1
|
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
|
2
|
Vašková J, Kováčová G, Pudelský J, Palenčár D, Mičková H. Methylglyoxal Formation-Metabolic Routes and Consequences. Antioxidants (Basel) 2025; 14:212. [PMID: 40002398 PMCID: PMC11852113 DOI: 10.3390/antiox14020212] [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: 12/18/2024] [Revised: 02/04/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Methylglyoxal (MGO), a by-product of glycolysis, plays a significant role in cellular metabolism, particularly under stress conditions. However, MGO is a potent glycotoxin, and its accumulation has been linked to the development of several pathological conditions due to oxidative stress, including diabetes mellitus and neurodegenerative diseases. This paper focuses on the biochemical mechanisms by which MGO contributes to oxidative stress, particularly through the formation of advanced glycation end products (AGEs), its interactions with antioxidant systems, and its involvement in chronic diseases like diabetes, neurodegeneration, and cardiovascular disorders. MGO exerts its effects through multiple signaling pathways, including NF-κB, MAPK, and Nrf2, which induce oxidative stress. Additionally, MGO triggers apoptosis primarily via intrinsic and extrinsic pathways, while endoplasmic reticulum (ER) stress is mediated through PERK-eIF2α and IRE1-JNK signaling. Moreover, the activation of inflammatory pathways, particularly through RAGE and NF-κB, plays a crucial role in the pathogenesis of these conditions. This study points out the connection between oxidative and carbonyl stress due to increased MGO formation, and it should be an incentive to search for a marker that could have prognostic significance or could be a targeted therapeutic intervention in various diseases.
Collapse
Affiliation(s)
- Janka Vašková
- Department of Medical Biology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 11 Košice, Slovakia
| | - Gabriela Kováčová
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik, 040 11 Košice, Slovakia; (G.K.)
| | - Jakub Pudelský
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik, 040 11 Košice, Slovakia; (G.K.)
| | - Drahomír Palenčár
- Department of Plastic Surgery, Faculty of Medicine, Comenius University Bratislava, 813 72 Bratislava, Slovakia
| | - Helena Mičková
- Department of Medical Biology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 11 Košice, Slovakia
| |
Collapse
|
3
|
Md Samsuzzaman, Hong SM, Lee JH, Park H, Chang KA, Kim HB, Park MG, Eo H, Oh MS, Kim SY. Depression like-behavior and memory loss induced by methylglyoxal is associated with tryptophan depletion and oxidative stress: a new in vivo model of neurodegeneration. Biol Res 2024; 57:87. [PMID: 39574138 PMCID: PMC11580208 DOI: 10.1186/s40659-024-00572-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 11/11/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Depression and memory loss are prevalent neurodegenerative disorders, with diabetic patients facing an elevated risk of brain dysfunction. Methylglyoxal (MGO) formation, which is heightened in diabetes owing to hyperglycemia and gut dysbiosis, may serve as a critical link between diabetes and brain diseases. Despite the high prevalence of MGO, the precise mechanisms underlying MGO-induced depression and memory loss remain unclear. RESULTS We investigated the effect of MGO stress on depression like-behavior and memory loss to elucidate the potential interplay between MGO-induced tryptophan (Trp) metabolism impairment and oxidative stress in the brain. It demonstrates that MGO induces depression-like behavior in mice, as confirmed by the OFT, TST, FST, SPT, and EPM behavioral tests. MGO led to the depletion of Trp and related neurotransmitters as 5-HT, EPI, and DA in the mouse brain. Additionally, MGO reduced the cell count in the DG, CA1, and CA3 hippocampal regions and modulated TPH2 levels in the brain. Notably, co-treatment with MGO and Trp mirrored the effects observed after Trp-null treatment in neurons, including reduced TPH1 and TPH2 levels and inhibition of neuronal outgrowth. Furthermore, MGO significantly altered the expression of key proteins associated with neurodegeneration, such as p-Tau, p-GSK-3β, APP, oAβ, BDNF, NGF, and p-TrkB. Concurrently, MGO activated MAPKs through ROS induction, triggering a redox imbalance by downregulating Nrf-2, Ho-1, TXNRD1, Trx, Sirt-3, and Sirt-5 expression levels, NAD+, and CAT activity in the mouse brain. This led to an accelerated neuroinflammatory response, as evidenced by increased expression of Iba-1, p-NF-κB, and the secretion of IL-6 and TNF-α. Importantly, Trp treatment ameliorated MGO-induced depression like-behavior and memory loss in mice and markedly mitigated increased expression of p-Tau, APP, p-ERK1/2, p-pJNK, and p-NF-κB in the brain. Likewise, Trp treatment also induced the expression of MGO detoxifying factors GLO-I and GLO-II and CAT activity, suggesting the induction of an antioxidant system and reduced inflammation by inhibiting IL-6 and TNF-α secretion. CONCLUSIONS Our data revealed that MGO-induced depression like-behavior and memory deficits resulted from disturbances in Trp, 5-HT, BDNF, and NGF levels, increased p-Tau and APP expression, neuroinflammation, and impaired redox status (Nrf-2/Ho-1/TXNRD1/Sirt3/5) in the brain.
Collapse
Affiliation(s)
- Md Samsuzzaman
- College of Pharmacy, Gachon University, #191, Hambakmoero, Yeonsu-gu, Incheon, 21936, Republic of Korea
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, MD, 21201, USA
| | - Seong-Min Hong
- College of Pharmacy, Gachon University, #191, Hambakmoero, Yeonsu-gu, Incheon, 21936, Republic of Korea
| | - Jae Hyuk Lee
- College of Pharmacy, Gachon University, #191, Hambakmoero, Yeonsu-gu, Incheon, 21936, Republic of Korea
| | - Hyunjun Park
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology (GAIHST), Gachon University, Incheon, Republic of Korea
- Neuroscience Research Institute, Gachon University, Incheon, Republic of Korea
| | - Keun-A Chang
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology (GAIHST), Gachon University, Incheon, Republic of Korea
- Neuroscience Research Institute, Gachon University, Incheon, Republic of Korea
- Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea
| | - Hyun-Bum Kim
- Department of East-West Medical Science, Graduate School of East-West Medical, Kyung Hee University, Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do, 446-701, Korea
| | - Myoung Gyu Park
- MetaCen Therapeutics Company, Changnyong-daero 256 Beon-gil, Yeongtong-gu, Suwon-si, Gyeonggi-do, 16229, Republic of Korea
| | - Hyeyoon Eo
- College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Myung Sook Oh
- College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Sun Yeou Kim
- College of Pharmacy, Gachon University, #191, Hambakmoero, Yeonsu-gu, Incheon, 21936, Republic of Korea.
- Gachon Institute of Pharmaceutical Science, Gachon University, #191, Hambakmoe-ro, Yeonsu-gu, Incheon, 21936, Republic of Korea.
| |
Collapse
|
4
|
Carone M, Premoli M, Bonini SA, Latsi R, Maccarinelli G, Memo M. Behavioral effects of two cannabidiol and cannabigerol-rich formulas on mice. Heliyon 2024; 10:e39938. [PMID: 39583801 PMCID: PMC11582752 DOI: 10.1016/j.heliyon.2024.e39938] [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: 08/01/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/26/2024] Open
Abstract
Cannabis sativa L. produces more than 100 specific bioactive compounds, known as cannabinoids. The major non-psychotropic Cannabis constituent is cannabidiol (CBD), which displays beneficial properties in a variety of medical conditions. However, the potential therapeutic role of other minor phytocannabinoids, such as cannabigerol (CBG), and their use in combination with CBD, has remained largely unexplored. In this study, we wanted to assess the in vivo effects of two novel non-psychotropic cannabinoid formulas, both containing relatively high percentages of CBD but differing mainly for CBG content, hereafter called CBG+ and CBG-formulas. We employed different behavioral tests to evaluate the effects of these formulas at three different dosages on mice locomotor activity, anxiety-related behaviors, short-term memory and sociability. We found that these two formulas display unique behavioral profiles: CBG + formula produced an increase in mice locomotor activity and displayed anxiolytic properties, whereas both formulas improved spatial short-term memory and social interactions. The results obtained suggest that different combinations of phytocannabinoids are able to determine different behavioral effects and highlight the importance of studying the effects of less known phytocannabinoids (like CBG), which used in combination with other phytocannabinoids can change the profile of action of other active compounds (such as CBD).
Collapse
Affiliation(s)
| | | | - Sara Anna Bonini
- Department of Molecular and Translational Medicine, University of Brescia, 25123, Brescia, Italy
| | - Rozana Latsi
- Department of Molecular and Translational Medicine, University of Brescia, 25123, Brescia, Italy
| | - Giuseppina Maccarinelli
- Department of Molecular and Translational Medicine, University of Brescia, 25123, Brescia, Italy
| | - Maurizio Memo
- Department of Molecular and Translational Medicine, University of Brescia, 25123, Brescia, Italy
| |
Collapse
|
5
|
Wang H, Boeren S, Bakker W, Rietjens IMCM, Saccenti E, Zheng L. An integrated proteomics and metabolomics analysis of methylglyoxal-induced neurotoxicity in a human neuroblastoma cell line. NPJ Sci Food 2024; 8:84. [PMID: 39448607 PMCID: PMC11502746 DOI: 10.1038/s41538-024-00328-0] [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: 07/03/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024] Open
Abstract
This study aimed to highlight the molecular and biochemical changes induced by methylglyoxal (MGO) exposure in SH-SY5Y human neuroblastoma cells, and to explore how these changes contribute to its neurotoxicity, utilizing an integrated proteomics and metabolomics approach. Using label-free quantitative nanoLC-MS/MS proteomics and targeted LC-TQ-MS/MS-based metabolomics, the results revealed that MGO exposure, particularly at cytotoxic levels, significantly altered the proteome and metabolome of SH-SY5Y cells. Analysis of proteomics data showed significant alterations in cellular functions including protein synthesis, cellular structural integrity, mitochondrial function, and oxidative stress responses. Analysis of metabolomics and integration of metabolomics and proteomics data highlighted significant changes in key metabolic pathways including arginine biosynthesis, glutathione metabolism, cysteine and methionine metabolism, and the tricarboxylic acid cycle. These results suggest that MGO exposure induced both toxic effects and adaptive responses in cells. MGO exposure led to increased endoplasmic reticulum stress, disruptions in cellular adhesion and extracellular matrix integrity, mitochondrial dysfunction, and amino acid metabolism disruption, contributing to cellular toxicity. Conversely, cells exhibited adaptive responses by upregulating protein synthesis, activating the Nrf2 pathway, and reprogramming metabolism to counteract dicarbonyl stress and maintain energy levels. Furthermore, a set of key proteins and metabolites associated with these changes were shown to exhibit a significant concentration-dependent decrease or increase in their expression levels with increasing MGO concentrations, suggesting their potential as biomarkers for MGO exposure. Taken together, these findings provide insight into the molecular mechanisms underlying MGO-induced neurotoxicity and potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Haomiao Wang
- Division of Toxicology, Wageningen University & Research, Wageningen, The Netherlands
| | - Sjef Boeren
- Laboratory of Biochemistry, Wageningen University & Research, Wageningen, The Netherlands
| | - Wouter Bakker
- Division of Toxicology, Wageningen University & Research, Wageningen, The Netherlands
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University & Research, Wageningen, The Netherlands
| | - Edoardo Saccenti
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, Wageningen, The Netherlands.
| | - Liang Zheng
- Division of Toxicology, Wageningen University & Research, Wageningen, The Netherlands.
| |
Collapse
|
6
|
Kirlioglu Balcioglu SS, Kurt Sabitay I, Uysal A, Yildirim Servi E, Yaman M, Mizrak OF, Ozturk N, Isiksacan N, Guclu O. Evaluation of changes in carbonyl stress markers with treatment in male patients with bipolar disorder manic episode: A controlled study. J Affect Disord 2024; 362:1-8. [PMID: 38944288 DOI: 10.1016/j.jad.2024.06.112] [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: 03/13/2024] [Revised: 05/26/2024] [Accepted: 06/26/2024] [Indexed: 07/01/2024]
Abstract
BACKGROUND Carbonyl stress, a metabolic state characterized by elevated production of reactive carbonyl compounds (RCCs), is closely related to oxidative stress and has been implicated in various diseases. This study aims to investigate carbonyl stress parameters in drug-free bipolar disorder (BD) patients compared to healthy controls, explore their relationship with clinical features, and assess the effect of treatment on these parameters. METHODS Patients with a primary diagnosis of a manic episode of BD and healthy controls were recruited. Exclusion criteria included intellectual disability, presence of neurological diseases, chronic medical conditions such as diabetes mellitus and metabolic syndrome, and clinical signs of inflammation. Levels of serum carbonyl stress parameters were determined using high-performance liquid chromatography. RESULTS Levels of glyoxal (GO) and methylglyoxal (MGO) did not differ between pre- and post-treatment patients, but malondialdehyde (MDA) levels decreased significantly post-treatment. Pre-treatment MGO and MDA levels were higher in patients compared to controls, and these differences persisted post-treatment. After adjusting for BMI and waist circumference, only MDA levels remained significantly higher in patients compared to controls. LIMITATIONS The study's limitations include the exclusion of female patients, which precluded any assessment of potential gender differences, and the lack of analysis of the effect of specific mood stabilizers or antipsychotic drugs. CONCLUSIONS This study is the first to focus on carbonyl stress markers in BD, specifically GO, MGO, and MDA. MDA levels remained significantly higher in patients, suggesting a potential role in BD pathophysiology. MGO levels were influenced by metabolic parameters, indicating a potential link to neurotoxicity in BD. Further research with larger cohorts is needed to better understand the role of RCCs in BD and their potential as therapeutic targets.
Collapse
Affiliation(s)
- Simge Seren Kirlioglu Balcioglu
- Department of Psychiatry, Basaksehir Cam and Sakura City Hospital, University of Health Sciences, Istanbul, Turkiye; Hamidiye Institute of Health Sciences, University of Health Sciences, Istanbul, Turkiye.
| | - Imren Kurt Sabitay
- Department of Psychiatry, Basaksehir Cam and Sakura City Hospital, University of Health Sciences, Istanbul, Turkiye
| | - Aybegum Uysal
- Department of Psychiatry, Basaksehir Cam and Sakura City Hospital, University of Health Sciences, Istanbul, Turkiye
| | - Esra Yildirim Servi
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Istanbul Sabahattin Zaim University, Istanbul, Turkiye
| | - Mustafa Yaman
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Istanbul Sabahattin Zaim University, Istanbul, Turkiye
| | - Omer Faruk Mizrak
- Sabri Ulker Food and Nutrition Center, Istanbul Sabahattin Zaim University, Istanbul, Turkiye
| | | | - Nilgun Isiksacan
- Hamidiye Institute of Health Sciences, University of Health Sciences, Istanbul, Turkiye; Department of Biochemistry, Dr Sadi Konuk Training and Research Hospital, Istanbul, Turkiye
| | - Oya Guclu
- Department of Psychiatry, Basaksehir Cam and Sakura City Hospital, University of Health Sciences, Istanbul, Turkiye
| |
Collapse
|
7
|
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
|
8
|
Abate G, Pezzotta A, Pucci M, Bortolotto V, Ribaudo G, Bonini SA, Mastinu A, Maccarinelli G, Ongaro A, Tirelli E, Zizioli D, Gianoncelli A, Memo M, Grilli M, Uberti D. The Bioactive Gamma-Oryzanol from Oryza sativa L. Promotes Neuronal Differentiation in Different In Vitro and In Vivo Models. Antioxidants (Basel) 2024; 13:969. [PMID: 39199215 PMCID: PMC11352202 DOI: 10.3390/antiox13080969] [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: 05/08/2024] [Revised: 07/28/2024] [Accepted: 08/02/2024] [Indexed: 09/01/2024] Open
Abstract
Gamma-oryzanol (ORY), found in rice (Oryza sativa L.), is a mixture of ferulic acid esters with triterpene alcohols, well-known for its antioxidant and anti-inflammatory properties. Our past research demonstrated its positive impact on cognitive function in adult mice, influencing synaptic plasticity and neuroprotection. In this study, we explored whether ORY can exert neuro-differentiating effects by using different experimental models. For this purpose, chemical characterization identified four components that are most abundant in ORY. In human neuroblastoma cells, we showed ORY's ability to stimulate neurite outgrowth, upregulating the expression of GAP43, BDNF, and TrkB genes. In addition, ORY was found to guide adult mouse hippocampal neural progenitor cells (NPCs) toward a neuronal commitment. Microinjection of ORY in zebrafish Tg (-3.1 neurog1:GFP) amplified neurog1-GFP signal, islet1, and bdnf mRNA levels. Zebrafish nrf2a and nrf2b morphants (MOs) were utilized to assess ORY effects in the presence or absence of Nrf2. Notably, ORY's ability to activate bdnf was nullified in nrf2a-MO and nrf2b-MO. Furthermore, computational analysis suggested ORY's single components have different affinities for the Keap1-Kelch domain. In conclusion, although more in-depth studies are needed, our findings position ORY as a potential source of bioactive molecules with neuro-differentiating potential involving the Nrf2 pathway.
Collapse
Affiliation(s)
- Giulia Abate
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (G.A.); (G.R.); (S.A.B.); (A.M.); (G.M.); (A.O.); (E.T.); (D.Z.); (A.G.); (M.M.); (D.U.)
| | - Alex Pezzotta
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20133 Milan, Italy;
| | - Mariachiara Pucci
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (G.A.); (G.R.); (S.A.B.); (A.M.); (G.M.); (A.O.); (E.T.); (D.Z.); (A.G.); (M.M.); (D.U.)
| | - Valeria Bortolotto
- Laboratory of Neuroplasticity, University of Piemonte Orientale, 28100 Novara, Italy; (V.B.); (M.G.)
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, 28100 Novara, Italy
| | - Giovanni Ribaudo
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (G.A.); (G.R.); (S.A.B.); (A.M.); (G.M.); (A.O.); (E.T.); (D.Z.); (A.G.); (M.M.); (D.U.)
| | - Sara A. Bonini
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (G.A.); (G.R.); (S.A.B.); (A.M.); (G.M.); (A.O.); (E.T.); (D.Z.); (A.G.); (M.M.); (D.U.)
| | - Andrea Mastinu
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (G.A.); (G.R.); (S.A.B.); (A.M.); (G.M.); (A.O.); (E.T.); (D.Z.); (A.G.); (M.M.); (D.U.)
| | - Giuseppina Maccarinelli
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (G.A.); (G.R.); (S.A.B.); (A.M.); (G.M.); (A.O.); (E.T.); (D.Z.); (A.G.); (M.M.); (D.U.)
| | - Alberto Ongaro
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (G.A.); (G.R.); (S.A.B.); (A.M.); (G.M.); (A.O.); (E.T.); (D.Z.); (A.G.); (M.M.); (D.U.)
| | - Emanuela Tirelli
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (G.A.); (G.R.); (S.A.B.); (A.M.); (G.M.); (A.O.); (E.T.); (D.Z.); (A.G.); (M.M.); (D.U.)
| | - Daniela Zizioli
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (G.A.); (G.R.); (S.A.B.); (A.M.); (G.M.); (A.O.); (E.T.); (D.Z.); (A.G.); (M.M.); (D.U.)
| | - Alessandra Gianoncelli
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (G.A.); (G.R.); (S.A.B.); (A.M.); (G.M.); (A.O.); (E.T.); (D.Z.); (A.G.); (M.M.); (D.U.)
| | - Maurizio Memo
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (G.A.); (G.R.); (S.A.B.); (A.M.); (G.M.); (A.O.); (E.T.); (D.Z.); (A.G.); (M.M.); (D.U.)
| | - Mariagrazia Grilli
- Laboratory of Neuroplasticity, University of Piemonte Orientale, 28100 Novara, Italy; (V.B.); (M.G.)
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, 28100 Novara, Italy
| | - Daniela Uberti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (G.A.); (G.R.); (S.A.B.); (A.M.); (G.M.); (A.O.); (E.T.); (D.Z.); (A.G.); (M.M.); (D.U.)
| |
Collapse
|
9
|
Khalid M, Adem A. The dynamic roles of advanced glycation end products. VITAMINS AND HORMONES 2024; 125:1-29. [PMID: 38997161 DOI: 10.1016/bs.vh.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2024]
Abstract
Advanced glycation end products (AGEs) are a heterogeneous group of potentially harmful molecules that can form as a result of a non-enzymatic reaction between reducing sugars and proteins, lipids, or nucleic acids. The total body pool of AGEs reflects endogenously produced AGEs as well as exogeneous AGEs that come from sources such as diet and the environment. Engagement of AGEs with their cellular receptor, the receptor for advanced glycation end products (RAGE), which is expressed on the surface of various cell types, converts a brief pulse of cellular activation to sustained cellular dysfunction and tissue destruction. The AGEs/RAGE interaction triggers a cascade of intracellular signaling pathways such as mitogen-activated protein kinase/extracellular signal-regulated kinase, phosphoinositide 3-kinases, transforming growth factor beta, c-Jun N-terminal kinases (JNK), and nuclear factor kappa B, which leads to the production of pro-inflammatory cytokines, chemokines, adhesion molecules, and oxidative stress. All these events contribute to the progression of several chronic diseases. This chapter will provide a comprehensive understanding of the dynamic roles of AGEs in health and disease which is crucial to develop interventions that prevent and mitigate the deleterious effects of AGEs accumulation.
Collapse
Affiliation(s)
- Mariyam Khalid
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Abdu Adem
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
10
|
Zhang WY, Zhao CM, Wang CS, Xie X, Li YQ, Chen BB, Feng L, Jiang P. Methylglyoxal accumulation contributes to accelerated brain aging in spontaneously hypertensive rats. Free Radic Biol Med 2024; 210:108-119. [PMID: 37984752 DOI: 10.1016/j.freeradbiomed.2023.11.012] [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: 08/19/2023] [Revised: 10/25/2023] [Accepted: 11/15/2023] [Indexed: 11/22/2023]
Abstract
While it is well-acknowledged that neurovascular dysfunction in hypertension is tightly associated with accelerated brain aging, we contend that the deleterious effects of hypertension may extend beyond affecting only the arteries. Methylglyoxal (MG) derived from glycolysis, is involved in the accumulation of advanced glycated end products (AGEs), which are the hallmarks of neurodegenerative disorders. Therefore, the present study aims to firstly investigate the role of MG metabolism in the hypertension-accelerated brain aging process. The results of our study indicate that the levels of MG increase with age in both the plasma and hippocampus of SHRs at 12, 16, and 30 weeks old. AGE methylglyoxal-hydro imidazoline-1 (MG-H1) is primarily localized in astrocytes, while its presence was not observed in neurons and microglia within the hypertensive hippocampus. Our observations also suggest that angiotensin II (Ang II) enhances glucose uptake and glycolysis while reducing the expression of Glo1 in cultured astrocytes. N-acetylcysteine (NAC) was found to counteract the increase in escape latency and inhibit the activation of the AGEs-RAGE axis in 30-week-old SHRs. NAC decreased Iba-1 immunofluorescence intensity, inhibited the levels of pro-inflammatory markers, and enhanced the abundance of anti-inflammatory markers in the hippocampus of SHRs. Moreover, NAC reduced the immunofluorescence signal of 4HNE and increased the content of GSH and SOD in SHRs. Finally, NAC was observed to inhibit apoptosis in the hippocampus of SHRs. Collectively, we firstly showed the enhanced accumulation of MG in the hypertensive brain, whereas the clearance of MG by NAC treatment mitigated the aging process and attenuated AGEs generation, neuroinflammation, and oxidative damage.
Collapse
Affiliation(s)
- Wen-Yuan Zhang
- Department of Pharmacy, Zhongshan City People's Hospital, Zhongshan, 528403, China; School of Pharmaceutical Sciences, Zunyi Medical University, Zunyi, 510006, China
| | - Cui-Mei Zhao
- Department of Pharmacy, Zhongshan City People's Hospital, Zhongshan, 528403, China; School of Pharmaceutical Sciences, Zunyi Medical University, Zunyi, 510006, China
| | - Chang-Shui Wang
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
| | - Xin Xie
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
| | - Yu-Qi Li
- Department of cardiology, Zhongshan City People's Hospital, Zhongshan, 528403, China
| | - Bei-Bei Chen
- ADFA School of Science, University of New South Wales, Canberra, Australia
| | - Lei Feng
- Department of Neurosurgery, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China.
| | - Pei Jiang
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China; Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, 272000, China.
| |
Collapse
|
11
|
Patil RS, Tupe RS. Communal interaction of glycation and gut microbes in diabetes mellitus, Alzheimer's disease, and Parkinson's disease pathogenesis. Med Res Rev 2024; 44:365-405. [PMID: 37589449 DOI: 10.1002/med.21987] [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: 09/23/2022] [Revised: 07/12/2023] [Accepted: 08/06/2023] [Indexed: 08/18/2023]
Abstract
Diabetes and its complications, Alzheimer's disease (AD), and Parkinson's disease (PD) are increasing gradually, reflecting a global threat vis-à-vis expressing the essentiality of a substantial paradigm shift in research and remedial actions. Protein glycation is influenced by several factors, like time, temperature, pH, metal ions, and the half-life of the protein. Surprisingly, most proteins associated with metabolic and neurodegenerative disorders are generally long-lived and hence susceptible to glycation. Remarkably, proteins linked with diabetes, AD, and PD share this characteristic. This modulates protein's structure, aggregation tendency, and toxicity, highlighting renovated attention. Gut microbes and microbial metabolites marked their importance in human health and diseases. Though many scientific shreds of evidence are proposed for possible change and dysbiosis in gut flora in these diseases, very little is known about the mechanisms. Screening and unfolding their functionality in metabolic and neurodegenerative disorders is essential in hunting the gut treasure. Therefore, it is imperative to evaluate the role of glycation as a common link in diabetes and neurodegenerative diseases, which helps to clarify if modulation of nonenzymatic glycation may act as a beneficial therapeutic strategy and gut microbes/metabolites may answer some of the crucial questions. This review briefly emphasizes the common functional attributes of glycation and gut microbes, the possible linkages, and discusses current treatment options and therapeutic challenges.
Collapse
Affiliation(s)
- Rahul Shivaji Patil
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Rashmi Santosh Tupe
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Pune, Maharashtra, India
| |
Collapse
|
12
|
Nikray N, Abharian N, Jafari Ashtiani S, Kobarfard F, Faizi M. Comparative Evaluation of Aminoguanidine, Semicarbazide and Thiosemicarbazide Treatment for Methylglyoxal-Induced Neurological Toxicity in Experimental Models. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2024; 23:e153322. [PMID: 39830657 PMCID: PMC11742376 DOI: 10.5812/ijpr-153322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 08/24/2024] [Accepted: 08/26/2024] [Indexed: 01/22/2025]
Abstract
Background Advanced glycation end products (AGEs) are complex compounds that play a critical role in neurological disorders, including the pathogenesis of Alzheimer's disease. Methylglyoxal (MG) is recognized as the primary precursor of AGEs. Methylglyoxal is produced endogenously and also introduced through dietary exposures. Objectives This study aimed to investigate and compare the effects of aminoguanidine (AG), semicarbazide (SC), and thiosemicarbazide (TSC) on MG-induced neurological toxicity in rats. Methods Male Wistar rats were exposed orally to MG, MG + AG, MG + SC, and MG + TSC for 70 days. Neurobehavioral, biochemical, and histopathological changes were evaluated. Results The findings indicated that oral administration of MG for 70 days resulted in memory impairment and increased anxiety in neurobehavioral tests. Additionally, MG elevated protein carbonylation in brain tissues. Semicarbazide was found to prevent MG-induced memory problems, while both SC and AG reduced carbonyl content in brain tissues. Aminoguanidine and TSC were effective in alleviating anxiety induced by MG exposure. Histopathological analysis revealed that MG caused cell damage and neuronal necrosis in the hippocampus, particularly in the cornu ammonis 1 and 3 (CA1 and CA3) and AG, SC, and TSC improved neuronal survival specifically in the CA1 and DG areas. Conclusions The data suggest that SC, AG, and TSC may offer neuroprotective effects against MG-induced neurobehavioral toxicity. Further studies are required to explore the mechanisms of action of these compounds.
Collapse
Affiliation(s)
- Noushin Nikray
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nikoo Abharian
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahin Jafari Ashtiani
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farzad Kobarfard
- Department of Medicinal Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehrdad Faizi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Yonamine CY, Michalani MLE, Moreira RJ, Machado UF. Glucose Transport and Utilization in the Hippocampus: From Neurophysiology to Diabetes-Related Development of Dementia. Int J Mol Sci 2023; 24:16480. [PMID: 38003671 PMCID: PMC10671460 DOI: 10.3390/ijms242216480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/12/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023] Open
Abstract
The association of diabetes with cognitive dysfunction has at least 60 years of history, which started with the observation that children with type 1 diabetes mellitus (T1D), who had recurrent episodes of hypoglycemia and consequently low glucose supply to the brain, showed a deficit of cognitive capacity. Later, the growing incidence of type 2 diabetes mellitus (T2D) and dementia in aged populations revealed their high association, in which a reduced neuronal glucose supply has also been considered as a key mechanism, despite hyperglycemia. Here, we discuss the role of glucose in neuronal functioning/preservation, and how peripheral blood glucose accesses the neuronal intracellular compartment, including the exquisite glucose flux across the blood-brain barrier (BBB) and the complex network of glucose transporters, in dementia-related areas such as the hippocampus. In addition, insulin resistance-induced abnormalities in the hippocampus of obese/T2D patients, such as inflammatory stress, oxidative stress, and mitochondrial stress, increased generation of advanced glycated end products and BBB dysfunction, as well as their association with dementia/Alzheimer's disease, are addressed. Finally, we discuss how these abnormalities are accompained by the reduction in the expression and translocation of the high capacity insulin-sensitive glucose transporter GLUT4 in hippocampal neurons, which leads to neurocytoglycopenia and eventually to cognitive dysfunction. This knowledge should further encourage investigations into the beneficial effects of promising therapeutic approaches which could improve central insulin sensitivity and GLUT4 expression, to fight diabetes-related cognitive dysfunctions.
Collapse
Affiliation(s)
- Caio Yogi Yonamine
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark;
| | - Maria Luiza Estimo Michalani
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil; (M.L.E.M.); (R.J.M.)
| | - Rafael Junges Moreira
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil; (M.L.E.M.); (R.J.M.)
| | - Ubiratan Fabres Machado
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil; (M.L.E.M.); (R.J.M.)
| |
Collapse
|
14
|
Işık M, Tunç A, Duran HE, Naldan ME, Yılmaz A, Koçak MN, Beydemir Ş. Evaluation of the relationship among gene expressions and enzyme activities with antioxidant role and presenilin 1 expression in Alzheimer's disease. J Cell Mol Med 2023; 27:3388-3394. [PMID: 37772794 PMCID: PMC10623531 DOI: 10.1111/jcmm.17953] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/15/2023] [Accepted: 09/01/2023] [Indexed: 09/30/2023] Open
Abstract
It is known that oxidative stress originating from reactive oxygen species plays a role in the pathogenesis of Alzheimer's disease. In this study, the role of antioxidant status associated with oxidative stress in Alzheimer's disease was investigated. Peripheral blood samples were obtained from 28 healthy individuals (as control) and 28 Alzheimer's patients who met the National Institute of Neurological and Communicative Diseases and Stroke/Alzheimer's Disease and Related Disorders Association criteria. Catalase, glutathione S-transferase and paraoxonase 1 enzyme activities in blood plasma and glutathione S-transferase enzyme activities in erythrocytes were determined by spectrophotometer. Catalase, glutathione S-transferase and presenilin 1 gene expressions in leukocytes were determined using qRT-PCR. Data were analysed with SPSS one-way anova, a LSD post hoc test at p < 0.05. The activity of each enzyme was significantly reduced in Alzheimer's patients compared to control. The catalase gene expression level did not change compared to the control. Glutathione S-transferase and presenilin 1 gene expression levels were increased compared to the control.
Collapse
Affiliation(s)
- Mesut Işık
- Department of Bioengineering, Faculty of EngineeringBilecik Şeyh Edebali UniversityBilecikTurkey
| | - Abdullah Tunç
- Department of Occupational Health and Safety, Faculty of Health SciencesBingöl UniversityBingölTurkey
| | - Hatice Esra Duran
- Department of Medical Biochemistry, Faculty of MedicineKafkas UniversityKarsTurkey
| | | | - Aslan Yılmaz
- Department of NeurologyÇekirge Public HospitalBursaTurkey
| | - Mehmet Nuri Koçak
- Department of Neurology, Faculty of MedicineAtatürk UniversityErzurumTurkey
| | - Şükrü Beydemir
- Department of Biochemistry, Faculty of PharmacyAnadolu UniversityEskişehirTurkey
| |
Collapse
|
15
|
Yonamine CY, Passarelli M, Suemoto CK, Pasqualucci CA, Jacob-Filho W, Alves VAF, Marie SKN, Correa-Giannella ML, Britto LR, Machado UF. Postmortem Brains from Subjects with Diabetes Mellitus Display Reduced GLUT4 Expression and Soma Area in Hippocampal Neurons: Potential Involvement of Inflammation. Cells 2023; 12:cells12091250. [PMID: 37174649 PMCID: PMC10177173 DOI: 10.3390/cells12091250] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 04/13/2023] [Accepted: 04/23/2023] [Indexed: 05/15/2023] Open
Abstract
Diabetes mellitus (DM) is an important risk factor for dementia, which is a common neurodegenerative disorder. DM is known to activate inflammation, oxidative stress, and advanced glycation end products (AGEs) generation, all capable of inducing neuronal dysfunctions, thus participating in the neurodegeneration progress. In that process, disturbed neuronal glucose supply plays a key role, which in hippocampal neurons is controlled by the insulin-sensitive glucose transporter type 4 (GLUT4). We investigated the expression of GLUT4, nuclear factor NF-kappa B subunit p65 [NFKB (p65)], carboxymethyllysine and synapsin1 (immunohistochemistry), and soma area in human postmortem hippocampal samples from control, obese, and obese+DM subjects (41 subjects). Moreover, in human SH-SY5Y neurons, tumor necrosis factor (TNF) and glycated albumin (GA) effects were investigated in GLUT4, synapsin-1 (SYN1), tyrosine hydroxylase (TH), synaptophysin (SYP) proteins, and respective genes; NFKB binding activity in the SLC2A4 promoter; effects of increased histone acetylation grade by histone deacetylase 3 (HDAC3) inhibition. Hippocampal neurons (CA4 area) of obese+DM subjects displayed reduced GLUT4 expression and neuronal soma area, associated with increased expression of NFKB (p65). Challenges with TNF and GA decreased the SLC2A4/GLUT4 expression in SH-SY5Y neurons. TNF decreased SYN1, TH, and SYP mRNAs and respective proteins, and increased NFKB binding activity in the SLC2A4 promoter. Inhibition of HDAC3 increased the SLC2A4 expression and the total neuronal content of CRE-binding proteins (CREB/ICER), and also counterbalanced the repressor effect of TNF upon these parameters. This study revealed reduced postmortem human hippocampal GLUT4 content and neuronal soma area accompanied by increased proinflammatory activity in the brains of DM subjects. In isolated human neurons, inflammatory activation by TNF reduced not only the SLC2A4/GLUT4 expression but also the expression of some genes related to neuronal function (SYN1, TH, SYP). These effects may be related to epigenetic regulations (H3Kac and H4Kac status) since they can be counterbalanced by inhibiting HDAC3. These results uncover the improvement in GLUT4 expression and/or the inhibition of HDAC3 as promising therapeutic targets to fight DM-related neurodegeneration.
Collapse
Affiliation(s)
- Caio Yogi Yonamine
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Marisa Passarelli
- Laboratório de Lipides (LIM-10) do HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-000, Brazil
- Programa de Pos-Graduação em Medicina, Universidade Nove de Julho (UNINOVE), São Paulo 01525-000, Brazil
| | - Claudia Kimie Suemoto
- Divisao de Geriatria, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo 01246-000, Brazil
| | | | - Wilson Jacob-Filho
- Divisao de Geriatria, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo 01246-000, Brazil
| | - Venâncio Avancini Ferreira Alves
- Laboratório de Investigação Médica em Patologia Hepática, (LIM14) do Hospital das Clínicas (HCFMUSP), Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-000, Brazil
| | | | - Maria Lucia Correa-Giannella
- Laboratorio de Carboidratos e Radioimunoensaio (LIM-18) do Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo 01246-000, Brazil
| | - Luiz Roberto Britto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Ubiratan Fabres Machado
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| |
Collapse
|
16
|
Croitoru DO, Piguet V. Methylglyoxal Autoimmunity: A Hidden Link in HS and Associated Diseases? J Invest Dermatol 2023; 143:183-185. [PMID: 36681420 DOI: 10.1016/j.jid.2022.09.649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 09/22/2022] [Accepted: 09/22/2022] [Indexed: 01/21/2023]
Affiliation(s)
- David O Croitoru
- Division of Dermatology, Department of Medicine, University of Toronto, Toronto, Canada; Division of Dermatology, Department of Medicine, Women's College Hospital, Toronto, Canada
| | - Vincent Piguet
- Division of Dermatology, Department of Medicine, University of Toronto, Toronto, Canada; Division of Dermatology, Department of Medicine, Women's College Hospital, Toronto, Canada.
| |
Collapse
|
17
|
Deng Y, Wang X, Zhang Y, Zhang C, Xie P, Huang L. Inhibitory effect of Ginkgo biloba seeds peptides on methylglyoxal-induced glycations. Food Chem Toxicol 2023; 172:113587. [PMID: 36596446 DOI: 10.1016/j.fct.2022.113587] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 01/01/2023]
Abstract
The aim of this study was to investigate the antiglycation activity and mechanism of two identified peptides, Valine-Valine-Phenylalanine-Proline-Glycine-Cysteine-Proline-Glutamic acid (VVFPGCPE) and Serine-Valine-Aspartic acid-Aspartic acid-Proline-Arginine-Threonine-Lysine (SVDDPRTL), from Ginkgo biloba seeds protein hydrolysates. Both VVFPGCPE and SVDDPRTL were efficient in bovine serum albumin (BSA)-methylglyoxal (MGO) model to inhibit BSA glycation, while VVFPGCPE showed higher antiglycation activity than SVDDPRTL. In antioxidant assays, VVFPGCPE scavenged more hydroxyl and super anion radicals, and chelated more Fe2+. Moreover, VVFPGCPE was more efficient in alleviating glycoxidation since it retained higher content of tryptophan and reduced dityrosine and kynurenine generation. Compared with SVDDPRTL, VVFPGCPE showed better performance in inhibiting protein aggregation and amyloid-like fibrillation formation. Therefore, VVFPGCPE was selected for further mechanism study. The circular dichroism analysis suggested VVFPGCPE could preserve α-helix structure and stabilize protein structure. The MGO trapping assay indicated VVFPGCPE (5 mg/mL) could capture 66.25% MGO within 24 h, and the mass spectrometry revealed VVFPGCPE could trap MGO by forming VVFPGCPE-mono-MGO adducts. Besides, molecular simulations suggested VVFPGCPE could interact with key glycation residues, arginine and lysine residues, of BSA mainly through van der Waals and hydrogen bonds. This study might supply a theoretical basis for the development of VVFPGCPE as an effective antiglycation agent.
Collapse
Affiliation(s)
- Yejun Deng
- Institute of Chemical Industry of Forest Products, CAF, National Engineering Lab. for Biomass Chemical Utilization, Key and Open Lab. of Forest Chemical Engineering, SFA, Key Lab. of Biomass Energy and Material, Jiangsu Province, Nanjing, 210042, China.
| | - Xiang Wang
- Institute of Chemical Industry of Forest Products, CAF, National Engineering Lab. for Biomass Chemical Utilization, Key and Open Lab. of Forest Chemical Engineering, SFA, Key Lab. of Biomass Energy and Material, Jiangsu Province, Nanjing, 210042, China.
| | - Yang Zhang
- Institute of Chemical Industry of Forest Products, CAF, National Engineering Lab. for Biomass Chemical Utilization, Key and Open Lab. of Forest Chemical Engineering, SFA, Key Lab. of Biomass Energy and Material, Jiangsu Province, Nanjing, 210042, China.
| | - Caihong Zhang
- Institute of Chemical Industry of Forest Products, CAF, National Engineering Lab. for Biomass Chemical Utilization, Key and Open Lab. of Forest Chemical Engineering, SFA, Key Lab. of Biomass Energy and Material, Jiangsu Province, Nanjing, 210042, China.
| | - Pujun Xie
- Institute of Chemical Industry of Forest Products, CAF, National Engineering Lab. for Biomass Chemical Utilization, Key and Open Lab. of Forest Chemical Engineering, SFA, Key Lab. of Biomass Energy and Material, Jiangsu Province, Nanjing, 210042, China.
| | - Lixin Huang
- Institute of Chemical Industry of Forest Products, CAF, National Engineering Lab. for Biomass Chemical Utilization, Key and Open Lab. of Forest Chemical Engineering, SFA, Key Lab. of Biomass Energy and Material, Jiangsu Province, Nanjing, 210042, China; Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University Nanjing, 210037, China.
| |
Collapse
|
18
|
Wei CC, Li SW, Wu CT, How CM, Pan MH. Dietary Methylglyoxal Exposure Induces Alzheimer's Disease by Promoting Amyloid β Accumulation and Disrupting Autophagy in Caenorhabditis elegans. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:10011-10021. [PMID: 35917150 DOI: 10.1021/acs.jafc.2c03411] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Methylglyoxal (MG) is a precursor of advanced glycation end products usually generated during cooking. The high level of MG in the brain is correlated to the pathogenesis of Alzheimer's disease (AD). However, it is not clear if MG consumed through the diet can cause AD-related toxicity. Herein, the Caenorhabditis elegans (C. elegans) AD model was used to investigate the neurotoxicity after long-term MG exposure at dietary levels. The results showed that C. elegans locomotive behaviors were significantly decreased after 0.1, 0.5, and 1 mM MG exposure (p < 0.001). In amyloid β (Aβ)-expressing transgenic C. elegans strains, 0.5 mM MG significantly promoted Aβ accumulation by around 50% in day-8 CL2006 (p < 0.001), enhanced paralysis in CL4176 (p < 0.001) and CL2006 (p < 0.01), and made CL2355 around 17% more vulnerable to 5-HT, indicating impaired serotonin reuptake (p < 0.05). Additionally, 0.5 mM MG significantly increased the reactive oxygen species level (p < 0.001) by inhibiting the expression of stress-response genes including sod-3, gst-4, and hsp-16.2 in day-8 aged worms. Moreover, the autophagic pathway was disrupted through lgg-1, vps-34, and bec-1 expression after MG exposure and Aβ accumulation. Treatment with the citrus flavonoid nobiletin reduced the MG-induced toxicity (p < 0.001). Overall, these findings imply that it is possible to exacerbate AD pathogenesis by MG exposure through the diet.
Collapse
Affiliation(s)
- Chia-Cheng Wei
- Institute of Food Safety and Health, College of Public Health, National Taiwan University, Taipei 10055, Taiwan
- Department of Public Health, College of Public Health, National Taiwan University, Taipei 10055, Taiwan
| | - Shang-Wei Li
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo 113-8656, Japan
| | - Chia-Tung Wu
- Institute of Food Safety and Health, College of Public Health, National Taiwan University, Taipei 10055, Taiwan
| | - Chun Ming How
- Department of Bioenvironmental Systems Engineering, College of Bio-Resources and Agriculture, National Taiwan University, Taipei 10617, Taiwan
| | - Min-Hsiung Pan
- Institute of Food Science and Technology, College of Bio-Resources and Agriculture, National Taiwan University, Taipei 10617, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan
- Department of Health and Nutrition Biotechnology, Asia University, Taichung 41354, Taiwan
| |
Collapse
|
19
|
Pucci M, Mandrone M, Chiocchio I, Sweeney EM, Tirelli E, Uberti D, Memo M, Poli F, Mastinu A, Abate G. Different Seasonal Collections of Ficus carica L. Leaves Diversely Modulate Lipid Metabolism and Adipogenesis in 3T3-L1 Adipocytes. Nutrients 2022; 14:nu14142833. [PMID: 35889791 PMCID: PMC9323846 DOI: 10.3390/nu14142833] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/29/2022] [Accepted: 07/05/2022] [Indexed: 12/15/2022] Open
Abstract
Due to the high prevalence of obesity and type 2 diabetes, adipogenesis dysfunction and metabolic disorders are common features in the elderly population. Thus, the identification of novel compounds with anti-adipogenic and lipolytic effects is highly desirable to reduce diabetes complications. Plants represent an important source of bioactive compounds. To date, the antidiabetic potential of several traditional plants has been reported, among which Ficus carica L. is one of the most promising. Considering that plant metabolome changes in response to a number of factors including seasonality, the aim of this study was to evaluate whether Ficus carica leaves extracts collected in autumn (FCa) and spring (FCs) differently modulate lipid metabolism and adipogenesis in 3T3-L1 adipocytes. The 1H-NMR profile of the extracts showed that FCs have a higher content of caffeic acid derivatives, glucose, and sucrose than FCa. In contrast, FCa showed a higher concentration of malic acid and furanocoumarins, identified as psoralen and bergapten. In vitro testing showed that only FCa treatments were able to significantly decrease the lipid content (Ctrl vs. FCa 25 μg/mL, 50 μg/mL and 80 μg/mL; p < 0.05, p < 0.01 and p < 0.001, respectively). Furthermore, FCa treatments were able to downregulate the transcriptional pathway of adipogenesis and insulin sensitivity in 3T3-L1 adipocytes. In more detail, FCa 80 μg/mL significantly decreased the gene expression of PPARγ (p < 0.05), C/EBPα (p < 0.05), Leptin (p < 0.0001), adiponectin (p < 0.05) and GLUT4 (p < 0.01). In conclusion, this study further supports an in-depth investigation of F. carica leaves extracts as a promising source of active compounds useful for targeting obesity and diabetes.
Collapse
Affiliation(s)
- Mariachiara Pucci
- Department of Molecular and Translational Medicine, Division of Pharmacology, University of Brescia, 25123 Brescia, Italy; (M.P.); (E.M.S.); (E.T.); (D.U.); (M.M.); (G.A.)
| | - Manuela Mandrone
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Irnerio 42, 40126 Bologna, Italy; (M.M.); (I.C.); (F.P.)
| | - Ilaria Chiocchio
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Irnerio 42, 40126 Bologna, Italy; (M.M.); (I.C.); (F.P.)
| | - Eileen Mac Sweeney
- Department of Molecular and Translational Medicine, Division of Pharmacology, University of Brescia, 25123 Brescia, Italy; (M.P.); (E.M.S.); (E.T.); (D.U.); (M.M.); (G.A.)
| | - Emanuela Tirelli
- Department of Molecular and Translational Medicine, Division of Pharmacology, University of Brescia, 25123 Brescia, Italy; (M.P.); (E.M.S.); (E.T.); (D.U.); (M.M.); (G.A.)
| | - Daniela Uberti
- Department of Molecular and Translational Medicine, Division of Pharmacology, University of Brescia, 25123 Brescia, Italy; (M.P.); (E.M.S.); (E.T.); (D.U.); (M.M.); (G.A.)
| | - Maurizio Memo
- Department of Molecular and Translational Medicine, Division of Pharmacology, University of Brescia, 25123 Brescia, Italy; (M.P.); (E.M.S.); (E.T.); (D.U.); (M.M.); (G.A.)
| | - Ferruccio Poli
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Irnerio 42, 40126 Bologna, Italy; (M.M.); (I.C.); (F.P.)
| | - Andrea Mastinu
- Department of Molecular and Translational Medicine, Division of Pharmacology, University of Brescia, 25123 Brescia, Italy; (M.P.); (E.M.S.); (E.T.); (D.U.); (M.M.); (G.A.)
- Correspondence: ; Tel.: +39-030-371-7509
| | - Giulia Abate
- Department of Molecular and Translational Medicine, Division of Pharmacology, University of Brescia, 25123 Brescia, Italy; (M.P.); (E.M.S.); (E.T.); (D.U.); (M.M.); (G.A.)
| |
Collapse
|
20
|
Neuroprotective Effects and Therapeutic Potential of the Citrus Flavonoid Hesperetin in Neurodegenerative Diseases. Nutrients 2022; 14:nu14112228. [PMID: 35684025 PMCID: PMC9183194 DOI: 10.3390/nu14112228] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 12/13/2022] Open
Abstract
Neurodegenerative disorders affect more than fifty million Americans each year and represent serious health threats as the population ages. Neuroinflammation and oxidative stress are critical in the onset, progression, and pathogenesis of neurodegenerative diseases such as Alzheimer’s (AD), Parkinson’s (PD), and amyotrophic lateral sclerosis (ALS). A wide range of natural compounds has been investigated because of their antioxidant, anti-inflammatory, and neuroprotective properties. The citrus flavonoid hesperetin (HPT), an aglycone of hesperidin found in oranges, mandarins, and lemons, has been extensively reported to exert neuroprotective effects in experimental models of neurogenerative diseases. This review has compiled multiple studies on HPT in both in vivo and in vitro models to study neurodegeneration. We focused on the modulatory effects of hesperetin on the release of cellular anti-inflammatory and antioxidative stress mediators. Additionally, this review discusses the hesperetin effect in maintaining the levels of microRNA (miRNA) and modulating autophagy as it relates to hesperetin’s protective mechanisms against neurodegeneration. Moreover, this review is focused on providing experimental data for hesperetin’s potential as a neuroprotective compound and discusses reported evidence that HPT crosses the blood–brain barrier. In summary, this review shows the evidence available in the literature to indicate the efficacy of hesperetin in delaying the onset of neurodegenerative diseases.
Collapse
|
21
|
Physiological, Biochemical, and Agronomic Trait Responses of Nigella sativa Genotypes to Water Stress. HORTICULTURAE 2022. [DOI: 10.3390/horticulturae8030193] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Water stress may affect the growth, physiology, morphology, biochemistry, and productivity of Nigella sativa (black cumin), a medicinal and aromatic plant. Measuring these parameters under various irrigation regimes could provide useful information for successful genotype selection and breeding. Therefore, these agronomically significant features were evaluated in ten black cumin genotypes (Afghanistan, Pakistan, Syria, India, Arak, Isfahan, Semirom, Shahreza, Shahrekord, and Mashhad) under three irrigation regimes (40% (I1), 60% (I2), and 80% (I3) of permissible moisture discharge) during the 2017 to 2018 growing seasons. Water stress was shown to increase the levels of carotenoids (Cars), proline, total soluble carbohydrates (TSC), malondialdehyde (MDA), hydrogen peroxide (H2O2), catalase (CAT), and ascorbate peroxidase (APX) activities but reduced the relative water content (RWC) and chlorophyll content. The highest increases in Cars, TSC, proline, CAT, and APX were noted in the Arak, Isfahan, Semirom, Shahreza, Shahrekord, and Mashhad genotypes under the I3 water regime, respectively. At the same time, the lowest decrease was observed in chlorophyll, H2O2, and relative water content (RWC) in Semirom. According to the stress susceptibility index, the most resistant genotypes were Shahrekord under I2 and Semirom under I3. These data demonstrate that the irrigation regimes affected the physiological, biochemical, and morphological features of black cumin both qualitatively and quantitatively, although the impact varied depending upon the genotype, irrigation regime, and traits. As such, the results presented represent valuable information with which to inform future selection and breeding programs for drought-tolerant black cumin. This is of particular significance considering global climate change.
Collapse
|