1
|
Wang Z, Liu S, Zhang M, Liu M. Dual roles of methylglyoxal in cancer. Front Oncol 2025; 15:1557162. [PMID: 40352588 PMCID: PMC12061732 DOI: 10.3389/fonc.2025.1557162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 04/04/2025] [Indexed: 05/14/2025] Open
Abstract
Cancer treatment currently includes a variety of approaches. Chemotherapy, targeted therapy, and immunotherapy are combined based on cancer characteristics to develop personalized treatment plans. However, drug resistance can hinder the progress of treatment over time. Methylglyoxal (MG) is a metabolite with hormesis, exhibiting both pro-tumor and anti-tumor actions depending on its concentration during cancer progression. The MG-related metabolic pathway is being explored in the development of anti-cancer drugs, focusing on reducing MG stress or exploiting its cytotoxic effects to inhibit cancer progression. This article investigates the dual role of MG in cancer, emphasizing its effects on cell metabolism and tumor progression. It proposes MG capture therapy for the pre-cancerous stage and MG toxicity therapy for the cancer stage, contributing to the development of precise and individualized cancer treatments.
Collapse
Affiliation(s)
| | | | | | - Min Liu
- Department of Oncology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| |
Collapse
|
2
|
Zhang X, Gschwind J, Erben V, Bennewitz K, Li X, Sticht C, Poschet G, Hausser I, Fleming T, Szendroedi J, Nawroth PP, Kroll J. Endogenous acrolein accumulation in akr7a3 mutants causes microvascular dysfunction due to increased arachidonic acid metabolism. Redox Biol 2025; 83:103639. [PMID: 40258306 PMCID: PMC12051060 DOI: 10.1016/j.redox.2025.103639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/14/2025] [Accepted: 04/14/2025] [Indexed: 04/23/2025] Open
Abstract
Acrolein (ACR) is an endogenous reactive unsaturated aldehyde that can be detoxified by the aldo-keto reductase (AKR) enzyme system. While it has been shown that accumulation of ACR is associated with several health problems, including inflammation, oxidative stress, and cardiovascular disease the study aimed to analyze whether an endogenous accumulation of ACR is causal for vascular dysfunction in an akr7a3 mutant zebrafish model. Enlargement of the hyaloid and retinal vasculature, as well as alterations in the larval pronephros and thickening of the glomerular basement membrane in the adult kidney were found upon ACR accumulation. Transcriptomic and metabolomic analyses, followed by functional validation, revealed that the up-regulation of genes controlling the arachidonic acid metabolism and activation of the leukotriene pathway are responsible for the observed microvascular changes. In conclusion, the data have identified an intrinsic function of ACR in akr7a3 mutants that activates the arachidonic acid metabolism and subsequently disrupts vascular integrity by promoting an inflammatory response. Thus, ACR is causal in the development of vascular disease.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Vascular Biology, ECAS, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Johannes Gschwind
- Department of Vascular Biology, ECAS, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Vanessa Erben
- Department of Vascular Biology, ECAS, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Katrin Bennewitz
- Department of Vascular Biology, ECAS, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Xiaogang Li
- Department of Vascular Biology, ECAS, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Carsten Sticht
- NGS Core Facility, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Gernot Poschet
- Metabolomics Core Technology Platform, Centre for Organismal Studies, Heidelberg University, Heidelberg, 69120, Germany
| | - Ingrid Hausser
- Institute of Pathology IPH, EM Lab, Heidelberg University Hospital, Heidelberg, 69120, Germany
| | - Thomas Fleming
- Department of Internal Medicine I and Clinical Chemistry, Heidelberg University Hospital, Heidelberg, 69120, Germany
| | - Julia Szendroedi
- Department of Internal Medicine I and Clinical Chemistry, Heidelberg University Hospital, Heidelberg, 69120, Germany
| | - Peter Paul Nawroth
- Medical Clinic and Polyclinic II, University Hospital Dresden, Dresden, 01307, Germany
| | - Jens Kroll
- Department of Vascular Biology, ECAS, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany.
| |
Collapse
|
3
|
Akbari MS, Joyce LR, Spencer BL, Brady A, McIver KS, Doran KS. Identification of glyoxalase A in group B Streptococcus and its contribution to methylglyoxal tolerance and virulence. Infect Immun 2025; 93:e0054024. [PMID: 40008888 PMCID: PMC11977320 DOI: 10.1128/iai.00540-24] [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: 12/18/2024] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Group B Streptococcus (GBS) is a Gram-positive pathobiont that commonly colonizes the gastrointestinal and lower female genital tracts but can cause sepsis and pneumonia in newborns and is a leading cause of neonatal meningitis. Despite the resulting disease severity, the pathogenesis of GBS is not completely understood, especially during the early phases of infection. To investigate GBS factors necessary for bloodstream survival, we performed a transposon (Tn) mutant screen in our bacteremia infection model using a GBS mariner transposon mutant library previously developed by our group. We identified significantly underrepresented mutations in 623 genes that contribute to survival in the blood, including those encoding known virulence factors such as capsule, the β-hemolysin, and inorganic metal ion transport systems. Most of the underrepresented genes have not been previously characterized or studied in GBS, including gloA and gloB, which are homologs for genes involved in methylglyoxal (MG) detoxification. MG is a byproduct of glycolysis and a highly reactive toxic aldehyde that is elevated in immune cells during infection. Here, we observed MG sensitivity across multiple GBS isolates and confirmed that gloA contributes to MG tolerance and invasive GBS infection. We show specifically that gloA contributes to GBS survival in the presence of neutrophils and depleting neutrophils in mice abrogates the decreased survival and infection of the gloA mutant. The requirement of the glyoxalase pathway during GBS infection suggests that MG detoxification is important for bacterial survival during host-pathogen interactions.IMPORTANCEA transposon-mutant screen of group B Streptococcus (GBS) in a bacteremia mouse model of infection revealed virulence factors known to be important for GBS survival such as the capsule, β-hemolysin/cytolysin, and genes involved in metal homeostasis. Many uncharacterized factors were also identified including genes that are part of the metabolic pathway that breaks down methylglyoxal (MG). The glyoxalase pathway is the most ubiquitous metabolic pathway for MG breakdown and is only a two-step process using glyoxalase A (gloA) and B (gloB) enzymes. MG is a highly reactive byproduct of glycolysis and is made by most cells. Here, we show that in GBS, the first enzyme in the glyoxalase pathway, encoded by gloA, contributes to MG resistance and blood survival. We further demonstrate that GloA contributes to GBS survival against neutrophils in vitro and in vivo and, therefore, is an important virulence factor required for invasive infection.
Collapse
Affiliation(s)
- Madeline S. Akbari
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Luke R. Joyce
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Brady L. Spencer
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Amanda Brady
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kevin S. McIver
- Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| | - Kelly S. Doran
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
4
|
Iozzo M, Pardella E, Giannoni E, Chiarugi P. The role of protein lactylation: A kaleidoscopic post-translational modification in cancer. Mol Cell 2025; 85:1263-1279. [PMID: 40073861 DOI: 10.1016/j.molcel.2025.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/18/2024] [Accepted: 02/14/2025] [Indexed: 03/14/2025]
Abstract
The recently discovered lysine lactylation represents a critical post-translational modification with widespread implications in epigenetics and cancer biology. Initially identified on histones, lysine lactylation has been also described on non-histone proteins, playing a pivotal role in transcriptional activation, protein function, and cellular processes. Two major sources of the lactyl moiety have been currently distinguished: L-lactyl-CoA (precursor of the L-lactyl moiety) and S-D-lactylglutathione (precursor of the D-lactyl moiety), which enable enzymatic and non-enzymatic mechanisms of lysine lactylation, respectively. Although the specific writers, erasers, and readers of this modification are still unclear, acetyltransferases and deacetylases have been proposed as crucial mediators of lysine lactylation. Remarkably, lactylation exerts significant influence on critical cancer-related pathways, thereby shaping cellular behavior during malignant transformation and the metastatic cascade. Hence, as recent insights into lysine lactylation underscore its growing potential in tumor biology, targeting this modification is emerging as a significant opportunity for cancer treatment.
Collapse
Affiliation(s)
- Marta Iozzo
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio," University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Elisa Pardella
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio," University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Elisa Giannoni
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio," University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Paola Chiarugi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio," University of Florence, Viale Morgagni 50, 50134 Florence, Italy.
| |
Collapse
|
5
|
Li S, Li H, Bennewitz K, Poschet G, Buettner M, Hausser I, Szendroedi J, Nawroth PP, Kroll J. Combined loss of glyoxalase 1 and aldehyde dehydrogenase 3a1 amplifies dicarbonyl stress, impairs proteasome activity resulting in hyperglycemia and activated retinal angiogenesis. Metabolism 2025; 165:156149. [PMID: 39892865 DOI: 10.1016/j.metabol.2025.156149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND & AIMS Any energy consumption results in the generation of highly reactive dicarbonyls and the need to prevent excessive dicarbonyls accumulation through the activity of several interdependent detoxification enzymes. Glyoxalase 1 (GLO1) knockout zebrafish showed only moderately elevated methylglyoxal (MG) levels, but increased Aldehyde Dehydrogenases (ALDH) activity and increased aldh3a1 expression. Elevated levels of 4-hydroxynonenal (4-HNE) but no MG increase were observed in ALDH3A1KO. The question of whether ALDH3A1 prevents MG formation as a compensatory mechanism in the absence of GLO1 remained unclear. METHODS To investigate whether ALDH3A1 detoxifies MG as a compensatory mechanism in the absence of GLO1, the GLO1/ALDH3A1 double knockout (DKO) zebrafish was first generated. Various metabolites including advanced glycation end products (AGEs), as well as glucose metabolism and hyaloid vasculature were analyzed in GLO1KO, ALDH3A1KO and GLO1/ALDH3A1DKO zebrafish. RESULTS In the absence of GLO1 and ALDH3A1, MG-H1 levels were increased. MG-H1 accumulation led to a severe deterioration of proteasome function, resulting in impaired glucose homeostasis and consequently amplified angiogenic activation of the hyaloid and retinal vasculature. Rescue of these pathological processes could be observed by using L-carnosine, and proteasome activator betulinic acid. CONCLUSION The present data, together with previous studies, suggest that ALDH3A1 and GLO1 are important detoxification enzymes that prevent the deleterious effects of MG-H1 accumulation on proteasome function, glucose homeostasis and vascular function.
Collapse
Affiliation(s)
- Shu Li
- Department of Vascular Biology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany
| | - Hao Li
- German Cancer Research Center (DKFZ), Unit D400, Heidelberg 69120, Germany
| | - Katrin Bennewitz
- Department of Vascular Biology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany
| | - Gernot Poschet
- Metabolomics Core Technology Platform, Centre for Organismal Studies, Heidelberg University, Heidelberg 69120, Germany
| | - Michael Buettner
- Metabolomics Core Technology Platform, Centre for Organismal Studies, Heidelberg University, Heidelberg 69120, Germany
| | - Ingrid Hausser
- Institute of Pathology IPH, EM Lab, Heidelberg University Hospital, Heidelberg 69120, Germany
| | - Julia Szendroedi
- Department of Internal Medicine I and Clinical Chemistry, Heidelberg University Hospital, Heidelberg 69120, Germany
| | - Peter Paul Nawroth
- Medical Clinic and Polyclinic II, University Hospital Dresden, Dresden 01307, Germany
| | - Jens Kroll
- Department of Vascular Biology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany.
| |
Collapse
|
6
|
Berends E, Vangrieken P, Amiri N, van de Waarenburg MPH, Scheijen JLJM, Hermes DJHP, Wouters K, van Oostenbrugge RJ, Schalkwijk CG, Foulquier S. Increased Levels of Circulating Methylglyoxal Have No Consequence for Cerebral Microvascular Integrity and Cognitive Function in Young Healthy Mice. Mol Neurobiol 2025; 62:4190-4202. [PMID: 39414727 PMCID: PMC11880179 DOI: 10.1007/s12035-024-04552-3] [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: 03/29/2024] [Accepted: 10/11/2024] [Indexed: 10/18/2024]
Abstract
Diabetes and other age-related diseases are associated with an increased risk of cognitive impairment, but the underlying mechanisms remain poorly understood. Methylglyoxal (MGO), a by-product of glycolysis and a major precursor in the formation of advanced glycation end-products (AGEs), is increased in individuals with diabetes and other age-related diseases and is associated with microvascular dysfunction. We now investigated whether increased levels of circulating MGO can lead to cerebral microvascular dysfunction, blood-brain barrier (BBB) dysfunction, and cognitive impairment. Mice were supplemented or not with 50 mM MGO in drinking water for 13 weeks. Plasma and cortical MGO and MGO-derived AGEs were measured with UPLC-MS/MS. Peripheral and cerebral microvascular integrity and inflammation were investigated. Cerebral blood flow and neurovascular coupling were investigated with laser speckle contrast imaging, and cognitive tests were performed. We found a 2-fold increase in plasma MGO and an increase in MGO-derived AGEs in plasma and cortex. Increased plasma MGO did not lead to cerebral microvascular dysfunction, inflammation, or cognitive decline. This study shows that increased concentrations of plasma MGO are not associated with cerebral microvascular dysfunction and cognitive impairment in healthy mice. Future research should focus on the role of endogenously formed MGO in cognitive impairment.
Collapse
Affiliation(s)
- Eline Berends
- Faculty of Health Medicine and Life Sciences, Department of Internal Medicine, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Philippe Vangrieken
- Faculty of Health Medicine and Life Sciences, Department of Internal Medicine, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Naima Amiri
- Faculty of Health Medicine and Life Sciences, Department of Internal Medicine, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Marjo P H van de Waarenburg
- Faculty of Health Medicine and Life Sciences, Department of Internal Medicine, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Jean L J M Scheijen
- Faculty of Health Medicine and Life Sciences, Department of Internal Medicine, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Denise J H P Hermes
- Department of Neuropsychology and Psychiatry, Maastricht University, Maastricht, the Netherlands
- MHeNs, School for Mental Health and Neurosciences, Maastricht University, Maastricht, the Netherlands
| | - Kristiaan Wouters
- Faculty of Health Medicine and Life Sciences, Department of Internal Medicine, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Robert J van Oostenbrugge
- CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
- MHeNs, School for Mental Health and Neurosciences, Maastricht University, Maastricht, the Netherlands
- Department of Neurology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Casper G Schalkwijk
- Faculty of Health Medicine and Life Sciences, Department of Internal Medicine, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands.
- CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands.
| | - Sébastien Foulquier
- CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands.
- MHeNs, School for Mental Health and Neurosciences, Maastricht University, Maastricht, the Netherlands.
- Faculty of Health Medicine and Life Sciences, Department of Pharmacology and Toxicology, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands.
| |
Collapse
|
7
|
Gao J, Liu R, Huang K, Li Z, Sheng X, Chakraborty K, Han C, Zhang D, Becker L, Zhao Y. Dynamic investigation of hypoxia-induced L-lactylation. Proc Natl Acad Sci U S A 2025; 122:e2404899122. [PMID: 40030031 PMCID: PMC11912421 DOI: 10.1073/pnas.2404899122] [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: 03/15/2024] [Accepted: 12/06/2024] [Indexed: 03/19/2025] Open
Abstract
The recently identified histone modification lysine lactylation can be stimulated by L-lactate and glycolysis. Although the chemical group added upon lysine lactylation was originally proposed to be the L-enantiomer of lactate (KL-la), two isomeric modifications, lysine D-lactylation (KD-la) and N-ε-(carboxyethyl) lysine (Kce), also exist in cells, with their precursors being metabolites of glycolysis. The dynamic regulation and differences among these three modifications in response to hypoxia remain poorly understood. In this study, we demonstrate that intracellular KL-la, but not KD-la or Kce, is up-regulated in response to hypoxia. Depletion of glyoxalase enzymes, GLO1 and GLO2, had minimal impact on KD-la, Kce, or hypoxia-induced KL-la. Conversely, blocking glycolytic flux to L-lactate under hypoxic conditions by knocking out lactate dehydrogenase A/B completely abolished the induction of KL-la but increased KD-la and Kce. We further observed a correlation between the level of KL-la and hypoxia-inducible factor 1 alpha (HIF-1α) expression under hypoxic conditions and when small molecules were used to stabilize HIF-1α in the normoxia condition. Our result demonstrated that there is a strong correlation between HIF-1α and KL-la in lung cancer tissues and that patient samples with higher grade tend to have higher KL-la levels. Using a proteomics approach, we quantified 66 KL-la sites that were up-regulated by hypoxia and demonstrated that p300/CBP contributes to hypoxia-induced KL-la. Collectively, our study demonstrates that KL-la, rather than KD-la or Kce, is the prevailing lysine lactylation in response to hypoxia. Our results therefore demonstrate a link between KL-la and the hypoxia-induced adaptation of tumor cells.
Collapse
Affiliation(s)
- Jinjun Gao
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL60637
| | - Ruilong Liu
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL60637
| | - Kevin Huang
- College of Agriculture and Life Science, Cornell University, Ithaca, NY14853
| | - Ziyuan Li
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI53706
| | - Xinlei Sheng
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL60637
| | - Kasturi Chakraborty
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL60637
| | - Chang Han
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL60637
| | - Di Zhang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing100871, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing100871, China
| | - Lev Becker
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL60637
| | - Yingming Zhao
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL60637
| |
Collapse
|
8
|
Kumar N, Samanta B, Km J, Raghunathan V, Sen P, Bhat R. Demonstration of Enhancement of Tumor Intravasation by Dicarbonyl Stress Using a Microfluidic Organ-on-chip. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2405998. [PMID: 39745135 DOI: 10.1002/smll.202405998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 11/28/2024] [Indexed: 02/13/2025]
Abstract
Cancer metastasis involves cell migration from their primary organ foci into vascular channels, followed by dissemination to prospective colonization sites. Vascular entry of tumor cells or intravasation involves their breaching stromal and endothelial extracellular matrix (ECM) and the endothelial barriers. How the kinetics of this breach are confounded by chronic inflammatory stresses seen in diabetes and aging remains ill-investigated. To study the problem, a histopathology-motivated, imaging-tractable, microfluidic multi-organ-on-chip platform is constructed, that seamlessly integrates a breast tumor-like compartment: invasive MDA-MB-231 in a 3D Collagen I scaffold, and a flow-implemented vascular channel: immortalized human aortic endothelia (TeloHAEC) on laminin-rich basement membrane (lrBM). The chip showcases the complexity of intravasation, wherein tumor cells and endothelia cooperate to form anastomotic structures, which facilitate cancer cell migration into the vascular channel. Upon entry, cancer cells adhere to and flow within the vascular channel. Exposure to methylglyoxal (MG), a dicarbonyl stressor associated with diabetic circulatory milieu increases cancer cell intravasation and adhesion through the vascular channel. This can be driven by MG-induced endothelial senescence and shedding, but also by the ability of MG to degrade lrBM and pathologically cross-link Collagen I, diminishing cell-ECM adhesion. Thus, dicarbonyl stress attenuates homeostatic barriers to cancer intravasation, exacerbating metastasis.
Collapse
Affiliation(s)
- Nilesh Kumar
- Centre for Nano Science and Engineering, Indian Institute of Science, Bengaluru, 560012, India
| | - Bidita Samanta
- Department of Bioengineering, Indian Institute of Science, Bengaluru, 560012, India
| | - Jyothsna Km
- Department of Electrical and Communications Engineering, Bengaluru, 560012, India
| | - Varun Raghunathan
- Department of Electrical and Communications Engineering, Bengaluru, 560012, India
| | - Prosenjit Sen
- Centre for Nano Science and Engineering, Indian Institute of Science, Bengaluru, 560012, India
- Department of Bioengineering, Indian Institute of Science, Bengaluru, 560012, India
| | - Ramray Bhat
- Department of Bioengineering, Indian Institute of Science, Bengaluru, 560012, India
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, 560012, India
| |
Collapse
|
9
|
Koike S, Kimura H, Ogasawara Y. Polysulfide and persulfide-mediated activation of the PERK-eIF2α-ATF4 pathway increases Sestrin2 expression and reduces methylglyoxal toxicity. Redox Biol 2025; 79:103450. [PMID: 39667306 PMCID: PMC11697784 DOI: 10.1016/j.redox.2024.103450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 11/29/2024] [Indexed: 12/14/2024] Open
Abstract
Unfolded protein response (UPR) is activated in cells under endoplasmic reticulum (ER) stress. One sensor protein involved in this response is PERK, which is activated through its redox-dependent oligomerization. Prolonged UPR activation is associated with the development and progression of various diseases, making it essential to understanding the redox regulation of PERK. Sulfane sulfur, such as polysulfides and persulfides, can modify the cysteine residues and regulate the function of various proteins. However, the regulatory mechanism and physiological effects of sulfane sulfur on the PERK-eIF2α-ATF4 pathway remain poorly understood. This study focuses on the persulfidation of PERK to elucidate the effects of polysulfides on the PERK-eIF2α-ATF4 pathway and investigate its cytoprotective mechanism. Here, we demonstrated that polysulfide treatment promoted the oligomerization of PERK and PTP1B in neuronal cells using western blotting under nonreducing conditions. We also observed that l-cysteine, a biological source of sulfane sulfur, promoted the oligomerization of PERK and the knockdown of CBS and 3-MST, two sulfane sulfur-producing enzymes, and reduced PERK oligomerization induced by l-cysteine treatment. Furthermore, the band shift assay and LC-MS/MS studies revealed that polysulfides and persulfides induce PTP1B and PERK persulfidation. Additionally, polysulfides promoted eIF2α phosphorylation and ATF4 accumulation in the nucleus, suggesting that polysulfides activate the PERK-eIF2α-ATF4 pathway in neuronal cells. Moreover, polysulfides protected neuronal cells from methylglyoxal-induced toxicity, and this protective effect was reduced when the expression of Sestrin2, regulated by ATF4 activity, was suppressed. This study identified a novel mechanism for the activation of the PERK-eIF2α-ATF4 pathway through persulfidation by polysulfides and persulfides. Interestingly, activation of this pathway overcame the toxicity of methylglyoxal in dependence on Sestrin2 expression. These findings deepen our understanding of neuronal diseases involving ER stress and UPR disturbance and may inspire new therapeutic strategies.
Collapse
Affiliation(s)
- Shin Koike
- Department of Analytical Biochemistry, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Hideo Kimura
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, 1-1-1 Daigaku-Dori, Sanyo-Onoda 756-0884, Yamaguchi, Japan
| | - Yuki Ogasawara
- Department of Analytical Biochemistry, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan.
| |
Collapse
|
10
|
Akbari MS, Joyce LR, Spencer BL, Brady A, McIver KS, Doran KS. Identification of Glyoxalase A in Group B Streptococcus and its contribution to methylglyoxal tolerance and virulence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.30.605887. [PMID: 39131367 PMCID: PMC11312555 DOI: 10.1101/2024.07.30.605887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Group B Streptococcus (GBS) is a Gram-positive pathobiont that commonly colonizes the gastrointestinal and lower female genital tracts but can cause sepsis and pneumonia in newborns and is a leading cause of neonatal meningitis. Despite the resulting disease severity, the pathogenesis of GBS is not completely understood, especially during the early phases of infection. To investigate GBS factors necessary for blood stream survival, we performed a transposon (Tn) mutant screen in our bacteremia infection model using a GBS mariner transposon mutant library previously developed by our group. We identified significantly underrepresented mutations in 623 genes that contribute to survival in the blood, including those encoding known virulence factors such as capsule, the β-hemolysin, and inorganic metal ion transport systems. Most of the underrepresented genes have not been previously characterized or studied in GBS, including gloA and gloB, which are homologs for genes involved in methylglyoxal (MG) detoxification. MG is a byproduct of glycolysis and a highly reactive toxic aldehyde that is elevated in immune cells during infection. Here, we observed MG sensitivity across multiple GBS isolates and confirm that gloA contributes to MG tolerance and invasive GBS infection. We show specifically that gloA contributes to GBS survival in the presence of neutrophils and depleting neutrophils in mice abrogates the decreased survival and infection of the gloA mutant. The requirement of the glyoxalase pathway during GBS infection suggests that MG detoxification is important for bacterial survival during host-pathogen interactions.
Collapse
Affiliation(s)
- Madeline S. Akbari
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado USA
| | - Luke R. Joyce
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado USA
| | - Brady L. Spencer
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado USA
| | - Amanda Brady
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado USA
| | - Kevin S. McIver
- Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland, USA
| | - Kelly S. Doran
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado USA
| |
Collapse
|
11
|
Prakoso NM, Sundari AM, Fadhilah, Abinawanto, Pustimbara A, Dwiranti A, Bowolaksono A. Methylglyoxal impairs human dermal fibroblast survival and migration by altering RAGE-hTERT mRNA expression in vitro. Toxicol Rep 2024; 13:101835. [PMID: 39687678 PMCID: PMC11646750 DOI: 10.1016/j.toxrep.2024.101835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 11/14/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Fibroblasts are native residents in dermal layer of human skin which are important for dermal regeneration and essential during cutaneous wound healing by releasing inflammatory markers and actively migrate to close an open wound. Premature skin ageing due to methylglyoxal (MGO) has recently caught the attention considering its potential to accelerate the emergence of skin ageing signs, however previous studies were only focused in primary neonatal dermal fibroblast and NIH3t3 fibroblast cell line. Therefore, thorough investigation is required to study the impact of MGO on primary human dermal fibroblast isolated from adult subject (HDFa). In our experiments, short exposure of MGO was observed to induced significant reductions in cell viability at concentrations of 7.5, 10, 12.5, 15, and 17.5 mM (p < 0.005) after 3 hours of treatment. The cellular death of HDFa at 10, 12.5 and 15 mM of MGO were also marked by increased in intracellular ROS level, indicating the involvement of oxidative stress-induced death in these cells. We also observed enlarge scratch areas of cells exposed with 7.5 and 10 mM MGO compared to control after 26 hours, thereby suggesting a decline in cell migration and viability in this group. We propose the increased ROS as the consequence of AGE-RAGE activation which was marked by significant elevation of RAGE mRNA on cells exposed to 10 mM MGO. Our data also suggest the occurrence of DNA damage events via ROS-induced oxidation or mediated by decline in hTERT mRNA expression.
Collapse
Affiliation(s)
- Nurul Muhammad Prakoso
- Master Program in Biology, Department of Biology, Faculty of Mathematics and Natural Science, University of Indonesia, Depok, West Java 16424, Indonesia
| | - Ayu Mulia Sundari
- Master Program in Biology, Department of Biology, Faculty of Mathematics and Natural Science, University of Indonesia, Depok, West Java 16424, Indonesia
| | - Fadhilah
- Cellular and Molecular Mechanisms in Biological System (CEMBIOS) Research Group, Department of Biology, Faculty of Mathematics and Natural Science, University of Indonesia, Depok, West Java 16424, Indonesia
| | - Abinawanto
- Cellular and Molecular Mechanisms in Biological System (CEMBIOS) Research Group, Department of Biology, Faculty of Mathematics and Natural Science, University of Indonesia, Depok, West Java 16424, Indonesia
| | - Anantya Pustimbara
- Tokyo Institute of Technology, School of Life Science and Technology, 4259 Midori-ku, Yokohama 2268501, Japan
- Center for Photodynamic Medicine, Kochi Medical School, Kohasu, Oko-cho, Nankoku-shi, Kochi 783-8505, Japan
| | - Astari Dwiranti
- Cellular and Molecular Mechanisms in Biological System (CEMBIOS) Research Group, Department of Biology, Faculty of Mathematics and Natural Science, University of Indonesia, Depok, West Java 16424, Indonesia
| | - Anom Bowolaksono
- Cellular and Molecular Mechanisms in Biological System (CEMBIOS) Research Group, Department of Biology, Faculty of Mathematics and Natural Science, University of Indonesia, Depok, West Java 16424, Indonesia
| |
Collapse
|
12
|
Belpomme D, Lacomme S, Poletti C, Bonesso L, Hinault-Boyer C, Barbier S, Irigaray P. Free Methylglyoxal as a Metabolic New Biomarker of Tumor Cell Proliferation in Cancers. Cancers (Basel) 2024; 16:3922. [PMID: 39682111 DOI: 10.3390/cancers16233922] [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: 10/24/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND A fundamental property of cancer cells is their metabolic reprogramming, allowing them to increase glucose uptake and glycolysis. Using a rat colon adenocarcinoma model, we previously showed that blood levels of free methylglyoxal (MG), a side-product of glycolysis, remained normal in animals grafted with a non-growing tumor cell clone, while MG levels were significantly increased and positively correlated with tumor growth in animals grafted with a tumorigenic cell clone issued from the same tumor. METHODS We measured free MG in the blood of cancerous non-diabetic patients and compared the results to healthy subjects and non-cancerous diabetic patients. We also measured free MG in tumors and in the corresponding non-cancer tissues, and the peripheral blood. RESULTS We show that free MG levels in the peripheral blood of cancer patients are significantly increased in comparison with free MG levels in the peripheral blood of healthy controls (p < 0.0001), and similar to those in the peripheral blood of hyperglycemic diabetic patients (p = 0.965). In addition, we show that repeated free MG level measurement could be used for the therapeutic monitoring of cancer patients. Moreover, we confirmed that free MG is produced by tumor cells at significantly higher levels than cells from their corresponding tissues (p < 0.0001), and is subsequently released in the peripheral blood. CONCLUSIONS Free MG measured in the blood could be a new metabolic biomarker useful for the diagnostic, prognostic and follow-up of non-diabetic patients with cancers, such as bronchus carcinoma, pancreatic carcinoma and glioblastoma, for which there are presently no available useful biomarkers.
Collapse
Affiliation(s)
- Dominique Belpomme
- Department of Cancer Clinical Research, Paris V University Hospital, 75005 Paris, France
- European Cancer and Environment Research Institute (ECERI), 1000 Brussels, Belgium
| | - Stéphanie Lacomme
- Centre de Ressources Biologiques, BB-0033-00035, CHRU, 54500 Nancy, France
| | | | - Laurent Bonesso
- Clinical Chemistry Laboratory, Pasteur University Hospital, 06000 Nice, France
| | - Charlotte Hinault-Boyer
- Clinical Chemistry Laboratory, Pasteur University Hospital, 06000 Nice, France
- Université Côte d'Azur, INSERM U1065, C3M, 06000 Nice, France
| | | | - Philippe Irigaray
- European Cancer and Environment Research Institute (ECERI), 1000 Brussels, Belgium
- Association for Research on Treatment Against Cancer (ARTAC), 75015 Paris, France
| |
Collapse
|
13
|
Koike S, Tsurudome S, Okano S, Kishida A, Ogasawara Y. Dimethyl Fumarate Reduces Methylglyoxal-derived Carbonyl Stress Through Nrf2/GSH Activation in SH-SY5Y Cells. Neurochem Res 2024; 50:28. [PMID: 39576418 DOI: 10.1007/s11064-024-04255-0] [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: 08/27/2024] [Revised: 10/04/2024] [Accepted: 10/15/2024] [Indexed: 11/24/2024]
Abstract
Carbonyl stress refers to the excessive accumulation of advanced glycation end products (AGEs) in mammalian tissues. This phenomenon plays a significant role in the pathogenesis of various diseases, including diabetes, chronic renal failure, arteriosclerosis, and central nervous system (CNS) disorders. We have previously demonstrated that an increase in glutathione concentration, dependent on the nuclear factor erythroid 2-related factor 2 (Nrf2) system, provides a potent cytoprotective effect against Methylglyoxal (MGO)-induced carbonyl stress. Meanwhile, dimethyl fumarate (DMF), known for its Nrf2-activating effects, was recently approved as a treatment for multiple sclerosis (MS), a neurodegenerative disease. DMF is a first line therapy for relapsing-remitting MS and may also be effective for other neurodegenerative conditions. However, the detailed mechanisms by which DMF mitigates neurodegenerative pathologies remain unclear. This study investigates the impact of DMF on anticarbonyl activity and its underlying mechanism focusing on the accumulation of carbonyl protein in the cell. MGO, a glucose metabolite, was used to induce carbonylation in the neuronal cell line. MGO is a typical carbonyl compound that readily reacts with arginine and lysine residues to form AGE-modified proteins. Methylglyoxal-derived hydroimidazolone 1 (MG-H1) often forms uncharged, hydrophobic residues on the protein surface, which can affect protein distribution and lead to misfolding. Our findings indicate that DMF increases levels of glutathione (GSH), glutamate cysteine ligase modifier subunit (GCLM), and nuclear Nrf2 in SH-SY5Y cells. Importantly, DMF pretreatment significantly reduced the accumulation of MG-H1-modified proteins. Furthermore, this effect of DMF was diminished when Nrf2 expression was suppressed and when GCL, a rate-limiting enzyme in GSH synthesis, was inhibited. Thus, the increase in GSH levels, leading to the activation of the Nrf2 pathway, a key factor in DMF's ability to suppress the accumulation of MG-H1-modified proteins. This study is the first to demonstrate that DMF possesses strong anticarbonyl stress activity in neuronal cells. Therefore, future research may extend the application of DMF to other CNS diseases associated with carbonyl stress, such as Alzheimer's and Parkinson's disease.
Collapse
Affiliation(s)
- Shin Koike
- Department of Analytical Biochemistry, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Satori Tsurudome
- Department of Analytical Biochemistry, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Saki Okano
- Department of Analytical Biochemistry, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Atsushi Kishida
- Graduate School of Pharmaceutical Sciences, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Yuki Ogasawara
- Department of Analytical Biochemistry, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan.
| |
Collapse
|
14
|
Rabbani N, Thornalley PJ. Unraveling the impaired incretin effect in obesity and type 2 diabetes: Key role of hyperglycemia-induced unscheduled glycolysis and glycolytic overload. Diabetes Res Clin Pract 2024; 217:111905. [PMID: 39447679 DOI: 10.1016/j.diabres.2024.111905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/01/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
Glucagon-like peptide-1 (GLP-1) agonists and GLP-1 and glucose-dependent insulinotropic polypeptide (GIP) co-agonists are major treatment options for subjects with obesity and patients with type 2 diabetes mellitus (T2DM). They counter without addressing the mechanistic cause of the impaired incretin effect associated with obesity and T2DM. Incretin effect impairment is characterized by decreased secretion of incretins from enteroendocrine cells and incretin resistance of pancreatic β-cells. It is linked to hyperglycemia. We present evidence that subversion of the gating of glucose entry into glycolysis, mainly by glucokinase (hexokinase-4), during persistent hyperglycemia in enteroendocrine cells, pancreatic β- and α-cells and appetite-regulating neurons contributes to the biochemical mechanism of the impaired incretin effect. Unscheduled glycolysis and glycolytic overload thereby produced decreases cell signalling of incretin secretion to glucose and other secretion stimuli and incretin receptor responses. This mechanism provides a guide for development of alternative therapies targeting recovery of the impaired incretin effect.
Collapse
Affiliation(s)
- Naila Rabbani
- QU Health, Qatar University, University Street, PO Box 2713, Doha, Qatar
| | - Paul J Thornalley
- College of Health and Life Sciences, Hamad Bin Khalifa University, Education City, PO Box 34110, Doha, Qatar.
| |
Collapse
|
15
|
Jensen SJ, Cuthbert BJ, Garza-Sánchez F, Helou CC, de Miranda R, Goulding CW, Hayes CS. Advanced glycation end-product crosslinking activates a type VI secretion system phospholipase effector protein. Nat Commun 2024; 15:8804. [PMID: 39394186 PMCID: PMC11470151 DOI: 10.1038/s41467-024-53075-x] [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: 04/09/2024] [Accepted: 10/01/2024] [Indexed: 10/13/2024] Open
Abstract
Advanced glycation end-products (AGE) are a pervasive form of protein damage implicated in the pathogenesis of neurodegenerative disease, atherosclerosis and diabetes mellitus. Glycation is typically mediated by reactive dicarbonyl compounds that accumulate in all cells as toxic byproducts of glucose metabolism. Here, we show that AGE crosslinking is harnessed to activate an antibacterial phospholipase effector protein deployed by the type VI secretion system of Enterobacter cloacae. Endogenous methylglyoxal reacts with a specific arginine-lysine pair to tether the N- and C-terminal α-helices of the phospholipase domain. Substitutions at these positions abrogate both crosslinking and toxic phospholipase activity, but in vitro enzyme function can be restored with an engineered disulfide that covalently links the N- and C-termini. Thus, AGE crosslinking serves as a bona fide post-translation modification to stabilize phospholipase structure. Given the ubiquity of methylglyoxal in prokaryotic and eukaryotic cells, these findings suggest that glycation may be exploited more generally to stabilize other proteins. This alternative strategy to fortify tertiary structure could be particularly advantageous in the cytoplasm, where redox potentials preclude disulfide bond formation.
Collapse
Affiliation(s)
- Steven J Jensen
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, 93106, USA
| | - Bonnie J Cuthbert
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, 92697, USA
| | - Fernando Garza-Sánchez
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, 93106, USA
| | - Colette C Helou
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, 92697, USA
| | - Rodger de Miranda
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, 92697, USA
| | - Celia W Goulding
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, 92697, USA
- Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, 92697, USA
| | - Christopher S Hayes
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, 93106, USA.
- Biomolecular Science and Engineering Program, University of California, Santa Barbara, Santa Barbara, 93106, USA.
| |
Collapse
|
16
|
Koike S, Mitsuhashi H, Kishida A, Ogasawara Y. Elucidating the Antiglycation Effect of Creatine on Methylglyoxal-Induced Carbonyl Stress In Vitro. Int J Mol Sci 2024; 25:10880. [PMID: 39456665 PMCID: PMC11506949 DOI: 10.3390/ijms252010880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
Advanced glycation end products (AGEs) with multiple structures are formed at the sites where carbonyl groups of reducing sugars bind to free amino groups of proteins through the Maillard reaction. In recent years, it has been highlighted that the accumulation of AGEs, which are generated when carbonyl compounds produced in the process of sugar metabolism react with proteins, is involved in various diseases. Creatine is a biocomponent that is homeostatically present throughout the body and is known to react nonenzymatically with α-dicarbonyl compounds. This study evaluated the antiglycation potential of creatine against methylglyoxal (MGO), a glucose metabolite that induces carbonyl stress with formation of AGEs in vitro. Further, to elucidate the mechanism of the cytoprotective action of creatine, its effect on the accumulation of carbonyl proteins in the cells and the MGO-induced cellular damage were investigated using neuroblastoma cells. The results revealed that creatine significantly inhibits protein carbonylation by directly reacting with MGO, and creatine added to the culture medium suppressed MGO-derived carbonylation of intracellular proteins and exerted a protective effect on MGO-induced cytotoxicity. These findings suggest that endogenous and supplemented creatine may contribute to the attenuation of carbonyl stress in vivo.
Collapse
Affiliation(s)
- Shin Koike
- Department of Analytical Biochemistry, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo 204-8588, Japan; (S.K.); (H.M.)
| | - Haruka Mitsuhashi
- Department of Analytical Biochemistry, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo 204-8588, Japan; (S.K.); (H.M.)
| | - Atsushi Kishida
- Graduate School of Pharmaceutical Sciences, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo 204-8588, Japan;
| | - Yuki Ogasawara
- Department of Analytical Biochemistry, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo 204-8588, Japan; (S.K.); (H.M.)
| |
Collapse
|
17
|
Zheng L, Li X, Widjaja F, Liu C, Rietjens IMCM. Use of physiologically based kinetic modeling to predict neurotoxicity and genotoxicity of methylglyoxal in humans. NPJ Sci Food 2024; 8:79. [PMID: 39368970 PMCID: PMC11455947 DOI: 10.1038/s41538-024-00322-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 09/27/2024] [Indexed: 10/07/2024] Open
Abstract
This study aimed to evaluate human neurotoxicity and genotoxicity risks from dietary and endogenous methylglyoxal (MGO), utilizing physiologically based kinetic (PBK) modeling-facilitated reverse dosimetry as a new approach methodology (NAM) to extrapolate in vitro toxicity data to in vivo dose-response predictions. A human PBK model was defined based on a newly developed and evaluated mouse model enabling the translation of in vitro toxicity data for MGO from human stem cell-derived neurons and WM-266-4 melanoma cells into quantitative human in vivo toxicity data and subsequent risk assessment by the margin of exposure (MOE) approach. The results show that the MOEs resulting from daily dietary intake did not raise a concern for endpoints for neurotoxicity including mitochondrial function, cytotoxicity, and apoptosis, while those for DNA adduct formation could not exclude a concern over genotoxicity. Endogenous MGO formation, especially under diabetic conditions, resulted in MOEs that raised concern not only for genotoxicity but also for some of the neurotoxicity endpoints evaluated. Thus, the results also point to the importance of taking the endogenous levels into account in the risk assessment of MGO.
Collapse
Affiliation(s)
- Liang Zheng
- Division of Toxicology, Wageningen University and Research, Wageningen, The Netherlands.
| | - Xiyu Li
- Division of Toxicology, Wageningen University and Research, Wageningen, The Netherlands
| | - Frances Widjaja
- Division of Toxicology, Wageningen University and Research, Wageningen, The Netherlands
| | - Chen Liu
- Division of Toxicology, Wageningen University and Research, Wageningen, The Netherlands
- Tea Refining and Innovation Key Laboratory of Sichuan Province, College of Horticulture, Sichuan Agricultural University, Chengdu, China
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University and Research, Wageningen, The Netherlands
| |
Collapse
|
18
|
Hanna GS, Findlay VJ, Turner DP, Hamann MT. Quantitative NMR Analysis of Marine Macroalgae for AGE Inhibition by Methylglyoxal Scavenging. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:21905-21911. [PMID: 39298668 DOI: 10.1021/acs.jafc.4c04367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Reactive carbonyl species (RCS) induce a fundamental form of biological stress that has driven the evolution of diverse mechanisms for minimizing its impact on organismal health. The complications that accompany uncontrolled hyperglycemia exemplify the health implications when RCS stress exceeds the body's capacity to prevent the excessive formation of advanced glycation end-products. Presented here is a novel quantitative NMR (qNMR) technique for evaluating scavengers of the prominent sugar-derived carbonyl methylglyoxal (MGO). This tool was employed to screen the chemical diversity of marine macroalgae extracts, with a focus on species that have a history of consumption by the World's healthiest populations and are subject to global scale aquacultural production. Fucus vesiculosus demonstrated the highest capacity for inhibiting glycation and scavenging MGO. Additionally, the Chondrus cripsus, Gracilaria vermiculophyla, and Gracilaria tikvahiae extracts had a high capacity for scavenging MGO, representing the first report of this activity. This new qNMR methodology presented is highly applicable for screening extracts and compounds from diverse sources, and the results highlight the potential of macroalgae extracts to be employed as RCS and AGE targeting therapeutics and food additives.
Collapse
Affiliation(s)
- George S Hanna
- Department of Biomedical Sciences and Drug Discovery, Medical University of South Carolina, Charleston, South Carolina 29425, United States
- Department of Public Health, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - Victoria J Findlay
- Department of Surgery, Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - David P Turner
- Department of Surgery, Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Mark T Hamann
- Department of Biomedical Sciences and Drug Discovery, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| |
Collapse
|
19
|
Chen Y, Schlotterer A, Lin J, Dietrich N, Fleming T, Lanzinger S, Holl RW, Hammes HP. Sex differences in the development of experimental diabetic retinopathy. Sci Rep 2024; 14:22812. [PMID: 39354039 PMCID: PMC11445250 DOI: 10.1038/s41598-024-73279-x] [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: 03/02/2024] [Accepted: 09/15/2024] [Indexed: 10/03/2024] Open
Abstract
This study aimed to characterize the role of female sex in the pathogenesis of diabetic retinopathy. In the retinae of female Ins2Akita-diabetic mice (F-IA), ovariectomized female Ins2Akita-diabetic mice (F-IA/OVX), male Ins2Akita-diabetic mice (M-IA), and female STZ-diabetic mice (F-STZ), the formation of reactive metabolites and post-translational modifications, damage to the neurovascular unit, and expression of cellular stress response genes were analyzed. Compared to the male diabetic retina, the concentrations of the glycation adduct fructosyl-lysine, the Maillard product 3-deoxyglucosone, and the reactive metabolite methylglyoxal were significantly reduced in females. In females, there was also less evidence of diabetic damage to the neurovascular unit, as shown by decreased pericyte loss and reduced microglial activation. In the male diabetic retina, the expression of several members of the crystallin gene family (Cryab, Cryaa, Crybb2, Crybb1, and Cryba4) was increased. Clinical data from type 1 diabetic females showed that premenopausal women had a significantly lower prevalence of diabetic retinopathy compared to postmenopausal women stratified for disease duration and glycemic control. These data emphasize the importance of estradiol in protecting the diabetic retina and highlight the pathogenic relevance of sex in diabetic retinopathy.
Collapse
Affiliation(s)
- Ying Chen
- Fifth Medical Department, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Andrea Schlotterer
- Fifth Medical Department, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Jihong Lin
- Fifth Medical Department, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Nadine Dietrich
- Fifth Medical Department, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Thomas Fleming
- Department of Internal Medicine I and Clinical Chemistry, University of Heidelberg, Heidelberg, Germany
| | - Stefanie Lanzinger
- Institute of Epidemiology and Medical Biometry, ZIBMT, University of Ulm, Ulm, Germany
- German Center for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Reinhard W Holl
- Institute of Epidemiology and Medical Biometry, ZIBMT, University of Ulm, Ulm, Germany
- German Center for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Hans-Peter Hammes
- Fifth Medical Department, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.
| |
Collapse
|
20
|
Huang Y, Huang L, Cheng M, Li C, Zhou X, Ullah A, Sarfraz S, Khatab A, Xie G. Progresses in biosynthesis pathway, regulation mechanism and potential application of 2-acetyl-1-pyrroline in fragrant rice. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2024; 215:109047. [PMID: 39153390 DOI: 10.1016/j.plaphy.2024.109047] [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: 05/14/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
The formation of rice aroma is a complex process that is influenced by genetic and environmental factors. More than 500 fragrance compounds have been documented in fragrant rice, among which 2-AP dominates the aroma of rice. This paper introduced the identification of OsBadh2 in the biosynthesis of 2-AP in rice. Then, non-enzymatic and enzymatic pathways of the 2-AP biosynthesis have been comprehensively investigated. In detail, 2-AP biosynthesis-associated enzyme, such as OsBADH2, OsP5CS, OsGAD, OsGAPDH, OsProDH, OsOAT, OsODC and OsDAO, have been summarized, while MG and fatty acids are also implicated in modulating the biosynthesis of 2-AP by providing the acetyl groups. Moreover, extensive collections of traditional fragrant rice varieties have been collated, together with the OsBadh2 haplotypes of 312 fragrant rice germplasm in China. And finally, genetic engineering of OsBadh2 and other genes in the 2-AP biosynthesis to develop fragrant rice are discussed.
Collapse
Affiliation(s)
- Yajing Huang
- MOA Key Laboratory of Crop Ecophysiology and Farming System in the Middle Reaches of the Yangtze River, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Lei Huang
- MOA Key Laboratory of Crop Ecophysiology and Farming System in the Middle Reaches of the Yangtze River, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China; The People's Government of Zougang Town, Xiaochang County, Xiaogan City, Hubei, 432910, China
| | - Maozhi Cheng
- MOA Key Laboratory of Crop Ecophysiology and Farming System in the Middle Reaches of the Yangtze River, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Chuanhao Li
- MOA Key Laboratory of Crop Ecophysiology and Farming System in the Middle Reaches of the Yangtze River, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xiaofeng Zhou
- MOA Key Laboratory of Crop Ecophysiology and Farming System in the Middle Reaches of the Yangtze River, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Aman Ullah
- MOA Key Laboratory of Crop Ecophysiology and Farming System in the Middle Reaches of the Yangtze River, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Samina Sarfraz
- MOA Key Laboratory of Crop Ecophysiology and Farming System in the Middle Reaches of the Yangtze River, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Ahmed Khatab
- MOA Key Laboratory of Crop Ecophysiology and Farming System in the Middle Reaches of the Yangtze River, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China; Rice Research and Training Center, 33717, Sakha, Kafr El-Sheikh, Egypt
| | - Guosheng Xie
- MOA Key Laboratory of Crop Ecophysiology and Farming System in the Middle Reaches of the Yangtze River, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
21
|
Hellwig M, Diel P, Eisenbrand G, Grune T, Guth S, Henle T, Humpf HU, Joost HG, Marko D, Raupbach J, Roth A, Vieths S, Mally A. Dietary glycation compounds - implications for human health. Crit Rev Toxicol 2024; 54:485-617. [PMID: 39150724 DOI: 10.1080/10408444.2024.2362985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/24/2024] [Accepted: 05/28/2024] [Indexed: 08/17/2024]
Abstract
The term "glycation compounds" comprises a wide range of structurally diverse compounds that are formed endogenously and in food via the Maillard reaction, a chemical reaction between reducing sugars and amino acids. Glycation compounds produced endogenously are considered to contribute to a range of diseases. This has led to the hypothesis that glycation compounds present in food may also cause adverse effects and thus pose a nutritional risk to human health. In this work, the Senate Commission on Food Safety (SKLM) of the German Research Foundation (DFG) summarized data on formation, occurrence, exposure and toxicity of glycation compounds (Part A) and systematically assessed potential associations between dietary intake of defined glycation compounds and disease, including allergy, diabetes, cardiovascular and renal disease, gut/gastrotoxicity, brain/cognitive impairment and cancer (Part B). A systematic search in Pubmed (Medline), Scopus and Web of Science using a combination of keywords defining individual glycation compounds and relevant disease patterns linked to the subject area of food, nutrition and diet retrieved 253 original publications relevant to the research question. Of these, only 192 were found to comply with previously defined quality criteria and were thus considered suitable to assess potential health risks of dietary glycation compounds. For each adverse health effect considered in this assessment, however, only limited numbers of human, animal and in vitro studies were identified. While studies in humans were often limited due to small cohort size, short study duration, and confounders, experimental studies in animals that allow for controlled exposure to individual glycation compounds provided some evidence for impaired glucose tolerance, insulin resistance, cardiovascular effects and renal injury in response to oral exposure to dicarbonyl compounds, albeit at dose levels by far exceeding estimated human exposures. The overall database was generally inconsistent or inconclusive. Based on this systematic review, the SKLM concludes that there is at present no convincing evidence for a causal association between dietary intake of glycation compounds and adverse health effects.
Collapse
Affiliation(s)
- Michael Hellwig
- Chair of Special Food Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Patrick Diel
- Department of Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | | | - Tilman Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition (DIfE), Nuthetal, Germany
| | - Sabine Guth
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Dortmund, Germany
| | - Thomas Henle
- Chair of Food Chemistry, TU Dresden, Dresden, Germany
| | | | - Hans-Georg Joost
- Department of Experimental Diabetology, German Institute of Human Nutrition (DIfE), Nuthetal, Germany
| | - Doris Marko
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Jana Raupbach
- Institute of Food Chemistry, Technische Universität Braunschweig, Braunschweig, Germany
| | - Angelika Roth
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Dortmund, Germany
| | | | - Angela Mally
- Department of Toxicology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
22
|
Pantano F, Simonetti S, Iuliani M, Guillen MJ, Cuevas C, Aviles P, Cavaliere S, Napolitano A, Cortellini A, Mazzocca A, Nibid L, Sabarese G, Perrone G, Gambarotti M, Righi A, Palmerini E, Stacchiotti S, Barisella M, Gronchi A, Valeri S, Sbaraglia M, Dei Tos AP, Tonini G, Vincenzi B. S-p-bromobenzyl-glutathione cyclopentyl diester (BBGC) as novel therapeutic strategy to enhance trabectedin anti-tumor effect in soft tissue sarcoma preclinical models. Oncogene 2024; 43:2986-2994. [PMID: 39198616 PMCID: PMC11436363 DOI: 10.1038/s41388-024-03143-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 08/01/2024] [Accepted: 08/22/2024] [Indexed: 09/01/2024]
Abstract
Trabectedin, approved for the treatment of soft tissue sarcoma (STS), interferes with cell division and genetic transcription processes. Due to its strong anti-tumor activity in only certain histotypes, several studies on trabectedin combinations are currently ongoing to improve its efficacy. In this study, we aimed to investigate novel potential therapeutic strategies to enhance the anti-tumor effect of trabectedin using integrated in silico, in vitro, and in vivo approaches. For in silico analysis, we screened two public datasets, GSEA M5190 and TCGA SARC. Fibrosarcoma, leiomyosarcoma, dedifferentiated, and myxoid liposarcoma cell lines were used for in vitro studies. For in vivo experiments, fibrosarcoma orthotopic murine model was developed. In silico analysis identified Glo1 as the only druggable target upregulated after trabectedin treatment and correlated with poor prognosis. The specific Glo1 inhibitor, S-p-bromobenzylglutathione cyclopentyl diester (BBGC), increased trabectedin cytotoxicity in STS cells, and restored drug sensitivity in myxoid liposarcoma cells resistant to trabectedin. Moreover, the combined treatment with BBGC and trabectedin had a synergistic antitumor effect in vivo without any additional toxicity to mice. Based on these results, we believe that BBGC warrants further investigation to evaluate its potential clinical use in combination with trabectedin.
Collapse
Affiliation(s)
- F Pantano
- Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Roma, Italy
| | - S Simonetti
- Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - M Iuliani
- Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy.
| | - M J Guillen
- Research Department, PharmaMar S.A, Madrid, Spain
| | - C Cuevas
- Research Department, PharmaMar S.A, Madrid, Spain
| | - P Aviles
- Research Department, PharmaMar S.A, Madrid, Spain
| | - S Cavaliere
- Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | | | - A Cortellini
- Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Roma, Italy
| | - A Mazzocca
- Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - L Nibid
- Research Unit of Anatomical Pathology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Roma, Italy
| | - G Sabarese
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - G Perrone
- Research Unit of Anatomical Pathology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Roma, Italy
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - M Gambarotti
- Department of Pathology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - A Righi
- Department of Pathology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - E Palmerini
- Osteoncology, Soft Tissue and Bone Sarcomas, Innovative Therapy Unit, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - S Stacchiotti
- Adult mesenchymal tumours and rare cancers unit, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - M Barisella
- Tissue Tumor Pathology Unit, Department of Advanced Diagnostics, Fondazione IRCSS Istituto Nazionale dei Tumori Milan, Milano, Italy
| | - A Gronchi
- Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - S Valeri
- Sarcoma Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - M Sbaraglia
- Department of Integrated Diagnostics, Azienda Ospedale-Università Padova; Department of Medicine-DIMED, University of Padua School of Medicine, Padua, Italy
| | - A P Dei Tos
- Department of Integrated Diagnostics, Azienda Ospedale-Università Padova; Department of Medicine-DIMED, University of Padua School of Medicine, Padua, Italy
| | - G Tonini
- Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Roma, Italy
| | - B Vincenzi
- Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Roma, Italy
| |
Collapse
|
23
|
Tusiewicz K, Wachełko O, Zawadzki M, Szpot P. Novel Technique for Simultaneous Ethylene Glycol and Its Metabolites Determination in Human Whole Blood and Urine Samples Using GC-QqQ-MS/MS. J Xenobiot 2024; 14:1143-1164. [PMID: 39311144 PMCID: PMC11417905 DOI: 10.3390/jox14030065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/05/2024] [Accepted: 08/20/2024] [Indexed: 09/26/2024] Open
Abstract
Toxicological analyses often necessitate the identification of compounds belonging to diverse functional groups. For GC-MS analyses, derivatization of compounds belonging to different functional groups can pose a challenge and requires the development of comprehensive methods of analysis. One example could be ethylene glycol, whose widespread use is related to possible unintentional or suicidal intoxications. This fact clearly indicates the need to develop sensitive methods for the determination of ethylene glycol and its metabolites in biological material, as only such complex analysis allows for proper toxicological expertise. A simultaneous GC-QqQ-MS/MS method for the determination of ethylene glycol together with its metabolites, glyoxal and glycolic acid, as well as the detection of glyoxylic acid and oxalic acid, was developed and fully validated. A novel approach for simultaneous derivatization of substances from different groups (alcohols, aldehydes, and carboxylic acids) was established. Sample preparation included the addition of three internal standards (BHB-d4, ethylene glycol-d4 and methylglyoxal), precipitation with acetonitrile and subsequent derivatization with N-tert-butyldimethylsilyl-N-methyltrifluoroacetamide (MTBSTFA), as well as pentafluorophenylhydrazine (PFPH). Detection was carried out with the use of triple quadrupole mass spectrometer. The ionization method was electron impact, and quantitative analysis was carried out in multiple reaction monitoring mode. The lower limit of quantification was 1 μg/mL, 0.1 μg/mL, and 500 μg/mL for ethylene glycol, glyoxal, and glycolic acid, respectively. The presented method was applied in three authentic postmortem cases of ethylene glycol intoxication.
Collapse
Affiliation(s)
- Kaja Tusiewicz
- Department of Forensic Medicine, Wroclaw Medical University, 4 J. Mikulicza-Radeckiego Street, 50345 Wroclaw, Poland
| | - Olga Wachełko
- Institute of Toxicology Research, 45 Kasztanowa Street, 55093 Borowa, Poland
| | - Marcin Zawadzki
- Faculty of Medicine, Department of Social Sciences and Infectious Diseases, Wroclaw University of Science and Technology, 27 Wybrzeże Wyspiańskiego, 50370 Wrocław, Poland
| | - Paweł Szpot
- Department of Forensic Medicine, Wroclaw Medical University, 4 J. Mikulicza-Radeckiego Street, 50345 Wroclaw, Poland
| |
Collapse
|
24
|
D’Haese S, Claes L, Jaeken E, Deluyker D, Evens L, Heeren E, Haesen S, Vastmans L, Lambrichts I, Wouters K, Schalkwijk CG, Hansen D, Eijnde BO, Bito V. Pyridoxamine Alleviates Cardiac Fibrosis and Oxidative Stress in Western Diet-Induced Prediabetic Rats. Int J Mol Sci 2024; 25:8508. [PMID: 39126079 PMCID: PMC11312841 DOI: 10.3390/ijms25158508] [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: 05/31/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024] Open
Abstract
Individuals with type 2 diabetes mellitus (T2DM) are at an increased risk for heart failure, yet preventive cardiac care is suboptimal in this population. Pyridoxamine (PM), a vitamin B6 analog, has been shown to exert protective effects in metabolic and cardiovascular diseases. In this study, we aimed to investigate whether PM limits adverse cardiac remodeling and dysfunction in rats who develop T2DM. Male rats received a standard chow diet or Western diet (WD) for 18 weeks to induce prediabetes. One WD group received additional PM (1 g/L) via drinking water. Glucose tolerance was assessed with a 1 h oral glucose tolerance test. Cardiac function was evaluated using echocardiography and hemodynamic measurements. Histology on left ventricular (LV) tissue was performed. Treatment with PM prevented the increase in fasting plasma glucose levels compared to WD-fed rats (p < 0.05). LV cardiac dilation tended to be prevented using PM supplementation. In LV tissue, PM limited an increase in interstitial collagen deposition (p < 0.05) seen in WD-fed rats. PM tended to decrease 3-nitrotyrosine and significantly lowered 4-hydroxynonenal content compared to WD-fed rats. We conclude that PM alleviates interstitial fibrosis and oxidative stress in the hearts of WD-induced prediabetic rats.
Collapse
Affiliation(s)
- Sarah D’Haese
- UHasselt, Cardio & Organ Systems (COST), Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (S.D.); (D.D.); (E.H.); (S.H.); (L.V.); (I.L.)
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands; (K.W.); (C.G.S.)
| | - Lisa Claes
- UHasselt, Cardio & Organ Systems (COST), Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (S.D.); (D.D.); (E.H.); (S.H.); (L.V.); (I.L.)
| | - Eva Jaeken
- UHasselt, Cardio & Organ Systems (COST), Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (S.D.); (D.D.); (E.H.); (S.H.); (L.V.); (I.L.)
| | - Dorien Deluyker
- UHasselt, Cardio & Organ Systems (COST), Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (S.D.); (D.D.); (E.H.); (S.H.); (L.V.); (I.L.)
| | - Lize Evens
- UHasselt, Cardio & Organ Systems (COST), Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (S.D.); (D.D.); (E.H.); (S.H.); (L.V.); (I.L.)
| | - Ellen Heeren
- UHasselt, Cardio & Organ Systems (COST), Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (S.D.); (D.D.); (E.H.); (S.H.); (L.V.); (I.L.)
| | - Sibren Haesen
- UHasselt, Cardio & Organ Systems (COST), Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (S.D.); (D.D.); (E.H.); (S.H.); (L.V.); (I.L.)
| | - Lotte Vastmans
- UHasselt, Cardio & Organ Systems (COST), Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (S.D.); (D.D.); (E.H.); (S.H.); (L.V.); (I.L.)
| | - Ivo Lambrichts
- UHasselt, Cardio & Organ Systems (COST), Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (S.D.); (D.D.); (E.H.); (S.H.); (L.V.); (I.L.)
| | - Kristiaan Wouters
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands; (K.W.); (C.G.S.)
| | - Casper G. Schalkwijk
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands; (K.W.); (C.G.S.)
| | - Dominique Hansen
- UHasselt, Faculty of Rehabilitation Sciences, REVAL Rehabilitation Research Centre, Agoralaan, 3590 Diepenbeek, Belgium;
- Department of Cardiology, Heart Centre Hasselt, Jessa Hospital, Stadsomvaart 11, 3500 Hasselt, Belgium
| | - BO Eijnde
- SMRc-Sports Medicine Research Center, BIOMED-Biomedical Research Institute, Faculty of Medicine & Life Sciences, Hasselt University, 3590 Diepenbeek, Belgium;
- Division of Sport Science, Stellenbosch University, Stellenbosch 7602, South Africa
| | - Virginie Bito
- UHasselt, Cardio & Organ Systems (COST), Biomedical Research Institute, Agoralaan, 3590 Diepenbeek, Belgium; (S.D.); (D.D.); (E.H.); (S.H.); (L.V.); (I.L.)
| |
Collapse
|
25
|
Hurben AK, Zhang Q, Galligan JJ, Tretyakova N, Erber L. Endogenous Cellular Metabolite Methylglyoxal Induces DNA-Protein Cross-Links in Living Cells. ACS Chem Biol 2024; 19:1291-1302. [PMID: 38752800 PMCID: PMC11353540 DOI: 10.1021/acschembio.4c00100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Methylglyoxal (MGO) is an electrophilic α-oxoaldehyde generated endogenously through metabolism of carbohydrates and exogenously due to autoxidation of sugars, degradation of lipids, and fermentation during food and drink processing. MGO can react with nucleophilic sites within proteins and DNA to form covalent adducts. MGO-induced advanced glycation end-products such as protein and DNA adducts are thought to be involved in oxidative stress, inflammation, diabetes, cancer, renal failure, and neurodegenerative diseases. Additionally, MGO has been hypothesized to form toxic DNA-protein cross-links (DPC), but the identities of proteins participating in such cross-linking in cells have not been determined. In the present work, we quantified DPC formation in human cells exposed to MGO and identified proteins trapped on DNA upon MGO exposure using mass spectrometry-based proteomics. A total of 265 proteins were found to participate in MGO-derived DPC formation including gene products engaged in telomere organization, nucleosome assembly, and gene expression. In vitro experiments confirmed DPC formation between DNA and glyceraldehyde-3-phosphate dehydrogenase (GAPDH), as well as histone proteins H3.1 and H4. Collectively, our study provides the first evidence for MGO-mediated DNA-protein cross-linking in living cells, prompting future studies regarding the relevance of these toxic lesions in cancer, diabetes, and other diseases linked to elevated MGO levels.
Collapse
Affiliation(s)
- Alexander K. Hurben
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States; Present Address: Department of Chemistry, University of California, Berkeley, California 94720, United States
| | - Qi Zhang
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - James J. Galligan
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
| | - Natalia Tretyakova
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States; Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Luke Erber
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
26
|
Peter A, Schleicher E, Kliemank E, Szendroedi J, Königsrainer A, Häring HU, Nawroth PP, Fleming T. Accumulation of Non-Pathological Liver Fat Is Associated with the Loss of Glyoxalase I Activity in Humans. Metabolites 2024; 14:209. [PMID: 38668337 PMCID: PMC11051733 DOI: 10.3390/metabo14040209] [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: 03/15/2024] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
The underlying molecular mechanisms for the development of non-alcoholic fatty liver (NAFL) and its progression to advanced liver diseases remain elusive. Glyoxalase 1 (Glo1) loss, leading to elevated methylglyoxal (MG) and dicarbonyl stress, has been implicated in various diseases, including obesity-related conditions. This study aimed to investigate changes in the glyoxalase system in individuals with non-pathological liver fat. Liver biopsies were obtained from 30 individuals with a narrow range of BMI (24.6-29.8 kg/m2). Whole-body insulin sensitivity was assessed using HOMA-IR. Liver biopsies were analyzed for total triglyceride content, Glo1 and Glo2 mRNA, protein expression, and activity. Liquid chromatography-tandem mass spectrometry determined liver dicarbonyl content and oxidation and glycation biomarkers. Liver Glo1 activity showed an inverse correlation with HOMA-IR and liver triglyceride content, but not BMI. Despite reduced Glo1 activity, no associations were found with elevated liver dicarbonyls or glycation markers. A sex dimorphism was observed in Glo1, with females exhibiting significantly lower liver Glo1 protein expression and activity, and higher liver MG-H1 content compared to males. This study demonstrates that increasing liver fat, even within a non-pathological range, is associated with reduced Glo1 activity.
Collapse
Affiliation(s)
- Andreas Peter
- German Centre for Diabetes Research (DZD), Helmholtz Centre Munich, 85764 Munich, Germany
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, 72016 Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Centre Munich, University of Tübingen, 72016 Tübingen, Germany
| | - Erwin Schleicher
- German Centre for Diabetes Research (DZD), Helmholtz Centre Munich, 85764 Munich, Germany
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, 72016 Tübingen, Germany
| | - Elisabeth Kliemank
- German Centre for Diabetes Research (DZD), Helmholtz Centre Munich, 85764 Munich, Germany
- Department of Medicine I and Clinical Chemistry, Heidelberg University Hospital, INF 410, 69120 Heidelberg, Germany
| | - Julia Szendroedi
- German Centre for Diabetes Research (DZD), Helmholtz Centre Munich, 85764 Munich, Germany
- Department of Medicine I and Clinical Chemistry, Heidelberg University Hospital, INF 410, 69120 Heidelberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Internal Medicine I, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Alfred Königsrainer
- Department of General, Visceral and Transplant Surgery, Eberhard-Karls-University Tübingen, 72016 Tübingen, Germany
| | - Hans-Ulrich Häring
- German Centre for Diabetes Research (DZD), Helmholtz Centre Munich, 85764 Munich, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Centre Munich, University of Tübingen, 72016 Tübingen, Germany
- Division of Diabetology, Endocrinology and Nephrology, Department of Internal Medicine IV, Eberhard-Karls-University Tübingen, 72016 Tübingen, Germany
| | - Peter P. Nawroth
- German Centre for Diabetes Research (DZD), Helmholtz Centre Munich, 85764 Munich, Germany
- Department of Medicine I and Clinical Chemistry, Heidelberg University Hospital, INF 410, 69120 Heidelberg, Germany
- Institute for Immunology, University Hospital of Heidelberg, INF 305, 69120 Heidelberg, Germany
| | - Thomas Fleming
- German Centre for Diabetes Research (DZD), Helmholtz Centre Munich, 85764 Munich, Germany
- Department of Medicine I and Clinical Chemistry, Heidelberg University Hospital, INF 410, 69120 Heidelberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Internal Medicine I, Heidelberg University Hospital, 69120 Heidelberg, Germany
| |
Collapse
|
27
|
Lai SWT, Hernandez-Castillo C, Gonzalez EDJL, Zoukari T, Talley M, Paquin N, Chen Z, Roep BO, Kaddis JS, Natarajan R, Termini J, Shuck SC. Methylglyoxal Adducts Are Prognostic Biomarkers for Diabetic Kidney Disease in Patients With Type 1 Diabetes. Diabetes 2024; 73:611-617. [PMID: 37967313 PMCID: PMC10958582 DOI: 10.2337/db23-0277] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 11/06/2023] [Indexed: 11/17/2023]
Abstract
More than 30% of patients with type 1 diabetes develop diabetic kidney disease (DKD), which significantly increases mortality risk. The Diabetes Control and Complications Trial (DCCT) and follow-up study, Epidemiology of Diabetes Interventions and Complications (EDIC), established that glycemic control measured by HbA1c predicts DKD risk. However, the continued high incidence of DKD reinforces the urgent need for additional biomarkers to supplement HbA1c. Here, we assessed biomarkers induced by methylglyoxal (MG), a metabolic by-product that forms covalent adducts on DNA, RNA, and proteins, called MG adducts. Urinary MG adducts were measured in samples from patients with type 1 diabetes enrolled in DCCT/EDIC who did (case patients; n = 90) or did not (control patients; n = 117) develop DKD. Univariate and multivariable analyses revealed that measurements of MG adducts independently predict DKD before established DKD biomarkers such as glomerular filtration rate and albumin excretion rate. Elevated levels of MG adducts bestowed the greatest risk of developing DKD in a multivariable model that included HbA1c and other clinical covariates. Our work establishes a novel class of biomarkers to predict DKD risk and suggests that inclusion of MG adducts may be a valuable tool to improve existing predictors of complications like DKD prior to overt disease, and to aid in identifying at-risk individuals and personalized risk management. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Seigmund Wai Tsuen Lai
- Department of Diabetes and Cancer Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Carlos Hernandez-Castillo
- Department of Diabetes and Cancer Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Edwin De Jesus Lopez Gonzalez
- Department of Diabetes and Cancer Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Tala Zoukari
- Department of Diabetes and Cancer Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Min Talley
- Biostatistics and Mathematical Oncology Core, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Nadia Paquin
- Department of Diabetes and Cancer Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Zhuo Chen
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Bart O. Roep
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - John S. Kaddis
- Department of Diabetes and Cancer Discovery Science, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Rama Natarajan
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - John Termini
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Sarah C. Shuck
- Department of Diabetes and Cancer Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA
| |
Collapse
|
28
|
Zambre S, Bangar N, Mistry A, Katarmal P, Khan MS, Ahmed I, Tupe R, Roy B. Aldosterone, Methylglyoxal, and Glycated Albumin Interaction with Macrophage Cells Affects Their Viability, Activation, and Differentiation. ACS OMEGA 2024; 9:11848-11859. [PMID: 38497023 PMCID: PMC10938338 DOI: 10.1021/acsomega.3c09420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/06/2024] [Accepted: 02/13/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND The inflammatory response in diabetes is strongly correlated with increasing amounts of advanced glycation end products (AGEs), methylglyoxal (MGO), aldosterone (Aldo), and activation of macrophages. Aldo is known to be associated with increased pro-inflammatory responses in general, but its significance in inflammatory responses under glycated circumstances has yet to be understood. In the current work, the aim of our study was to study the macrophage immune response in the presence of AGEs, MGO, and Aldo to comprehend their combined impact on diabetes-associated complications. METHODS AND RESULTS The viability of macrophages upon treatment with glycated HSA (Gly-HSA) promoted cell growth as the concentration increased from 100 to 500 μg/mL, whereas MGO at a high concentration (≥300 μM) significantly hampered cell growth. At lower concentrations (0.5-5 nM), Aldo strongly promoted cell growth, whereas at higher concentrations (50 nM), it was seen to inhibit growth when used for cell treatment for 24 h. Aldo had no effect on MGO-induced cell growth inhibition after 24 h of treatment. However, compared to MGO or Aldo treatment alone, an additional decrease in viability could be seen after 48 h of treatment with a combination of MGO and Aldo. Treatment with Aldo and MGO induced expression of TNF-α independently and when combined. However, when combined, Aldo and MGO significantly suppressed the expression of TGF-β. Aldo, Gly-HSA, and MGO strongly induced the transcription of NF-κB and RAGE mRNA and, as expected, also promoted the formation of reactive oxygen species. Also, by inducing iNOS and MHC-II and suppressing CD206 transcript expression, Gly-HSA strongly favored the differentiation of macrophages into M1 type (pro-inflammatory). On the other hand, the combination of Aldo and MGO strongly induced the expression of MHC-II, CD206, and ARG1 (M2 macrophage marker). These findings suggest that Gly-HSA, MGO, and Aldo differently influence macrophage survival, activation, and differentiation. CONCLUSIONS Overall, this study gives an insight into the effects of glycated protein and MGO in the presence of Aldo on macrophage survival, activation, differentiation, and inflammatory response.
Collapse
Affiliation(s)
- Saee Zambre
- Symbiosis
School of Biological Sciences (SSBS), Symbiosis
International (Deemed University) (SIU), Lavale, Pune 412115, Maharashtra, India
| | - Nilima Bangar
- Symbiosis
School of Biological Sciences (SSBS), Symbiosis
International (Deemed University) (SIU), Lavale, Pune 412115, Maharashtra, India
| | - Armaan Mistry
- Symbiosis
School of Biological Sciences (SSBS), Symbiosis
International (Deemed University) (SIU), Lavale, Pune 412115, Maharashtra, India
| | - Poonam Katarmal
- Symbiosis
School of Biological Sciences (SSBS), Symbiosis
International (Deemed University) (SIU), Lavale, Pune 412115, Maharashtra, India
| | - Mohd Shahnawaz Khan
- Department
of Biochemistry, College of Science, King
Saud University, Riyadh 11451, Saudi Arabia
| | - Irshad Ahmed
- Department
of Biochemistry and Structural Biology, School of Medicine, UT Health Science Center, San Antonio, Texas 78229, United States
| | - Rashmi Tupe
- Symbiosis
School of Biological Sciences (SSBS), Symbiosis
International (Deemed University) (SIU), Lavale, Pune 412115, Maharashtra, India
| | - Bishnudeo Roy
- Symbiosis
School of Biological Sciences (SSBS), Symbiosis
International (Deemed University) (SIU), Lavale, Pune 412115, Maharashtra, India
| |
Collapse
|
29
|
Harrer P, Inderhees J, Zhao C, Schormair B, Tilch E, Gieger C, Peters A, Jöhren O, Fleming T, Nawroth PP, Berger K, Hermesdorf M, Winkelmann J, Schwaninger M, Oexle K. Phenotypic and genome-wide studies on dicarbonyls: major associations to glomerular filtration rate and gamma-glutamyltransferase activity. EBioMedicine 2024; 101:105007. [PMID: 38354534 PMCID: PMC10875252 DOI: 10.1016/j.ebiom.2024.105007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND The dicarbonyl compounds methylglyoxal (MG), glyoxal (GO) and 3-deoxyglucosone (3-DG) have been linked to various diseases. However, disease-independent phenotypic and genotypic association studies with phenome-wide and genome-wide reach, respectively, have not been provided. METHODS MG, GO and 3-DG were measured by LC-MS in 1304 serum samples of two populations (KORA, n = 482; BiDirect, n = 822) and assessed for associations with genome-wide SNPs (GWAS) and with phenome-wide traits. Redundancy analysis (RDA) was used to identify major independent trait associations. FINDINGS Mutual correlations of dicarbonyls were highly significant, being stronger between MG and GO (ρ = 0.6) than between 3-DG and MG or GO (ρ = 0.4). Significant phenotypic results included associations of all dicarbonyls with sex, waist-to-hip ratio, glomerular filtration rate (GFR), gamma-glutamyltransferase (GGT), and hypertension, of MG and GO with age and C-reactive protein, of GO and 3-DG with glucose and antidiabetics, of MG with contraceptives, of GO with ferritin, and of 3-DG with smoking. RDA revealed GFR, GGT and, in case of 3-DG, glucose as major contributors to dicarbonyl variance. GWAS did not identify genome-wide significant loci. SNPs previously associated with glyoxalase activity did not reach nominal significance. When multiple testing was restricted to the lead SNPs of GWASs on the traits selected by RDA, 3-DG was found to be associated (p = 2.3 × 10-5) with rs1741177, an eQTL of NF-κB inhibitor NFKBIA. INTERPRETATION This large-scale, population-based study has identified numerous associations, with GFR and GGT being of pivotal importance, providing unbiased perspectives on dicarbonyls beyond the current state. FUNDING Deutsche Forschungsgemeinschaft, Helmholtz Munich, German Centre for Cardiovascular Research (DZHK), German Federal Ministry of Research and Education (BMBF).
Collapse
Affiliation(s)
- Philip Harrer
- Institute of Neurogenomics, Helmholtz Munich, Neuherberg, Germany; Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
| | - Julica Inderhees
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lubeck, Lubeck, Germany; Bioanalytic Core Facility, Center for Brain, Behavior and Metabolism, University of Lübeck, Germany; German Centre for Cardiovascular Research (DZHK), Hamburg-Lübeck-Kiel, Germany
| | - Chen Zhao
- Institute of Neurogenomics, Helmholtz Munich, Neuherberg, Germany; Neurogenetic Systems Analysis Group, Institute of Neurogenomics, Helmholtz Munich, Neuherberg, Germany
| | - Barbara Schormair
- Institute of Neurogenomics, Helmholtz Munich, Neuherberg, Germany; Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
| | - Erik Tilch
- Institute of Neurogenomics, Helmholtz Munich, Neuherberg, Germany; Neurogenetic Systems Analysis Group, Institute of Neurogenomics, Helmholtz Munich, Neuherberg, Germany
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Munich, Neuherberg, Germany; Institute of Epidemiology, Helmholtz Munich, Neuherberg, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Chair of Epidemiology, Institute for Medical Information Processing, Biometry and Epidemiology, Medical Faculty, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Olaf Jöhren
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lubeck, Lubeck, Germany; Bioanalytic Core Facility, Center for Brain, Behavior and Metabolism, University of Lübeck, Germany
| | - Thomas Fleming
- Department of Internal Medicine, University of Heidelberg, Heidelberg, Germany
| | - Peter P Nawroth
- Department of Internal Medicine, University of Heidelberg, Heidelberg, Germany
| | - Klaus Berger
- Institute of Epidemiology and Social Medicine, University of Münster, Münster, Germany
| | - Marco Hermesdorf
- Institute of Epidemiology and Social Medicine, University of Münster, Münster, Germany
| | - Juliane Winkelmann
- Institute of Neurogenomics, Helmholtz Munich, Neuherberg, Germany; Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; German Centre for Mental Health (DZPG), Munich-Augsburg, Germany
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lubeck, Lubeck, Germany; German Centre for Cardiovascular Research (DZHK), Hamburg-Lübeck-Kiel, Germany
| | - Konrad Oexle
- Institute of Neurogenomics, Helmholtz Munich, Neuherberg, Germany; Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany; Neurogenetic Systems Analysis Group, Institute of Neurogenomics, Helmholtz Munich, Neuherberg, Germany.
| |
Collapse
|
30
|
Blencowe LA, Božović A, Wong E, Kulasingam V, Cheung AM. Total serum pentosidine quantification using liquid chromatography-tandem mass spectrometry. Bone Rep 2024; 20:101737. [PMID: 38317648 PMCID: PMC10839865 DOI: 10.1016/j.bonr.2024.101737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 02/07/2024] Open
Abstract
Pentosidine (PEN) is an Advanced Glycation End-product (AGE) that is known to accumulate in bone collagen with aging and contribute to fracture risk. The PEN content in bone is correlated with serum PEN, making it an attractive, potential osteoporosis biomarker. We sought to develop a method for quantifying PEN in stored serum. After conducting a systematic narrative review of PEN quantification methodologies, we developed a liquid chromatography tandem mass spectrometry (LC-MS/MS) method for quantifying total serum PEN. Our method is both sensitive and precise (LOD 2 nM, LOQ 5 nM, %CV < 6.5 % and recovery 91.2-100.7 %). Our method is also equivalent or better than other methods identified in our review. Additionally, LC-MS/MS avoids the pitfalls and limitations of using fluorescence as a means of detection and could be adapted to investigate a broad range of AGE compounds.
Collapse
Affiliation(s)
- Lindsie A. Blencowe
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Andrea Božović
- Department of Clinical Biochemistry, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Evelyn Wong
- Department of Medicine, University Health Network, Toronto, ON, Canada
| | - Vathany Kulasingam
- Department of Clinical Biochemistry, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Angela M. Cheung
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Department of Medicine, University Health Network, Toronto, ON, Canada
| |
Collapse
|
31
|
Kumar P, Roy A, Mukul SJ, Singh AK, Singh DK, Nalli A, Banerjee P, Babu KSD, Raman B, Kruparani SP, Siddiqi I, Sankaranarayanan R. A translation proofreader of archaeal origin imparts multi-aldehyde stress tolerance to land plants. eLife 2024; 12:RP92827. [PMID: 38372335 PMCID: PMC10942605 DOI: 10.7554/elife.92827] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2024] Open
Abstract
Aldehydes, being an integral part of carbon metabolism, energy generation, and signalling pathways, are ingrained in plant physiology. Land plants have developed intricate metabolic pathways which involve production of reactive aldehydes and its detoxification to survive harsh terrestrial environments. Here, we show that physiologically produced aldehydes, i.e., formaldehyde and methylglyoxal in addition to acetaldehyde, generate adducts with aminoacyl-tRNAs, a substrate for protein synthesis. Plants are unique in possessing two distinct chiral proofreading systems, D-aminoacyl-tRNA deacylase1 (DTD1) and DTD2, of bacterial and archaeal origins, respectively. Extensive biochemical analysis revealed that only archaeal DTD2 can remove the stable D-aminoacyl adducts on tRNA thereby shielding archaea and plants from these system-generated aldehydes. Using Arabidopsis as a model system, we have shown that the loss of DTD2 gene renders plants susceptible to these toxic aldehydes as they generate stable alkyl modification on D-aminoacyl-tRNAs, which are recycled only by DTD2. Bioinformatic analysis identifies the expansion of aldehyde metabolising repertoire in land plant ancestors which strongly correlates with the recruitment of archaeal DTD2. Finally, we demonstrate that the overexpression of DTD2 offers better protection against aldehydes than in wild type Arabidopsis highlighting its role as a multi-aldehyde detoxifier that can be explored as a transgenic crop development strategy.
Collapse
Affiliation(s)
- Pradeep Kumar
- CSIR–Centre for Cellular and Molecular BiologyHyderabadIndia
- Academy of Scientific and Innovative Research (AcSIR), CSIR–CCMB CampusHyderabadIndia
- Academy of Scientific and Innovative Research (AcSIR)GhaziabadIndia
| | - Ankit Roy
- CSIR–Centre for Cellular and Molecular BiologyHyderabadIndia
| | - Shivapura Jagadeesha Mukul
- CSIR–Centre for Cellular and Molecular BiologyHyderabadIndia
- Academy of Scientific and Innovative Research (AcSIR), CSIR–CCMB CampusHyderabadIndia
- Academy of Scientific and Innovative Research (AcSIR)GhaziabadIndia
| | | | | | - Aswan Nalli
- CSIR–Centre for Cellular and Molecular BiologyHyderabadIndia
| | | | | | | | | | - Imran Siddiqi
- CSIR–Centre for Cellular and Molecular BiologyHyderabadIndia
- Academy of Scientific and Innovative Research (AcSIR), CSIR–CCMB CampusHyderabadIndia
| | - Rajan Sankaranarayanan
- CSIR–Centre for Cellular and Molecular BiologyHyderabadIndia
- Academy of Scientific and Innovative Research (AcSIR), CSIR–CCMB CampusHyderabadIndia
- Academy of Scientific and Innovative Research (AcSIR)GhaziabadIndia
| |
Collapse
|
32
|
John T, Saffoon N, Walsby-Tickle J, Hester SS, Dingler FA, Millington CL, McCullagh JSO, Patel KJ, Hopkinson RJ, Schofield CJ. Aldehyde-mediated inhibition of asparagine biosynthesis has implications for diabetes and alcoholism. Chem Sci 2024; 15:2509-2517. [PMID: 38362406 PMCID: PMC10866355 DOI: 10.1039/d3sc06551k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 01/01/2024] [Indexed: 02/17/2024] Open
Abstract
Patients with alcoholism and type 2 diabetes manifest altered metabolism, including elevated aldehyde levels and unusually low asparagine levels. We show that asparagine synthetase B (ASNS), the only human asparagine-forming enzyme, is inhibited by disease-relevant reactive aldehydes, including formaldehyde and acetaldehyde. Cellular studies show non-cytotoxic amounts of reactive aldehydes induce a decrease in asparagine levels. Biochemical analyses reveal inhibition results from reaction of the aldehydes with the catalytically important N-terminal cysteine of ASNS. The combined cellular and biochemical results suggest a possible mechanism underlying the low asparagine levels in alcoholism and diabetes. The results will stimulate research on the biological consequences of the reactions of aldehydes with nucleophilic residues.
Collapse
Affiliation(s)
- Tobias John
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| | - Nadia Saffoon
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| | - John Walsby-Tickle
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| | - Svenja S Hester
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford Oxford UK
| | - Felix A Dingler
- MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital/Headley Way Oxford OX3 9DS UK
| | - Christopher L Millington
- MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital/Headley Way Oxford OX3 9DS UK
| | - James S O McCullagh
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| | - Ketan J Patel
- MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital/Headley Way Oxford OX3 9DS UK
| | - Richard J Hopkinson
- Leicester Institute for Structural and Chemical Biology and School of Chemistry, University of Leicester, Henry Wellcome Building Lancaster Road Leicester LE1 7RH UK
| | - Christopher J Schofield
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| |
Collapse
|
33
|
Xue M, Irshad Z, Rabbani N, Thornalley PJ. Increased cellular protein modification by methylglyoxal activates endoplasmic reticulum-based sensors of the unfolded protein response. Redox Biol 2024; 69:103025. [PMID: 38199038 PMCID: PMC10821617 DOI: 10.1016/j.redox.2024.103025] [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: 12/03/2023] [Revised: 12/30/2023] [Accepted: 01/01/2024] [Indexed: 01/12/2024] Open
Abstract
The unfolded protein response (UPR) detects increased misfolded proteins and activates protein refolding, protein degradation and inflammatory responses. UPR sensors in the endoplasmic reticulum, IRE1α and PERK, bind and are activated by proteins with unexpected surface hydrophobicity, whereas sensor ATF6 is activated by proteolytic cleavage when released from complexation with protein disulfide isomerases (PDIs). Metabolic dysfunction leading to the formation of misfolded proteins with surface hydrophobicity and disruption of ATF6-PDI complexes leading to activation of UPR sensors remains unclear. The cellular concentration of reactive dicarbonyl metabolite, methylglyoxal (MG), is increased in impaired metabolic health, producing increased MG-modified cellular proteins. Herein we assessed the effect of high glucose concentration and related increased cellular MG on activation status of IRE1α, PERK and ATF6. Human aortal endothelial cells and HMEC-1 microvascular endothelial cells were incubated in low and high glucose concentration to model blood glucose control, with increase or decrease of MG by silencing or increasing expression of glyoxalase 1 (Glo1), which metabolizes MG. Increased MG induced by high glucose concentration activated IRE1α, PERK and ATF6 and related downstream signalling leading to increased chaperone, apoptotic and inflammatory gene expression. Correction of increased MG by increasing Glo1 expression prevented UPR activation. MG modification of proteins produces surface hydrophobicity through arginine-derived hydroimidazolone MG-H1 formation, with related protein unfolding and preferentially targets PDIs and chaperone pathways for modification. It thereby poses a major challenge to proteostasis and activates UPR sensors. Pharmacological decrease of MG with Glo1 inducer, trans-resveratrol and hesperetin in combination, offers a novel treatment strategy to counter UPR-related cell dysfunction, particularly in hyperglycemia associated with diabetes.
Collapse
Affiliation(s)
- Mingzhan Xue
- Diabetes Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University (HBKU), Qatar Foundation, P.O. Box 34110, Doha, Qatar
| | - Zehra Irshad
- Clinical Sciences Research Laboratories, Warwick Medical School, University of Warwick, University Hospital, Coventry, CV2 2DX, UK
| | - Naila Rabbani
- Department of Basic Medical Science, College of Medicine, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Paul J Thornalley
- Diabetes Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University (HBKU), Qatar Foundation, P.O. Box 34110, Doha, Qatar; Clinical Sciences Research Laboratories, Warwick Medical School, University of Warwick, University Hospital, Coventry, CV2 2DX, UK; College of Health and Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, P.O. Box 34110, Doha, Qatar.
| |
Collapse
|
34
|
Kotian NP, Prabhu A, Tender T, Raghu Chandrashekar H. Methylglyoxal Induced Modifications to Stabilize Therapeutic Proteins: A Review. Protein J 2024; 43:39-47. [PMID: 38017314 DOI: 10.1007/s10930-023-10166-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2023] [Indexed: 11/30/2023]
Abstract
Therapeutic proteins are potent, fast-acting drugs that are highly effective in treating various conditions. Medicinal protein usage has increased in the past 10 years, and it will evolve further as we better understand disease molecular pathways. However, it is associated with high processing costs, limited stability, difficulty in being administered as an oral medication, and the inability of large proteins to penetrate tissue and reach their target locations. Many methods have been developed to overcome the problems with the stability and chaperone activity of therapeutic proteins, viz., the addition of external agents (changing the properties of the surrounding solvent by using stabilizing excipients, e.g., amino acids, sugars, polyols) and internal agents (chemical modifications that influence its structural properties, e.g., mutations, glycosylation). However, these methods must completely clear protein instability and chaperone issues. There is still much work to be done on finetuning chaperone proteins to increase their biological efficacy and stability. Methylglyoxal (MGO), a potent dicarbonyl compound, reacts with proteins and forms covalent cross-links. Much research on MGO scavengers has been conducted since they are known to alter protein structure, which may result in alterations in biological activity and stability. MGO is naturally produced within our body, however, its impact on chaperones and protein stability needs to be better understood and seems to vary based on concentration. This review highlights the efforts of several research groups on the effect of MGO on various proteins. It also addresses the impact of MGO on a client protein, α-crystallin, to understand the potential solutions to the protein's chaperone and stability problems.
Collapse
Affiliation(s)
- Nainika Prashant Kotian
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Anusha Prabhu
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Tenzin Tender
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Hariharapura Raghu Chandrashekar
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India.
| |
Collapse
|
35
|
Brings S, Mier W, Beijer B, Kliemank E, Herzig S, Szendroedi J, Nawroth PP, Fleming T. Non-cross-linking advanced glycation end products affect prohormone processing. Biochem J 2024; 481:33-44. [PMID: 38112318 DOI: 10.1042/bcj20230321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/01/2023] [Accepted: 12/19/2023] [Indexed: 12/21/2023]
Abstract
Advanced glycation end products (AGEs) are non-enzymatic post-translational modifications of amino acids and are associated with diabetic complications. One proposed pathomechanism is the impaired processing of AGE-modified proteins or peptides including prohormones. Two approaches were applied to investigate whether substrate modification with AGEs affects the processing of substrates like prohormones to the active hormones. First, we employed solid-phase peptide synthesis to generate unmodified as well as AGE-modified protease substrates. Activity of proteases towards these substrates was quantified. Second, we tested the effect of AGE-modified proinsulin on the processing to insulin. Proteases showed the expected activity towards the unmodified peptide substrates containing arginine or lysine at the C-terminal cleavage site. Indeed, modification with Nε-carboxymethyllysine (CML) or methylglyoxal-hydroimidazolone 1 (MG-H1) affected all proteases tested. Cysteine cathepsins displayed a reduction in activity by ∼50% towards CML and MG-H1 modified substrates. The specific proteases trypsin, proprotein convertases subtilisin-kexins (PCSKs) type proteases, and carboxypeptidase E (CPE) were completely inactive towards modified substrates. Proinsulin incubation with methylglyoxal at physiological concentrations for 24 h resulted in the formation of MG-modified proinsulin. The formation of insulin was reduced by up to 80% in a concentration-dependent manner. Here, we demonstrate the inhibitory effect of substrate-AGE modifications on proteases. The finding that PCSKs and CPE, which are essential for prohormone processing, are inactive towards modified substrates could point to a yet unrecognized pathomechanism resulting from AGE modification relevant for the etiopathogenesis of diabetes and the development of obesity.
Collapse
Affiliation(s)
- Sebastian Brings
- Department of Endocrinology, Metabolism and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Walter Mier
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Barbro Beijer
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Elisabeth Kliemank
- Department of Endocrinology, Metabolism and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany
| | - Stephan Herzig
- German Centre of Diabetes Research (DZD), Munich, Germany
- Institute for Diabetes and Cancer IDC Helmholtz Center Munich and Joint Heidelberg-IDC Translational Diabetes Program, Munich, Germany
| | - Julia Szendroedi
- Department of Endocrinology, Metabolism and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany
- German Centre of Diabetes Research (DZD), Munich, Germany
- Institute for Diabetes and Cancer IDC Helmholtz Center Munich and Joint Heidelberg-IDC Translational Diabetes Program, Munich, Germany
- Center for Molecular Biology Heidelberg (ZMBH), Heidelberg, Germany
- Joint Division Molecular Metabolic Control, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter P Nawroth
- Department of Immunology, University Hospital Heidelberg, Heidelberg, Germany
| | - Thomas Fleming
- Department of Endocrinology, Metabolism and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany
- German Centre of Diabetes Research (DZD), Munich, Germany
- Institute for Diabetes and Cancer IDC Helmholtz Center Munich and Joint Heidelberg-IDC Translational Diabetes Program, Munich, Germany
- Center for Molecular Biology Heidelberg (ZMBH), Heidelberg, Germany
- Joint Division Molecular Metabolic Control, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
36
|
König A, Outeiro TF. Diabetes and Parkinson's Disease: Understanding Shared Molecular Mechanisms. JOURNAL OF PARKINSON'S DISEASE 2024; 14:917-924. [PMID: 38995799 PMCID: PMC11307096 DOI: 10.3233/jpd-230104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/19/2024] [Indexed: 07/14/2024]
Abstract
Aging is a major risk factor for Parkinson's disease (PD). Genetic mutations account for a small percentage of cases and the majority appears to be sporadic, with yet unclear causes. However, various environmental factors have been linked to an increased risk of developing PD and, therefore, understanding the complex interplay between genetic and environmental factors is crucial for developing effective disease-modifying therapies. Several studies identified a connection between type 2 diabetes (T2DM) and PD. T2DM is characterized by insulin resistance and failure of β-cells to compensate, leading to hyperglycemia and serious comorbidities. Both PD and T2DM share misregulated processes, including mitochondrial dysfunction, oxidative stress, chronic inflammation, altered proteostasis, protein aggregation, and misregulation of glucose metabolism. Chronic or recurring hyperglycemia is a T2DM hallmark and can lead to increased methylglyoxal (MGO) production, which is responsible for protein glycation. Glycation of alpha-synuclein (aSyn), a central player in PD pathogenesis, accelerates the deleterious aSyn effects. Interestingly, MGO blood plasma levels and aSyn glycation are significantly elevated in T2DM patients, suggesting a molecular mechanism underlying the T2DM - PD link. Compared to high constant glucose levels, glycemic variability (fluctuations in blood glucose levels), can be more detrimental for diabetic patients, causing oxidative stress, inflammation, and endothelial damage. Accordingly, it is imperative for future research to prioritize the exploration of glucose variability's influence on PD development and progression. This involves moving beyond the binary classification of patients as diabetic or non-diabetic, aiming to pave the way for the development of enhanced therapeutic interventions.
Collapse
Affiliation(s)
- Annekatrin König
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Tiago F. Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
- Max Planck Institute for Multidisciplinary Science, Göttingen, Germany
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle Upon Tyne, UK
| |
Collapse
|
37
|
Koike S, Saito Y, Ogasawara Y. Novel Fluorometric Assay of Antiglycation Activity Based on Methylglyoxal-Induced Protein Carbonylation. Antioxidants (Basel) 2023; 12:2030. [PMID: 38136150 PMCID: PMC10740428 DOI: 10.3390/antiox12122030] [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: 10/27/2023] [Revised: 11/18/2023] [Accepted: 11/20/2023] [Indexed: 12/24/2023] Open
Abstract
Advanced glycation end products (AGEs), which can have multiple structures, are formed at the sites where the carbonyl groups of reducing sugars bind to the free amino groups of proteins through the Maillard reaction. Some AGE structures exhibit fluorescence, and this fluorescence has been used to measure the formation and quantitative changes in carbonylated proteins. Recently, fluorescent AGEs have also been used as an index for the evaluation of compounds that inhibit protein glycation. However, the systems used to generate fluorescent AGEs from the reaction of reducing sugars and proteins used for the evaluation of antiglycation activity have not been determined through appropriate research; thus, problems remain regarding sensitivity, quantification, and precision. In the present study, using methylglyoxal (MGO), a reactive carbonyl compound to induce glycation, a comparative analysis of the mechanisms of formation of fluorescent substances from several types of proteins was conducted. The analysis identified hen egg lysozyme (HEL) as a protein that produces stronger fluorescent AGEs faster in the Maillard reaction with MGO. It was also found that the AGE structure produced in MGO-induced in HEL was argpyrimidine. By optimizing the reaction system, we developed a new evaluation method for compounds with antiglycation activity and established an efficient evaluation method (HEL-MGO assay) with greater sensitivity and accuracy than the conventional method, which requires high concentrations of bovine serum albumin and glucose. Furthermore, when compounds known to inhibit glycation were evaluated using this method, their antiglycation activities were clearly and significantly measured, demonstrating the practicality of this method.
Collapse
Affiliation(s)
| | | | - Yuki Ogasawara
- Department of Analytical Biochemistry, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo 204-8588, Japan; (S.K.); (Y.S.)
| |
Collapse
|
38
|
Coukos JS, Lee CW, Pillai KS, Shah H, Moellering RE. PARK7 Catalyzes Stereospecific Detoxification of Methylglyoxal Consistent with Glyoxalase and Not Deglycase Function. Biochemistry 2023; 62:3126-3133. [PMID: 37884446 PMCID: PMC10634309 DOI: 10.1021/acs.biochem.3c00325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 10/04/2023] [Accepted: 10/09/2023] [Indexed: 10/28/2023]
Abstract
The protein PARK7 (also known as DJ-1) has been implicated in several diseases, with the most notable being Parkinson's disease. While several molecular and cellular roles have been ascribed to DJ-1, there is no real consensus on what its true cellular functions are and how the loss of DJ-1 function may contribute to the pathogenesis of Parkinson's disease. Recent reports have implicated DJ-1 in the detoxification of several reactive metabolites that are produced during glycolytic metabolism, with the most notable being the α-oxoaldehyde species methylglyoxal. While it is generally agreed that DJ-1 is able to metabolize methylglyoxal to lactate, the mechanism by which it does so is hotly debated with potential implications for cellular function. In this work, we provide definitive evidence that recombinant DJ-1 produced in human cells prevents the stable glycation of other proteins through the conversion of methylglyoxal or a related alkynyl dicarbonyl probe to their corresponding α-hydroxy carboxylic acid products. This protective action of DJ-1 does not require a physical interaction with a target protein, providing direct evidence for a glutathione-free glyoxalase and not a deglycase mechanism of methylglyoxal detoxification. Stereospecific liquid chromatography-mass spectrometry (LC-MS) measurements further uncovered the existence of nonenzymatic production of racemic lactate from MGO under physiological buffer conditions, whereas incubation with DJ-1 predominantly produces l-lactate. Collectively, these studies provide direct support for the stereospecific conversion of MGO to l-lactate by DJ-1 in solution with negligible or no contribution of direct protein deglycation.
Collapse
Affiliation(s)
- John S. Coukos
- Department
of Chemistry, The University of Chicago, 929 E. 57th Street, Chicago, Illinois 60637, United States
| | - Chris W. Lee
- Department
of Chemistry, The University of Chicago, 929 E. 57th Street, Chicago, Illinois 60637, United States
| | - Kavya S. Pillai
- Department
of Chemistry, The University of Chicago, 929 E. 57th Street, Chicago, Illinois 60637, United States
| | - Hardik Shah
- University
of Chicago Medicine Comprehensive Cancer Center Metabolomics Platform, The University of Chicago, 900 E. 57th Street, Chicago, Illinois 60637, United States
| | - Raymond E. Moellering
- Department
of Chemistry, The University of Chicago, 929 E. 57th Street, Chicago, Illinois 60637, United States
- University
of Chicago Medicine Comprehensive Cancer Center Metabolomics Platform, The University of Chicago, 900 E. 57th Street, Chicago, Illinois 60637, United States
| |
Collapse
|
39
|
Sutkowska E, Fecka I, Marciniak D, Bednarska K, Sutkowska M, Hap K. Analysis of Methylglyoxal Concentration in a Group of Patients with Newly Diagnosed Prediabetes. Biomedicines 2023; 11:2968. [PMID: 38001968 PMCID: PMC10669086 DOI: 10.3390/biomedicines11112968] [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: 10/19/2023] [Accepted: 11/01/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND The abnormal serum concentration of methylglyoxal (MGO) has been presented as an indicator of chronic complications in diabetes (DM). Because such complications are also found in pre-DM, we decided to assess the concentration of this compound in individuals with pre-DM, without cardio-vascular diseases. METHODS Frozen samples from individuals newly diagnosed with pre-DM (N = 31) and healthy subjects (N = 11) were prepared and MGO concentration was determined using UHPLC-ESI-QqTOF-MS. RESULTS Statistical significance was established when the groups were compared for body weight, BMI, fasting glucose level, fatty liver and use of statins but not for the other descriptive parameters. The positive linear correlation showed that the higher HbA1c, the higher MGO concentration (p = 0.01). The values of MGO were within the normal range in both groups (mean value for pre-DM: 135.44 nM (±SD = 32.67) and for the control group: 143.25 nM (±SD = 17.93); p = 0.46 (±95% CI)), with no statistical significance between the groups. CONCLUSIONS We did not confirm the elevated MGO levels in the group of patients with pre-DM. The available data suggests a possible effect of statin intake on MGO levels. This thesis requires confirmation on a larger number of patients with an assessment of MGO levels before and after the introduction of statins.
Collapse
Affiliation(s)
- Edyta Sutkowska
- University Rehabilitation Centre, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland;
| | - Izabela Fecka
- Department of Pharmacognosy, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland; (I.F.); (K.B.)
- The Committee on Therapeutics and Pharmaceutical Sciences, The Polish Academy of Sciences, pl. Defilad 1, 00-901 Warszawa, Poland
| | - Dominik Marciniak
- Department of Drugs Form Technology, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland;
| | - Katarzyna Bednarska
- Department of Pharmacognosy, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland; (I.F.); (K.B.)
| | - Magdalena Sutkowska
- Faculty of Medicine, Wroclaw Medical University, Wybrzeże Ludwika Pasteura 1, 50-367 Wroclaw, Poland;
| | - Katarzyna Hap
- University Rehabilitation Centre, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland;
| |
Collapse
|
40
|
Oezer K, Kolibabka M, Gassenhuber J, Dietrich N, Fleming T, Schlotterer A, Morcos M, Wohlfart P, Hammes HP. The effect of GLP-1 receptor agonist lixisenatide on experimental diabetic retinopathy. Acta Diabetol 2023; 60:1551-1565. [PMID: 37423944 PMCID: PMC10520173 DOI: 10.1007/s00592-023-02135-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 06/10/2023] [Indexed: 07/11/2023]
Abstract
AIMS Glucagon-like peptide-1 receptor agonists are effective treatments for type 2 diabetes, effectively lowering glucose without weight gain and with low risk for hypoglycemia. However, their influence on the retinal neurovascular unit remains unclear. In this study, we analyzed the effects of the GLP-1 RA lixisenatide on diabetic retinopathy. METHODS Vasculo- and neuroprotective effects were assessed in experimental diabetic retinopathy and high glucose-cultivated C. elegans, respectively. In STZ-diabetic Wistar rats, acellular capillaries and pericytes (quantitative retinal morphometry), neuroretinal function (mfERG), macroglia (GFAP western blot) and microglia (immunohistochemistry) quantification, methylglyoxal (LC-MS/MS) and retinal gene expressions (RNA-sequencing) were determined. The antioxidant properties of lixisenatide were tested in C. elegans. RESULTS Lixisenatide had no effect on glucose metabolism. Lixisenatide preserved the retinal vasculature and neuroretinal function. The macro- and microglial activation was mitigated. Lixisenatide normalized some gene expression changes in diabetic animals to control levels. Ets2 was identified as a regulator of inflammatory genes. In C. elegans, lixisenatide showed the antioxidative property. CONCLUSIONS Our data suggest that lixisenatide has a protective effect on the diabetic retina, most likely due to a combination of neuroprotective, anti-inflammatory and antioxidative effects of lixisenatide on the neurovascular unit.
Collapse
Affiliation(s)
- Kuebra Oezer
- 5th Medical Department, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| | - Matthias Kolibabka
- 5th Medical Department, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | - Nadine Dietrich
- 5th Medical Department, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Thomas Fleming
- Department of Medicine I and Clinical Chemistry, Heidelberg University, Heidelberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Andrea Schlotterer
- 5th Medical Department, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Michael Morcos
- 5th Medical Department, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Stoffwechselzentrum Rhein-Pfalz, Belchenstraße 1-5, 68163, Mannheim, Germany
| | - Paulus Wohlfart
- Sanofi, MSAT M&I Bioassays and Compliance, Frankfurt, Germany
| | - Hans-Peter Hammes
- 5th Medical Department, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
41
|
Berends E, van Oostenbrugge RJ, Foulquier S, Schalkwijk CG. Methylglyoxal, a highly reactive dicarbonyl compound, as a threat for blood brain barrier integrity. Fluids Barriers CNS 2023; 20:75. [PMID: 37875994 PMCID: PMC10594715 DOI: 10.1186/s12987-023-00477-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/10/2023] [Indexed: 10/26/2023] Open
Abstract
The brain is a highly metabolically active organ requiring a large amount of glucose. Methylglyoxal (MGO), a by-product of glucose metabolism, is known to be involved in microvascular dysfunction and is associated with reduced cognitive function. Maintenance of the blood-brain barrier (BBB) is essential to maintain optimal brain function and a large amount of evidence indicates negative effects of MGO on BBB integrity. In this review, we summarized the current literature on the effect of MGO on the different cell types forming the BBB. BBB damage by MGO most likely occurs in brain endothelial cells and mural cells, while astrocytes are most resistant to MGO. Microglia on the other hand appear to be not directly influenced by MGO but rather produce MGO upon activation. Although there is clear evidence that MGO affects components of the BBB, the impact of MGO on the BBB as a multicellular system warrants further investigation. Diminishing MGO stress can potentially form the basis for new treatment strategies for maintaining optimal brain function.
Collapse
Affiliation(s)
- Eline Berends
- Department of Internal Medicine, Maastricht University, Universiteitssingel, Maastricht, 50 6229ER, The Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, Maastricht, 6229ER, The Netherlands
| | - Robert J van Oostenbrugge
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, Maastricht, 6229ER, The Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Universiteitssingel 40, Maastricht, 6229ER, The Netherlands
- Department of Neurology, Maastricht University Medical Centre (MUMC+), P. Debyelaan 25 6202AZ, Maastricht, The Netherlands
| | - Sébastien Foulquier
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, Maastricht, 6229ER, The Netherlands.
- Department of Neurology, Maastricht University Medical Centre (MUMC+), P. Debyelaan 25 6202AZ, Maastricht, The Netherlands.
- Department of Pharmacology and Toxicology, Maastricht University, Universiteitssingel 50 6229ER, Maastricht, The Netherlands.
| | - Casper G Schalkwijk
- Department of Internal Medicine, Maastricht University, Universiteitssingel, Maastricht, 50 6229ER, The Netherlands.
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, Maastricht, 6229ER, The Netherlands.
| |
Collapse
|
42
|
Miranda ER, Haus JM. Glyoxalase I is a novel target for the prevention of metabolic derangement. Pharmacol Ther 2023; 250:108524. [PMID: 37722607 DOI: 10.1016/j.pharmthera.2023.108524] [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: 03/01/2023] [Revised: 08/07/2023] [Accepted: 08/29/2023] [Indexed: 09/20/2023]
Abstract
Obesity prevalence in the US has nearly tripled since 1975 and a parallel increase in prevalence of type 2 diabetes (T2D). Obesity promotes a myriad of metabolic derangements with insulin resistance (IR) being perhaps the most responsible for the development of T2D and other related diseases such as cardiovascular disease. The precarious nature of IR development is such that it provides a valuable target for the prevention of further disease development. However, the mechanisms driving IR are numerous and complex making the development of viable interventions difficult. The development of metabolic derangement in the context of obesity promotes accumulation of reactive metabolites such as the reactive alpha-dicarbonyl methylglyoxal (MG). MG accumulation has long been appreciated as a marker of disease progression in patients with T2D as well as the development of diabetic complications. However, recent evidence suggests that the accumulation of MG occurs with obesity prior to T2D onset and may be a primary driving factor for the development of IR and T2D. Further, emerging evidence also suggests that this accumulation of MG with obesity may be a result in a loss of MG detoxifying capacity of glyoxalase I. In this review, we will discuss the evidence that posits MG accumulation because of GLO1 attenuation is a novel target mechanism of the development of metabolic derangement. In addition, we will also explore the regulation of GLO1 and the strategies that have been investigated so far to target GLO1 regulation for the prevention and treatment of metabolic derangement.
Collapse
Affiliation(s)
- Edwin R Miranda
- School of Kinesiology, University of Michigan, Ann Arbor, MI, United States of America; Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, United States of America
| | - Jacob M Haus
- School of Kinesiology, University of Michigan, Ann Arbor, MI, United States of America.
| |
Collapse
|
43
|
Muthaiyan Shanmugam M, Chaudhuri J, Sellegounder D, Sahu AK, Guha S, Chamoli M, Hodge B, Bose N, Amber C, Farrera DO, Lithgow G, Sarpong R, Galligan JJ, Kapahi P. Methylglyoxal-derived hydroimidazolone, MG-H1, increases food intake by altering tyramine signaling via the GATA transcription factor ELT-3 in Caenorhabditis elegans. eLife 2023; 12:e82446. [PMID: 37728328 PMCID: PMC10611433 DOI: 10.7554/elife.82446] [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: 08/04/2022] [Accepted: 09/19/2023] [Indexed: 09/21/2023] Open
Abstract
The Maillard reaction, a chemical reaction between amino acids and sugars, is exploited to produce flavorful food ubiquitously, from the baking industry to our everyday lives. However, the Maillard reaction also occurs in all cells, from prokaryotes to eukaryotes, forming advanced glycation end-products (AGEs). AGEs are a heterogeneous group of compounds resulting from the irreversible reaction between biomolecules and α-dicarbonyls (α-DCs), including methylglyoxal (MGO), an unavoidable byproduct of anaerobic glycolysis and lipid peroxidation. We previously demonstrated that Caenorhabditis elegans mutants lacking the glod-4 glyoxalase enzyme displayed enhanced accumulation of α-DCs, reduced lifespan, increased neuronal damage, and touch hypersensitivity. Here, we demonstrate that glod-4 mutation increased food intake and identify that MGO-derived hydroimidazolone, MG-H1, is a mediator of the observed increase in food intake. RNAseq analysis in glod-4 knockdown worms identified upregulation of several neurotransmitters and feeding genes. Suppressor screening of the overfeeding phenotype identified the tdc-1-tyramine-tyra-2/ser-2 signaling as an essential pathway mediating AGE (MG-H1)-induced feeding in glod-4 mutants. We also identified the elt-3 GATA transcription factor as an essential upstream regulator for increased feeding upon accumulation of AGEs by partially controlling the expression of tdc-1 gene. Furthermore, the lack of either tdc-1 or tyra-2/ser-2 receptors suppresses the reduced lifespan and rescues neuronal damage observed in glod-4 mutants. Thus, in C. elegans, we identified an elt-3 regulated tyramine-dependent pathway mediating the toxic effects of MG-H1 AGE. Understanding this signaling pathway may help understand hedonistic overfeeding behavior observed due to modern AGE-rich diets.
Collapse
Affiliation(s)
| | | | | | | | - Sanjib Guha
- The Buck Institute for Research on AgingNovatoUnited States
| | - Manish Chamoli
- The Buck Institute for Research on AgingNovatoUnited States
| | - Brian Hodge
- The Buck Institute for Research on AgingNovatoUnited States
| | - Neelanjan Bose
- The Buck Institute for Research on AgingNovatoUnited States
| | - Charis Amber
- Department of Chemistry, University of California, BerkeleyBerkeleyUnited States
| | - Dominique O Farrera
- Department of Pharmacology and Toxicology, College of Pharmacy, University of ArizonaTucsonUnited States
| | - Gordon Lithgow
- The Buck Institute for Research on AgingNovatoUnited States
| | - Richmond Sarpong
- Department of Chemistry, University of California, BerkeleyBerkeleyUnited States
| | - James J Galligan
- Department of Pharmacology and Toxicology, College of Pharmacy, University of ArizonaTucsonUnited States
| | - Pankaj Kapahi
- The Buck Institute for Research on AgingNovatoUnited States
- Department of Urology, University of California, San FranciscoSan FranciscoUnited States
| |
Collapse
|
44
|
Wei SL, Yang Y, Si WY, Zhou Y, Li T, Du T, Zhang P, Li XL, Duan RN, Duan RS, Yang CL. Methylglyoxal suppresses microglia inflammatory response through NRF2-IκBζ pathway. Redox Biol 2023; 65:102843. [PMID: 37573838 PMCID: PMC10440576 DOI: 10.1016/j.redox.2023.102843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/05/2023] [Accepted: 08/06/2023] [Indexed: 08/15/2023] Open
Abstract
Methylglyoxal (MGO) is a highly reactive metabolite generated by glycolysis. Although abnormal accumulation of MGO has been reported in several autoimmune diseases such as multiple sclerosis and rheumatoid arthritis, the role of MGO in autoimmune diseases has not yet been fully investigated. In this study, we found that the intracellular MGO levels increased in activated immune cells, such as microglia and lymphocytes. Treatment with MGO inhibited inflammatory cell accumulation in the spinal cord and ameliorated the clinical symptoms in EAE mice. Further analysis indicated that MGO suppressed M1-polarization of microglia cells and diminished their inflammatory cytokine production. MGO also inhibited the ability of microglial cells to recruit and activate lymphocytes by decreasing chemokine secretion and expression of co-stimulatory molecules. Furthermore, MGO negatively regulated glycolysis by suppressing glucose transporter 1 expression. Mechanically, we found that MGO could activate nuclear factor erythroid 2-related factor 2 (NRF2) pathway and NRF2 could bind to the promoter of IκBζ gene and suppressed its transcription and subsequently pro-inflammatory cytokine production. In conclusion, our results showed that MGO acts as an immunosuppressive metabolite by activating the NRF2-IκBζ.
Collapse
Affiliation(s)
- Shu-Li Wei
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250011, PR China
| | - Ying Yang
- Department of Pharmacy, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Wei-Yue Si
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250011, PR China
| | - Yang Zhou
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, PR China
| | - Tao Li
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, PR China
| | - Tong Du
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, PR China; Shandong Institute of Neuroimmunology, Jinan 250014, PR China
| | - Peng Zhang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, PR China; Shandong Institute of Neuroimmunology, Jinan 250014, PR China
| | - Xiao-Li Li
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, PR China; Shandong Institute of Neuroimmunology, Jinan 250014, PR China
| | - Ruo-Nan Duan
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Rui-Sheng Duan
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250011, PR China; Department of Neurology, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, PR China; Shandong Institute of Neuroimmunology, Jinan 250014, PR China.
| | - Chun-Lin Yang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, PR China; Shandong Institute of Neuroimmunology, Jinan 250014, PR China.
| |
Collapse
|
45
|
Bednarska K, Fecka I, Scheijen JLJM, Ahles S, Vangrieken P, Schalkwijk CG. A Citrus and Pomegranate Complex Reduces Methylglyoxal in Healthy Elderly Subjects: Secondary Analysis of a Double-Blind Randomized Cross-Over Clinical Trial. Int J Mol Sci 2023; 24:13168. [PMID: 37685975 PMCID: PMC10488144 DOI: 10.3390/ijms241713168] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 08/18/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
Reactive α-dicarbonyls (α-DCs), such as methylglyoxal (MGO), glyoxal (GO), and 3-deoxyglucosone (3-DG), are potent precursors in the formation of advanced glycation end products (AGEs). In particular, MGO and MGO-derived AGEs are thought to be involved in the development of vascular complications in diabetes. Experimental studies showed that citrus and pomegranate polyphenols can scavenge α-DCs. Therefore, the aim of this study was to evaluate the effect of a citrus and pomegranate complex (CPC) on the α-DCs plasma levels in a double-blind, placebo-controlled cross-over trial, where thirty-six elderly subjects were enrolled. They received either 500 mg of Citrus sinensis peel extract and 200 mg of Punica granatum concentrate in CPC capsules or placebo capsules for 4 weeks, with a 4-week washout period in between. For the determination of α-DCs concentrations, liquid chromatography tandem mass spectrometry was used. Following four weeks of CPC supplementation, plasma levels of MGO decreased by 9.8% (-18.7 nmol/L; 95% CI: -36.7, -0.7 nmol/L; p = 0.042). Our findings suggest that CPC supplementation may represent a promising strategy for mitigating the conditions associated with MGO involvement. This study was registered on clinicaltrials.gov as NCT03781999.
Collapse
Affiliation(s)
- Katarzyna Bednarska
- Department of Pharmacognosy, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland
| | - Izabela Fecka
- Department of Pharmacognosy, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland
- The Committee on Therapeutics and Pharmaceutical Sciences, The Polish Academy of Sciences, Pl. Defilad 1, 00-901 Warsaw, Poland
| | - Jean L. J. M. Scheijen
- Department of Internal Medicine, Maastricht University Medical Center+, 6229 ER Maastricht, The Netherlands; (J.L.J.M.S.); (P.V.); (C.G.S.)
- CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Sanne Ahles
- Department of Nutrition and Movement Sciences, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
- BioActor BV, 6229 GS Maastricht, The Netherlands
| | - Philippe Vangrieken
- Department of Internal Medicine, Maastricht University Medical Center+, 6229 ER Maastricht, The Netherlands; (J.L.J.M.S.); (P.V.); (C.G.S.)
- CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Casper G. Schalkwijk
- Department of Internal Medicine, Maastricht University Medical Center+, 6229 ER Maastricht, The Netherlands; (J.L.J.M.S.); (P.V.); (C.G.S.)
- CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
46
|
Rhein S, Inderhees J, Herrmann O, Othman A, Begemann K, Fleming T, Nawroth PP, Klika KD, Isa R, König IR, Royl G, Schwaninger M. Glyoxal in hyperglycaemic ischemic stroke - a cohort study. Cardiovasc Diabetol 2023; 22:173. [PMID: 37438755 DOI: 10.1186/s12933-023-01892-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/17/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND Hyperglycaemia is frequent in acute ischemic stroke and denotes a bad prognosis, even in the absence of pre-existing diabetes. However, in clinical trials treatment of elevated glucose levels with insulin did not improve stroke outcome, suggesting that collateral effects rather than hyperglycaemia itself aggravate ischemic brain damage. As reactive glucose metabolites, glyoxal and methylglyoxal are candidates for mediating the deleterious effects of hyperglycaemia in acute stroke. METHODS In 135 patients with acute stroke, we used liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS) to measure glyoxal, methylglyoxal and several of their glycated amino acid derivatives in serum. Results were verified in a second cohort of 61 stroke patients. The association of serum concentrations with standard stroke outcome scales (NIHSS, mRS) was tested. RESULTS Glucose, glyoxal, methylglyoxal, and the glyoxal-derived glycated amino acid Nδ-(5-hydro-4-imidazolon-2-yl)ornithine (G-H1) were positively correlated with a bad stroke outcome at 3 months as measured by mRS90, at least in one of the two cohorts. However, the glycated amino acids Nε-carboxyethyllysine (CEL) and in one cohort pyrraline showed an inverse correlation with stroke outcome probably reflecting lower food intake in severe stroke. Patients with a poor outcome had higher serum concentrations of glyoxal and methylglyoxal. CONCLUSIONS The glucose-derived α-dicarbonyl glyoxal and glycated amino acids arising from a reaction with glyoxal are associated with a poor outcome in ischemic stroke. Thus, lowering α-dicarbonyls or counteracting their action could be a therapeutic strategy for hyperglycaemic stroke.
Collapse
Affiliation(s)
- Sina Rhein
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center for Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
- German Centre for Cardiovascular Research, (DZHK), Hamburg-Lübeck-Kiel, Germany
| | - Julica Inderhees
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center for Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
- German Centre for Cardiovascular Research, (DZHK), Hamburg-Lübeck-Kiel, Germany
- Bioanalytic Core Facility, Center for Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
| | - Oliver Herrmann
- Department of Neurology, University of Heidelberg, Heidelberg, Germany
| | - Alaa Othman
- Bioanalytic Core Facility, Center for Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
| | - Kimberly Begemann
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center for Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
| | - Thomas Fleming
- Department of Internal Medicine, University of Heidelberg, Heidelberg, Germany
- German Research Centre for Diabetes Research, Düsseldorf, Germany
| | - Peter P Nawroth
- Department of Internal Medicine, University of Heidelberg, Heidelberg, Germany
| | - Karel D Klika
- Molecular Structure Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rakad Isa
- Department of Neurology, Center for Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
| | - Inke R König
- German Centre for Cardiovascular Research, (DZHK), Hamburg-Lübeck-Kiel, Germany
- Institute of Medical Biometry and Statistics, University of Lübeck, Lübeck, Germany
| | - Georg Royl
- Department of Neurology, Center for Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center for Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany.
- German Centre for Cardiovascular Research, (DZHK), Hamburg-Lübeck-Kiel, Germany.
| |
Collapse
|
47
|
Li Y, Hook JS, Ding Q, Xiao X, Chung SS, Mettlen M, Xu L, Moreland JG, Agathocleous M. Neutrophil metabolomics in severe COVID-19 reveal GAPDH as a suppressor of neutrophil extracellular trap formation. Nat Commun 2023; 14:2610. [PMID: 37147288 PMCID: PMC10162006 DOI: 10.1038/s41467-023-37567-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/20/2023] [Indexed: 05/07/2023] Open
Abstract
Severe COVID-19 is characterized by an increase in the number and changes in the function of innate immune cells including neutrophils. However, it is not known how the metabolome of immune cells changes in patients with COVID-19. To address these questions, we analyzed the metabolome of neutrophils from patients with severe or mild COVID-19 and healthy controls. We identified widespread dysregulation of neutrophil metabolism with disease progression including in amino acid, redox, and central carbon metabolism. Metabolic changes in neutrophils from patients with severe COVID-19 were consistent with reduced activity of the glycolytic enzyme GAPDH. Inhibition of GAPDH blocked glycolysis and promoted pentose phosphate pathway activity but blunted the neutrophil respiratory burst. Inhibition of GAPDH was sufficient to cause neutrophil extracellular trap (NET) formation which required neutrophil elastase activity. GAPDH inhibition increased neutrophil pH, and blocking this increase prevented cell death and NET formation. These findings indicate that neutrophils in severe COVID-19 have an aberrant metabolism which can contribute to their dysfunction. Our work also shows that NET formation, a pathogenic feature of many inflammatory diseases, is actively suppressed in neutrophils by a cell-intrinsic mechanism controlled by GAPDH.
Collapse
Affiliation(s)
- Yafeng Li
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jessica S Hook
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Qing Ding
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xue Xiao
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Stephen S Chung
- Department of Internal Medicine, Division of Hematology and Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Marcel Mettlen
- Department of Cell Biology, Quantitative Light Microscopy Core, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lin Xu
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jessica G Moreland
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Michalis Agathocleous
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
48
|
Knörlein A, Xiao Y, David Y. Leveraging histone glycation for cancer diagnostics and therapeutics. Trends Cancer 2023; 9:410-420. [PMID: 36804508 PMCID: PMC10121827 DOI: 10.1016/j.trecan.2023.01.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 02/22/2023]
Abstract
Cancer cells undergo metabolic reprogramming to rely mostly on aerobic glycolysis (the Warburg effect). The increased glycolytic intake enhances the intracellular levels of reactive sugars and sugar metabolites. These reactive species can covalently modify macromolecules in a process termed glycation. Histones are particularly susceptible to glycation, resulting in substantial alterations to chromatin structure, function, and transcriptional output. Growing evidence suggests a link between dysregulated metabolism of tumors and cancer proliferation through epigenetic changes. This review discusses recent advances in the understanding of histone glycation, its impact on the epigenetic landscape and cellular fate, and its role in cancer. In addition, we investigate the possibility of using histone glycation as biomarkers and targets for anticancer therapeutics.
Collapse
Affiliation(s)
- Anna Knörlein
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yang Xiao
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Tri-Institutional PhD Program in Chemical Biology, New York, NY, USA
| | - Yael David
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Tri-Institutional PhD Program in Chemical Biology, New York, NY, USA; Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA; Department of Physiology, Biophysics, and Systems Biology, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
49
|
Wang Q, Ai Z, Guo Q, Wang X, Dai C, Wang H, Sun J, Tang Y, Jiang D, Pei X, Chen R, Gou J, Yu L, Ding J, Wee ATS, Liu Y, Wei D. Photo-Enhanced Chemo-Transistor Platform for Ultrasensitive Assay of Small Molecules. J Am Chem Soc 2023; 145:10035-10044. [PMID: 37097713 DOI: 10.1021/jacs.2c13655] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
Compared with traditional assay techniques, field-effect transistors (FETs) have advantages such as fast response, high sensitivity, being label-free, and point-of-care detection, while lacking generality to detect a wide range of small molecules since most of them are electrically neutral with a weak doping effect. Here, we demonstrate a photo-enhanced chemo-transistor platform based on a synergistic photo-chemical gating effect in order to overcome the aforementioned limitation. Under light irradiation, accumulated photoelectrons generated from covalent organic frameworks offer a photo-gating modulation, amplifying the response to small molecule adsorption including methylglyoxal, p-nitroaniline, nitrobenzene, aniline, and glyoxal when measuring the photocurrent. We perform testing in buffer, artificial urine, sweat, saliva, and diabetic mouse serum. The limit of detection is down to 10-19 M methylglyoxal, about 5 orders of magnitude lower than existing assay technologies. This work develops a photo-enhanced FET platform to detect small molecules or other neutral species with enhanced sensitivity for applications in fields such as biochemical research, health monitoring, and disease diagnosis.
Collapse
Affiliation(s)
- Qiankun Wang
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
- Department of Macromolecular Science, Fudan University, Shanghai 200433, China
- Laboratory of Molecular Materials and Devices, Fudan University, Shanghai 200433, China
| | - Zhaolin Ai
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
- Department of Macromolecular Science, Fudan University, Shanghai 200433, China
- Laboratory of Molecular Materials and Devices, Fudan University, Shanghai 200433, China
| | - Qianying Guo
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
- Department of Macromolecular Science, Fudan University, Shanghai 200433, China
- Laboratory of Molecular Materials and Devices, Fudan University, Shanghai 200433, China
| | - Xuejun Wang
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
- Department of Macromolecular Science, Fudan University, Shanghai 200433, China
- Laboratory of Molecular Materials and Devices, Fudan University, Shanghai 200433, China
| | - Changhao Dai
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
- Department of Macromolecular Science, Fudan University, Shanghai 200433, China
- Laboratory of Molecular Materials and Devices, Fudan University, Shanghai 200433, China
| | - Hancheng Wang
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
- Department of Macromolecular Science, Fudan University, Shanghai 200433, China
| | - Jiang Sun
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
- Department of Macromolecular Science, Fudan University, Shanghai 200433, China
- Laboratory of Molecular Materials and Devices, Fudan University, Shanghai 200433, China
| | - Yanan Tang
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
- Department of Macromolecular Science, Fudan University, Shanghai 200433, China
- Laboratory of Molecular Materials and Devices, Fudan University, Shanghai 200433, China
| | - Dingding Jiang
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
- Department of Macromolecular Science, Fudan University, Shanghai 200433, China
- Laboratory of Molecular Materials and Devices, Fudan University, Shanghai 200433, China
| | - Xinjie Pei
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
- Department of Macromolecular Science, Fudan University, Shanghai 200433, China
- Laboratory of Molecular Materials and Devices, Fudan University, Shanghai 200433, China
| | - Renzhong Chen
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
- Department of Macromolecular Science, Fudan University, Shanghai 200433, China
- Laboratory of Molecular Materials and Devices, Fudan University, Shanghai 200433, China
| | - Jian Gou
- Department of Physics, National University of Singapore, 2 Science Drive 3, Singapore 117542, Singapore
| | - Lin Yu
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
- Department of Macromolecular Science, Fudan University, Shanghai 200433, China
| | - Jiandong Ding
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
- Department of Macromolecular Science, Fudan University, Shanghai 200433, China
| | - Andrew T S Wee
- Department of Physics, National University of Singapore, 2 Science Drive 3, Singapore 117542, Singapore
| | - Yunqi Liu
- Laboratory of Molecular Materials and Devices, Fudan University, Shanghai 200433, China
- Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Dacheng Wei
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
- Department of Macromolecular Science, Fudan University, Shanghai 200433, China
- Laboratory of Molecular Materials and Devices, Fudan University, Shanghai 200433, China
- Research Institute of Intelligent Complex Systems, Fudan University, Shanghai 200433, China
| |
Collapse
|
50
|
Abstract
Diabetes mellitus is the ninth leading cause of mortality worldwide. It is a complex disease that manifests as chronic hyperglycemia. Glucose exposure causes biochemical changes at the proteome level as reflected in accumulation of glycated proteins. A prominent example is hemoglobin A1c (HbA1c), a glycated protein widely accepted as a diabetic indicator. Another emerging biomarker is glycated albumin which has demonstrated utility in situations where HbA1c cannot be used. Other proteins undergo glycation as well thus impacting cellular function, transport and immune response. Accordingly, these glycated counterparts may serve as predictors for diabetic complications and thus warrant further inquiry. Fortunately, modern proteomics has provided unique analytic capability to enable improved and more comprehensive exploration of glycating agents and glycated proteins. This review broadly covers topics from epidemiology of diabetes to modern analytical tools such as mass spectrometry to facilitate a better understanding of diabetes pathophysiology. This serves as an attempt to connect clinically relevant questions with findings of recent proteomic studies to suggest future avenues of diabetes research.
Collapse
Affiliation(s)
- Aleks Shin
- Department of Pathology & Anatomical Sciences, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Shawn Connolly
- Department of Pathology & Anatomical Sciences, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Kuanysh Kabytaev
- Department of Pathology & Anatomical Sciences, School of Medicine, University of Missouri, Columbia, MO, United States.
| |
Collapse
|