1
|
Szabo C. Role of cystathionine-β-synthase and hydrogen sulfide in down syndrome. Neurotherapeutics 2025:e00584. [PMID: 40187942 DOI: 10.1016/j.neurot.2025.e00584] [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: 01/23/2025] [Revised: 03/15/2025] [Accepted: 03/25/2025] [Indexed: 04/07/2025] Open
Abstract
Down syndrome (DS) is a genetic condition where the person affected by it is born with an additional - full or partial - copy of chromosome 21. DS presents with characteristic morphological features and is associated with a wide range of biochemical alterations and maladaptations. Cystathionine-β-synthase (CBS) - one of the key mammalian enzymes responsible for the biogenesis of the gaseous transmitter hydrogen sulfide (H2S) - is located on chromosome 21, and people with DS exhibit a significant upregulation of this enzyme in their brain and other organs. Even though 3-mercaptopyruvate sulfurtransferase - another key mammalian enzyme responsible for the biogenesis of H2S and of reactive polysulfides - is not located on chromosome 21, there is also evidence for the upregulation of this enzyme in DS cells. The hypothesis that excess H2S in DS impairs mitochondrial function and cellular bioenergetics was first proposed in the 1990s and has been substantiated and expanded upon over the past 25 years. DS cells are in a state of metabolic suppression due to H2S-induced, reversible inhibition of mitochondrial Complex IV activity. The impairment of aerobic ATP generation in DS cells is partially compensated by an upregulation of glycolysis. The DS-associated metabolic impairment can be reversed by pharmacological CBS inhibition or CBS silencing. In rodent models of DS, CBS upregulation and H2S overproduction contribute to the development of cognitive dysfunction, alter brain electrical activity, and promote reactive gliosis: pharmacological inhibition or genetic correction of CBS overactivation reverses these alterations. CBS can be considered a preclinically validated drug target for the experimental therapy of DS.
Collapse
Affiliation(s)
- Csaba Szabo
- Section of Pharmacology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Switzerland.
| |
Collapse
|
2
|
Liu B, Dong K, Zhao Y, Wang X, Sun Z, Xie F, Qian L. Depletion of MGO or Its Derivatives Ameliorate CUMS-Induced Neuroinflammation. Cells 2025; 14:397. [PMID: 40136646 PMCID: PMC11941696 DOI: 10.3390/cells14060397] [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: 01/03/2025] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/27/2025] Open
Abstract
Advanced glycation end products (AGEs) are a series of structurally complex and harmful compounds formed through the reaction between the carbonyl group of reducing sugars (such as glucose and fructose) and the free amino groups of proteins, lipids, or nucleic acids. Excessive accumulation of AGEs in the body can trigger oxidative stress, induce inflammatory responses, and contribute to the development of diabetes, atherosclerosis, and neurological disorders. Within the category of dicarbonyl compounds, methylglyoxal (MGO)-a byproduct resulting from glucose degradation-serves as a pivotal precursor in the formation of AGEs and the induction of neurotoxicity. Specifically, AGEs generated from MGO display significant cytotoxicity toward cells in the central nervous system. Therefore, we aimed to investigate the role of MGO-AGEs in neuroinflammation mediated by CUMS. Interestingly, we found that the overexpression of glyoxalase 1 (GLO1) reduced the levels of MGO in corticosterone-treated microglia, thereby alleviating the inflammatory response. Furthermore, overexpression of GLO1 in the hippocampus of chronically stressed mice reduced MGO levels, mitigating CUMS-induced neuroinflammation and cognitive impairment. Additionally, when using the receptor for advanced glycation end products (RAGE) inhibitor FPS-ZM1 in primary microglia cells, we observed that despite corticosterone-induced elevation of MGO, no significant inflammatory response occurred. This suggests that RAGE clearance can reduce MGO-AGE-mediated neurotoxicity. Subsequently, we used FPS-ZM1 to treat chronically stressed mice and found that it significantly ameliorated neuroinflammation and cognitive dysfunction. These results suggest that targeting MGO metabolism could serve as a therapeutic approach to manage neuroinflammation in stress-related mental disorders.
Collapse
Affiliation(s)
- Bing Liu
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100191, China; (B.L.); (K.D.); (Y.Z.); (X.W.); (Z.S.)
| | - Ke Dong
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100191, China; (B.L.); (K.D.); (Y.Z.); (X.W.); (Z.S.)
- School of Medicine, South China University of Technology, Guangzhou 511442, China
| | - Yun Zhao
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100191, China; (B.L.); (K.D.); (Y.Z.); (X.W.); (Z.S.)
| | - Xue Wang
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100191, China; (B.L.); (K.D.); (Y.Z.); (X.W.); (Z.S.)
| | - Zhaowei Sun
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100191, China; (B.L.); (K.D.); (Y.Z.); (X.W.); (Z.S.)
| | - Fang Xie
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100191, China; (B.L.); (K.D.); (Y.Z.); (X.W.); (Z.S.)
| | - Lingjia Qian
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100191, China; (B.L.); (K.D.); (Y.Z.); (X.W.); (Z.S.)
| |
Collapse
|
3
|
Rao SP, Imam-Fulani AO, Xie W, Phillip S, Chennavajula K, Lind EB, Zhang Y, Vince R, Lee MK, More SS. Oral prodrug of a novel glutathione surrogate reverses metabolic dysregulation and attenuates neurodegenerative process in APP/PS1 mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.15.633247. [PMID: 39868172 PMCID: PMC11761491 DOI: 10.1101/2025.01.15.633247] [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: 01/28/2025]
Abstract
Glycation-induced oxidative stress underlies the numerous metabolic ravages of Alzheimer's disease (AD). Reduced glutathione levels in AD lead to increased oxidative stress, including glycation-induced pathology. Previously, we showed that the accumulation of reactive 1,2-dicarbonyls such as methylglyoxal, the major precursor of non-enzymatic glycation products, was reduced by the increased function of GSH-dependent glyoxalase-1 enzyme in the brain. In this two-pronged study, we evaluate the therapeutic efficacy of an orally bioavailable prodrug of our lead glyoxalase substrate, pro-ψ-GSH, for the first time in a transgenic Alzheimer's disease mouse model. This prodrug delivers pharmacodynamically relevant brain concentrations of ψ-GSH upon oral delivery. Chronic oral dosing of pro-ψ-GSH effectively reverses the cognitive decline observed in the APP/PS1 mouse model. The prodrug successfully mirrors the robust effects of the parent drug i.e., reducing amyloid pathology, glycation stress, neuroinflammation, and the resultant neurodegeneration in these mice. We also report the first metabolomics study of such a treatment, which yields key biomarkers linked to the reversal of AD-related metabolic dysregulation. Collectively, this study establishes pro-ψ-GSH as a viable, disease-modifying therapy for AD and paves the way for further preclinical advancement of such therapeutics. Metabolomic signatures identified could prove beneficial in the development of treatment-specific clinically translatable biomarkers.
Collapse
Affiliation(s)
- Swetha Pavani Rao
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Aminat O. Imam-Fulani
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Wei Xie
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Samuel Phillip
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Krishna Chennavajula
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Erin B. Lind
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Ying Zhang
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN, USA
| | - Robert Vince
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Michael K. Lee
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Swati S. More
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
4
|
Sian-Hulsmann J, Riederer P, Michel TM. Metabolic Dysfunction in Parkinson's Disease: Unraveling the Glucose-Lipid Connection. Biomedicines 2024; 12:2841. [PMID: 39767747 PMCID: PMC11673947 DOI: 10.3390/biomedicines12122841] [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: 09/24/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
Despite many years of research into the complex neurobiology of Parkinson's disease, the precise aetiology cannot be pinpointed down to one causative agent but rather a multitude of mechanisms. Current treatment options can alleviate symptomsbut only slightly slow down the progression and not cure the disease and its underlying causes. Factors that play a role in causing the debilitating neurodegenerative psycho-motoric symptoms include genetic alterations, oxidative stress, neuroinflammation, general inflammation, neurotoxins, iron toxicity, environmental influences, and mitochondrial dysfunction. Recent findings suggest that the characteristic abnormal protein aggregation of alpha-synuclein and destruction of substantia nigra neurons might be due to mitochondrial dysfunction related to disturbances in lipid and glucose metabolism along with insulin resistance. The latter mechanism of action might be mediated by insulin receptor substrate docking to proteins that are involved in neuronal survival and signaling related to cell destruction. The increased risk of developing Type 2 Diabetes Mellitus endorses a connection between metabolic dysfunction and neurodegeneration. Here, we explore and highlight the potential role of glycolipid cellular insults in the pathophysiology of the disorder, opening up new promising avenues for the treatment of PD. Thus, antidiabetic drugs may be employed as neuromodulators to hinder the progression of the disorder.
Collapse
Affiliation(s)
- Jeswinder Sian-Hulsmann
- Department of Human Anatomy and Medical Physiology, University of Nairobi, P.O. Box 30197, Nairobi 00100, Kenya;
| | - Peter Riederer
- Research Unit of Psychiatry, Department of Psychiatry, Odense, Region of Southern Denmark, University Hospital of Southern Denmark, 5000 Odense, Denmark;
| | - Tanja Maria Michel
- Research Unit of Psychiatry, Department of Psychiatry, Odense, Region of Southern Denmark, University Hospital of Southern Denmark, 5000 Odense, Denmark;
| |
Collapse
|
5
|
Erukainure OL, Chukwuma CI. African walnut ( Plukenetia conophora) oil promotes glucose uptake while improving energy metabolism and steroidogenesis and maintaining surface architecture in rat testes. Front Nutr 2024; 11:1505453. [PMID: 39628465 PMCID: PMC11611562 DOI: 10.3389/fnut.2024.1505453] [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: 10/02/2024] [Accepted: 10/30/2024] [Indexed: 12/06/2024] Open
Abstract
Background African walnut (Plukenetia conophora) oil (AWO) has been reported for its nutritional and medicinal properties and has been employed for the management of metabolic diseases including hyperglycemia-mediated ailments. Objective In the present study, AWO was investigated for its ability to stimulate glucose uptake and its effect on energy metabolism, steroidogenesis, and tissue morphology in isolated testes of Wistar rats. Methods Isolated testes were incubated with AWO (30-240 μg/mL) in the presence of 11.1 mMol glucose at 37°C for 2 h. Control consisted of testes incubated with glucose only, while normal control consisted of testes not incubated with AWO and/or glucose. The standard antidiabetic drug was metformin. Results and conclusion Incubation with AWO led to significant increase in glucose uptake, hexokinase, glyoxalase 1, glutathione reductase, glutathione peroxidase, 3β-hydroxysteroid dehydrogenase, 17β-hydroxysteroid dehydrogenase activities, GLUT4, glutathione, and ATP levels while concomitantly suppressing glucose-6-phosphatase, fructose-1,6-biphosphatase, glycogen phosphorylase, aldose reductase, polyol dehydrogenase, E-NTPDase, and ATPase activities. Furthermore, incubation with AWO led to improved testicular morphology while elevating testicular levels of magnesium, sulfur, potassium, calcium, and iron. Fatty acid profiling with GC-MS revealed linoleic acid and linolenic acid as the predominant essential fatty acids in AWO. Molecular docking analysis revealed potent molecular interactions of linoleic acid and linolenic acid with GLUT4. These results suggest the ability of AWO to improve testicular glucose metabolism and steroidogenesis and can be explored in the management of male infertility.
Collapse
Affiliation(s)
- Ochuko L. Erukainure
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Doornfontein, South Africa
| | - Chika I. Chukwuma
- Centre for Quality of Health and Living, Faculty of Health and Environmental Sciences, Central University of Technology, Bloemfontein, South Africa
| |
Collapse
|
6
|
Mariño L, Belén Uceda A, Leal F, Adrover M. Insight into the Effect of Methylglyoxal on the Conformation, Function, and Aggregation Propensity of α-Synuclein. Chemistry 2024; 30:e202400890. [PMID: 38687053 DOI: 10.1002/chem.202400890] [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: 03/03/2024] [Revised: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 05/02/2024]
Abstract
It is well-known that people suffering from hyperglycemia have a higher propensity to develop Parkinson's disease (PD). One of the most plausible mechanisms linking these two pathologies is the glycation of neuronal proteins and the pathological consequences of it. α-Synuclein, a key component in PD, can be glycated at its fifteen lysine. In fact, the end products of this process have been detected on aggregated α-synuclein isolated from in vivo. However, the consequences of glycation are not entirely clear, which are of crucial importance to understand the mechanism underlying the connection between diabetes and PD. To better clarify this, we have here examined how methylglyoxal (the most important carbonyl compound found in the cytoplasm) affects the conformation and aggregation propensity of α-synuclein, as well as its ability to cluster and fuse synaptic-like vesicles. The obtained data prove that methylglyoxal induces the Lys-Lys crosslinking through the formation of MOLD. However, this does not have a remarkable effect on the averaged conformational ensemble of α-synuclein, although it completely depletes its native propensity to form soluble oligomers and insoluble amyloid fibrils. Moreover, methylglyoxal has a disrupting effect on the ability of α-synuclein to bind, cluster and fusion synaptic-like vesicles.
Collapse
Affiliation(s)
- Laura Mariño
- Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Institut de Recerca en Ciències de la Salut (IdISBa), Departament de Química, Universitat de les Illes Balears, Ctra, Valldemossa km 7.5, E-07122, Palma de Mallorca, Spain
| | - Ana Belén Uceda
- Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Institut de Recerca en Ciències de la Salut (IdISBa), Departament de Química, Universitat de les Illes Balears, Ctra, Valldemossa km 7.5, E-07122, Palma de Mallorca, Spain
| | - Francisco Leal
- Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Institut de Recerca en Ciències de la Salut (IdISBa), Departament de Química, Universitat de les Illes Balears, Ctra, Valldemossa km 7.5, E-07122, Palma de Mallorca, Spain
| | - Miquel Adrover
- Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Institut de Recerca en Ciències de la Salut (IdISBa), Departament de Química, Universitat de les Illes Balears, Ctra, Valldemossa km 7.5, E-07122, Palma de Mallorca, Spain
| |
Collapse
|
7
|
Coccini T, Caloni F, Russo LA, Villani L, Lonati D, De Simone U. 3D human stem-cell-derived neuronal spheroids for in vitro neurotoxicity testing of methylglyoxal, highly reactive glycolysis byproduct and potent glycating agent. Curr Res Toxicol 2024; 7:100176. [PMID: 38975063 PMCID: PMC11225170 DOI: 10.1016/j.crtox.2024.100176] [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: 12/13/2023] [Revised: 03/27/2024] [Accepted: 06/05/2024] [Indexed: 07/09/2024] Open
Abstract
Human-derived three-dimensional (3D) in vitro models are advanced human cell-based model for their complexity, relevance and application in toxicity testing. Intracellular accumulation of methylglyoxal (MGO), the most potent glycating agent in humans, mainly generated as a by-product of glycolysis, is associated with age-related diseases including neurodegenerative disorders. In our study, 3D human stem-cell-derived neuronal spheroids were set up and applied to evaluate cytotoxic effects after short-term (5 to 48 h) treatments with different MGO concentrations, including low levels, taking into consideration several biochemical endpoints. In MGO-treated neurospheroids, reduced cell growth proliferation and decreased cell viability occurred early from 5-10 μM, and their compactness diminished starting from 100 μM, apparently without affecting spheroid size. MGO markedly caused loss of the neuronal markers MAP-2 and NSE from 10-50 μM, decreased the detoxifying Glo1 enzyme from 50 μM, and activated NF-kB by nuclear translocation. The cytochemical evaluation of the 3D sections showed the presence of necrotic cells with loss of nuclei. Apoptotic cells were observed from 50 μM MGO after 48 h, and from 100 μM after 24 h. MGO (50-10 µM) also induced modifications of the cell-cell and cell-ECM interactions. These effects worsened at the higher concentrations (300-500 µM). In 3D neuronal spheroids, MGO tested concentrations comparable to human samples levels measured in MGO-associated diseases, altered neuronal key signalling endpoints relevant for the pathogenesis of neurodegenerative diseases and aging. The findings also demonstrated that the use of 3D neuronal spheroids of human origin can be useful in a strategy in vitro for testing MGO and other dicarbonyls evaluation.
Collapse
Affiliation(s)
- Teresa Coccini
- Istituti Clinici Scientifici Maugeri IRCCS, Laboratory of Clinical and Experimental Toxicology, and Pavia Poison Centre-National Toxicology Information Centre, Toxicology Unit, Pavia, Italy
| | - Francesca Caloni
- Dipartimento di Scienze e Politiche Ambientali (ESP), Università degli Studi di Milano, Milan, Italy
| | | | - Laura Villani
- Istituti Clinici Scientifici Maugeri IRCCS, Pathology Unit, Pavia, Italy
| | - Davide Lonati
- Istituti Clinici Scientifici Maugeri IRCCS, Laboratory of Clinical and Experimental Toxicology, and Pavia Poison Centre-National Toxicology Information Centre, Toxicology Unit, Pavia, Italy
| | - Uliana De Simone
- Istituti Clinici Scientifici Maugeri IRCCS, Laboratory of Clinical and Experimental Toxicology, and Pavia Poison Centre-National Toxicology Information Centre, Toxicology Unit, Pavia, Italy
| |
Collapse
|
8
|
Alhujaily M. Glyoxalase System in Breast and Ovarian Cancers: Role of MEK/ERK/SMAD1 Pathway. Biomolecules 2024; 14:584. [PMID: 38785990 PMCID: PMC11117840 DOI: 10.3390/biom14050584] [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: 05/03/2024] [Accepted: 05/05/2024] [Indexed: 05/25/2024] Open
Abstract
The glyoxalase system, comprising GLO1 and GLO2 enzymes, is integral in detoxifying methylglyoxal (MGO) generated during glycolysis, with dysregulation implicated in various cancer types. The MEK/ERK/SMAD1 signaling pathway, crucial in cellular processes, influences tumorigenesis, metastasis, and angiogenesis. Altered GLO1 expression in cancer showcases its complex role in cellular adaptation and cancer aggressiveness. GLO2 exhibits context-dependent functions, contributing to both proapoptotic and antiapoptotic effects in different cancer scenarios. Research highlights the interconnected nature of these systems, particularly in ovarian cancer and breast cancer. The glyoxalase system's involvement in drug resistance and its impact on the MEK/ERK/SMAD1 signaling cascade underscore their clinical significance. Furthermore, this review delves into the urgent need for effective biomarkers, exemplified in ovarian cancer, where the RAGE-ligand pathway emerges as a potential diagnostic tool. While therapeutic strategies targeting these pathways hold promise, this review emphasizes the challenges posed by context-dependent effects and intricate crosstalk within the cellular milieu. Insights into the molecular intricacies of these pathways offer a foundation for developing innovative therapeutic approaches, providing hope for enhanced cancer diagnostics and tailored treatment strategies.
Collapse
Affiliation(s)
- Muhanad Alhujaily
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| |
Collapse
|
9
|
Alhujaily M. Molecular Assessment of Methylglyoxal-Induced Toxicity and Therapeutic Approaches in Various Diseases: Exploring the Interplay with the Glyoxalase System. Life (Basel) 2024; 14:263. [PMID: 38398772 PMCID: PMC10890012 DOI: 10.3390/life14020263] [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: 12/16/2023] [Revised: 01/31/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
This comprehensive exploration delves into the intricate interplay of methylglyoxal (MG) and glyoxalase 1 (GLO I) in various physiological and pathological contexts. The linchpin of the narrative revolves around the role of these small molecules in age-related issues, diabetes, obesity, cardiovascular diseases, and neurodegenerative disorders. Methylglyoxal, a reactive dicarbonyl metabolite, takes center stage, becoming a principal player in the development of AGEs and contributing to cell and tissue dysfunction. The dual facets of GLO I-activation and inhibition-unfold as potential therapeutic avenues. Activators, spanning synthetic drugs like candesartan to natural compounds like polyphenols and isothiocyanates, aim to restore GLO I function. These molecular enhancers showcase promising outcomes in conditions such as diabetic retinopathy, kidney disease, and beyond. On the contrary, GLO I inhibitors emerge as crucial players in cancer treatment, offering new possibilities in diseases associated with inflammation and multidrug resistance. The symphony of small molecules, from GLO I activators to inhibitors, presents a nuanced understanding of MG regulation. From natural compounds to synthetic drugs, each element contributes to a molecular orchestra, promising novel interventions and personalized approaches in the pursuit of health and wellbeing. The abstract concludes with an emphasis on the necessity of rigorous clinical trials to validate these findings and acknowledges the importance of individual variability in the complex landscape of health.
Collapse
Affiliation(s)
- Muhanad Alhujaily
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| |
Collapse
|
10
|
Chi H, Sun Y, Lin P, Zhou J, Zhang J, Yang Y, Qiao Y, Liu D. Glucose Fluctuation Inhibits Nrf2 Signaling Pathway in Hippocampal Tissues and Exacerbates Cognitive Impairment in Streptozotocin-Induced Diabetic Rats. J Diabetes Res 2024; 2024:5584761. [PMID: 38282656 PMCID: PMC10817812 DOI: 10.1155/2024/5584761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 12/25/2023] [Accepted: 01/09/2024] [Indexed: 01/30/2024] Open
Abstract
Background This research investigated whether glucose fluctuation (GF) can exacerbate cognitive impairment in streptozotocin-induced diabetic rats and explored the related mechanism. Methods After 4 weeks of feeding with diets containing high fats plus sugar, the rat model of diabetes mellitus (DM) was established by intraperitoneal injection of streptozotocin (STZ). Then, GF was triggered by means of alternating satiety and starvation for 24 h. The weight, blood glucose level, and water intake of the rats were recorded. The Morris water maze (MWM) test was carried out to appraise the cognitive function at the end of week 12. Moreover, the morphological structure of hippocampal neurons was viewed through HE and Nissl staining, and transmission electron microscopy (TEM) was performed for ultrastructure observation. The protein expression levels of Nrf2, HO-1, NQO-1, Bax, Bcl-2, and Caspase-3 in the hippocampal tissues of rats were measured via Western blotting, and the mRNA expressions of Nrf2, HO-1, and NQO-1 were examined using qRT-PCR. Finally, Western blotting and immunohistochemistry were conducted to detect BDNF levels. Results It was manifested that GF not only aggravated the impairment of spatial memory in rats with STZ-induced type 2 DM but also stimulated the loss, shrinkage, and apoptosis of hippocampal neurons. Regarding the expressions in murine hippocampal tissues, GF depressed Nrf2, HO-1, NQO-1, Bcl-2, and BDNF but boosted Caspase-3 and Bax. Conclusions GF aggravates cognitive impairment by inhibiting the Nrf2 signaling pathway and inducing oxidative stress and apoptosis in the hippocampal tissues.
Collapse
Affiliation(s)
- Haiyan Chi
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- Department of Endocrinology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, China
| | - Yujing Sun
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Peng Lin
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Junyu Zhou
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jinbiao Zhang
- Department of Neurology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, China
| | - Yachao Yang
- Department of Endocrinology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, China
| | - Yun Qiao
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Deshan Liu
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
11
|
Qian X, Klatt S, Bennewitz K, Wohlfart DP, Lou B, Meng Y, Buettner M, Poschet G, Morgenstern J, Fleming T, Sticht C, Hausser I, Fleming I, Szendroedi J, Nawroth PP, Kroll J. Impaired Detoxification of Trans, Trans-2,4-Decadienal, an Oxidation Product from Omega-6 Fatty Acids, Alters Insulin Signaling, Gluconeogenesis and Promotes Microvascular Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2302325. [PMID: 38059818 PMCID: PMC10811472 DOI: 10.1002/advs.202302325] [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: 04/12/2023] [Revised: 11/22/2023] [Indexed: 12/08/2023]
Abstract
Omega-6 fatty acids are the primary polyunsaturated fatty acids in most Western diets, while their role in diabetes remains controversial. Exposure of omega-6 fatty acids to an oxidative environment results in the generation of a highly reactive carbonyl species known as trans, trans-2,4-decadienal (tt-DDE). The timely and efficient detoxification of this metabolite, which has actions comparable to other reactive carbonyl species, such as 4-hydroxynonenal, acrolein, acetaldehyde, and methylglyoxal, is essential for disease prevention. However, the detoxification mechanism for tt-DDE remains elusive. In this study, the enzyme Aldh9a1b is identified as having a key role in the detoxification of tt-DDE. Loss of Aldh9a1b increased tt-DDE levels and resulted in an abnormal retinal vasculature and glucose intolerance in aldh9a1b-/- zebrafish. Transcriptomic and metabolomic analyses revealed that tt-DDE and aldh9a1b deficiency in larval and adult zebrafish induced insulin resistance and impaired glucose homeostasis. Moreover, alterations in hyaloid vasculature is induced by aldh9a1b knockout or by tt-DDE treatment can be rescued by the insulin receptor sensitizers metformin and rosiglitazone. Collectively, these results demonstrated that tt-DDE is the substrate of Aldh9a1b which causes microvascular damage and impaired glucose metabolism through insulin resistance.
Collapse
Affiliation(s)
- Xin Qian
- Department of Vascular BiologyEuropean Center for Angioscience (ECAS)Medical Faculty MannheimHeidelberg University68167MannheimGermany
| | - Stephan Klatt
- Institute for Vascular SignalingCentre for Molecular MedicineGoethe‐Universityam Main60590FrankfurtGermany
- The German Centre for Cardiovascular Research (DZHK)Partner site RheinMain60590FrankfurtGermany
| | - Katrin Bennewitz
- Department of Vascular BiologyEuropean Center for Angioscience (ECAS)Medical Faculty MannheimHeidelberg University68167MannheimGermany
| | - David Philipp Wohlfart
- Department of Vascular BiologyEuropean Center for Angioscience (ECAS)Medical Faculty MannheimHeidelberg University68167MannheimGermany
| | - Bowen Lou
- Department of Vascular BiologyEuropean Center for Angioscience (ECAS)Medical Faculty MannheimHeidelberg University68167MannheimGermany
- Present address:
Cardiovascular Department, the First Affiliated Hospital of Xi'an Jiaotong University277 West Yanta RoadXi'an710061China
| | - Ye Meng
- Bone Marrow Transplantation CenterThe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
| | - Michael Buettner
- Metabolomics Core Technology PlatformCentre for Organismal StudiesHeidelberg University69120HeidelbergGermany
| | - Gernot Poschet
- Metabolomics Core Technology PlatformCentre for Organismal StudiesHeidelberg University69120HeidelbergGermany
| | - Jakob Morgenstern
- Department of Internal Medicine I and Clinical ChemistryHeidelberg University Hospital69120HeidelbergGermany
| | - Thomas Fleming
- Department of Internal Medicine I and Clinical ChemistryHeidelberg University Hospital69120HeidelbergGermany
| | - Carsten Sticht
- NGS Core FacilityMedical Faculty MannheimHeidelberg University68167MannheimGermany
| | - Ingrid Hausser
- Institute of Pathology IPHEM LabHeidelberg University Hospital69120HeidelbergGermany
| | - Ingrid Fleming
- Institute for Vascular SignalingCentre for Molecular MedicineGoethe‐Universityam Main60590FrankfurtGermany
- The German Centre for Cardiovascular Research (DZHK)Partner site RheinMain60590FrankfurtGermany
| | - Julia Szendroedi
- Department of Internal Medicine I and Clinical ChemistryHeidelberg University Hospital69120HeidelbergGermany
| | - Peter Paul Nawroth
- Department of Internal Medicine I and Clinical ChemistryHeidelberg University Hospital69120HeidelbergGermany
| | - Jens Kroll
- Department of Vascular BiologyEuropean Center for Angioscience (ECAS)Medical Faculty MannheimHeidelberg University68167MannheimGermany
| |
Collapse
|
12
|
Coccini T, Schicchi A, Locatelli CA, Caloni F, Negri S, Grignani E, De Simone U. Methylglyoxal-induced neurotoxic effects in primary neuronal-like cells transdifferentiated from human mesenchymal stem cells: Impact of low concentrations. J Appl Toxicol 2023; 43:1819-1839. [PMID: 37431083 DOI: 10.1002/jat.4515] [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: 05/16/2023] [Revised: 06/12/2023] [Accepted: 06/26/2023] [Indexed: 07/12/2023]
Abstract
In the last decades, advanced glycation end-products (AGEs) have aroused the interest of the scientific community due to the increasing evidence of their involvement in many pathophysiological processes including various neurological disorders and cognitive decline age related. Methylglyoxal (MG) is one of the reactive dicarbonyl precursors of AGEs, mainly generated as a by-product of glycolysis, whose accumulation induces neurotoxicity. In our study, MG cytotoxicity was evaluated employing a human stem cell-derived model, namely, neuron-like cells (hNLCs) transdifferentiated from mesenchymal stem/stromal cells, which served as a source of human based species-specific "healthy" cells. MG increased ROS production and induced the first characteristic apoptotic hallmarks already at low concentrations (≥10 μM), decreased the cell growth (≥5-10 μM) and viability (≥25 μM), altered Glo-1 and Glo-2 enzymes (≥25 μM), and markedly affected the neuronal markers MAP-2 and NSE causing their loss at low MG concentrations (≥10 μM). Morphological alterations started at 100 μM, followed by even more marked effects and cell death after few hours (5 h) from 200 μM MG addition. Substantially, most effects occurred as low as 10 μM, concentration much lower than that reported from previous observations using different in vitro cell-based models (e.g., human neuroblastoma cell lines, primary animal cells, and human iPSCs). Remarkably, this low effective concentration approaches the level range measured in biological samples of pathological subjects. The use of a suitable cellular model, that is, human primary neurons, can provide an additional valuable tool, mimicking better the physiological and biochemical properties of brain cells, in order to evaluate the mechanistic basis of molecular and cellular alterations in CNS.
Collapse
Affiliation(s)
- Teresa Coccini
- Laboratory of Clinical and Experimental Toxicology, and Pavia Poison Centre-National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Azzurra Schicchi
- Laboratory of Clinical and Experimental Toxicology, and Pavia Poison Centre-National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Carlo Alessandro Locatelli
- Laboratory of Clinical and Experimental Toxicology, and Pavia Poison Centre-National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Francesca Caloni
- Dipartimento di Scienze e Politiche Ambientali (ESP), Università degli Studi di Milano, Milan, Italy
| | - Sara Negri
- Environmental Research Center, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Elena Grignani
- Environmental Research Center, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Uliana De Simone
- Laboratory of Clinical and Experimental Toxicology, and Pavia Poison Centre-National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| |
Collapse
|
13
|
Kolotyeva NA, Gilmiyarova FN, Averchuk AS, Baranich TI, Rozanova NA, Kukla MV, Tregub PP, Salmina AB. Novel Approaches to the Establishment of Local Microenvironment from Resorbable Biomaterials in the Brain In Vitro Models. Int J Mol Sci 2023; 24:14709. [PMID: 37834155 PMCID: PMC10572431 DOI: 10.3390/ijms241914709] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/19/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
The development of brain in vitro models requires the application of novel biocompatible materials and biopolymers as scaffolds for controllable and effective cell growth and functioning. The "ideal" brain in vitro model should demonstrate the principal features of brain plasticity like synaptic transmission and remodeling, neurogenesis and angiogenesis, and changes in the metabolism associated with the establishment of new intercellular connections. Therefore, the extracellular scaffolds that are helpful in the establishment and maintenance of local microenvironments supporting brain plasticity mechanisms are of critical importance. In this review, we will focus on some carbohydrate metabolites-lactate, pyruvate, oxaloacetate, malate-that greatly contribute to the regulation of cell-to-cell communications and metabolic plasticity of brain cells and on some resorbable biopolymers that may reproduce the local microenvironment enriched in particular cell metabolites.
Collapse
Affiliation(s)
| | - Frida N. Gilmiyarova
- Department of Fundamental and Clinical Biochemistry with Laboratory Diagnostics, Samara State Medical University, 443099 Samara, Russia
| | - Anton S. Averchuk
- Brain Science Institute, Research Center of Neurology, 125367 Moscow, Russia
| | - Tatiana I. Baranich
- Brain Science Institute, Research Center of Neurology, 125367 Moscow, Russia
| | | | - Maria V. Kukla
- Brain Science Institute, Research Center of Neurology, 125367 Moscow, Russia
| | - Pavel P. Tregub
- Brain Science Institute, Research Center of Neurology, 125367 Moscow, Russia
- Department of Pathophysiology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Alla B. Salmina
- Brain Science Institute, Research Center of Neurology, 125367 Moscow, Russia
| |
Collapse
|
14
|
Amaro A, Sousa D, Sá-Rocha M, Ferreira-Júnior MD, Barra C, Monteiro T, Mathias P, Gomes RM, Baptista FI, Matafome P. Sex-specificities in offspring neurodevelopment and behaviour upon maternal glycation: Putative underlying neurometabolic and synaptic changes. Life Sci 2023; 321:121597. [PMID: 36948389 DOI: 10.1016/j.lfs.2023.121597] [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: 01/28/2023] [Revised: 03/07/2023] [Accepted: 03/15/2023] [Indexed: 03/24/2023]
Abstract
AIM Lactation is an important programming window for metabolic disease and neuronal alterations later in life. We aimed to study the effect of maternal glycation during lactation on offspring neurodevelopment and behaviour, assessing possible sex differences and underpinning molecular players. METHODS Female Wistar rats were treated with the Glyoxalase-1 inhibitor S-p-Bromobenzylguthione cyclopentyl diester (BBGC 5 mg/kg). A control and vehicle group treated with dimethyl sulfoxide were considered. Male and female offspring were tested at infancy for neurodevelopment hallmarks. After weaning, triglycerides and total antioxidant capacity were measured in breast milk. At adolescence, offspring were tested for locomotor ability, anxious-like behaviour, and recognition memory. Metabolic parameters were assessed, and the hippocampus and prefrontal cortex were collected for molecular analysis. KEY FINDINGS Maternal glycation reduced triglycerides and total antioxidant capacity levels in breast milk. At infancy, both male and female offspring presented an anticipation on the achievement of neurodevelopmental milestones. At adolescence, male offspring exposed to maternal glycation presented hyperlocomotion, whereas offspring of both sexes presented a risk-taking phenotype, accompanied by GABAA receptor upregulation in the hippocampus. Females also demonstrated GABAA and PSD-95 changes in prefrontal cortex. Furthermore, lower levels of GLO1 and consequently higher accumulation of AGES were also observed in both male and female offspring hippocampus. SIGNIFICANCE Early exposure to maternal glycation induces changes in milk composition leading to neurodevelopment changes at infancy, and sex-specific behavioural and neurometabolic changes at adolescence, further evidencing that lactation period is a critical metabolic programming window and in sculpting behaviour.
Collapse
Affiliation(s)
- Andreia Amaro
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal; Clinical-Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Diana Sousa
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal; Clinical-Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Mariana Sá-Rocha
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal; Clinical-Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Marcos D Ferreira-Júnior
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Portugal
| | - Cátia Barra
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal; Clinical-Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Tamaeh Monteiro
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal; Clinical-Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Paulo Mathias
- Department of Physiological Sciences (DCiF), Institute of Biological Sciences, University Federal of Goiás (UFG), Goiânia, Brazil
| | - Rodrigo Mello Gomes
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cellular Biology, State University of Maringá, Maringá, Brazil
| | - Filipa I Baptista
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal; Clinical-Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Paulo Matafome
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal; Clinical-Academic Center of Coimbra (CACC), Coimbra, Portugal; Polytechnic Institute of Coimbra, Coimbra Health School (ESTeSC), Coimbra, Portugal.
| |
Collapse
|
15
|
Oyagbemi AA, Adebayo AK, Adebiyi OE, Adigun KO, Folarin OR, Esan OO, Ajibade TO, Ogunpolu BS, Falayi OO, Ogunmiluyi IO, Olutayo Omobowale T, Ola-Davies OE, Olopade JO, Saba AB, Adedapo AA, Nkadimeng SM, McGaw LJ, Yakubu MA, Nwulia E, Oguntibeju OO. Leaf extract of Anacardium occidentale ameliorates biomarkers of neuroinflammation, memory loss, and neurobehavioral deficit in N(ω)-nitro-L-arginine methyl ester (L-NAME) treated rats. Biomarkers 2023; 28:263-272. [PMID: 36632742 DOI: 10.1080/1354750x.2022.2164354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
PURPOSE Anacardium occidentale commonly known as Cashew is a plant that is widely used in African traditional medicine. It is endowed with phytochemical constituents that are responsible for its medicinal properties. METHODS Twenty-five male Wistar rats were grouped as follows: Control (Group A), Group B (L-NAME 40 mg/kg), Group C (100 mg/kg Anacardium occidentale extract plus 40 mg/kg L-NAME), Group D (200 mg/kg extract plus 40 mg/kg L-NAME) and Group E (10 mg/kg of Lisinopril plus 40 mg/kg L-NAME). The animals were treated with oral administration of either the extracts or Lisnopril daily for 4 weeks. Neuro-behavioural tests such as the Morris Water Maze and Hanging Wire Grip tests were carried out to evaluate memory/spatial learning and muscular strength, respectively. Makers of oxidative stress, antioxidant enzymes and immunohistochemical staining of Glial Fibrillary Acidic Protein and Ionised Calcium Binding Adaptor molecule 1 were assessed. RESULTS L-NAME administration caused significant increases in biomarkers of oxidative stress, decreased antioxidant status, acetylcholinesterase activity, altered neuro-behavioural changes, astrocytosis, and microgliosis. However, Anacardium occidentale reversed exaggerated oxidative stress biomarkers and improved neuro-behavioural changes. CONCLUSIONS Combining all, Anacardium occidentale enhanced brain antioxidant defence status, improved memory and muscular strength, thus, suggesting the neuroprotective properties of Anacardium occidentale.
Collapse
Affiliation(s)
- Ademola Adetokunbo Oyagbemi
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adedeji Kolawole Adebayo
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olamide Elizabeth Adebiyi
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Kabirat Oluwaseun Adigun
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Oluwabusayo Racheal Folarin
- Department of Biomedical Laboratory Sciences, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Oluwaseun Olanrewaju Esan
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Temitayo Olabisi Ajibade
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Blessing Seun Ogunpolu
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olufunke Olubunmi Falayi
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Iyanuoluwa Omolola Ogunmiluyi
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Temidayo Olutayo Omobowale
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olufunke Eunice Ola-Davies
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - James Olukayode Olopade
- Department of Veterinary Anatomy, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adebowale Benard Saba
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adeolu Alex Adedapo
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Sanah Malomile Nkadimeng
- Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa Florida Campus, University of South Africa, Roodepoort, South Africa
| | - Lyndy Joy McGaw
- Phytomedicine Programme, Department of Paraclinical Science, University of Pretoria, Faculty of Veterinary Science, Pretoria, South Africa
| | - Momoh Audu Yakubu
- Department of Environmental & Interdisciplinary Sciences, College of Science, Engineering & Technology, Vascular Biology Unit, Center for Cardiovascular Diseases, COPHS, Texas Southern University, Houston, Texas, USA
| | - Evaristus Nwulia
- Department of Psychiatry and Behavioral Sciences, College of Medicine, Howard University Hospital, Howard University, Washington, District of Columbia, USA
| | - Oluwafemi Omoniyi Oguntibeju
- Phytomedicine and Phytochemistry Group, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Bellville, South Africa
| |
Collapse
|