitization, we tested the hypothesis that CLAV inhibits acute and sensitized locomotor responses to METH in mice and investigated effects of CLAV on METH-induced changes in glutaminase, the major glutamate-producing enzyme in the brain. Acute METH (3 mg/kg) produced hyperlocomotion that was reduced by CLAV (20 mg/kg but not 10 mg/kg). Mice injected with METH (3 mg/kg) every other day for 9 d and then challenged with METH 27 d later displayed locomotor sensitization. CLAV (10 mg/kg), when injected 15 min before each METH injection during the 9-d exposure interval, blocked locomotor sensitization induced by METH challenge. In METH-sensitized mice, mRNA levels of both isoforms of glutaminase (GLS and GLS2) were altered in the nucleus accumbens compared to mice exposed to a single injection of METH (i.e., GLS decreased and GLS2 increased). CLAV normalized the METH-induced GLS deficit but not the increase in GLS2. In summary, CLAV reduced acute and sensitized locomotor responses to METH and normalized the METH-induced reduction of GLS gene expression in the NAC. Given that glutaminases belong to the β-lactamase superfamily and CLAV is a β-lactamase inhibitor, our data point toward studying glutaminase as a therapeutic target of CLAV.
Collapse
Affiliation(s)
- Taylor Hawthorne Walters
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Sonita Wiah
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Aryan Shekarabi
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Mia Milton
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Samhitha Reddy
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Pingwei Zhao
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Prateek S Mokkarala
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Raghava Potula
- Department of Pathology and Laboratory Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Scott M Rawls
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA; Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA.
| |
Collapse
|
2
|
Cooper AJL, Denton TT. ω-Amidase and Its Substrate α-Ketoglutaramate (the α-Keto Acid Analogue of Glutamine) as Biomarkers in Health and Disease. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1660-1680. [PMID: 39523108 DOI: 10.1134/s000629792410002x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/10/2024] [Accepted: 09/15/2024] [Indexed: 11/16/2024]
Abstract
A large literature exists on the biochemistry, chemistry, metabolism, and clinical importance of the α-keto acid analogues of many amino acids. However, although glutamine is the most abundant amino acid in human tissues, and transamination of glutamine to its α-keto acid analogue (α-ketoglutaramate; KGM) was described more than seventy years ago, little information is available on the biological importance of KGM. Herein, we summarize the metabolic importance of KGM as an intermediate in the glutamine transaminase - ω-amidase (GTωA) pathway for the conversion of glutamine to anaplerotic α-ketoglutarate. We describe some properties of KGM, notably its occurrence as a lactam (2-hydroxy-5-oxoproline; 99.7% at pH 7.2), and its presence in normal tissues and body fluids. We note that the concentration of KGM is elevated in the cerebrospinal fluid of liver disease patients and that the urinary KGM/creatinine ratio is elevated in patients with an inborn error of the urea cycle and in patients with citrin deficiency. Recently, of the 607 urinary metabolites measured in a kidney disease study, KGM was noted to be one of five metabolites that was most significantly associated with uromodulin (a potential biomarker for tubular functional mass). Finally, we note that KGM is an intermediate in the breakdown of nicotine in certain organisms and is an important factor in nitrogen homeostasis in some microorganisms and plants. In conclusion, we suggest that biochemists and clinicians should consider KGM as (i) a key intermediate in nitrogen metabolism in all branches of life, and (ii) a biomarker, along with ω-amidase, in several diseases.
Collapse
Affiliation(s)
- Arthur J L Cooper
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, 10595, USA
| | - Travis T Denton
- LiT Biosciences, Spokane, WA, 99202-5029, USA. ARRAY(0x5d17383a0090)
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University Health Sciences Spokane, Spokane, WA, USA
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University Health Sciences Spokane, Spokane, WA, USA
- Steve Gleason Institute for Neuroscience, Washington State University Health Sciences Spokane, Spokane, WA, USA
| |
Collapse
|
3
|
Gao Y, Ma L, Yuan J, Huang Y, Ban Y, Zhang P, Tan D, Liang M, Li Z, Gong C, Xu T, Yang X, Chen Y. GLS2 reduces the occurrence of epilepsy by affecting mitophagy function in mouse hippocampal neurons. CNS Neurosci Ther 2024; 30:e70036. [PMID: 39404053 PMCID: PMC11474837 DOI: 10.1111/cns.70036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/21/2024] [Accepted: 07/26/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Altered mitophagy has been observed in various neurological disorders, such as epilepsy. The role of mitophagy in causing neuronal damage during epileptic episodes is significant, and recent research has indicated that GLS2 plays a crucial role in regulating autophagy. However, exactly how GLS2 affects epilepsy is still unclear. AIMS To investigate the expression and distribution characteristics of GLS2 in epilepsy, and then observed the changes in behavior and electrophysiology caused by overexpression of GLS2 in epileptic mice, and determined whether GLS2 regulated seizure-like changes in the mouse model through the protective mechanism of mitophagy. RESULTS The expression of GLS2 in a kainic acid (KA)-induced epileptic mouse model and aglutamate-inducedneuronal excitatory damage in HT22 cells model was downregulation. In brief, overexpression of GLS2 can alleviate epileptic activity. Subsequently, we demonstrated that GLS2 interacts with mitophagy-related proteins in a KA-induced epilepsy mouse model. Mechanistically, overexpression of GLS2 inhibited mitophagy in epileptic mice, downregulating the expression of LC3 and reducing ROS production. CONCLUSIONS This study proves the GLS2 expression pattern is abnormal in epileptic mice. The function of mitophagy in hippocampal neurons is affected by GLS2, and overexpression of GLS2 can reduce the occurrence of seizure-like events (SLEs) by altering mitophagy function. Thus, GLS2 might control seizures, and our findings provide a fresh avenue for antiepileptic treatment and offer novel insights into treating and preventing epilepsy.
Collapse
Affiliation(s)
- Yuan Gao
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Limin Ma
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Department of GerontologyChongqing University Three Gorges HospitalChongqingChina
| | - Jinxian Yuan
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Yunyi Huang
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Yuenan Ban
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Peng Zhang
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Dandan Tan
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Minxue Liang
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Zhipeng Li
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Chen Gong
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Tao Xu
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Xiaolan Yang
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Department of NeurologyChongqing Medical University Affiliated Second Hospital Affiliated Fengjie HospitalChongqingChina
| | - Yangmei Chen
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| |
Collapse
|
4
|
Erb HHH, Polishchuk N, Stasyk O, Kahya U, Weigel MM, Dubrovska A. Glutamine Metabolism and Prostate Cancer. Cancers (Basel) 2024; 16:2871. [PMID: 39199642 PMCID: PMC11352381 DOI: 10.3390/cancers16162871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 09/01/2024] Open
Abstract
Glutamine (Gln) is a non-essential amino acid that is involved in the development and progression of several malignancies, including prostate cancer (PCa). While Gln is non-essential for non-malignant prostate epithelial cells, PCa cells become highly dependent on an exogenous source of Gln. The Gln metabolism in PCa is tightly controlled by well-described oncogenes such as MYC, AR, and mTOR. These oncogenes contribute to therapy resistance and progression to the aggressive castration-resistant PCa. Inhibition of Gln catabolism impedes PCa growth, survival, and tumor-initiating potential while sensitizing the cells to radiotherapy. Therefore, given its significant role in tumor growth, targeting Gln metabolism is a promising approach for developing new therapeutic strategies. Ongoing clinical trials evaluate the safety and efficacy of Gln catabolism inhibitors in combination with conventional and targeted therapies in patients with various solid tumors, including PCa. Further understanding of how PCa cells metabolically interact with their microenvironment will facilitate the clinical translation of Gln inhibitors and help improve therapeutic outcomes. This review focuses on the role of Gln in PCa progression and therapy resistance and provides insights into current clinical trials.
Collapse
Affiliation(s)
- Holger H. H. Erb
- Department of Urology, Technische Universität Dresden, 01307 Dresden, Germany;
| | - Nikita Polishchuk
- Department of Cell Signaling, Institute of Cell Biology, National Academy of Sciences of Ukraine, 79000 Lviv, Ukraine; (N.P.); (O.S.)
| | - Oleh Stasyk
- Department of Cell Signaling, Institute of Cell Biology, National Academy of Sciences of Ukraine, 79000 Lviv, Ukraine; (N.P.); (O.S.)
| | - Uğur Kahya
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (U.K.); (M.M.W.)
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, 01328 Dresden, Germany
| | - Matthias M. Weigel
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (U.K.); (M.M.W.)
| | - Anna Dubrovska
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (U.K.); (M.M.W.)
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, 01328 Dresden, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, 01309 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany
| |
Collapse
|
5
|
Zhang D, Hua Z, Li Z. The role of glutamate and glutamine metabolism and related transporters in nerve cells. CNS Neurosci Ther 2024; 30:e14617. [PMID: 38358002 PMCID: PMC10867874 DOI: 10.1111/cns.14617] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/15/2023] [Accepted: 01/10/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND Glutamate and glutamine are the most abundant amino acids in the blood and play a crucial role in cell survival in the nervous system. Various transporters found in cell and mitochondrial membranes, such as the solute carriers (SLCs) superfamily, are responsible for maintaining the balance of glutamate and glutamine in the synaptic cleft and within cells. This balance affects the metabolism of glutamate and glutamine as non-essential amino acids. AIMS This review aims to provide an overview of the transporters and enzymes associated with glutamate and glutamine in neuronal cells. DISCUSSION We delve into the function of glutamate and glutamine in the nervous system by discussing the transporters involved in the glutamate-glutamine cycle and the key enzymes responsible for their mutual conversion. Additionally, we highlight the role of glutamate and glutamine as carbon and nitrogen donors, as well as their significance as precursors for the synthesis of reduced glutathione (GSH). CONCLUSION Glutamate and glutamine play a crucial role in the brain due to their special effects. It is essential to focus on understanding glutamate and glutamine metabolism to comprehend the physiological behavior of nerve cells and to treat nervous system disorders and cancer.
Collapse
Affiliation(s)
- Dongyang Zhang
- Department of PediatricsShengjing Hospital of China Medical UniversityShenyangLiaoningChina
- Medical Research Center, Liaoning Key Laboratory of Research and Application of Animal Models for Environment and Metabolic DiseasesShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Zhongyan Hua
- Department of PediatricsShengjing Hospital of China Medical UniversityShenyangLiaoningChina
- Medical Research Center, Liaoning Key Laboratory of Research and Application of Animal Models for Environment and Metabolic DiseasesShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Zhijie Li
- Department of PediatricsShengjing Hospital of China Medical UniversityShenyangLiaoningChina
- Medical Research Center, Liaoning Key Laboratory of Research and Application of Animal Models for Environment and Metabolic DiseasesShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| |
Collapse
|
6
|
Buczkowska J, Szeliga M. Two Faces of Glutaminase GLS2 in Carcinogenesis. Cancers (Basel) 2023; 15:5566. [PMID: 38067269 PMCID: PMC10705333 DOI: 10.3390/cancers15235566] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 04/29/2025] Open
Abstract
In rapidly proliferating cancer cells, glutamine is a major source of energy and building blocks. Increased glutamine uptake and enhanced glutaminolysis are key metabolic features of many cancers. Glutamine is metabolized by glutaminase (GA), which is encoded by two genes: GLS and GLS2. In contrast to isoforms arising from the GLS gene, which clearly act as oncoproteins, the role of GLS2 products in tumorigenesis is far from well understood. While in some cancer types GLS2 is overexpressed and drives cancer development, in some other types it is downregulated and behaves as a tumor suppressor gene. In this review, we describe the essential functions and regulatory mechanisms of human GLS2 and the cellular compartments in which GLS2 has been localized. Furthermore, we present the context-dependent oncogenic and tumor-suppressor properties of GLS2, and delve into the mechanisms underlying these phenomena.
Collapse
Affiliation(s)
| | - Monika Szeliga
- Department of Neurotoxicology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106 Warsaw, Poland;
| |
Collapse
|
7
|
Costa RK, Brancaglion GA, Pinheiro MP, Meira DA, da Silva BN, de V. Negrao CZ, de A. Gonçalves K, Rodrigues CT, Ambrósio AL, Guido RV, Pastre JC, Dias SM. Discovery of aminothiazole derivatives as a chemical scaffold for glutaminase inhibition. RESULTS IN CHEMISTRY 2023. [DOI: 10.1016/j.rechem.2023.100842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023] Open
|
8
|
Lauria G, Curcio R, Lunetti P, Tiziani S, Coppola V, Dolce V, Fiermonte G, Ahmed A. Role of Mitochondrial Transporters on Metabolic Rewiring of Pancreatic Adenocarcinoma: A Comprehensive Review. Cancers (Basel) 2023; 15:411. [PMID: 36672360 PMCID: PMC9857038 DOI: 10.3390/cancers15020411] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
Pancreatic cancer is among the deadliest cancers worldwide and commonly presents as pancreatic ductal adenocarcinoma (PDAC). Metabolic reprogramming is a hallmark of PDAC. Glucose and glutamine metabolism are extensively rewired in order to fulfil both energetic and synthetic demands of this aggressive tumour and maintain favorable redox homeostasis. The mitochondrial pyruvate carrier (MPC), the glutamine carrier (SLC1A5_Var), the glutamate carrier (GC), the aspartate/glutamate carrier (AGC), and the uncoupling protein 2 (UCP2) have all been shown to influence PDAC cell growth and progression. The expression of MPC is downregulated in PDAC and its overexpression reduces cell growth rate, whereas the other four transporters are usually overexpressed and the loss of one or more of them renders PDAC cells unable to grow and proliferate by altering the levels of crucial metabolites such as aspartate. The aim of this review is to comprehensively evaluate the current experimental evidence about the function of these carriers in PDAC metabolic rewiring. Dissecting the precise role of these transporters in the context of the tumour microenvironment is necessary for targeted drug development.
Collapse
Affiliation(s)
- Graziantonio Lauria
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Rosita Curcio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Paola Lunetti
- Department of Bioscience, Biotechnology and Environment, University of Bari, 70125 Bari, Italy
| | - Stefano Tiziani
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX 78712, USA
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX 78723, USA
- Department of Oncology, Dell Medical School, LiveSTRONG Cancer Institutes, The University of Texas at Austin, Austin, TX 78723, USA
| | - Vincenzo Coppola
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Vincenza Dolce
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Giuseppe Fiermonte
- Department of Bioscience, Biotechnology and Environment, University of Bari, 70125 Bari, Italy
| | - Amer Ahmed
- Department of Bioscience, Biotechnology and Environment, University of Bari, 70125 Bari, Italy
| |
Collapse
|
9
|
β-Escin reduces cancer progression in aggressive MDA-MB-231 cells by inhibiting glutamine metabolism through downregulation of c-myc oncogene. Mol Biol Rep 2022; 49:7409-7415. [PMID: 35655054 DOI: 10.1007/s11033-022-07536-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 04/22/2022] [Accepted: 04/27/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND The c-myc oncogene, which causes glutamine dependence in triple negative breast cancers (TNBC), is also the target of one of the signaling pathways affected by β-Escin. METHODS AND RESULTS We sought to determine how c-myc protein affects glutamine metabolism and the proteins, glutamine transporter alanine-serine-cysteine 2 (ASCT2) and glutaminase (GLS1), in β-Escin-treated MDA-MB-231 cells using glutamine uptake and western blot analysis. Cell viability, colony formation, migration and apoptosis were also evaluated in MDA-MB-231 cells in response to β-Escin treatment using MTS, colony forming, wound healing, and Annexin-V assay. We determined that β-Escin decreased glutamine uptake and reduced c-myc and GLS1 protein expressions and increased the expression of ASCT2. In addition, this inhibition of glutamine metabolism decreased cell proliferation, colony formation and migration, and induced apoptosis. CONCLUSIONS In this study, it was suggested that β-Escin inhibits glutamine metabolism via c-myc in MDA-MB-231 cells, and it is thought that as a result of interrupting the energy supply in these cells via c-myc, it results in a decrease in the carcinogenic properties of the cells. Consequently, β-Escin may be promising as a therapeutic agent for glutamine-dependent cancers.
Collapse
|
10
|
Huang Q, Lian C, Dong Y, Zeng H, Liu B, Xu N, He Z, Guo H. SNAP25 Inhibits Glioma Progression by Regulating Synapse Plasticity via GLS-Mediated Glutaminolysis. Front Oncol 2021; 11:698835. [PMID: 34490096 PMCID: PMC8416623 DOI: 10.3389/fonc.2021.698835] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022] Open
Abstract
Background Neuronal activity regulated by synaptic communication exerts an important role in tumorigenesis and progression in brain tumors. Genes for soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) annotated with the function ‘vesicle’ about synaptic connectivity were identified, and synaptosomal-associated protein 25 (SNAP25), one of those proteins, was found to have discrepant expression levels in neuropathies. However, the specific mechanism and prognostic value of SNAP25 during glioma progression remain unclear. Methods Using RNA sequencing data from The Cancer Genome Atlas (TCGA) database, the differential synaptosis-related genes between low grade glioma (LGG) and glioblastoma (GBM) were identified as highly correlated. Cox proportional hazards regression analysis and survival analysis were used to differentiate the outcome of low- and high-risk patients, and the Chinese Glioma Genome Atlas (CGGA) cohort was used for validation of the data set. RT-qPCR, western blot, and immunohistochemistry assays were performed to examine the expression level of SNAP25 in glioma cells and samples. Functional assays were performed to identify the effects of SNAP25 knockdown and overexpression on cell viability, migration, and invasion. Liquid chromatography-high resolution mass spectrometry (LC-MS)-based metabolomics approach was presented for identifying crucial metabolic disturbances in glioma cells. In situ mouse xenograft model was used to investigate the role of SNAP25 in vivo. Then, an immunofluorescence assay of the xenograft tissue was applied to evaluate the expression of the neuronal dendron formation marker-Microtubule Associated Protein 2 (MAP2). Results SNAP25 was decreased in level of expression in glioma tissues and cell lines, and low-level SNAP25 indicated an unfavorable prognosis of glioma patients. SNAP25 inhibited cell proliferation, migration, invasion and fostered glutamine metabolism of glioma cells, exerting a tumor suppressor role. Overexpressed SNAP25 exerted a lower expression level of MAP2, indicating poor neuronal plasticity and connectivity. SNAP25 could regulate glutaminase (GLS)-mediated glutaminolysis, and GLS knockdown could rescue the anti-tumor effect of SNAP25 in glioma cells. Moreover, upregulated SNAP25 also decreased tumor volume and prolonged the overall survival (OS) of the xenograft mouse. Conclusion SNAP25, a tumor suppressor inhibited carcinogenesis of glioma via limiting glutamate metabolism by regulating GLS expression, as well as inhibiting dendritic formation, which could be considered as a novel molecular therapeutic target for glioma.
Collapse
Affiliation(s)
- Qiongzhen Huang
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Department of Neurosurgery, Guangzhou, China
| | - Changlin Lian
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Department of Neurosurgery, Guangzhou, China
| | - Yaoyuan Dong
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Department of Neurosurgery, Guangzhou, China
| | - Huijun Zeng
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Department of Neurosurgery, Guangzhou, China
| | - Boyang Liu
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Department of Neurosurgery, Guangzhou, China
| | - Ningbo Xu
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Department of Neurosurgery, Guangzhou, China
| | - Zhenyan He
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Department of Neurosurgery, Guangzhou, China
| | - Hongbo Guo
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Department of Neurosurgery, Guangzhou, China
| |
Collapse
|
11
|
Ferreira IM, Quesñay JEN, Bastos AC, Rodrigues CT, Vollmar M, Krojer T, Strain-Damerell C, Burgess-Brown NA, von Delft F, Yue WW, Dias SM, Ambrosio AL. Structure and activation mechanism of the human liver-type glutaminase GLS2. Biochimie 2021; 185:96-104. [PMID: 33746066 PMCID: PMC8130709 DOI: 10.1016/j.biochi.2021.03.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 02/23/2021] [Accepted: 03/14/2021] [Indexed: 11/24/2022]
Abstract
Cancer cells exhibit an altered metabolic phenotype, consuming higher levels of the amino acid glutamine. This metabolic reprogramming depends on increased mitochondrial glutaminase activity to convert glutamine to glutamate, an essential precursor for bioenergetic and biosynthetic processes in cells. Mammals encode the kidney-type (GLS) and liver-type (GLS2) glutaminase isozymes. GLS is overexpressed in cancer and associated with enhanced malignancy. On the other hand, GLS2 is either a tumor suppressor or an oncogene, depending on the tumor type. The GLS structure and activation mechanism are well known, while the structural determinants for GLS2 activation remain elusive. Here, we describe the structure of the human glutaminase domain of GLS2, followed by the functional characterization of the residues critical for its activity. Increasing concentrations of GLS2 lead to tetramer stabilization, a process enhanced by phosphate. In GLS2, the so-called “lid loop” is in a rigid open conformation, which may be related to its higher affinity for phosphate and lower affinity for glutamine; hence, it has lower glutaminase activity than GLS. The lower affinity of GLS2 for glutamine is also related to its less electropositive catalytic site than GLS, as indicated by a Thr225Lys substitution within the catalytic site decreasing the GLS2 glutamine concentration corresponding to half-maximal velocity (K0.5). Finally, we show that the Lys253Ala substitution (corresponding to the Lys320Ala in the GLS “activation” loop, formerly known as the “gating” loop) renders a highly active protein in stable tetrameric form. We conclude that the “activation” loop, a known target for GLS inhibition, may also be a drug target for GLS2. A positive cooperative mechanism of activation is demonstrated for the liver-type glutaminase. The novel crystal structure for the glutaminase domain of human GLS2 is presented. Site-directed mutagenesis suggest the activation loop as a hotspot for inhibitor design.
Collapse
Affiliation(s)
- Igor M Ferreira
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, Sao Paulo, Zip Code, 13083-970, Brazil
| | - José Edwin N Quesñay
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, Sao Paulo, Zip Code, 13083-970, Brazil; Sao Carlos Institute of Physics, University of Sao Paulo, Sao Carlos, SP, Zip Code, 13563-120, Brazil
| | - Alliny Cs Bastos
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, Sao Paulo, Zip Code, 13083-970, Brazil; Graduate Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Camila T Rodrigues
- Sao Carlos Institute of Physics, University of Sao Paulo, Sao Carlos, SP, Zip Code, 13563-120, Brazil
| | - Melanie Vollmar
- Structural Genomics Consortium, University of Oxford, Old Road Campus, Roosevelt Drive, Headington, OX3 7DQ, UK
| | - Tobias Krojer
- Structural Genomics Consortium, University of Oxford, Old Road Campus, Roosevelt Drive, Headington, OX3 7DQ, UK
| | - Claire Strain-Damerell
- Structural Genomics Consortium, University of Oxford, Old Road Campus, Roosevelt Drive, Headington, OX3 7DQ, UK; Diamond Light Source Ltd., The Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0FA, UK
| | - Nicola A Burgess-Brown
- Structural Genomics Consortium, University of Oxford, Old Road Campus, Roosevelt Drive, Headington, OX3 7DQ, UK
| | - Frank von Delft
- Structural Genomics Consortium, University of Oxford, Old Road Campus, Roosevelt Drive, Headington, OX3 7DQ, UK; Diamond Light Source Ltd., The Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0FA, UK; Department of Biochemistry, University of Johannesburg, Auckland Park, 2006, South Africa; Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, OX11 0FA, UK
| | - Wyatt W Yue
- Structural Genomics Consortium, University of Oxford, Old Road Campus, Roosevelt Drive, Headington, OX3 7DQ, UK.
| | - Sandra Mg Dias
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, Sao Paulo, Zip Code, 13083-970, Brazil.
| | - Andre Lb Ambrosio
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, Sao Paulo, Zip Code, 13083-970, Brazil; Sao Carlos Institute of Physics, University of Sao Paulo, Sao Carlos, SP, Zip Code, 13563-120, Brazil.
| |
Collapse
|
12
|
Ding L, Xu X, Li C, Wang Y, Xia X, Zheng JC. Glutaminase in microglia: A novel regulator of neuroinflammation. Brain Behav Immun 2021; 92:139-156. [PMID: 33278560 DOI: 10.1016/j.bbi.2020.11.038] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/11/2020] [Accepted: 11/28/2020] [Indexed: 12/15/2022] Open
Abstract
Neuroinflammation is the inflammatory responses that are involved in the pathogenesis of most neurological disorders. Glutaminase (GLS) is the enzyme that catalyzes the hydrolysis of glutamine to produce glutamate. Besides its well-known role in cellular metabolism and excitatory neurotransmission, GLS has recently been increasingly noticed to be up-regulated in activated microglia under pathological conditions. Furthermore, GLS overexpression induces microglial activation, extracellular vesicle secretion, and neuroinflammatory microenvironment formation, which, are compromised by GLS inhibitors in vitro and in vivo. These results indicate that GLS has more complicated implications in brain disease etiology than what are previously known. In this review, we introduce GLS isoforms, expression patterns in the body and the brain, and expression/activities regulation. Next, we discuss the metabolic and neurotransmission functions of GLS. Afterwards, we summarize recent findings of GLS-mediated microglial activation and pro-inflammatory extracellular vesicle secretion, which, in turns, induces neuroinflammation. Lastly, we provide a comprehensive discussion for the involvement of microglial GLS in the pathogenesis of various neurological disorders, indicating microglial GLS as a promising target to treat these diseases.
Collapse
Affiliation(s)
- Lu Ding
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai 200072, China
| | - Xiaonan Xu
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai 200072, China
| | - Congcong Li
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai 200072, China
| | - Yi Wang
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai 200072, China; Collaborative Innovation Center for Brain Science, Tongji University, Shanghai 200092, China; Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital affiliated to Tongji University School of Medicine, Shanghai 200434, China.
| | - Xiaohuan Xia
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai 200072, China; Collaborative Innovation Center for Brain Science, Tongji University, Shanghai 200092, China; Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital affiliated to Tongji University School of Medicine, Shanghai 200434, China.
| | - Jialin C Zheng
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai 200072, China; Collaborative Innovation Center for Brain Science, Tongji University, Shanghai 200092, China; Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital affiliated to Tongji University School of Medicine, Shanghai 200434, China; Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930, USA.
| |
Collapse
|
13
|
Matés JM, Campos-Sandoval JA, de Los Santos-Jiménez J, Segura JA, Alonso FJ, Márquez J. Metabolic Reprogramming of Cancer by Chemicals that Target Glutaminase Isoenzymes. Curr Med Chem 2020; 27:5317-5339. [PMID: 31038055 DOI: 10.2174/0929867326666190416165004] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 03/19/2019] [Accepted: 03/31/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Metabolic reprogramming of tumours is a hallmark of cancer. Among the changes in the metabolic network of cancer cells, glutaminolysis is a key reaction altered in neoplasms. Glutaminase proteins control the first step in glutamine metabolism and their expression correlates with malignancy and growth rate of a great variety of cancers. The two types of glutaminase isoenzymes, GLS and GLS2, differ in their expression patterns and functional roles: GLS has oncogenic properties and GLS2 has been described as a tumour suppressor factor. RESULTS We have focused on glutaminase connections with key oncogenes and tumour suppressor genes. Targeting glutaminase isoenzymes includes different strategies aimed at deactivating the rewiring of cancer metabolism. In addition, we found a long list of metabolic enzymes, transcription factors and signalling pathways dealing with glutaminase. On the other hand, a number of chemicals have been described as isoenzyme-specific inhibitors of GLS and/or GLS2 isoforms. These molecules are being characterized as synergic and therapeutic agents in many types of tumours. CONCLUSION This review states the metabolic pathways that are rewired in cancer, the roles of glutaminase isoforms in cancer, as well as the metabolic circuits regulated by glutaminases. We also show the plethora of anticancer drugs that specifically inhibit glutaminase isoenzymes for treating several sets of cancer.
Collapse
Affiliation(s)
- José M Matés
- Instituto de Investigacion Biomedica de Malaga (IBIMA), Department of Molecular Biology and Biochemistry, Canceromics Lab, Faculty of Sciences, Campus de Teatinos, University of Malaga, 29071 Malaga, Spain
| | - José A Campos-Sandoval
- Instituto de Investigacion Biomedica de Malaga (IBIMA), Department of Molecular Biology and Biochemistry, Canceromics Lab, Faculty of Sciences, Campus de Teatinos, University of Malaga, 29071 Malaga, Spain
| | - Juan de Los Santos-Jiménez
- Instituto de Investigacion Biomedica de Malaga (IBIMA), Department of Molecular Biology and Biochemistry, Canceromics Lab, Faculty of Sciences, Campus de Teatinos, University of Malaga, 29071 Malaga, Spain
| | - Juan A Segura
- Instituto de Investigacion Biomedica de Malaga (IBIMA), Department of Molecular Biology and Biochemistry, Canceromics Lab, Faculty of Sciences, Campus de Teatinos, University of Malaga, 29071 Malaga, Spain
| | - Francisco J Alonso
- Instituto de Investigacion Biomedica de Malaga (IBIMA), Department of Molecular Biology and Biochemistry, Canceromics Lab, Faculty of Sciences, Campus de Teatinos, University of Malaga, 29071 Malaga, Spain
| | - Javier Márquez
- Instituto de Investigacion Biomedica de Malaga (IBIMA), Department of Molecular Biology and Biochemistry, Canceromics Lab, Faculty of Sciences, Campus de Teatinos, University of Malaga, 29071 Malaga, Spain
| |
Collapse
|
14
|
Nuclear Translocation of Glutaminase GLS2 in Human Cancer Cells Associates with Proliferation Arrest and Differentiation. Sci Rep 2020; 10:2259. [PMID: 32042057 PMCID: PMC7010782 DOI: 10.1038/s41598-020-58264-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 01/08/2020] [Indexed: 11/08/2022] Open
Abstract
Glutaminase (GA) catalyzes the first step in mitochondrial glutaminolysis playing a key role in cancer metabolic reprogramming. Humans express two types of GA isoforms: GLS and GLS2. GLS isozymes have been consistently related to cell proliferation, but the role of GLS2 in cancer remains poorly understood. GLS2 is repressed in many tumor cells and a better understanding of its function in tumorigenesis may further the development of new therapeutic approaches. We analyzed GLS2 expression in HCC, GBM and neuroblastoma cells, as well as in monkey COS-7 cells. We studied GLS2 expression after induction of differentiation with phorbol ester (PMA) and transduction with the full-length cDNA of GLS2. In parallel, we investigated cell cycle progression and levels of p53, p21 and c-Myc proteins. Using the baculovirus system, human GLS2 protein was overexpressed, purified and analyzed for posttranslational modifications employing a proteomics LC-MS/MS platform. We have demonstrated a dual targeting of GLS2 in human cancer cells. Immunocytochemistry and subcellular fractionation gave consistent results demonstrating nuclear and mitochondrial locations, with the latter being predominant. Nuclear targeting was confirmed in cancer cells overexpressing c-Myc- and GFP-tagged GLS2 proteins. We assessed the subnuclear location finding a widespread distribution of GLS2 in the nucleoplasm without clear overlapping with specific nuclear substructures. GLS2 expression and nuclear accrual notably increased by treatment of SH-SY5Y cells with PMA and it correlated with cell cycle arrest at G2/M, upregulation of tumor suppressor p53 and p21 protein. A similar response was obtained by overexpression of GLS2 in T98G glioma cells, including downregulation of oncogene c-Myc. Furthermore, human GLS2 was identified as being hypusinated by MS analysis, a posttranslational modification which may be relevant for its nuclear targeting and/or function. Our studies provide evidence for a tumor suppressor role of GLS2 in certain types of cancer. The data imply that GLS2 can be regarded as a highly mobile and multilocalizing protein translocated to both mitochondria and nuclei. Upregulation of GLS2 in cancer cells induced an antiproliferative response with cell cycle arrest at the G2/M phase.
Collapse
|
15
|
Obara-Michlewska M, Szeliga M. Targeting Glutamine Addiction in Gliomas. Cancers (Basel) 2020; 12:cancers12020310. [PMID: 32013066 PMCID: PMC7072559 DOI: 10.3390/cancers12020310] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/16/2020] [Accepted: 01/27/2020] [Indexed: 12/12/2022] Open
Abstract
The most common malignant brain tumors are those of astrocytic origin, gliomas, with the most aggressive glioblastoma (WHO grade IV) among them. Despite efforts, medicine has not made progress in terms of the prognosis and life expectancy of glioma patients. Behind the malignant phenotype of gliomas lies multiple genetic mutations leading to reprogramming of their metabolism, which gives those highly proliferating cells an advantage over healthy ones. The so-called glutamine addiction is a metabolic adaptation that supplements oxidative glycolysis in order to secure neoplastic cells with nutrients and energy in unfavorable conditions of hypoxia. The present review aims at presenting the research and clinical attempts targeting the different metabolic pathways involved in glutamine metabolism in gliomas. A brief description of the biochemistry of glutamine transport, synthesis, and glutaminolysis, etc. will forego a detailed comparison of the therapeutic strategies undertaken to inhibit glutamine utilization by gliomas.
Collapse
|
16
|
Dias MM, Adamoski D, Dos Reis LM, Ascenção CFR, de Oliveira KRS, Mafra ACP, da Silva Bastos AC, Quintero M, de G Cassago C, Ferreira IM, Fidelis CHV, Rocco SA, Bajgelman MC, Stine Z, Berindan-Neagoe I, Calin GA, Ambrosio ALB, Dias SMG. GLS2 is protumorigenic in breast cancers. Oncogene 2019; 39:690-702. [PMID: 31541193 DOI: 10.1038/s41388-019-1007-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 08/21/2019] [Accepted: 08/26/2019] [Indexed: 12/16/2022]
Abstract
Many types of cancers have a well-established dependence on glutamine metabolism to support survival and growth, a process linked to glutaminase 1 (GLS) isoforms. Conversely, GLS2 variants often have tumor-suppressing activity. Triple-negative (TN) breast cancer (testing negative for estrogen, progesterone, and Her2 receptors) has elevated GLS protein levels and reportedly depends on exogenous glutamine and GLS activity for survival. Despite having high GLS levels, we verified that several breast cancer cells (including TN cells) express endogenous GLS2, defying its role as a bona fide tumor suppressor. Moreover, ectopic GLS2 expression rescued cell proliferation, TCA anaplerosis, redox balance, and mitochondrial function after GLS inhibition by the small molecule currently in clinical trials CB-839 or GLS knockdown of GLS-dependent cell lines. In several cell lines, GLS2 knockdown decreased cell proliferation and glutamine-linked metabolic phenotypes. Strikingly, long-term treatment of TN cells with another GLS-exclusive inhibitor bis-2'-(5-phenylacetamide-1,3,4-thiadiazol-2-yl)ethyl sulfide (BPTES) selected for a drug-resistant population with increased endogenous GLS2 and restored proliferative capacity. GLS2 was linked to enhanced in vitro cell migration and invasion, mesenchymal markers (through the ERK-ZEB1-vimentin axis under certain conditions) and in vivo lung metastasis. Of concern, GLS2 amplification or overexpression is linked to an overall, disease-free and distant metastasis-free worse survival prognosis in breast cancer. Altogether, these data establish an unforeseen role of GLS2 in sustaining tumor proliferation and underlying metastasis in breast cancer and provide an initial framework for exploring GLS2 as a novel therapeutic target.
Collapse
Affiliation(s)
- Marilia M Dias
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Sao Paulo, 13083-970, Brazil.,Graduate Program in Genetics and Molecular Biology, Institute of Biology University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| | - Douglas Adamoski
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Sao Paulo, 13083-970, Brazil.,Graduate Program in Genetics and Molecular Biology, Institute of Biology University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| | - Larissa M Dos Reis
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Sao Paulo, 13083-970, Brazil.,Graduate Program in Genetics and Molecular Biology, Institute of Biology University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| | - Carolline F R Ascenção
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Sao Paulo, 13083-970, Brazil.,Graduate Program in Genetics and Molecular Biology, Institute of Biology University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| | - Krishina R S de Oliveira
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Sao Paulo, 13083-970, Brazil.,Graduate Program in Genetics and Molecular Biology, Institute of Biology University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| | - Ana Carolina Paschoalini Mafra
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Sao Paulo, 13083-970, Brazil.,Graduate Program in Genetics and Molecular Biology, Institute of Biology University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| | - Alliny Cristiny da Silva Bastos
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Sao Paulo, 13083-970, Brazil.,Graduate Program in Genetics and Molecular Biology, Institute of Biology University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| | - Melissa Quintero
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Sao Paulo, 13083-970, Brazil
| | - Carolina de G Cassago
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Sao Paulo, 13083-970, Brazil
| | - Igor M Ferreira
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Sao Paulo, 13083-970, Brazil
| | - Carlos H V Fidelis
- ThoMSon Mass Spectrometry Laboratory, Institute of Chemistry, University of Campinas, Campinas, Sao Paulo, 13083-970, Brazil
| | - Silvana A Rocco
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Sao Paulo, 13083-970, Brazil
| | - Marcio Chaim Bajgelman
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Sao Paulo, 13083-970, Brazil
| | - Zachary Stine
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, 19104, USA
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, University of Medicine and Pharmacy "Iuliu-Hatieganu", 400337, Cluj-Napoca, Romania.,MedFuture Research Center for Advanced Medicine, University of Medicine and Pharmacy "Iuliu-Hatieganu", 400349, Cluj-Napoca, Romania.,Department of Functional Genomics and Experimental Pathology, The Oncology Institute "Prof. Dr. Ion Chiricuţă", 400015, Cluj-Napoca, Romania
| | - George A Calin
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd. Unit 1950, Houston, TX, 77030, USA.,Center for RNA Inference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd. Unit 1950, Houston, TX, 77030, USA
| | - Andre Luis Berteli Ambrosio
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Sao Paulo, 13083-970, Brazil. .,Sao Carlos Institute of Physics (IFSC), University of Sao Paulo (USP), Sao Carlos, Sao Paulo, 13563-120, Brazil.
| | - Sandra Martha Gomes Dias
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Sao Paulo, 13083-970, Brazil.
| |
Collapse
|
17
|
Matés JM, Di Paola FJ, Campos-Sandoval JA, Mazurek S, Márquez J. Therapeutic targeting of glutaminolysis as an essential strategy to combat cancer. Semin Cell Dev Biol 2019; 98:34-43. [PMID: 31100352 DOI: 10.1016/j.semcdb.2019.05.012] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/11/2019] [Accepted: 05/13/2019] [Indexed: 01/08/2023]
Abstract
Metabolic reprogramming in cancer targets glutamine metabolism as a key mechanism to provide energy, biosynthetic precursors and redox requirements to allow the massive proliferation of tumor cells. Glutamine is also a signaling molecule involved in essential pathways regulated by oncogenes and tumor suppressor factors. Glutaminase isoenzymes are critical proteins to control glutaminolysis, a key metabolic pathway for cell proliferation and survival that directs neoplasms' fate. Adaptive glutamine metabolism can be altered by different metabolic therapies, including the use of specific allosteric inhibitors of glutaminase that can evoke synergistic effects for the therapy of cancer patients. We also review other clinical applications of in vivo assessment of glutaminolysis by metabolomic approaches, including diagnosis and monitoring of cancer.
Collapse
Affiliation(s)
- José M Matés
- Instituto de Investigación Biomédica de Málaga (IBIMA), Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Málaga, E-29071 Málaga, Spain
| | - Floriana J Di Paola
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University of Giessen, D-35392 Giessen, Germany
| | - José A Campos-Sandoval
- Instituto de Investigación Biomédica de Málaga (IBIMA), Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Málaga, E-29071 Málaga, Spain
| | - Sybille Mazurek
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University of Giessen, D-35392 Giessen, Germany
| | - Javier Márquez
- Instituto de Investigación Biomédica de Málaga (IBIMA), Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Málaga, E-29071 Málaga, Spain.
| |
Collapse
|
18
|
Transfection with GLS2 Glutaminase (GAB) Sensitizes Human Glioblastoma Cell Lines to Oxidative Stress by a Common Mechanism Involving Suppression of the PI3K/AKT Pathway. Cancers (Basel) 2019; 11:cancers11010115. [PMID: 30669455 PMCID: PMC6356507 DOI: 10.3390/cancers11010115] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 01/16/2019] [Accepted: 01/17/2019] [Indexed: 01/23/2023] Open
Abstract
GLS-encoded glutaminase promotes tumorigenesis, while GLS2-encoded glutaminase displays tumor-suppressive properties. In glioblastoma (GBM), the most aggressive brain tumor, GLS is highly expressed and in most cases GLS2 is silenced. Previously, it was shown that transfection with a sequence encoding GAB, the main GLS2 isoform, decreased the survival, growth, and ability to migrate of human GBM cells T98G and increased their sensitivity towards an alkylating agent temozolomide (TMZ) and oxidative stress compared to the controls, by a not well-defined mechanism. In this study we report that GAB transfection inhibits growth and increases susceptibility towards TMZ and H2O2-mediated oxidative stress of two other GBM cell lines, U87MG and LN229. We also show that in GAB-transfected cells treated with H2O2, the PI3K/AKT pathway is less induced compared to the pcDNA-transfected counterparts and that pretreatment with PDGF-BB, an activator of AKT, protects GAB-transfected cells from death caused by the H2O2 treatment. In conclusion, our results show that (i) GAB suppresses the malignant phenotype of the GBM cells of different tumorigenic potentials and genetic backgrounds and (ii) the GAB-mediated increase of sensitivity to oxidative stress is causally related to the inhibition of the PI3K/AKT pathway. The upregulation of the GLS2 expression and the inhibition of the PI3K/AKT pathway may become a novel combined therapeutic strategy for anti-glioma preclinical investigations.
Collapse
|
19
|
Matés JM, Campos-Sandoval JA, Márquez J. Glutaminase isoenzymes in the metabolic therapy of cancer. Biochim Biophys Acta Rev Cancer 2018; 1870:158-164. [DOI: 10.1016/j.bbcan.2018.07.007] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/14/2018] [Accepted: 07/15/2018] [Indexed: 12/11/2022]
|
20
|
de Guzzi Cassago CA, Dias MM, Pinheiro MP, Pasquali CC, Bastos ACS, Islam Z, Consonni SR, de Oliveira JF, Gomes EM, Ascenção CFR, Honorato R, Pauletti BA, Indolfo NDC, Filho HVR, de Oliveira PSL, Figueira ACM, Paes Leme AF, Ambrosio ALB, Dias SMG. Glutaminase Affects the Transcriptional Activity of Peroxisome Proliferator-Activated Receptor γ (PPARγ) via Direct Interaction. Biochemistry 2018; 57:6293-6307. [PMID: 30295466 DOI: 10.1021/acs.biochem.8b00773] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Phosphate-activated glutaminases catalyze the deamidation of glutamine to glutamate and play key roles in several physiological and pathological processes. In humans, GLS encodes two multidomain splicing isoforms: KGA and GAC. In both isoforms, the canonical glutaminase domain is flanked by an N-terminal region that is folded into an EF-hand-like four-helix bundle. However, the splicing event replaces a well-structured three-repeat ankyrin domain in KGA with a shorter, unordered C-terminal stretch in GAC. The multidomain architecture, which contains putative protein-protein binding motifs, has led to speculation that glutaminases are involved in cellular processes other than glutamine metabolism; in fact, some proteins have been identified as binding partners of KGA and the isoforms of its paralogue gene, GLS2. Here, a yeast two-hybrid assay identified nuclear receptor peroxisome proliferator-activated receptor γ (PPARγ) as a new binding partner of the glutaminase. We show that KGA and GAC directly bind PPARγ with a low-micromolar dissociation constant; the interaction involves the N-terminal and catalytic domains of glutaminases as well as the ligand-binding domain of the nuclear receptor. The interaction occurs within the nucleus, and by sequestering PPARγ from its responsive element DR1, the glutaminases decreased nuclear receptor activity as assessed by a luciferase reporter assay. Altogether, our findings reveal an unexpected glutaminase-binding partner and, for the first time, directly link mitochondrial glutaminases to an unanticipated role in gene regulation.
Collapse
Affiliation(s)
- Carolina Aparecida de Guzzi Cassago
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Marília Meira Dias
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Matheus Pinto Pinheiro
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Camila Cristina Pasquali
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Alliny Cristiny Silva Bastos
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil.,Graduate Program in Genetics and Molecular Biology, Institute of Biology , University of Campinas (UNICAMP) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Zeyaul Islam
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Sílvio Roberto Consonni
- Department of Biochemistry and Tissue Biology , University of Campinas , Campinas , Sao Paulo 13083-872 , Brazil
| | - Juliana Ferreira de Oliveira
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Emerson Machi Gomes
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Carolline Fernanda Rodrigues Ascenção
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil.,Graduate Program in Genetics and Molecular Biology, Institute of Biology , University of Campinas (UNICAMP) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Rodrigo Honorato
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Bianca Alves Pauletti
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Nathalia de Carvalho Indolfo
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Helder Veras Ribeiro Filho
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Paulo Sergio Lopes de Oliveira
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Ana Carolina Migliorini Figueira
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Adriana Franco Paes Leme
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Andre Luis Berteli Ambrosio
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| | - Sandra Martha Gomes Dias
- Brazilian Biosciences National Laboratory (LNBio) , Brazilian Center for Research in Energy and Materials (CNPEM) , Campinas , Sao Paulo 13083-970 , Brazil
| |
Collapse
|
21
|
Ahn J, Kim DH, Suh Y, Lee JW, Lee K. Adipose-specific expression of mouse Rbp7 gene and its developmental and metabolic changes. Gene 2018; 670:38-45. [DOI: 10.1016/j.gene.2018.05.101] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/07/2018] [Accepted: 05/23/2018] [Indexed: 11/16/2022]
|
22
|
Sadam H, Pihlak A, Kivil A, Pihelgas S, Jaago M, Adler P, Vilo J, Vapalahti O, Neuman T, Lindholm D, Partinen M, Vaheri A, Palm K. Prostaglandin D2 Receptor DP1 Antibodies Predict Vaccine-induced and Spontaneous Narcolepsy Type 1: Large-scale Study of Antibody Profiling. EBioMedicine 2018; 29:47-59. [PMID: 29449194 PMCID: PMC5925455 DOI: 10.1016/j.ebiom.2018.01.043] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 01/23/2018] [Accepted: 01/31/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Neuropathological findings support an autoimmune etiology as an underlying factor for loss of orexin-producing neurons in spontaneous narcolepsy type 1 (narcolepsy with cataplexy; sNT1) as well as in Pandemrix influenza vaccine-induced narcolepsy type 1 (Pdmx-NT1). The precise molecular target or antigens for the immune response have, however, remained elusive. METHODS Here we have performed a comprehensive antigenic repertoire analysis of sera using the next-generation phage display method - mimotope variation analysis (MVA). Samples from 64 children and adolescents were analyzed: 10 with Pdmx-NT1, 6 with sNT1, 16 Pandemrix-vaccinated, 16 H1N1 infected, and 16 unvaccinated healthy individuals. The diagnosis of NT1 was defined by the American Academy of Sleep Medicine international criteria of sleep disorders v3. FINDINGS Our data showed that although the immunoprofiles toward vaccination were generally similar in study groups, there were also striking differences in immunoprofiles between sNT1 and Pdmx-NT1 groups as compared with controls. Prominent immune response was observed to a peptide epitope derived from prostaglandin D2 receptor (DP1), as well as peptides homologous to B cell lymphoma 6 protein. Further validation confirmed that these can act as true antigenic targets in discriminating NT1 diseased along with a novel epitope of hemagglutinin of H1N1 to delineate exposure to H1N1. INTERPRETATION We propose that DP1 is a novel molecular target of autoimmune response and presents a potential diagnostic biomarker for NT1. DP1 is involved in the regulation of non-rapid eye movement (NREM) sleep and thus alterations in its functions could contribute to the disturbed sleep regulation in NT1 that warrants further studies. Together our results also show that MVA is a helpful method for finding novel peptide antigens to classify human autoimmune diseases, possibly facilitating the design of better therapies.
Collapse
Affiliation(s)
- Helle Sadam
- Protobios Llc, Mäealuse 4, 12618 Tallinn, Estonia; Department of Gene Technology, Tallinn University of Technology, Akadeemia Tee 15, 12618 Tallinn, Estonia
| | - Arno Pihlak
- Protobios Llc, Mäealuse 4, 12618 Tallinn, Estonia; Department of Gene Technology, Tallinn University of Technology, Akadeemia Tee 15, 12618 Tallinn, Estonia
| | - Anri Kivil
- Protobios Llc, Mäealuse 4, 12618 Tallinn, Estonia
| | | | | | - Priit Adler
- Institute of Computer Science, University of Tartu, Liivi 2-314, 50409 Tartu, Estonia; Quretec LLC, Ülikooli 6a, 51003 Tartu, Estonia
| | - Jaak Vilo
- Institute of Computer Science, University of Tartu, Liivi 2-314, 50409 Tartu, Estonia; Quretec LLC, Ülikooli 6a, 51003 Tartu, Estonia
| | - Olli Vapalahti
- Department of Virology, Medicum, Haartmaninkatu 3, 00014 University of Helsinki, Finland; Department of Veterinary Biosciences, University of Helsinki, Agnes Sjöbergin Katu 2, 00014 University of Helsinki, Finland; Virology and Immunology, HUSLAB, Helsinki University Hospital, 00290 Helsinki, Finland
| | - Toomas Neuman
- Protobios Llc, Mäealuse 4, 12618 Tallinn, Estonia; IPDx Immunoprofiling Diagnostics GmbH, Deutscher Platz 5e, 04103 Leipzig, Germany
| | - Dan Lindholm
- Department of Biochemistry and Developmental Biology, Medicum, Haartmaninkatu 8, 00014 University of Helsinki, Finland; Minerva Foundation Medical Research Institute, Tukholmankatu 8, 00290 Helsinki, Finland
| | - Markku Partinen
- Finnish Narcolepsy Research Center, Helsinki Sleep Clinic, Vitalmed Research Center, Valimotie 21, 00380, Helsinki, Finland
| | - Antti Vaheri
- Department of Virology, Medicum, Haartmaninkatu 3, 00014 University of Helsinki, Finland
| | - Kaia Palm
- Protobios Llc, Mäealuse 4, 12618 Tallinn, Estonia; Department of Gene Technology, Tallinn University of Technology, Akadeemia Tee 15, 12618 Tallinn, Estonia.
| |
Collapse
|
23
|
Fazzari J, Linher-Melville K, Singh G. Tumour-Derived Glutamate: Linking Aberrant Cancer Cell Metabolism to Peripheral Sensory Pain Pathways. Curr Neuropharmacol 2018; 15:620-636. [PMID: 27157265 PMCID: PMC5543678 DOI: 10.2174/1570159x14666160509123042] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/16/2016] [Accepted: 04/17/2016] [Indexed: 01/22/2023] Open
Abstract
Background Chronic pain is a major symptom that develops in cancer patients, most commonly emerging during advanced stages of the disease. The nature of cancer-induced pain is complex, and the efficacy of current therapeutic interventions is restricted by the dose-limiting side-effects that accompany common centrally targeted analgesics. Methods This review focuses on how up-regulated glutamate production and export by the tumour converge at peripheral afferent nerve terminals to transmit nociceptive signals through the transient receptor cation channel, TRPV1, thereby initiating central sensitization in response to peripheral disease-mediated stimuli. Results Cancer cells undergo numerous metabolic changes that include increased glutamine catabolism and over-expression of enzymes involved in glutaminolysis, including glutaminase. This mitochondrial enzyme mediates glutaminolysis, producing large pools of intracellular glutamate. Up-regulation of the plasma membrane cystine/glutamate antiporter, system xc-, promotes aberrant glutamate release from cancer cells. Increased levels of extracellular glutamate have been associated with the progression of cancer-induced pain and we discuss how this can be mediated by activation of TRPV1. Conclusion With a growing population of patients receiving inadequate treatment for intractable pain, new targets need to be considered to better address this largely unmet clinical need for improving their quality of life. A better understanding of the mechanisms that underlie the unique qualities of cancer pain will help to identify novel targets that are able to limit the initiation of pain from a peripheral source–the tumour.
Collapse
Affiliation(s)
| | | | - Gurmit Singh
- Department of Pathology and Molecular Medicine; Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON. Canada
| |
Collapse
|
24
|
Peñalver A, Campos-Sandoval JA, Blanco E, Cardona C, Castilla L, Martín-Rufián M, Estivill-Torrús G, Sánchez-Varo R, Alonso FJ, Pérez-Hernández M, Colado MI, Gutiérrez A, de Fonseca FR, Márquez J. Glutaminase and MMP-9 Downregulation in Cortex and Hippocampus of LPA 1 Receptor Null Mice Correlate with Altered Dendritic Spine Plasticity. Front Mol Neurosci 2017; 10:278. [PMID: 28928633 PMCID: PMC5591874 DOI: 10.3389/fnmol.2017.00278] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 08/17/2017] [Indexed: 12/03/2022] Open
Abstract
Lysophosphatidic acid (LPA) is an extracellular lipid mediator that regulates nervous system development and functions acting through G protein-coupled receptors (GPCRs). Here we explore the crosstalk between LPA1 receptor and glutamatergic transmission by examining expression of glutaminase (GA) isoforms in different brain areas isolated from wild-type (WT) and KOLPA1 mice. Silencing of LPA1 receptor induced a severe down-regulation of Gls-encoded long glutaminase protein variant (KGA) (glutaminase gene encoding the kidney-type isoforms, GLS) protein expression in several brain regions, particularly in brain cortex and hippocampus. Immunohistochemical assessment of protein levels for the second type of glutaminase (GA) isoform, glutaminase gene encoding the liver-type isoforms (GLS2), did not detect substantial differences with regard to WT animals. The regional mRNA levels of GLS were determined by real time RT-PCR and did not show significant variations, except for prefrontal and motor cortex values which clearly diminished in KO mice. Total GA activity was also significantly reduced in prefrontal and motor cortex, but remained essentially unchanged in the hippocampus and rest of brain regions examined, suggesting activation of genetic compensatory mechanisms and/or post-translational modifications to compensate for KGA protein deficit. Remarkably, Golgi staining of hippocampal regions showed an altered morphology of glutamatergic pyramidal cells dendritic spines towards a less mature filopodia-like phenotype, as compared with WT littermates. This structural change correlated with a strong decrease of active matrix-metalloproteinase (MMP) 9 in cerebral cortex and hippocampus of KOLPA1 mice. Taken together, these results demonstrate that LPA signaling through LPA1 influence expression of the main isoenzyme of glutamate biosynthesis with strong repercussions on dendritic spines maturation, which may partially explain the cognitive and learning defects previously reported for this colony of KOLPA1 mice.
Collapse
Affiliation(s)
- Ana Peñalver
- Canceromics Laboratory, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Campus de TeatinosMálaga, Spain
| | - José A Campos-Sandoval
- Canceromics Laboratory, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Campus de TeatinosMálaga, Spain
| | - Eduardo Blanco
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de MálagaMálaga, Spain
| | - Carolina Cardona
- Canceromics Laboratory, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Campus de TeatinosMálaga, Spain
| | - Laura Castilla
- Canceromics Laboratory, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Campus de TeatinosMálaga, Spain
| | - Mercedes Martín-Rufián
- Canceromics Laboratory, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Campus de TeatinosMálaga, Spain
| | - Guillermo Estivill-Torrús
- Unidad de Gestión Clínica de Neurociencias, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de MálagaMálaga, Spain
| | - Raquel Sánchez-Varo
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Instituto de Investigación Biomédica de Málaga (IBIMA), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universidad de Málaga, Campus de TeatinosMálaga, Spain
| | - Francisco J Alonso
- Canceromics Laboratory, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Campus de TeatinosMálaga, Spain
| | - Mercedes Pérez-Hernández
- Departamento de Farmacología, Facultad de Medicina, Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de OctubreMadrid, Spain
| | - María I Colado
- Departamento de Farmacología, Facultad de Medicina, Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de OctubreMadrid, Spain
| | - Antonia Gutiérrez
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Instituto de Investigación Biomédica de Málaga (IBIMA), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universidad de Málaga, Campus de TeatinosMálaga, Spain
| | - Fernando Rodríguez de Fonseca
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de MálagaMálaga, Spain
| | - Javier Márquez
- Canceromics Laboratory, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Campus de TeatinosMálaga, Spain
| |
Collapse
|
25
|
Pasquali CC, Islam Z, Adamoski D, Ferreira IM, Righeto RD, Bettini J, Portugal RV, Yue WWY, Gonzalez A, Dias SMG, Ambrosio ALB. The origin and evolution of human glutaminases and their atypical C-terminal ankyrin repeats. J Biol Chem 2017; 292:11572-11585. [PMID: 28526749 DOI: 10.1074/jbc.m117.787291] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 05/11/2017] [Indexed: 12/14/2022] Open
Abstract
On the basis of tissue-specific enzyme activity and inhibition by catalytic products, Hans Krebs first demonstrated the existence of multiple glutaminases in mammals. Currently, two human genes are known to encode at least four glutaminase isoforms. However, the phylogeny of these medically relevant enzymes remains unclear, prompting us to investigate their origin and evolution. Using prokaryotic and eukaryotic glutaminase sequences, we built a phylogenetic tree whose topology suggested that the multidomain architecture was inherited from bacterial ancestors, probably simultaneously with the hosting of the proto-mitochondrion endosymbiont. We propose an evolutionary model wherein the appearance of the most active enzyme isoform, glutaminase C (GAC), which is expressed in many cancers, was a late retrotransposition event that occurred in fishes from the Chondrichthyes class. The ankyrin (ANK) repeats in the glutaminases were acquired early in their evolution. To obtain information on ANK folding, we solved two high-resolution structures of the ANK repeat-containing C termini of both kidney-type glutaminase (KGA) and GLS2 isoforms (glutaminase B and liver-type glutaminase). We found that the glutaminase ANK repeats form unique intramolecular contacts through two highly conserved motifs; curiously, this arrangement occludes a region usually involved in ANK-mediated protein-protein interactions. We also solved the crystal structure of full-length KGA and present a small-angle X-ray scattering model for full-length GLS2. These structures explain these proteins' compromised ability to assemble into catalytically active supra-tetrameric filaments, as previously shown for GAC. Collectively, these results provide information about glutaminases that may aid in the design of isoform-specific glutaminase inhibitors.
Collapse
Affiliation(s)
- Camila Cristina Pasquali
- From the Laboratório Nacional de Biociências, Centro Nacional de Pesquisa em Energia e Materiais, Campinas, São Paulo 13083-970, Brazil
| | - Zeyaul Islam
- From the Laboratório Nacional de Biociências, Centro Nacional de Pesquisa em Energia e Materiais, Campinas, São Paulo 13083-970, Brazil
| | - Douglas Adamoski
- From the Laboratório Nacional de Biociências, Centro Nacional de Pesquisa em Energia e Materiais, Campinas, São Paulo 13083-970, Brazil
| | - Igor Monteze Ferreira
- From the Laboratório Nacional de Biociências, Centro Nacional de Pesquisa em Energia e Materiais, Campinas, São Paulo 13083-970, Brazil.,the Structural Genomics Consortium, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Ricardo Diogo Righeto
- the Laboratório Nacional de Nanotecnologia, Centro Nacional de Pesquisa em Energia e Materiais, Campinas, São Paulo 13083-970, Brazil.,the School of Electrical and Computer Engineering, University of Campinas, São Paulo 13083-852, Brazil, and
| | - Jefferson Bettini
- the Laboratório Nacional de Nanotecnologia, Centro Nacional de Pesquisa em Energia e Materiais, Campinas, São Paulo 13083-970, Brazil
| | - Rodrigo Villares Portugal
- the Laboratório Nacional de Nanotecnologia, Centro Nacional de Pesquisa em Energia e Materiais, Campinas, São Paulo 13083-970, Brazil
| | - Wyatt Wai-Yin Yue
- the Structural Genomics Consortium, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Ana Gonzalez
- the Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Menlo Park, California 94025
| | - Sandra Martha Gomes Dias
- From the Laboratório Nacional de Biociências, Centro Nacional de Pesquisa em Energia e Materiais, Campinas, São Paulo 13083-970, Brazil,
| | - Andre Luis Berteli Ambrosio
- From the Laboratório Nacional de Biociências, Centro Nacional de Pesquisa em Energia e Materiais, Campinas, São Paulo 13083-970, Brazil,
| |
Collapse
|
26
|
Márquez J, Alonso FJ, Matés JM, Segura JA, Martín-Rufián M, Campos-Sandoval JA. Glutamine Addiction In Gliomas. Neurochem Res 2017; 42:1735-1746. [PMID: 28281102 DOI: 10.1007/s11064-017-2212-1] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 02/15/2017] [Accepted: 02/17/2017] [Indexed: 10/20/2022]
Abstract
Cancer cells develop and succeed by shifting to different metabolic programs compared with their normal cell counterparts. One of the classical hallmarks of cancer cells is their higher glycolysis rate and lactate production even in the presence of abundant O2 (Warburg effect). Another common metabolic feature of cancer cells is a high rate of glutamine (Gln) consumption normally exceeding their biosynthetic and energetic needs. The term Gln addiction is now widely used to reflect the strong dependence shown by most cancer cells for this essential nitrogen substrate after metabolic reprogramming. A Gln/glutamate (Glu) cycle occurs between host tissues and the tumor in order to maximize its growth and proliferation rates. The mechanistic basis for this deregulated tumor metabolism and how these changes are connected to oncogenic and tumor suppressor pathways are becoming increasingly understood. Based on these advances, new avenues of research have been initiated to find novel therapeutic targets and to explore strategies that interfere with glutamine metabolism as anticancer therapies. In this review, we provided an updated overview of glutamine addiction in glioma, the most prevalent type of brain tumor.
Collapse
Affiliation(s)
- Javier Márquez
- Canceromics lab, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Instituto de Biomedicina de Málaga (IBIMA), Universidad de Málaga, 29071, Málaga, Spain.
| | - Francisco J Alonso
- Canceromics lab, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Instituto de Biomedicina de Málaga (IBIMA), Universidad de Málaga, 29071, Málaga, Spain
| | - José M Matés
- Canceromics lab, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Instituto de Biomedicina de Málaga (IBIMA), Universidad de Málaga, 29071, Málaga, Spain
| | - Juan A Segura
- Canceromics lab, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Instituto de Biomedicina de Málaga (IBIMA), Universidad de Málaga, 29071, Málaga, Spain
| | - Mercedes Martín-Rufián
- Canceromics lab, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Instituto de Biomedicina de Málaga (IBIMA), Universidad de Málaga, 29071, Málaga, Spain
| | - José A Campos-Sandoval
- Canceromics lab, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Instituto de Biomedicina de Málaga (IBIMA), Universidad de Málaga, 29071, Málaga, Spain
| |
Collapse
|
27
|
Katt WP, Lukey MJ, Cerione RA. A tale of two glutaminases: homologous enzymes with distinct roles in tumorigenesis. Future Med Chem 2017; 9:223-243. [PMID: 28111979 PMCID: PMC5558546 DOI: 10.4155/fmc-2016-0190] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 12/01/2016] [Indexed: 01/17/2023] Open
Abstract
Many cancer cells exhibit an altered metabolic phenotype, in which glutamine consumption is upregulated relative to healthy cells. This metabolic reprogramming often depends upon mitochondrial glutaminase activity, which converts glutamine to glutamate, a key precursor for biosynthetic and bioenergetic processes. Two isozymes of glutaminase exist, a kidney-type (GLS) and a liver-type enzyme (GLS2 or LGA). While a majority of studies have focused on GLS, here we summarize key findings on both glutaminases, describing their structure and function, their roles in cancer and pharmacological approaches to inhibiting their activities.
Collapse
Affiliation(s)
- William P Katt
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Michael J Lukey
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Richard A Cerione
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA
- Department of Chemistry & Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
28
|
Márquez J, Campos-Sandoval JA, Peñalver A, Matés JM, Segura JA, Blanco E, Alonso FJ, de Fonseca FR. Glutamate and Brain Glutaminases in Drug Addiction. Neurochem Res 2016; 42:846-857. [DOI: 10.1007/s11064-016-2137-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 09/12/2016] [Accepted: 12/08/2016] [Indexed: 10/20/2022]
|
29
|
Abstract
Mammalian glutaminases catalyze the stoichiometric conversion of L-glutamine to L-glutamate and ammonium ions. In brain, glutaminase is considered the prevailing pathway for synthesis of the neurotransmitter pool of glutamate. Besides neurotransmission, the products of glutaminase reaction also fulfill crucial roles in energy and metabolic homeostasis in mammalian brain. In the last years, new functional roles for brain glutaminases are being uncovered by using functional genomic and proteomic approaches. Glutaminases may act as multifunctional proteins able to perform different tasks: the discovery of multiple transcript variants in neurons and glial cells, novel extramitochondrial localizations, and isoform-specific proteininteracting partners strongly support possible moonlighting functions for these proteins. In this chapter, we present a critical account of essential works on brain glutaminase 80 years after its discovery. We will highlight the impact of recent findings and thoughts in the context of the glutamate/glutamine brain homeostasis.
Collapse
|
30
|
Glutaminases in slowly proliferating gastroenteropancreatic neuroendocrine neoplasms/tumors (GEP-NETs): Selective overexpression of mRNA coding for the KGA isoform. Exp Mol Pathol 2015; 100:74-8. [PMID: 26581715 DOI: 10.1016/j.yexmp.2015.11.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 11/11/2015] [Indexed: 11/22/2022]
Abstract
Glutamine (Gln) is a crucial metabolite in cancer cells of different origin, and the expression and activity of different isoforms of the Gln-degrading enzyme, glutaminase (GA), have variable implications for tumor growth and metabolism. Human glutaminases are encoded by two genes: the GLS gene encodes the kidney-type glutaminases, KGA and GAC, while the GLS2 gene encodes the liver-type glutaminases, GAB and LGA. Recent studies suggest that the GAC isoform and thus high GAC/KGA ratio, are characteristic of highly proliferating tumors, while GLS2 proteins have an inhibitory effect on tumor growth. Here we analyzed the expression levels of distinct GA transcripts in 7 gastroenteropancreatic neuroendocrine tumors (GEP-NETs) with low proliferation index and 7 non-neoplastic tissues. GEP-NETs overexpressed KGA, while GAC, which was the most abundant isoform, was not different from control. The expression of the GLS2 gene showed tendency towards elevation in GEP-NETs compared to control. Collectively, the expression pattern of GA isoforms conforms to the low proliferative capacity of GEP-NETs encompassed in this study.
Collapse
|
31
|
Szeliga M, Bogacińska-Karaś M, Kuźmicz K, Rola R, Albrecht J. Downregulation of GLS2 in glioblastoma cells is related to DNA hypermethylation but not to the p53 status. Mol Carcinog 2015; 55:1309-16. [PMID: 26258493 DOI: 10.1002/mc.22372] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 06/29/2015] [Accepted: 07/06/2015] [Indexed: 12/19/2022]
Abstract
Human phosphate-activated glutaminase (GA) is encoded by two genes: GLS and GLS2. Glioblastomas (GB) usually lack GLS2 transcripts, and their reintroduction inhibits GB growth. The GLS2 gene in peripheral tumors may be i) methylation- controlled and ii) a target of tumor suppressor p53 often mutated in gliomas. Here we assessed the relation of GLS2 downregulation in GB to its methylation and TP53 status. DNA demethylation with 5-aza-2'-deoxycytidine restored GLS2 mRNA and protein content in human GB cell lines with both mutated (T98G) and wild-type (U87MG) p53 and reduced the methylation of CpG1 (promoter region island), and CpG2 (first intron island) in both cell lines. In cell lines and clinical GB samples alike, methylated CpG islands were detected both in the GLS2 promoter (as reported earlier) and in the first intron of this gene. CpG methylation of either island was absent in GLS2-expressing non-tumoros brain tissues. Screening for mutation in the exons 5-8 of TP53 revealed a point mutation in only one out of seven GB examined. In conclusion, aberrant methylation of CpG islands, appear to contribute to silencing of GLS2 in GB by a mechanism bypassing TP53 mutations. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Monika Szeliga
- Department of Neurotoxicology, Mossakowski Medical Research Centre, Warsaw, Poland
| | | | | | - Radosław Rola
- Department of Neurosurgery and Paediatric Neurosurgery of the Lublin Medical University, Lublin, Poland.,Department of Physiopathology, Institute of Agricultural Medicine, Lublin, Poland
| | - Jan Albrecht
- Department of Neurotoxicology, Mossakowski Medical Research Centre, Warsaw, Poland
| |
Collapse
|
32
|
Targeting glutaminolysis has antileukemic activity in acute myeloid leukemia and synergizes with BCL-2 inhibition. Blood 2015; 126:1346-56. [PMID: 26186940 DOI: 10.1182/blood-2015-01-621870] [Citation(s) in RCA: 286] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 07/09/2015] [Indexed: 12/20/2022] Open
Abstract
Cancer cells require glutamine to adapt to increased biosynthetic activity. The limiting step in intracellular glutamine catabolism involves its conversion to glutamate by glutaminase (GA). Different GA isoforms are encoded by the genes GLS1 and GLS2 in humans. Herein, we show that glutamine levels control mitochondrial oxidative phosphorylation (OXPHOS) in acute myeloid leukemia (AML) cells. Glutaminase C (GAC) is the GA isoform that is most abundantly expressed in AML. Both knockdown of GLS1 expression and pharmacologic GLS1 inhibition by the drug CB-839 can reduce OXPHOS, leading to leukemic cell proliferation arrest and apoptosis without causing cytotoxic activity against normal human CD34(+) progenitors. Strikingly, GLS1 knockdown dramatically inhibited AML development in NSG mice. The antileukemic activity of CB-839 was abrogated by both the expression of a hyperactive GAC(K320A) allele and the addition of the tricarboxyclic acid cycle product α-ketoglutarate, indicating the critical function of GLS1 in AML cell survival. Finally, glutaminolysis inhibition activated mitochondrial apoptosis and synergistically sensitized leukemic cells to priming with the BCL-2 inhibitor ABT-199. These findings show that targeting glutamine addiction via GLS1 inhibition offers a potential novel therapeutic strategy for AML.
Collapse
|
33
|
Campos-Sandoval JA, Martín-Rufián M, Cardona C, Lobo C, Peñalver A, Márquez J. Glutaminases in brain: Multiple isoforms for many purposes. Neurochem Int 2015; 88:1-5. [PMID: 25837287 DOI: 10.1016/j.neuint.2015.03.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 03/12/2015] [Accepted: 03/16/2015] [Indexed: 10/23/2022]
Abstract
Glutaminase is expressed in most mammalian tissues and cancer cells, but recent studies are now revealing a considerably degree of complexity in its pattern of expression and functional regulation. Novel transcript variants of the mammalian glutaminase Gls2 gene have been recently found and characterized in brain. Co-expression of different isoforms in the same cell type would allow cells to fine-tune their Gln/Glu levels under a wide range of metabolic states. Moreover, the discovery of protein interacting partners and novel subcellular localizations, for example nucleocytoplasmic in neurons and astrocytes, strongly suggest non-neurotransmission roles for Gls2 isoforms associated with transcriptional regulation and cellular differentiation. Of note, Gls isoforms have been considered as an important trophic factor for neuronal differentiation and postnatal development of brain regions. On the other hand, glutaminases are taking center stage in tumor biology as new therapeutic targets to inhibit metabolic reprogramming of cancer cells. Interestingly, glutaminase isoenzymes play seemingly opposing roles in cancer cell growth and proliferation; this issue will be also succinctly discussed with special emphasis on brain tumors.
Collapse
Affiliation(s)
- José A Campos-Sandoval
- Departamento de Biología Molecular y Bioquímica, Canceromics Lab. Facultad de Ciencias, Universidad de Málaga, 29071 Málaga, Spain; Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
| | | | - Carolina Cardona
- Departamento de Biología Molecular y Bioquímica, Canceromics Lab. Facultad de Ciencias, Universidad de Málaga, 29071 Málaga, Spain; Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
| | - Carolina Lobo
- Proteomics Lab, Central Facility Core, Universidad de Málaga, 29071 Málaga, Spain
| | - Ana Peñalver
- Departamento de Biología Molecular y Bioquímica, Canceromics Lab. Facultad de Ciencias, Universidad de Málaga, 29071 Málaga, Spain; Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
| | - Javier Márquez
- Departamento de Biología Molecular y Bioquímica, Canceromics Lab. Facultad de Ciencias, Universidad de Málaga, 29071 Málaga, Spain; Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain.
| |
Collapse
|
34
|
Cardona C, Sánchez-Mejías E, Dávila JC, Martín-Rufián M, Campos-Sandoval JA, Vitorica J, Alonso FJ, Matés JM, Segura JA, Norenberg MD, Rama Rao KV, Jayakumar AR, Gutiérrez A, Márquez J. Expression of Gls and Gls2 glutaminase isoforms in astrocytes. Glia 2014; 63:365-82. [PMID: 25297978 DOI: 10.1002/glia.22758] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 09/24/2014] [Indexed: 01/10/2023]
Abstract
The expression of glutaminase in glial cells has been a controversial issue and matter of debate for many years. Actually, glutaminase is essentially considered as a neuronal marker in brain. Astrocytes are endowed with efficient and high capacity transport systems to recapture synaptic glutamate which seems to be consistent with the absence of glutaminase in these glial cells. In this work, a comprehensive study was devised to elucidate expression of glutaminase in neuroglia and, more concretely, in astrocytes. Immunocytochemistry in rat and human brain tissues employing isoform-specific antibodies revealed expression of both Gls and Gls2 glutaminase isozymes in glutamatergic and GABAergic neuronal populations as well as in astrocytes. Nevertheless, there was a different subcellular distribution: Gls isoform was always present in mitochondria while Gls2 appeared in two different locations, mitochondria and nucleus. Confocal microscopy and double immunofluorescence labeling in cultured astrocytes confirmed the same pattern previously seen in brain tissue samples. Astrocytic glutaminase expression was also assessed at the mRNA level, real-time quantitative RT-PCR detected transcripts of four glutaminase isozymes but with marked differences on their absolute copy number: the predominance of Gls isoforms over Gls2 transcripts was remarkable (ratio of 144:1). Finally, we proved that astrocytic glutaminase proteins possess enzymatic activity by in situ activity staining: concrete populations of astrocytes were labeled in the cortex, cerebellum and hippocampus of rat brain demonstrating functional catalytic activity. These results are relevant for the stoichiometry of the Glu/Gln cycle at the tripartite synapse and suggest novel functions for these classical metabolic enzymes.
Collapse
Affiliation(s)
- Carolina Cardona
- Canceromics Lab. Facultad de Ciencias, Departamento de Biología Molecular y Bioquímica, Universidad de Málaga, 29071, Málaga, Spain; Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Botman D, Tigchelaar W, Van Noorden CJF. Determination of phosphate-activated glutaminase activity and its kinetics in mouse tissues using metabolic mapping (quantitative enzyme histochemistry). J Histochem Cytochem 2014; 62:813-26. [PMID: 25163927 PMCID: PMC4230542 DOI: 10.1369/0022155414551177] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Phosphate-activated glutaminase (PAG) converts glutamine to glutamate as part of the glutaminolysis pathway in mitochondria. Two genes, GLS1 and GLS2, which encode for kidney-type PAG and liver-type PAG, respectively, differ in their tissue-specific activities and kinetics. Tissue-specific PAG activity and its kinetics were determined by metabolic mapping using a tetrazolium salt and glutamate dehydrogenase as an auxiliary enzyme in the presence of various glutamine concentrations. In kidney and brain, PAG showed Michaelis-Menten kinetics with a Km of 0.6 mM glutamine and a Vmax of 1.1 µmol/mL/min when using 5 mM glutamine. PAG activity was high in the kidney cortex and inner medulla but low in the outer medulla, papillary tip, glomeruli and the lis of Henle. In brain tissue sections, PAG was active in the grey matter and inactive in myelin-rich regions. Liver PAG showed allosteric regulation with a Km of 11.6 mM glutamine and a Vmax of 0.5 µmol/mL/min when using 20 mM glutamine. The specificity of the method was shown after incubation of brain tissue sections with the PAG inhibitor 6-diazo-5-oxo-L-norleucine. PAG activity was decreased to 22% in the presence of 2 mM 6-diazo-5-oxo-L-norleucine. At low glutamine concentrations, PAG activity was higher in periportal regions, indicating a lower Km for periportal PAG. When compared with liver, kidney and brain, other tissues showed 3-fold to 6-fold less PAG activity. In conclusion, PAG is mainly active in mouse kidney, brain and liver, and shows different kinetics depending on which type of PAG is expressed.
Collapse
Affiliation(s)
- Dennis Botman
- Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands (DB, WT, CJFVN)
| | - Wikky Tigchelaar
- Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands (DB, WT, CJFVN)
| | - Cornelis J F Van Noorden
- Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands (DB, WT, CJFVN)
| |
Collapse
|
36
|
Martín-Rufián M, Nascimento-Gomes R, Higuero A, Crisma AR, Campos-Sandoval JA, Gómez-García MC, Cardona C, Cheng T, Lobo C, Segura JA, Alonso FJ, Szeliga M, Albrecht J, Curi R, Márquez J, Colquhoun A, Deberardinis RJ, Matés JM. Both GLS silencing and GLS2 overexpression synergize with oxidative stress against proliferation of glioma cells. J Mol Med (Berl) 2013; 92:277-90. [PMID: 24276018 DOI: 10.1007/s00109-013-1105-2] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 11/12/2013] [Accepted: 11/13/2013] [Indexed: 12/30/2022]
Abstract
UNLABELLED Mitochondrial glutaminase (GA) plays an essential role in cancer cell metabolism, contributing to biosynthesis, bioenergetics, and redox balance. Humans contain several GA isozymes encoded by the GLS and GLS2 genes, but the specific roles of each in cancer metabolism are still unclear. In this study, glioma SFxL and LN229 cells with silenced isoenzyme glutaminase KGA (encoded by GLS) showed lower survival ratios and a reduced GSH-dependent antioxidant capacity. These GLS-silenced cells also demonstrated induction of apoptosis indicated by enhanced annexin V binding capacity and caspase 3 activity. GLS silencing was associated with decreased mitochondrial membrane potential (ΔΨm) (JC-1 dye test), indicating that apoptosis was mediated by mitochondrial dysfunction. Similar observations were made in T98 glioma cells overexpressing glutaminase isoenzyme GAB, encoded by GLS2, though some characteristics (GSH/GSSG ratio) were different in the differently treated cell lines. Thus, control of GA isoenzyme expression may prove to be a key tool to alter both metabolic and oxidative stress in cancer therapy. Interestingly, reactive oxygen species (ROS) generation by treatment with oxidizing agents: arsenic trioxide or hydrogen peroxide, synergizes with either KGA silencing or GAB overexpression to suppress malignant properties of glioma cells, including the reduction of cellular motility. Of note, negative modulation of GLS isoforms or GAB overexpression evoked lower c-myc and bcl-2 expression, as well as higher pro-apoptotic bid expression. Combination of modulation of GA expression and treatment with oxidizing agents may become a therapeutic strategy for intractable cancers and provides a multi-angle evaluation system for anti-glioma pre-clinical investigations. KEY MESSAGE Silencing GLS or overexpressing GLS2 induces growth inhibition in glioma cell lines. Inhibition is synergistically enhanced after arsenic trioxide (ATO) or H2O2 treatment. Glutatione levels decrease in GLS-silenced cells but augment if GLS2 is overexpressed. ROS synergistically inhibit cell migration by GLS silencing or GLS2 overexpression. c-myc, bid, and bcl-2 mediate apoptosis resulting from GLS silencing or GLS2 overexpression.
Collapse
Affiliation(s)
- Mercedes Martín-Rufián
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, Campus de Teatinos, University of Málaga, 29071, Málaga, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Szeliga M, Bogacińska-Karaś M, Różycka A, Hilgier W, Marquez J, Albrecht J. Silencing of GLS and overexpression of GLS2 genes cooperate in decreasing the proliferation and viability of glioblastoma cells. Tumour Biol 2013; 35:1855-62. [PMID: 24096582 PMCID: PMC3967065 DOI: 10.1007/s13277-013-1247-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 09/23/2013] [Indexed: 12/25/2022] Open
Abstract
Glutamine (Gln) metabolism, initiated by its degradation by glutaminases (GA), is elevated in neoplastic cells and tissues. In malignant glia-derived tumors, GA isoforms, KGA and GAC, coded by the GLS gene, are overexpressed, whereas the GLS2-coded GAB and LGA isoforms, are hardly detectable in there. Our previous study revealed that transfection of T98G glioblastoma cells with GAB reduced cell proliferation and migration, by a yet unknown mechanism not related to Gln degradation. The question arose how simultaneous overexpression of GAB and inhibition of KGA would affect glioblastoma cell growth. Here, we used siRNA to silence the expression of Gls in T98G cells which were or were not stably transfected with GAB (TGAB cells). In both T98G and TGAB cell lines, silencing of Gls with siRNAs targeted at different sequences decreased cell viability and proliferation in a different, sequence-dependent degree, and the observed decreases were in either cell line highly correlated with increase of intracellular Gln (r > 0.9), a parameter manifesting decreased Gln degradation. The results show that combination of negative modulation of GA isoforms arising from GLS gene with the introduction of the GLS2 gene product, GAB, may in the future provide a useful means to curb glioblastoma growth in situ. At the same time, the results underscore the critical role of Gln degradation mediated by KGA in the manifestations of aggressive glial tumor phenotype.
Collapse
Affiliation(s)
- Monika Szeliga
- Department of Neurotoxicology, Mossakowski Medical Research Centre Polish Academy of Sciences, 5 Pawińskiego Str., 02-106, Warsaw, Poland,
| | | | | | | | | | | |
Collapse
|
38
|
Márquez J, Cardona C, Campos-Sandoval JA, Peñalver A, Tosina M, Matés JM, Martín-Rufián M. Mammalian glutaminase isozymes in brain. Metab Brain Dis 2013; 28:133-7. [PMID: 23149879 DOI: 10.1007/s11011-012-9356-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 10/30/2012] [Indexed: 11/28/2022]
Abstract
Glutamine/glutamate homeostasis must be exquisitely regulated in mammalian brain and glutaminase (GA, E.C. 3.5.1.2) is one of the main enzymes involved. The products of GA reaction, glutamate and ammonia, are essential metabolites for energy and biosynthetic purposes but they are also hazardous compounds at concentrations beyond their normal physiological thresholds. The classical pattern of GA expression in mammals has been recently challenged by the discovery of novel transcript variants and protein isoforms. Furthermore, the interactome of brain GA is also starting to be uncovered adding a new level of regulatory complexity. GA may traffic in brain and unexpected locations, like cytosol and nucleus, have been found for GA isoforms. Finally, the expression of GA in glial cells has been reported and its potential implications in ammonia homeostasis are discussed.
Collapse
Affiliation(s)
- Javier Márquez
- Departamento de Biología Molecular y Bioquímica, Laboratorio de Química de Proteínas, Facultad de Ciencias, Universidad de Málaga, Málaga 29071, Spain.
| | | | | | | | | | | | | |
Collapse
|
39
|
Giacobbe A, Bongiorno-Borbone L, Bernassola F, Terrinoni A, Markert EK, Levine AJ, Feng Z, Agostini M, Zolla L, Agrò AF, Notterman DA, Melino G, Peschiaroli A. p63 regulates glutaminase 2 expression. Cell Cycle 2013; 12:1395-405. [PMID: 23574722 DOI: 10.4161/cc.24478] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The transcription factor p63 is critical for many biological processes, including development and maintenance of epidermal tissues and tumorigenesis. Here, we report that the TAp63 isoforms regulate cell metabolism through the induction of the mitochondrial glutaminase 2 (GLS2) gene both in primary cells and tumor cell lines. By ChIP analysis and luciferase assay, we confirmed that TAp63 binds directly to the p53/p63 consensus DNA binding sequence within the GLS2 promoter region. Given the critical role of p63 in epidermal differentiation, we have investigated the regulation of GLS2 expression during this process. GLS2 and TAp63 expression increases during the in vitro differentiation of primary human keratinocytes, and depletion of GLS2 inhibits skin differentiation both at molecular and cellular levels. We found that GLS2 and TAp63 expression are concomitantly induced in cancer cells exposed to oxidative stresses. siRNA-mediated depletion of GLS2 sensitizes cells to ROS-induced apoptosis, suggesting that the TAp63/GLS2 axis can be functionally important as a cellular antioxidant pathway in the absence of p53. Accordingly, we found that GLS2 is upregulated in colon adenocarcinoma. Altogether, our findings demonstrate that GLS2 is a bona fide TAp63 target gene, and that the TAp63-dependent regulation of GLS2 is important for both physiological and pathological processes.
Collapse
Affiliation(s)
- Arianna Giacobbe
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Laulederkind SJF, Hayman GT, Wang SJ, Smith JR, Lowry TF, Nigam R, Petri V, de Pons J, Dwinell MR, Shimoyama M, Munzenmaier DH, Worthey EA, Jacob HJ. The Rat Genome Database 2013--data, tools and users. Brief Bioinform 2013; 14:520-6. [PMID: 23434633 PMCID: PMC3713714 DOI: 10.1093/bib/bbt007] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The Rat Genome Database (RGD) was started >10 years ago to provide a core genomic resource for rat researchers. Currently, RGD combines genetic, genomic, pathway, phenotype and strain information with a focus on disease. RGD users are provided with access to structured and curated data from the molecular level through the organismal level. Those users access RGD from all over the world. End users are not only rat researchers but also researchers working with mouse and human data. Translational research is supported by RGD’s comparative genetics/genomics data in disease portals, in GBrowse, in VCMap and on gene report pages. The impact of RGD also goes beyond the traditional biomedical researcher, as the influence of RGD reaches bioinformaticians, tool developers and curators. Import of RGD data into other publicly available databases expands the influence of RGD to a larger set of end users than those who avail themselves of the RGD website. The value of RGD continues to grow as more types of data and more tools are added, while reaching more types of end users.
Collapse
Affiliation(s)
- Stanley J F Laulederkind
- Human and Molecular Genetics Center, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226-3548, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Network of brain protein level changes in glutaminase deficient fetal mice. J Proteomics 2013; 80:236-49. [PMID: 23376484 DOI: 10.1016/j.jprot.2013.01.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 12/17/2012] [Accepted: 01/05/2013] [Indexed: 02/06/2023]
Abstract
Glutaminase is a multifunctional enzyme encoded by gene Gls involved in energy metabolism, ammonia trafficking and regeneration of neurotransmitter glutamate. To address the proteomic basis for the neurophenotypes of glutaminase-deficient mice, brain proteins from late gestation wild type, Gls+/- and Gls-/- male mice were subjected to two-dimensional gel electrophoresis, with subsequent identification by mass spectrometry using nano-LC-ESI-MS/MS. Protein spots that showed differential genotypic variation were quantified by immunoblotting. Differentially expressed proteins unambiguously identified by MS/MS included neurocalcin delta, retinol binding protein-1, reticulocalbin-3, cytoskeleton proteins fascin and tropomyosin alpha-4-chain, dihydropyrimidinase-related protein-5, apolipoprotein IV and proteins from protein metabolism proteasome subunits alpha type 2, type 7, heterogeneous nuclear ribonucleoprotein C1/C2 and H, voltage-gated anion-selective channel proteins 1 and 2, ATP synthase subunit β and transitional endoplasmic reticulum ATPase. An interaction network determined by Ingenuity Pathway Analysis revealed a link between glutaminase and calcium, Akt and retinol signaling, cytoskeletal elements, ATPases, ion channels, protein synthesis and the proteasome system, intermediary, nucleic acid and lipid metabolism, huntingtin, guidance cues, transforming growth factor beta-1 and hepatocyte nuclear factor 4-alpha. The network identified involves (a) cellular assembly and organization and (b) cell signaling and cell cycle, suggesting that Gls is crucial for neuronal maturation.
Collapse
|
42
|
Hallen A, Jamie JF, Cooper AJL. Imine reductases: a comparison of glutamate dehydrogenase to ketimine reductases in the brain. Neurochem Res 2013; 39:527-41. [PMID: 23314864 DOI: 10.1007/s11064-012-0964-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 12/12/2012] [Accepted: 12/22/2012] [Indexed: 02/06/2023]
Abstract
A key intermediate in the glutamate dehydrogenase (GDH)-catalyzed reaction is an imine. Mechanistically, therefore, GDH exhibits similarities to the ketimine reductases. In the current review, we briefly discuss (a) the metabolic importance of the GDH reaction in liver and brain, (b) the mechanistic similarities between GDH and the ketimine reductases, (c) the metabolic importance of the brain ketimine reductases, and (d) the neurochemical consequences of defective ketimine reductases. Our review contains many historical references to the early work on amino acid metabolism. This work tends to be overlooked nowadays, but is crucial for a contemporary understanding of the central importance of ketimines in nitrogen and intermediary metabolism. The ketimine reductases are important enzymes linking nitrogen flow among several key amino acids, yet have been little studied. The cerebral importance of the ketimine reductases is an area of biomedical research that deserves far more attention.
Collapse
Affiliation(s)
- André Hallen
- Department of Chemistry and Biomolecular Sciences, Macquarie University, Balaclava Road, North Ryde, NSW, 2109, Australia,
| | | | | |
Collapse
|
43
|
Shukla K, Ferraris DV, Thomas AG, Stathis M, Duvall B, Delahanty G, Alt J, Rais R, Rojas C, Gao P, Xiang Y, Dang CV, Slusher BS, Tsukamoto T. Design, synthesis, and pharmacological evaluation of bis-2-(5-phenylacetamido-1,2,4-thiadiazol-2-yl)ethyl sulfide 3 (BPTES) analogs as glutaminase inhibitors. J Med Chem 2012; 55:10551-63. [PMID: 23151085 DOI: 10.1021/jm301191p] [Citation(s) in RCA: 160] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Bis-2-(5-phenylacetamido-1,2,4-thiadiazol-2-yl)ethyl sulfide (BPTES) is a potent and selective allosteric inhibitor of kidney-type glutaminase (GLS) that has served as a molecular probe to determine the therapeutic potential of GLS inhibition. In an attempt to identify more potent GLS inhibitors with improved drug-like molecular properties, a series of BPTES analogs were synthesized and evaluated. Our structure-activity relationship (SAR) studies revealed that some truncated analogs retained the potency of BPTES, presenting an opportunity to improve its aqueous solubility. One of the analogs, N-(5-{2-[2-(5-amino-[1,3,4]thiadiazol-2-yl)-ethylsulfanyl]-ethyl}-[1,3,4]thiadiazol-2-yl)-2-phenyl-acetamide 6, exhibited similar potency and better solubility relative to BPTES and attenuated the growth of P493 human lymphoma B cells in vitro as well as in a mouse xenograft model.
Collapse
Affiliation(s)
- Krupa Shukla
- Department of Neurology and Brain Science Institute, Johns Hopkins University, Baltimore, Maryland 21205, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|