1
|
Overexpression of glyoxalase system enzymes in human kidney tumor: Erratum. Cancer J 2024; 30:297. [PMID: 39042782 DOI: 10.1097/ppo.0000000000000739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
|
2
|
Beyond metabolic waste: lysine lactylation and its potential roles in cancer progression and cell fate determination. Cell Oncol (Dordr) 2023; 46:465-480. [PMID: 36656507 DOI: 10.1007/s13402-023-00775-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/21/2022] [Accepted: 11/26/2022] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Lactate is an important metabolite derived from glycolysis under physiological and pathological conditions. The Warburg effect reveals the vital role of lactate in cancer progression. Numerous studies have reported crucial roles for lactate in cancer progression and cell fate determination. Lactylation, a novel posttranslational modification (PTM), has provided a new opportunity to investigate metabolic epigenetic regulation, and studies of this process have been initiated in a wide range of cancer cells, cancer-associated immune cells, and embryonic stem cells. CONCLUSION Lactylation is a novel and interesting mechanism of lactate metabolism linked to metabolic rewiring and epigenetic remodeling. It is a potential and hopeful target for cancer therapy. Here, we summarize the discovery of lactylation, the mechanisms of site modification, and progress in research on nonhistone lactylation. We focus on the potential roles of lactylation in cancer progression and cell fate determination and the possible therapeutic strategies for targeting lysine lactylation. Finally, we suggest some future research topics on lactylation to inspire some interesting ideas.
Collapse
|
3
|
Abstract
The glyoxalase gene family consists of six structurally and functionally diverse enzymes with broad roles in metabolism. The common feature that defines this family is based on structural motifs that coordinate divalent cations which are required for activity. These family members have been implicated in a variety of physiological processes, including amino-acid metabolism (4-hydroxyphenylpyruvate dioxygenase; HPD), primary metabolism (methylmalonyl-CoA epimerase; MCEE), and aldehyde detoxication (glyoxalase 1; GLO1) and therefore have significant associations with disease. A central function of this family is the detoxification of reactive dicarbonyls (e.g., methylglyoxal), which react with cellular nucleophiles, resulting in the modification of lipids, proteins, and DNA. These damaging modifications activate canonical stress responses such as heat shock, unfolded protein, antioxidant, and DNA damage responses. Thus, glyoxalases serve an important role in homeostasis, preventing the pathogenesis of metabolic disease states, including obesity, diabetes, cardiovascular disease, renal failure, and aging. This review presents a thorough overview of the literature surrounding this diverse enzyme class. Although extensive literature exists for some members of this family (e.g., GLO1), little is known about the physiological role of glyoxalase domain-containing protein 4 (GLOD4) and 5 (GLOD5), paving the way for exciting avenues for future research.
Collapse
Affiliation(s)
- Dominique O Farrera
- Department of Pharmacology and College of Pharmacy, University of Arizona, Tucson, Arizona85721, United States
| | - James J Galligan
- Department of Pharmacology and College of Pharmacy, University of Arizona, Tucson, Arizona85721, United States
| |
Collapse
|
4
|
Alfarouk KO, Alqahtani SS, Alshahrani S, Morgenstern J, Supuran CT, Reshkin SJ. The possible role of methylglyoxal metabolism in cancer. J Enzyme Inhib Med Chem 2021; 36:2010-2015. [PMID: 34517737 PMCID: PMC8451662 DOI: 10.1080/14756366.2021.1972994] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Tumours reprogram their metabolism to acquire an evolutionary advantage over normal cells. However, not all such metabolic pathways support energy production. An example of these metabolic pathways is the Methylglyoxal (MG) one. This pathway helps maintain the redox state, and it might act as a phosphate sensor that monitors the intracellular phosphate levels. In this work, we discuss the biochemical step of the MG pathway and interrelate it with cancer.
Collapse
Affiliation(s)
- Khalid O Alfarouk
- Department of Evolutionary Pharmacology, and Tumor Metabolism, Hala Alfarouk Cancer Center, Khartoum, Sudan
| | - Saad S Alqahtani
- Pharmacy Practice Research Unit, Clinical Pharmacy Department, College of Pharmacy, Jazan University, Jazan, KSA
| | - Saeed Alshahrani
- Pharmacology and Toxicology Department, College of Pharmacy, Jazan University, Jazan, KSA
| | - Jakob Morgenstern
- Department of Internal Medicine I, Endocrinology and Metabolism, Heidelberg University, Germany
| | - Claudiu T Supuran
- Neurofarba Department, Universita Degli Studi di Firenze, Florence, Italy
| | - Stephan J Reshkin
- Department of Bioscience, Biotechnology and Biopharmaceutics, University of Bari, Bari, Italy
| |
Collapse
|
5
|
Rounds L, Nagle RB, Muranyi A, Jandova J, Gill S, Vela E, Wondrak GT. Glyoxalase 1 Expression as a Novel Diagnostic Marker of High-Grade Prostatic Intraepithelial Neoplasia in Prostate Cancer. Cancers (Basel) 2021; 13:3608. [PMID: 34298821 PMCID: PMC8304603 DOI: 10.3390/cancers13143608] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/12/2021] [Accepted: 07/15/2021] [Indexed: 12/03/2022] Open
Abstract
Glyoxalase 1 (GLO1) is an enzyme involved in the detoxification of methylglyoxal (MG), a reactive oncometabolite formed in the context of energy metabolism as a result of high glycolytic flux. Prior clinical evidence has documented GLO1 upregulation in various tumor types including prostate cancer (PCa). However, GLO1 expression has not been explored in the context of PCa progression with a focus on high-grade prostatic intraepithelial neoplasia (HGPIN), a frequent precursor to invasive cancer. Here, we have evaluated GLO1 expression by immunohistochemistry in archival tumor samples from 187 PCa patients (stage 2 and 3). Immunohistochemical analysis revealed GLO1 upregulation during tumor progression, observable in HGPIN and PCa versus normal prostatic tissue. GLO1 upregulation was identified as a novel hallmark of HGPIN lesions, displaying the highest staining intensity in all clinical patient specimens. GLO1 expression correlated with intermediate-high risk Gleason grade but not with patient age, biochemical recurrence, or pathological stage. Our data identify upregulated GLO1 expression as a molecular hallmark of HGPIN lesions detectable by immunohistochemical analysis. Since current pathological assessment of HGPIN status solely depends on morphological features, GLO1 may serve as a novel diagnostic marker that identifies this precancerous lesion.
Collapse
Affiliation(s)
- Liliana Rounds
- Department of Pharmacology and Toxicology, College of Pharmacy & UA Cancer Center, University of Arizona, Tucson, AZ 85724, USA; (L.R.); (J.J.)
- Roche Diagnostics Solutions, Tucson, AZ 85755, USA; (A.M.); (S.G.); (E.V.)
| | - Ray B. Nagle
- Department of Pathology, University of Arizona, Tucson, AZ 85724, USA;
| | - Andrea Muranyi
- Roche Diagnostics Solutions, Tucson, AZ 85755, USA; (A.M.); (S.G.); (E.V.)
| | - Jana Jandova
- Department of Pharmacology and Toxicology, College of Pharmacy & UA Cancer Center, University of Arizona, Tucson, AZ 85724, USA; (L.R.); (J.J.)
| | - Scott Gill
- Roche Diagnostics Solutions, Tucson, AZ 85755, USA; (A.M.); (S.G.); (E.V.)
| | - Elizabeth Vela
- Roche Diagnostics Solutions, Tucson, AZ 85755, USA; (A.M.); (S.G.); (E.V.)
| | - Georg T. Wondrak
- Department of Pharmacology and Toxicology, College of Pharmacy & UA Cancer Center, University of Arizona, Tucson, AZ 85724, USA; (L.R.); (J.J.)
| |
Collapse
|
6
|
Al-Balas QA, Al-Sha'er MA, Hassan MA, Al Zu'bi E. Identification of the first "two digit nano-molar" inhibitors of the human glyoxalase-I enzyme as potential anticancer agents. Med Chem 2021; 18:473-483. [PMID: 34264188 DOI: 10.2174/1573406417666210714170403] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 04/23/2021] [Accepted: 05/12/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Glyoxalase-I (Glo-I) enzyme is recognized as an indispensable druggable target in cancer treatment. Its inhibition will lead to the accumulation of toxic aldehyde metabolites and cell death. Paramount efforts were spent to discover potential competitive inhibitors to eradicate cancer. OBJECTIVE Based on our previously work on this target for discovering potent inhibitors of this enzyme, herein, we address the discovery of the most potent Glo-I inhibitors reported in literature with two digits nano-molar activity. METHODS Molecular docking and in vitro assay were performed to discover these inhibitors and explore the active site's binding pattern. A detailed SAR scheme was generated, which identifies the significant functionalities responsible for the observed activity. RESULTS Compound 1 with an IC50 of 16.5 nM exhibited the highest activity, catechol moiety as an essential zinc chelating functionality. It has been shown by using molecular modeling techniques that the catechol moiety is responsible for the chelation zinc atom at the active site, an essential feature for enzyme inhibition. CONCLUSION Catechol derivatives are successful zinc chelators in the Glo-I enzyme while showing exceptional activity against the enzyme to the nanomolar level.
Collapse
Affiliation(s)
- Qosay A Al-Balas
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | | | - Mohammad A Hassan
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Esra'a Al Zu'bi
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
7
|
Al-Oudat BA, Jaradat HM, Al-Balas QA, Al-Shar'i NA, Bryant-Friedrich A, Bedi MF. Design, synthesis and biological evaluation of novel glyoxalase I inhibitors possessing diazenylbenzenesulfonamide moiety as potential anticancer agents. Bioorg Med Chem 2020; 28:115608. [PMID: 32690268 DOI: 10.1016/j.bmc.2020.115608] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 06/19/2020] [Accepted: 06/22/2020] [Indexed: 10/23/2022]
Abstract
The enzyme glyoxalase-I (Glo-I) is an essential therapeutic target in cancer treatment. Significant efforts have been made to discover competitive inhibitors of Glo-I as potential anticancer agents. Herein, we report the synthesis of a series of diazenylbenzenesulfonamide derivatives, their in vitro evaluation against Glo-I and the resulting structure-activity relationships. Among the compounds tested, compounds 9h and 9j exhibited the highest activity with IC50 1.28 µM and 1.13 µM, respectively. Docking studies to explore the binding mode of the compounds identified key moieties that may contribute to the observed activities. The active compounds will serve as suitable leads for further chemical optimization.
Collapse
Affiliation(s)
- Buthina A Al-Oudat
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan.
| | - Hana'a M Jaradat
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan
| | - Qosay A Al-Balas
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan
| | - Nizar A Al-Shar'i
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan
| | - Amanda Bryant-Friedrich
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43606, USA
| | - Mel F Bedi
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43606, USA
| |
Collapse
|
8
|
Circulating free methylglyoxal as a metabolic tumor biomarker in a rat colon adenocarcinoma model. Mol Clin Oncol 2020; 12:311-316. [PMID: 32190311 PMCID: PMC7058004 DOI: 10.3892/mco.2020.2000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 12/11/2019] [Indexed: 11/05/2022] Open
Abstract
Since the 1956 hypothesis of Otto Warburg, aerobic glycolysis has been recognized as a metabolic hallmark of cancer. Because methylglyoxal (MG) is a naturally occurring waste metabolite of glycolysis, we measured blood levels of this molecule in colon cancer-bearing rats. To compare the blood levels of free MG in cancerous and healthy animals,the present study used a dedicated tumor graft model consisting of the subcutaneous administration in syngenic BD-IX rats of a tumorigenic cell clone (PROb) and another non-tumorigenic clone (REGb) derived from the same tumor. Rats grafted with the PROb growing tumor cell clone exhibited a statistically significant increase in free MG blood levels (P=0.003), whereas rats transplanted with the REGb non-growing tumor cell clone exhibited normal MG values. The present study (first of three parts) suggests that cancer cells can produce and release free MG at higher levels than normal cells, making MG a putative novel metabolic biomarker of cancer.
Collapse
|
9
|
Dicarbonyl Stress and S-Glutathionylation in Cerebrovascular Diseases: A Focus on Cerebral Cavernous Malformations. Antioxidants (Basel) 2020; 9:antiox9020124. [PMID: 32024152 PMCID: PMC7071005 DOI: 10.3390/antiox9020124] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/25/2020] [Accepted: 01/29/2020] [Indexed: 02/07/2023] Open
Abstract
Dicarbonyl stress is a dysfunctional state consisting in the abnormal accumulation of reactive α-oxaldehydes leading to increased protein modification. In cells, post-translational changes can also occur through S-glutathionylation, a highly conserved oxidative post-translational modification consisting of the formation of a mixed disulfide between glutathione and a protein cysteine residue. This review recapitulates the main findings supporting a role for dicarbonyl stress and S-glutathionylation in the pathogenesis of cerebrovascular diseases, with specific emphasis on cerebral cavernous malformations (CCM), a vascular disease of proven genetic origin that may give rise to various clinical signs and symptoms at any age, including recurrent headaches, seizures, focal neurological deficits, and intracerebral hemorrhage. A possible interplay between dicarbonyl stress and S-glutathionylation in CCM is also discussed.
Collapse
|
10
|
de Bari L, Atlante A, Armeni T, Kalapos MP. Synthesis and metabolism of methylglyoxal, S-D-lactoylglutathione and D-lactate in cancer and Alzheimer's disease. Exploring the crossroad of eternal youth and premature aging. Ageing Res Rev 2019; 53:100915. [PMID: 31173890 DOI: 10.1016/j.arr.2019.100915] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 02/27/2019] [Accepted: 05/31/2019] [Indexed: 12/15/2022]
Abstract
Both cancer and Alzheimer's disease (AD) are emerging as metabolic diseases in which aberrant/dysregulated glucose metabolism and bioenergetics occur, and play a key role in disease progression. Interestingly, an enhancement of glucose uptake, glycolysis and pentose phosphate pathway occurs in both cancer cells and amyloid-β-resistant neurons in the early phase of AD. However, this metabolic shift has its adverse effects. One of them is the increase in methylglyoxal production, a physiological cytotoxic by-product of glucose catabolism. Methylglyoxal is mainly detoxified via cytosolic glyoxalase route comprising glyoxalase 1 and glyoxalase 2 with the production of S-D-lactoylglutathione and D-lactate as intermediate and end-product, respectively. Due to the existence of mitochondrial carriers and intramitochondrial glyoxalase 2 and D-lactate dehydrogenase, the transport and metabolism of both S-D-lactoylglutathione and D-lactate in mitochondria can contribute to methylglyoxal elimination, cellular antioxidant power and energy production. In this review, it is supposed that the different ability of cancer cells and AD neurons to metabolize methylglyoxal, S-D-lactoylglutathione and D-lactate scores cell fate, therefore being at the very crossroad of the "eternal youth" of cancer and the "premature death" of AD neurons. Understanding of these processes would help to elaborate novel metabolism-based therapies for cancer and AD treatment.
Collapse
|
11
|
Antognelli C, Moretti S, Frosini R, Puxeddu E, Sidoni A, Talesa VN. Methylglyoxal Acts as a Tumor-Promoting Factor in Anaplastic Thyroid Cancer. Cells 2019; 8:cells8060547. [PMID: 31174324 PMCID: PMC6627963 DOI: 10.3390/cells8060547] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 06/03/2019] [Accepted: 06/03/2019] [Indexed: 02/06/2023] Open
Abstract
Methylglyoxal (MG) is a potent inducer of advanced glycation end products (AGEs). MG, long considered a highly cytotoxic molecule with potential anticancer value, is now being re-evaluated to a protumorigenic agent in some malignancies. Anaplastic thyroid cancer (ATC) is an extremely aggressive and highly lethal cancer for which conventional therapies have proved ineffective. Successful therapeutic intervention in ATC is undermined by our poor understanding of its molecular etiology. In the attempt to understand the role of MG in ATC aggressiveness, we used immunohistochemistry to examine the level of MG protein adducts in ATC and slow-growing papillary thyroid cancer (PTC). We detected a high level of MG adducts in ATC compared to PTC ones, suggesting a protumor role for MG-mediated dicarbonyl stress in ATC. Accordingly, MG adduct accumulation in ATC cells in vitro was associated with a marked mesenchymal phenotype and increased migration/invasion, which were both reversed by aminoguanidine (AG)—a scavenger of MG—and resveratrol—an activator of Glyoxalase 1 (Glo1), the key metabolizing enzyme of MG. Our study represents the first demonstration that MG, via AGEs, acts as a tumor-promoting factor in ATC and suggests that MG scavengers and/or Glo1 activators merit investigations as potential therapeutic strategies for this malignancy.
Collapse
Affiliation(s)
- Cinzia Antognelli
- Department of Experimental Medicine, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy.
| | - Sonia Moretti
- Department of Medicine, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy.
| | - Roberta Frosini
- Department of Experimental Medicine, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy.
| | - Efisio Puxeddu
- Department of Medicine, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy.
| | - Angelo Sidoni
- Department of Experimental Medicine, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy.
| | - Vincenzo N Talesa
- Department of Experimental Medicine, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy.
| |
Collapse
|
12
|
Jafari S, Kazemi N, Ryde U, Irani M. Higher Flexibility of Glu-172 Explains the Unusual Stereospecificity of Glyoxalase I. Inorg Chem 2018; 57:4944-4958. [PMID: 29634252 DOI: 10.1021/acs.inorgchem.7b03215] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Despite many studies during the latest two decades, the reason for the unusual stereospecificity of glyoxalase I (GlxI) is still unknown. This metalloenzyme converts both enantiomers of its natural substrate to only one enantiomer of its product. In addition, GlxI catalyzes reactions involving some substrate and product analogues with a stereospecificity similar to that of its natural substrate reaction. For example, the enzyme exchanges the pro- S, but not the pro- R, hydroxymethyl proton of glutathiohydroxyacetone (HOC-SG) with a deuterium from D2O. To find some clues to the unusual stereospecificity of GlxI, we have studied the stereospecific proton exchange of the hydroxymethyl proton of HOC-SG by this enzyme. We employed density functional theory and molecular dynamics (MD) simulations to study the proton exchange mechanism and origin of the stereospecificity. The results show that a rigid cluster model with the same flexibility for the two active-site glutamate residues cannot explain the unusual stereospecificity of GlxI. However, using a cluster model with full flexibility of Glu-172 or a larger model with the entire glutamates, extending the backbone into the neighboring residues, the results showed that there is no way for HOC-SG to exchange its protons if the alcoholic proton is directed toward Glu-99. However, if the hydroxymethyl proton instead is directed toward the more flexible Glu-172, we find a catalytic reaction mechanism for the exchange of the HS proton by a deuterium, in accordance with experimental findings. Thus, our results indicate that the special stereospecificity of GlxI is caused by the more flexible environment of Glu-172 in comparison to that of Glu-99. This higher flexibility of Glu-172 is also confirmed by MD simulations. We propose a reaction mechanism for the stereospecific proton exchange of the hydroxymethyl proton of HOC-SG by GlxI with an overall energy barrier of 15 kcal/mol.
Collapse
Affiliation(s)
- Sonia Jafari
- Department of Chemistry , University of Kurdistan , P.O. Box 66175-416, Sanandaj , Iran.,Department of Theoretical Chemistry , Lund University , P.O. Box 124, SE-221 00 Lund , Sweden
| | - Nadia Kazemi
- Department of Chemistry , University of Kurdistan , P.O. Box 66175-416, Sanandaj , Iran
| | - Ulf Ryde
- Department of Theoretical Chemistry , Lund University , P.O. Box 124, SE-221 00 Lund , Sweden
| | - Mehdi Irani
- Department of Chemistry , University of Kurdistan , P.O. Box 66175-416, Sanandaj , Iran
| |
Collapse
|
13
|
Antognelli C, Talesa VN. Glyoxalases in Urological Malignancies. Int J Mol Sci 2018; 19:ijms19020415. [PMID: 29385039 PMCID: PMC5855637 DOI: 10.3390/ijms19020415] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/25/2018] [Accepted: 01/26/2018] [Indexed: 12/16/2022] Open
Abstract
Urological cancers include a spectrum of malignancies affecting organs of the reproductive and/or urinary systems, such as prostate, kidney, bladder, and testis. Despite improved primary prevention, detection and treatment, urological cancers are still characterized by an increasing incidence and mortality worldwide. While advances have been made towards understanding the molecular bases of these diseases, a complete understanding of the pathological mechanisms remains an unmet research goal that is essential for defining safer pharmacological therapies and prognostic factors, especially for the metastatic stage of these malignancies for which no effective therapies are currently being used. Glyoxalases, consisting of glyoxalase 1 (Glo1) and glyoxalase 2 (Glo2), are enzymes that catalyze the glutathione-dependent metabolism of cytotoxic methylglyoxal (MG), thus protecting against cellular damage and apoptosis. They are generally overexpressed in numerous cancers as a survival strategy by providing a safeguard through enhancement of MG detoxification. Increasing evidence suggests that glyoxalases, especially Glo1, play an important role in the initiation and progression of urological malignancies. In this review, we highlight the critical role of glyoxalases as regulators of tumorigenesis in the prostate through modulation of various critical signaling pathways, and provide an overview of the current knowledge on glyoxalases in bladder, kidney and testis cancers. We also discuss the promise and challenges for Glo1 inhibitors as future anti-prostate cancer (PCa) therapeutics and the potential of glyoxalases as biomarkers for PCa diagnosis.
Collapse
Affiliation(s)
- Cinzia Antognelli
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy.
| | | |
Collapse
|
14
|
Inhibition of GLO1 in Glioblastoma Multiforme Increases DNA-AGEs, Stimulates RAGE Expression, and Inhibits Brain Tumor Growth in Orthotopic Mouse Models. Int J Mol Sci 2018; 19:ijms19020406. [PMID: 29385725 PMCID: PMC5855628 DOI: 10.3390/ijms19020406] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 01/22/2018] [Accepted: 01/26/2018] [Indexed: 12/22/2022] Open
Abstract
Cancers that exhibit the Warburg effect may elevate expression of glyoxylase 1 (GLO1) to detoxify the toxic glycolytic byproduct methylglyoxal (MG) and inhibit the formation of pro-apoptotic advanced glycation endproducts (AGEs). Inhibition of GLO1 in cancers that up-regulate glycolysis has been proposed as a therapeutic targeting strategy, but this approach has not been evaluated for glioblastoma multiforme (GBM), the most aggressive and difficult to treat malignancy of the brain. Elevated GLO1 expression in GBM was established in patient tumors and cell lines using bioinformatics tools and biochemical approaches. GLO1 inhibition in GBM cell lines and in an orthotopic xenograft GBM mouse model was examined using both small molecule and short hairpin RNA (shRNA) approaches. Inhibition of GLO1 with S-(p-bromobenzyl) glutathione dicyclopentyl ester (p-BrBzGSH(Cp)2) increased levels of the DNA-AGE N2-1-(carboxyethyl)-2′-deoxyguanosine (CEdG), a surrogate biomarker for nuclear MG exposure; substantially elevated expression of the immunoglobulin-like receptor for AGEs (RAGE); and induced apoptosis in GBM cell lines. Targeting GLO1 with shRNA similarly increased CEdG levels and RAGE expression, and was cytotoxic to glioma cells. Mice bearing orthotopic GBM xenografts treated systemically with p-BrBzGSH(Cp)2 exhibited tumor regression without significant off-target effects suggesting that GLO1 inhibition may have value in the therapeutic management of these drug-resistant tumors.
Collapse
|
15
|
Abdul-Maksoud RS, Elsayed WS, Elsayed RS. The influence of glyoxalase 1 gene polymorphism on its expression at different stages of breast cancer in Egyptian women. Genes Cancer 2017; 8:799-807. [PMID: 29321821 PMCID: PMC5755725 DOI: 10.18632/genesandcancer.163] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aim To assess the association of GLO1 C332C gene polymorphism with breast cancer risk at different stages of the disease and to investigate the effect of this gene polymorphism on its mRNA expression and enzyme activity. Methods GLO1 C332C gene polymorphism was analyzed by PCR-RFLP in 100 healthy controls and 200 patients with breast cancer (100 patients with stage I & II and 100 patients with stage III & IV). GLO1 mRNA expression was measured by real time PCR. Serum GLO1 enzyme activity was measured colorimetrically. Results GLO1 A allele was associated with increased risk of breast cancer [OR (95%CI)= 2.8(1.9-4.1), P < 0.001]. Its frequency was significantly higher among advanced stages of breast cancer compared with localized tumors (OR (95%CI)= 1.9(1.3-2.9), p < 0.001). GLO1 mRNA expression and enzyme activity were significantly higher in breast cancer patients compared to controls and they were much higher in the advanced stages of the disease (P < 0.001). Carriers of AA genotype showed higher GLO1 expression and enzyme activity compared with carriers of CC genotype. Conclusion GLO1 C332C SNP was associated with overexpression of GLO1 mRNA and higher enzyme activity in breast cancer patients suggesting its role in the development of breast cancer and its progression from localized to advanced.
Collapse
Affiliation(s)
| | - Walid Sh Elsayed
- Pathology Department, Faculty of Medicine, Zagazig University, Egypt
| | - Rasha S Elsayed
- General Surgery Department, Faculty of Medicine, Zagazig University, Egypt
| |
Collapse
|
16
|
Antognelli C, Ferri I, Bellezza G, Siccu P, Love HD, Talesa VN, Sidoni A. Glyoxalase 2 drives tumorigenesis in human prostate cells in a mechanism involving androgen receptor and p53-p21 axis. Mol Carcinog 2017; 56:2112-2126. [PMID: 28470764 DOI: 10.1002/mc.22668] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 04/26/2017] [Accepted: 05/01/2017] [Indexed: 12/20/2022]
Abstract
Glyoxalase 2 (Glo2), a metabolic enzyme, is overexpressed in some human cancers which suggests this enzyme may play a role in human tumorigenesis. In prostate cancer (PCa), the role of Glo2 has been scarcely investigated and there are no studies addressing a causative involvement of this protein in this neoplasia. Here, we examined the immunohistochemical profile of Glo2 in human PCa and benign adjacent tissues and investigated Glo2 involvement in PCa development in human prostate cell lines. PCa and matched adjacent normal tissues were obtained from paraffin sections of primary PCa from 20 patients who had undergone radical prostatectomy. Histopathological diagnosis was confirmed for each sample. Glo2 expression analysis was performed by immunohistochemistry in prostate tissues, and by qRT-PCR and immunoblotting in prostate cell lines. The causative and mechanistic role of Glo2 in prostate tumorigenesis was demonstrated by Glo2 ectopic expression/silencing and employing specific activators/inhibitors. Our results showed that Glo2 was selectively expressed in PCa but not in the luminal compartment of the adjacent benign epithelium consistently in all the examined 20 cases. Glo2 expression in PCa was dependent on androgen receptor (AR) and was aimed at stimulating cell proliferation and eluding apoptosis through a mechanism involving the p53-p21 axis. Glo2 was intensely expressed in the basal cells of benign glands but was not involved in PCa genesis. Our results demonstrate for the first time that Glo2 drives prostate tumorigenesis and suggest that it may represent a novel adjuvant marker in the pathological diagnosis of early PCa.
Collapse
Affiliation(s)
- Cinzia Antognelli
- Division of Biosciences and Medical Embryology, Department of Experimental Medicine, School of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Ivana Ferri
- Division of Anatomic Pathology and Histology, Department of Experimental Medicine, School of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Guido Bellezza
- Division of Anatomic Pathology and Histology, Department of Experimental Medicine, School of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Paola Siccu
- Division of Anatomic Pathology and Histology, Department of Experimental Medicine, School of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Harold D Love
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Vincenzo N Talesa
- Division of Biosciences and Medical Embryology, Department of Experimental Medicine, School of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Angelo Sidoni
- Division of Anatomic Pathology and Histology, Department of Experimental Medicine, School of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
17
|
Chen Y, Fang L, Li G, Zhang J, Li C, Ma M, Guan C, Bai F, Lyu J, Meng QH. Synergistic inhibition of colon cancer growth by the combination of methylglyoxal and silencing of glyoxalase I mediated by the STAT1 pathway. Oncotarget 2017; 8:54838-54857. [PMID: 28903386 PMCID: PMC5589625 DOI: 10.18632/oncotarget.18601] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 05/31/2017] [Indexed: 12/31/2022] Open
Abstract
Methylglyoxal (MG), an extremely reactive glucose metabolite, exhibits antitumor activity. Glyoxalase I (GLOI), which catalyzes MG metabolism, is associated with the progression of human malignancies. While the roles of MG or GLOI have been demonstrated in some types of cancer, their effects in colon cancer and the mechanisms underlying these effects remain largely unknown. For this study, MG and GLOI levels were manipulated in colon cancer cells and the effects on their viability, proliferation, apoptosis, migration, and invasion in vitro were quantified by Cell Counting Kit-8, colony formation assay, flow cytometry, and transwell assays. The expression levels of STAT1 pathway–associated proteins and mRNAs in these cells were quantified by western blot and qRT-PCR, respectively. The antitumor effects of MG and silencing of GLOI were investigated in vivo in a SW620 colon cancer xenograft model in BALB/c nude mice. Our findings demonstrate that MG in combination with silencing of GLOI synergistically inhibited the cancer cells’ proliferation, colony formation, migration, and invasion and induced apoptosis in vitro compared with the controls. Furthermore, these treatments up-regulated STAT1 and Bax while down-regulating Bcl-2 in vitro. MG treatment alone or in combination with silencing of GLOI also reduced the growth of the SW620 tumors in mice by up-regulation of STAT1 and Bax and down-regulation of Bcl-2. Taken together, our findings suggest that MG in combination with silencing of GLOI merits further evaluation as a targeted therapeutic strategy for colon cancer.
Collapse
Affiliation(s)
- Yuan Chen
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Lei Fang
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Gefei Li
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jiali Zhang
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Changxi Li
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Mengni Ma
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Chen Guan
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Fumao Bai
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jianxin Lyu
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Qing H Meng
- Department of Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
18
|
Chen Y, Fang L, Zhang J, Li G, Ma M, Li C, Lyu J, Meng QH. Blockage of Glyoxalase I Inhibits Colorectal Tumorigenesis and Tumor Growth via Upregulation of STAT1, p53, and Bax and Downregulation of c-Myc and Bcl-2. Int J Mol Sci 2017; 18:ijms18030570. [PMID: 28282916 PMCID: PMC5372586 DOI: 10.3390/ijms18030570] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 02/26/2017] [Accepted: 03/02/2017] [Indexed: 12/14/2022] Open
Abstract
GlyoxalaseI (GLOI) is an enzyme that catalyzes methylglyoxal metabolism. Overexpression of GLOI has been documented in numerous tumor tissues, including colorectal cancer (CRC). The antitumor effects of GLOI depletion have been demonstrated in some types of cancer, but its role in CRC and the mechanisms underlying this activity remain largely unknown. Our purpose was to investigate the antitumor effects of depleted GLOI on CRC in vitro and in vivo. RNA interference was used to deplete GLOI activity in four CRC cell lines. The cells' proliferation, apoptosis, migration, and invasion were assessed by using the Cell Counting Kit-8, plate colony formation assay, flow cytometry, and transwell assays. Protein and mRNA levels were analyzed by western blot and quantitative real-time PCR (qRT-PCR), respectively. The antitumor effect of GLOI depletion in vivo was investigated in a SW620 xenograft tumor model in BALB/c nude mice. Our results show that GLOI is over-expressed in the CRC cell lines. GLOI depletion inhibited the proliferation, colony formation, migration, and invasion and induced apoptosis of all CRC cells compared with the controls. The levels of signal transducer and activator of transcription 1 (STAT1), p53, and Bcl-2 assaciated X protein (Bax) were upregulated by GLOI depletion, while cellular homologue of avian myelocytomatosis virus oncogene (c-Myc) and B cell lymphoma/lewkmia-2 (Bcl-2) were downregulated. Moreover, the growth of SW620-induced CRC tumors in BALB/c nude mice was significantly attenuated by GLOI depletion. The expression levels of STAT1, p53, and Bax were increased and those of c-Myc and Bcl-2 were decreased in the GLOI-depleted tumors. Our findings demonstrate that GLOI depletion has an antitumor effect through the STAT1 or p53 signaling pathways in CRC, suggesting that GLOI is a potential therapeutic target.
Collapse
Affiliation(s)
- Yuan Chen
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Lei Fang
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Jiali Zhang
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Gefei Li
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Mengni Ma
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Changxi Li
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Jianxin Lyu
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Qing H Meng
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China.
- Department of Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
19
|
Talesa VN, Ferri I, Bellezza G, Love HD, Sidoni A, Antognelli C. Glyoxalase 2 Is Involved in Human Prostate Cancer Progression as Part of a Mechanism Driven By PTEN/PI3K/AKT/mTOR Signaling With Involvement of PKM2 and ERα. Prostate 2017; 77:196-210. [PMID: 27696457 DOI: 10.1002/pros.23261] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 09/12/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND Glyoxalase 2 (Glo2), together with glyoxalase 1 (Glo1), forms the main scavenging system of methylglyoxal, a potent pro-apoptotic agent mainly generated by glycolysis. An increased rate of glycolysis is a well known signature of cancer cells. As a survival strategy, Glo1 is overexpressed in many human malignant cells, including prostate cancer (PCa), where it plays a crucial role in progression. No information is available on the role of Glo2 in the same ambit. PCa is the most common malignancy affecting men in the western world. Progression to a lethal hormone-refractory PCa represents the major concern in this pathology. Therefore, a deeper understanding of the molecular mechanisms underlying PCa invasiveness and metastasis is urgently needed in order to develop novel therapeutic targets for this incurable state of the malignancy. METHODS Glo2 and Glo1 expression was examined in clinical samples of PCa by immunohistochemistry and in different PCa cell models by western blotting and quantitative real-time polymerase chain reaction. Gene silencing/overexpression and scavenging/inhibitory agents were used for functional analyses. RESULTS We demonstrated that Glo2, together with Glo1, represents a novel mechanism in PCa progression as part of a pathway driven by PTEN/PI3K/AKT/mTOR signaling with involvement of PKM2 and ERα. Importantly, Glo1/Glo2 silencing did not alter the behavior of benign cells. CONCLUSIONS Targeting glyoxalases metabolic pathway may represent a strategy to selectively inhibit advanced PCa. Prostate 77:196-210, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Vincenzo N Talesa
- Division of Biosciences and Medical Embryology, Department of Experimental Medicine, School of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Ivana Ferri
- Division of Anatomic Pathology and Histology, Department of Experimental Medicine, School of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Guido Bellezza
- Division of Anatomic Pathology and Histology, Department of Experimental Medicine, School of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Harold D Love
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Angelo Sidoni
- Division of Anatomic Pathology and Histology, Department of Experimental Medicine, School of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Cinzia Antognelli
- Division of Biosciences and Medical Embryology, Department of Experimental Medicine, School of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
20
|
Falone S, Santini S, di Loreto S, Cordone V, Grannonico M, Cesare P, Cacchio M, Amicarelli F. Improved Mitochondrial and Methylglyoxal-Related Metabolisms Support Hyperproliferation Induced by 50 Hz Magnetic Field in Neuroblastoma Cells. J Cell Physiol 2016; 231:2014-25. [PMID: 26757151 DOI: 10.1002/jcp.25310] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 01/08/2016] [Indexed: 12/27/2022]
Abstract
Extremely low frequency magnetic fields (ELF-MF) are common environmental agents that are suspected to promote later stages of tumorigenesis, especially in brain-derived malignancies. Even though ELF magnetic fields have been previously linked to increased proliferation in neuroblastoma cells, no previous work has studied whether ELF-MF exposure may change key biomolecular features, such as anti-glycative defence and energy re-programming, both of which are currently considered as crucial factors involved in the phenotype and progression of many malignancies. Our study investigated whether the hyperproliferation that is induced in SH-SY5Y human neuroblastoma cells by a 50 Hz, 1 mT ELF magnetic field is supported by an improved defense towards methylglyoxal (MG), which is an endogenous cancer-static and glycating α-oxoaldehyde, and by rewiring of energy metabolism. Our findings show that not only the ELF magnetic field interfered with the biology of neuron-derived malignant cells, by de-differentiating further the cellular phenotype and by increasing the proliferative activity, but also triggered cytoprotective mechanisms through the enhancement of the defense against MG, along with a more efficient management of metabolic energy, presumably to support the rapid cell outgrowth. Intriguingly, we also revealed that the MF-induced bioeffects took place after an initial imbalance of the cellular homeostasis, which most likely created a transient unstable milieu. The biochemical pathways and molecular targets revealed in this research could be exploited for future approaches aimed at limiting or suppressing the deleterious effects of ELF magnetic fields. J. Cell. Physiol. 231: 2014-2025, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Stefano Falone
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Silvano Santini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Silvia di Loreto
- Institute of Translational Pharmacology (IFT)-CNR, L'Aquila, Italy
| | - Valeria Cordone
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Marta Grannonico
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Patrizia Cesare
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Marisa Cacchio
- Department of Biomedical Sciences, University "G. d'Annunzio", Via dei Vestini, Chieti Scalo (CH), Italy
| | - Fernanda Amicarelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.,Institute of Translational Pharmacology (IFT)-CNR, L'Aquila, Italy
| |
Collapse
|
21
|
Chen CC, Wu ML, Doerksen RJ, Ho CT, Huang TC. Andrographolide induces apoptosis via down-regulation of glyoxalase 1 and HMG-CoA reductase in HL-60 cells. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.01.048] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
22
|
Polymorphisms of the receptor for advanced glycation end-products and glyoxalase I in patients with renal cancer. Tumour Biol 2014; 36:2121-6. [DOI: 10.1007/s13277-014-2821-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Accepted: 11/06/2014] [Indexed: 10/24/2022] Open
|
23
|
Inhibition by active site directed covalent modification of human glyoxalase I. Bioorg Med Chem 2014; 22:3301-8. [DOI: 10.1016/j.bmc.2014.04.055] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 04/18/2014] [Accepted: 04/28/2014] [Indexed: 11/22/2022]
|
24
|
Zhou Y, Guo T, Li X, Dong Y, Galatsis P, Johnson DS, Pan Z. Discovery of selective 2,4-diaminopyrimidine-based photoaffinity probes for glyoxalase I. MEDCHEMCOMM 2014. [DOI: 10.1039/c3md00286a] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
L1-Bpyne was discovered as a potent inhibitor and cell permeable probe of glyoxalase I.
Collapse
Affiliation(s)
- Yiqing Zhou
- Key laboratory of Chemical Genomics
- School of Chemical Biology and Biotechnology
- Peking University
- Xili University Town
- Shenzhen
| | - Tianlin Guo
- Key laboratory of Chemical Genomics
- School of Chemical Biology and Biotechnology
- Peking University
- Xili University Town
- Shenzhen
| | - Xitao Li
- Key laboratory of Chemical Genomics
- School of Chemical Biology and Biotechnology
- Peking University
- Xili University Town
- Shenzhen
| | - Yi Dong
- Key laboratory of Chemical Genomics
- School of Chemical Biology and Biotechnology
- Peking University
- Xili University Town
- Shenzhen
| | - Paul Galatsis
- Neuroscience Medicinal Chemistry and Chemical Biology
- Pfizer Worldwide Research and Development
- Cambridge
- USA
| | - Douglas S. Johnson
- Neuroscience Medicinal Chemistry and Chemical Biology
- Pfizer Worldwide Research and Development
- Cambridge
- USA
| | - Zhengying Pan
- Key laboratory of Chemical Genomics
- School of Chemical Biology and Biotechnology
- Peking University
- Xili University Town
- Shenzhen
| |
Collapse
|
25
|
Baunacke M, Horn LC, Trettner S, Engel KMY, Hemdan NYA, Wiechmann V, Stolzenburg JU, Bigl M, Birkenmeier G. Exploring glyoxalase 1 expression in prostate cancer tissues: targeting the enzyme by ethyl pyruvate defangs some malignancy-associated properties. Prostate 2014; 74:48-60. [PMID: 24105621 DOI: 10.1002/pros.22728] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Accepted: 08/19/2013] [Indexed: 11/06/2022]
Abstract
BACKGROUND The glyoxalase (GLO)1 is part of a ubiquitous detoxification system in the glycolytic pathway of normal and tumor cells. It protects against cellular damage caused by cytotoxic metabolites. METHODS Aiming at exploring the role of GLO1 in prostate cancer, we evaluated and targeted the expression of GLO1 in prostate cancer tissues and cell lines and analyzed its correlation with grading systems and tumor growth indices. RESULTS Immunohistochemical studies on 37 prostate cancer specimens revealed a positive correlation between Helpap-grading and the cytoplasmic (P = 0.002)/nuclear (P = 0.006) GLO1 level. A positive correlation between Ki-67 proliferation marker and the cytoplasmic GLO1 (P = 0.006) was evident. Furthermore, the highest GLO1 level was detected in the androgen-sensitive LNCaP compared to the androgen-independent Du-145 and PC-3 prostate cell lines and the breast cancer cell MCF-7, both at protein and mRNA level. Treating cancer cells with ethyl pyruvate was found to defang some malignancy-associated properties of cancer cells including proliferation, invasion and anchorage-independent growth. In vitro results revealed that the potency of ethyl pyruvate is increased when cells are metabolically activated by growth stimulators, for example, by fetal calf serum, dihydrotestosterone, tumor growth factor-β1 and leptin. CONCLUSIONS The positive correlation of GLO1 expression level in prostate cancer tissues with the pathological grade and proliferation rate may assign GLO1 as a risk factor for prostate cancer development and progression. Furthermore, our data indicate that inhibitors of GLO1 might be useful to decelerate the cancer cell growth by a novel therapeutic approach that we may call "induced metabolic catastrophe."
Collapse
Affiliation(s)
- Martin Baunacke
- Institute of Biochemistry, University of Leipzig, Leipzig, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Antognelli C, Mezzasoma L, Fettucciari K, Mearini E, Talesa VN. Role of glyoxalase I in the proliferation and apoptosis control of human LNCaP and PC3 prostate cancer cells. Prostate 2013; 73:121-32. [PMID: 22653787 DOI: 10.1002/pros.22547] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Accepted: 05/14/2012] [Indexed: 11/10/2022]
Abstract
BACKGROUND Glyoxalase I (GLOI) detoxifies reactive dicarbonyls, as methylglyoxal (MG) that, directly or through the formation of MG-derived adducts, is a growth inhibitor and apoptosis inducer. GLOI has been considered a general marker of cell proliferation, but a direct link between the two has yet to be demonstrated. The aim of the present work was to clarify whether GLOI was involved in the proliferation control of LNCaP and PC3 human prostate cancer cells or might play a different role in the growth regulation of these cells. METHODS RNA interference was used to study the role of GLOI in cell proliferation or apoptosis. Cell proliferation was evaluated by [3H]thymidine incorporation assay and flow cytometry, that was also used to analyze apoptosis. Real-time TaqMan polymerase chain reaction and spectrophotometric analyses were used to study transcript levels or specific activity, respectively. Proteins levels were analyzed by Western blot. MG was measured by high-performance liquid chromatography. RESULTS We found that GLOI is not implicated in the proliferation control of LNCaP and PC3 cells but plays a role in the apoptosis of invasive prostate cancer PC3 cells, through a mechanism involving a specific MG-adduct and NF-kB signaling pathway. CONCLUSIONS Our data represent the first systematic demonstration that GLOI cannot be considered a general marker of cell proliferation and that acts as a pro-survival factor in invasive PC3 cells by elusing apoptosis. GLOI may be involved in prostate cancer progression, via the control of key molecules in the mitochondrial apoptotic mechanism, through NF-kB signaling pathway.
Collapse
Affiliation(s)
- Cinzia Antognelli
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| | | | | | | | | |
Collapse
|
27
|
Ferreira L, Fuentes-Calvo I, Muñoz-Félix JM, Muñiz-Martín C, Sánchez-Juanes F, Raposo C, González-Buitrago JM, López-Novoa JM, Martínez-Salgado C. Functional specific roles of H-ras and N-ras. A proteomic approach using knockout cell lines. Electrophoresis 2012; 33:1385-96. [PMID: 22648805 DOI: 10.1002/elps.201100606] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Ras small GTPases function as transducers of extracellular signals regulating cell survival, growth and differentiation. There are three major ras isoforms: H-, N- and K-Ras. To improve the understanding of H- and N-Ras protein signalling networks, we compared total proteome changes in mouse embryonic fibroblasts knock out for H-ras and/or N-ras, using proteomics tools combining 2DE, semi-quantitative image analysis, in-gel trypsin digestion and mass spectrometry. There are four up-regulated proteins due to the loss of expression of H-Ras (including cyclin-dependent kinase inhibitor 2A) and eight down-regulated (including stress-70 protein, dihydropyrimidinase-related-protein 3, heat shock cognate 71 kDa protein, tropomyosin beta chain, Rho GDP-dissociation inhibitor 1) and six up-regulated proteins (e.g. leukocyte elastase inhibitor A, L-lactate dehydrogenase B chain, c-Myc-responsive protein Rcl, interleukin-1 receptor antagonist protein) due to the loss of expression of both N- and H-Ras. Most of these proteins are related to Ras signalling in one way or another. Changes in expression of some of these proteins were further confirmed by Western blot. This proteomic comparative analysis from loss of function of H- and N-Ras knockout fibroblasts yields interpretable data to elucidate the differential protein expression, and contributes to evaluate the possibilities for physiological and therapeutic targets.
Collapse
Affiliation(s)
- Laura Ferreira
- Unidad de Investigación, Hospital Universitario de Salamanca, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Dong L, Zhou Q, Zhang Z, Zhu Y, Duan T, Feng Y. Metformin sensitizes endometrial cancer cells to chemotherapy by repressing glyoxalase I expression. J Obstet Gynaecol Res 2012; 38:1077-85. [PMID: 22540333 DOI: 10.1111/j.1447-0756.2011.01839.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
AIM Metformin plays an important role in the inhibition of cancer cell growth and prolongs remission durations. It reverses progestin-resistance in endometrial cancer cells by downregulating glyoxalase I (GloI) expression. This study aimed to investigate the effect of metformin on endometrial cancer cell chemotherapeutic sensitivity and explore the underlying molecular mechanisms. MATERIAL AND METHODS MTT assay was performed to determine the rate of cell death after cisplatin and paclitaxel with or without metformin. Western blot was carried out to analyze GloI expression. SiRNA-targeting of GloI was used to knockdown GloI expression before further treatment with chemotherapeutic agents to examine the effect of GloI downregulation on chemotherapy-induced cell killing. In addition, plasmid transfection was used to overexpress GloI and determine whether high GloI levels blocked metformin-enhanced cell sensitivity to chemotherapy. PCR was used to analyze the efficiency of RNA interference and plasmid transfection. RESULTS The addition of metformin enhanced the sensitivity of endometrial cells to cisplatin and paclitaxel, which was associated with reduced levels of GloI expression. Moreover, low-dose chemotherapeutic drugs alone could not significantly reduce GloI expression, whereas the addition of metformin potently downregulated GloI protein levels. Cisplatin and paclitaxel markedly inhibited the proliferative ability of GloI-depleted endometrial cancer cells. However, the overexpression of GloI abolished the effect of metformin-enhanced cell sensitivity to chemotherapeutic drugs. CONCLUSION Metformin enhances the rate of cell-killing induced by chemotherapeutic agents by repressing GloI expression.
Collapse
Affiliation(s)
- Lingling Dong
- Graduate School, Shanghai Medical College, Fudan University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
29
|
Notarnicola M, Caruso MG, Tutino V, Guerra V, Misciagna G. Low red blood cell levels of deglycating enzymes in colorectal cancer patients. World J Gastroenterol 2011; 17:329-33. [PMID: 21253391 PMCID: PMC3022292 DOI: 10.3748/wjg.v17.i3.329] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Revised: 07/28/2010] [Accepted: 08/05/2010] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate Glyoxalase I and fructosamine-3-kinase (FN3K) activity in red blood cells from patients with colorectal adenomas and cancer.
METHODS: Thirty three consecutive subjects with one or more histologically confirmed colorectal adenomatous polyps, 16 colorectal cancer patients and a group of 11 control subjects with normal colonoscopy were included in the study. Glyoxalase I and FN3K activities were measured in red blood cells using a spectrophotometric and radiometric assay, respectively.
RESULTS: A significant reduction in both Glyoxalase I and FN3K activity was detected in patients with tumors compared to patients with adenomas and the controls. Erythrocyte Glyoxalase I activity in colorectal cancer was approximately 6 times lower than that detected in patients with adenoma (0.022 ± 0.01 mmol/min per milliliter vs 0.128 ± 0.19 mmol/min per milliliter of red blood cells, P = 0.003, Tukey’s test). FN3K activity in red blood cells from patients with colon cancer was approximately 2 times lower than that detected in adenoma patients (19.55 ± 6.4 pmol/min per milliliter vs 38.6 ± 31.7 pmol/min per milliliter of red blood cells, P = 0.04, Tukey’s test).
CONCLUSION: These findings suggest that deglycating enzymes may be involved in the malignant transformation of colon mucosa.
Collapse
|
30
|
Rodríguez-Ulloa A, Ramos Y, Gil J, Perera Y, Castellanos-Serra L, García Y, Betancourt L, Besada V, González LJ, Fernández-de-Cossio J, Sanchez A, Serrano JM, Farina H, Alonso DF, Acevedo BE, Padrón G, Musacchio A, Perea SE. Proteomic profile regulated by the anticancer peptide CIGB-300 in non-small cell lung cancer (NSCLC) cells. J Proteome Res 2010; 9:5473-83. [PMID: 20804217 DOI: 10.1021/pr100728v] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
CIGB-300 is a proapoptotic peptide-based drug that abrogates the CK2-mediated phosphorylation. This peptide has antineoplastic effect on lung cancer cells in vitro and in vivo. To understand the mechanisms involved on such anticancer activity, the NCI-H125 cell line proteomic profile after short-term incubation (45 min) with CIGB-300 was investigated. As determined by 2-DE or 2D-LC-MS/MS, 137 proteins changed their abundances more than 2-fold in response to the CIGB-300 treatment. The expression levels of proteins related to ribosome biogenesis, metastasis, cell survival and proliferation, apoptosis, and drug resistance were significantly modulated by the presence of CIGB-300. The protein translation process was the most affected (23% of the identified proteins). From the proteome analysis of the NCI-H125 cell line, novel potentialities for CIGB-300 as anticancer agent were evidenced.
Collapse
Affiliation(s)
- Arielis Rodríguez-Ulloa
- Department of Bioinformatics, Center for Genetic Engineering and Biotechnology, Havana, Cuba.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Naidu R, Har YC, Taib NAM. Glyoxalase I Ala111Glu gene polymorphism: No association with breast cancer risk but correlated with absence of progesterone receptor. Pathol Int 2010; 60:614-20. [DOI: 10.1111/j.1440-1827.2010.02568.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
32
|
Kim ST, Moon W, Chae Y, Kim YJ, Lee H, Park HJ. The effect of electroaucpuncture for 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced proteomic changes in the mouse striatum. J Physiol Sci 2010; 60:27-34. [PMID: 19760485 PMCID: PMC10717007 DOI: 10.1007/s12576-009-0061-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Accepted: 08/23/2009] [Indexed: 11/28/2022]
Abstract
Using a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) Parkinson's disease mouse model, we investigated protein expression changes associated with the action of electroacupuncture (EA) in the mouse striatum. Twelve-week-old male C57BL/6 mice were injected intraperitoneally with 30 mg/kg of MPTP at 24-h intervals for 5 days, and the 100-Hz EA stimulation was performed at GB34 and GB39 once a day for 12 days consecutively from the first injection. With the EA, the MPTP-induced dopaminergic neuronal destruction was reduced. Of the 13 proteins that were differentially expressed between control and MPTP treated mice, cytosolic malate dehydrogenase, munc18-1, and hydroxyacylglutathione hydrolase, which were increased by MPTP, and cytochrome c oxidase subunit Vb, which was decreased by MPTP, were restored to the level of the saline group after EA treatment. These proteins are likely related to cellular metabolism. Altogether, we propose that the EA may exert neuroprotective effects in mice striatum through reducing MPTP-induced toxicity such as oxidative stress.
Collapse
Affiliation(s)
- Seung-Tae Kim
- Division of Meridian and Structural Medicine, School of Korean Medicine, Pusan National University, Beomeo-ri, Mulgeum-eup, Yangsan-si, Gyeongsagnam-do 626-870 Republic of Korea
| | - Woongjoon Moon
- Acupuncture and Meridian Science Research Center (AMSRC), Kyung Hee University, 1 Hoegi-dong, Dongdaemoon-gu, Seoul, 130-701 Republic of Korea
| | - Younbyoung Chae
- Acupuncture and Meridian Science Research Center (AMSRC), Kyung Hee University, 1 Hoegi-dong, Dongdaemoon-gu, Seoul, 130-701 Republic of Korea
| | - Youn Jung Kim
- East-West Nursing Research Institute, Kyung Hee University, 1 Hoegi-dong, Dongdaemoon-gu, Seoul, 130-701 Republic of Korea
| | - Hyejung Lee
- Acupuncture and Meridian Science Research Center (AMSRC), Kyung Hee University, 1 Hoegi-dong, Dongdaemoon-gu, Seoul, 130-701 Republic of Korea
| | - Hi-Joon Park
- Acupuncture and Meridian Science Research Center (AMSRC), Kyung Hee University, 1 Hoegi-dong, Dongdaemoon-gu, Seoul, 130-701 Republic of Korea
| |
Collapse
|
33
|
Limphong P, McKinney RM, Adams NE, Bennett B, Makaroff CA, Gunasekera T, Crowder MW. Human glyoxalase II contains an Fe(II)Zn(II) center but is active as a mononuclear Zn(II) enzyme. Biochemistry 2009; 48:5426-34. [PMID: 19413286 DOI: 10.1021/bi9001375] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Human glyoxalase II (Glx2) was overexpressed in rich medium and in minimal medium containing zinc, iron, or cobalt, and the resulting Glx2 analogues were characterized using metal analyses, steady-state and pre-steady-state kinetics, and NMR and EPR spectroscopies to determine the nature of the metal center in the enzyme. Recombinant human Glx2 tightly binds nearly 1 equiv each of Zn(II) and Fe. In contrast to previous reports, this study demonstrates that an analogue containing 2 equiv of Zn(II) cannot be prepared. EPR studies suggest that most of the iron in recombinant Glx2 is Fe(II). NMR studies show that Fe(II) binds to the consensus Zn(2) site in Glx2 and that this site can also bind Co(II) and Ni(II), suggesting that Zn(II) binds to the consensus Zn(1) site. The NMR studies also reveal the presence of a dinuclear Co(II) center in Co(II)-substituted Glx2. Steady-state and pre-steady-state kinetic studies show that Glx2 containing only 1 equiv of Zn(II) is catalytically active and that the metal ion in the consensus Zn(2) site has little effect on catalytic activity. Taken together, these studies suggest that Glx2 contains a Fe(II)Zn(II) center in vivo but that the catalytic activity is due to Zn(II) in the Zn(1) site.
Collapse
Affiliation(s)
- Pattraranee Limphong
- Department of Chemistry and Biochemistry, 160 Hughes Hall, Miami University, Oxford, Ohio 45056, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
More SS, Vince R. Inhibition of Glyoxalase I: The First Low-Nanomolar Tight-Binding Inhibitors. J Med Chem 2009; 52:4650-6. [DOI: 10.1021/jm900382u] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Swati S. More
- Center for Drug Design, Academic Health Center, and Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, 8-123A Weaver Densford Hall, 308 Harvard Street SE, Minneapolis, Minnesota 55455
| | - Robert Vince
- Center for Drug Design, Academic Health Center, and Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, 8-123A Weaver Densford Hall, 308 Harvard Street SE, Minneapolis, Minnesota 55455
| |
Collapse
|
35
|
Germanová A, Germanová A, Tesarová P, Jáchymová M, Zvára K, Zima T, Kalousová M. Glyoxalase I Glu111Ala polymorphism in patients with breast cancer. Cancer Invest 2009; 27:655-60. [PMID: 19452310 DOI: 10.1080/07357900802350822] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Effect of advanced glycation end products (AGEs) in the pathogenesis of cancer could be diminished by interaction with soluble RAGE or by reducing AGE-precursors via glyoxalase I. Glu111Ala polymorphism of glyoxalase I gene, AGEs, and sRAGE serum levels were studied in 113 breast cancer patients and in 58 controls. Higher frequency of the mutated C allele was found in patients with negative estrogen receptors and in patients in clinical stage III compared to controls (P< 0.05). The presence of the C allele could represent a negative prognostic factor; however, further studies are needed to confirm this hypothesis.
Collapse
Affiliation(s)
- Alexandra Germanová
- Institute of Clinical Chemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | | | | | | | | | | | | |
Collapse
|
36
|
Arabidopsis thaliana GLX2-1 contains a dinuclear metal binding site, but is not a glyoxalase 2. Biochem J 2009; 417:323-30. [PMID: 18782082 DOI: 10.1042/bj20081151] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In an effort to probe the structure and function of a predicted mitochondrial glyoxalase 2, GLX2-1, from Arabidopsis thaliana, GLX2-1 was cloned, overexpressed, purified and characterized using metal analyses, kinetics, and UV-visible, EPR, and (1)H-NMR spectroscopies. The purified enzyme was purple and contained substoichiometric amounts of iron and zinc; however, metal-binding studies reveal that GLX2-1 can bind nearly two equivalents of either iron or zinc and that the most stable analogue of GLX2-1 is the iron-containing form. UV-visible spectra of the purified enzyme suggest the presence of Fe(II) in the protein, but the Fe(II) can be oxidized over time or by the addition of metal ions to the protein. EPR spectra revealed the presence of an anti-ferromagnetically-coupled Fe(III)Fe(II) centre and the presence of a protein-bound high-spin Fe(III) centre, perhaps as part of a FeZn centre. No paramagnetically shifted peaks were observed in (1)H-NMR spectra of the GLX2-1 analogues, suggesting low amounts of the paramagnetic, anti-ferromagnetically coupled centre. Steady-state kinetic studies with several thiolester substrates indicate that GLX2-1 is not a GLX2. In contrast with all of the other GLX2 proteins characterized, GLX2-1 contains an arginine in place of one of the metal-binding histidine residues at position 246. In order to evaluate further whether Arg(246) binds metal, the R246L mutant was prepared. The metal binding results are very similar to those of native GLX2-1, suggesting that a different amino acid is recruited as a metal-binding ligand. These results demonstrate that Arabidopsis GLX2-1 is a novel member of the metallo-beta-lactamase superfamily.
Collapse
|
37
|
Su Z, Sukdeo N, Honek JF. 15N−1H HSQC NMR Evidence for Distinct Specificity of Two Active Sites in Escherichia coli Glyoxalase I. Biochemistry 2008; 47:13232-41. [DOI: 10.1021/bi8013278] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Zhengding Su
- Department of Chemistry, University of Waterloo, Waterloo ON N2L 3G1, Canada
| | - Nicole Sukdeo
- Department of Chemistry, University of Waterloo, Waterloo ON N2L 3G1, Canada
| | - John F. Honek
- Department of Chemistry, University of Waterloo, Waterloo ON N2L 3G1, Canada
| |
Collapse
|
38
|
Kueper T, Grune T, Muhr GM, Lenz H, Wittern KP, Wenck H, Stäb F, Blatt T. Modification of vimentin: a general mechanism of nonenzymatic glycation in human skin. Ann N Y Acad Sci 2008; 1126:328-32. [PMID: 18448838 DOI: 10.1196/annals.1433.039] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
In a recent study, we were able to show that the intermediate filament protein vimentin aggregates in human dermal fibroblasts because of modification by the advanced glycation endproduct carboxymethyllysine (CML). In this work, we investigated the formation of intracellular CML in relation to the concentration of glucose in the culture medium. The natural degradation product of glucose, methylglyoxal, was able to induce the aggregation of vimentin. This dicarbonyl leads to the formation of the modifications MG-H1 and carboxyethyllysine (CEL) as a result of the reaction with arginine and lysine residues of proteins. Furthermore, we found that the protein vimentin was modified, not only by CML and CEL, but also by pentosidine and pyrraline. These findings underline the special position of vimentin as a preferential target of the Maillard reaction in human skin.
Collapse
|
39
|
More SS, Vince R. A metabolically stable tight-binding transition-state inhibitor of glyoxalase-I. Bioorg Med Chem Lett 2006; 16:6039-42. [PMID: 16997560 DOI: 10.1016/j.bmcl.2006.08.121] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2006] [Revised: 08/29/2006] [Accepted: 08/29/2006] [Indexed: 10/24/2022]
Abstract
The design, synthesis, and enzyme kinetics evaluation of a transition-state inhibitor of glyoxalase-I is described. The union of the hydroxamic acid zinc-chelator with a urea isostere for the glu-cys amide bond led to a glutathione analog which retained inhibitory potency toward glyoxalase-I while possessing resistance toward gamma-glutamyltranspeptidase mediated breakdown. This compound is viewed as a potential lead for the development of second-generation glyoxalase-I inhibitors wherein, the problems pertaining to metabolism and selectivity are overcome.
Collapse
Affiliation(s)
- Swati S More
- Center for Drug Design, Academic Health Center, and Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, 8-123A WDH, 308 Harvard St SE, Minneapolis, MN 55455, USA
| | | |
Collapse
|