1
|
Bano N, Khan S, Ahamad S, Dar NJ, Alanazi HH, Nazir A, Bhat SA. Microglial NOX2 as a therapeutic target in traumatic brain injury: Mechanisms, consequences, and potential for neuroprotection. Ageing Res Rev 2025; 108:102735. [PMID: 40122395 DOI: 10.1016/j.arr.2025.102735] [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: 01/24/2025] [Revised: 03/08/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
Traumatic brain injury (TBI) is a leading cause of long-term disability worldwide, with secondary injury mechanisms, including neuroinflammation and oxidative stress, driving much of its chronic pathology. While NADPH oxidase 2 (NOX2)-mediated reactive oxygen species (ROS) production is a recognized factor in TBI, the specific role of microglial NOX2 in perpetuating oxidative and inflammatory damage remains underexplored. Addressing this gap is critical, as current therapeutic approaches primarily target acute symptoms and fail to interrupt the persistent neuroinflammation that contributes to progressive neurodegeneration. Besides NOX, other ROS-generating enzymes, such as CYP1B1, COX2, and XO, also play crucial roles in triggering oxidative stress and neuroinflammatory conditions in TBI. However, this review highlights the pathophysiological role of microglial NOX2 in TBI, focusing on its activation following injury and its impact on ROS generation, neuroinflammatory signaling, and neuronal loss. These insights reveal NOX2 as a critical driver of secondary injury, linked to worsened outcomes, particularly in aged individuals where NOX2 activation is more pronounced. In addition, this review evaluates emerging therapeutic approaches targeting NOX2, such as GSK2795039 and other selective NOX2 inhibitors, which show potential in reducing ROS levels, limiting neuroinflammation, and preserving neurological functions. By highlighting the specific role of NOX2 in microglial ROS production and secondary neurodegeneration, this study advocates for NOX2 inhibition as a promising strategy to improve TBI outcomes by addressing the unmet need for therapies targeting long-term inflammation and neuroprotection. Our review highlights the potential of NOX2-targeted interventions to disrupt the cycle of oxidative stress and inflammation, ultimately offering a pathway to mitigate the chronic impact of TBI.
Collapse
Affiliation(s)
- Nargis Bano
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Sameera Khan
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India
| | - Nawab John Dar
- CNB, SALK Institute of Biological Sciences, La Jolla, CA 92037, USA
| | - Hamad H Alanazi
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, Al Jouf University 77455, Saudi Arabia
| | - Aamir Nazir
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India; Academy of Scientific and Innovative Research, New Delhi, India.
| | - Shahnawaz Ali Bhat
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India.
| |
Collapse
|
2
|
Bortolotti M, Polito L, Battelli MG, Bolognesi A. Xanthine Oxidoreductase: A Double-Edged Sword in Neurological Diseases. Antioxidants (Basel) 2025; 14:483. [PMID: 40298821 PMCID: PMC12024114 DOI: 10.3390/antiox14040483] [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: 03/18/2025] [Revised: 04/08/2025] [Accepted: 04/15/2025] [Indexed: 04/30/2025] Open
Abstract
Non-communicable neurological disorders are the second leading cause of death, and their burden continues to increase as the world population grows and ages. Oxidative stress and inflammation are crucially implicated in the triggering and progression of multiple sclerosis, Alzheimer's disease, amyotrophic lateral sclerosis, Huntington's disease, Parkinson's disease, and even stroke. In this narrative review, we examine the role of xanthine oxidoreductase (XOR) activities and products in all the above-cited neurological diseases. The redox imbalance responsible for oxidative stress could arise from excess reactive oxygen and nitrogen species resulting from the activities of XOR, as well as from the deficiency of its main product, uric acid (UA), which is the pivotal antioxidant system in the blood. In fact, with the exception of stroke, serum UA levels are inversely related to the onset and progression of these neurological disorders. The inverse correlation observed between the level of uricemia and the presence of neurological diseases suggests a neuroprotective role for UA. Oxidative stress and inflammation are also caused by ischemia and reperfusion, a condition in which XOR action has been recognized as a contributing factor to tissue damage. The findings reported in this review could be useful for addressing clinical decision-making and treatment optimization.
Collapse
|
3
|
Aryal B, Kwakye J, Ariyo OW, Ghareeb AFA, Milfort MC, Fuller AL, Khatiwada S, Rekaya R, Aggrey SE. Major Oxidative and Antioxidant Mechanisms During Heat Stress-Induced Oxidative Stress in Chickens. Antioxidants (Basel) 2025; 14:471. [PMID: 40298812 PMCID: PMC12023971 DOI: 10.3390/antiox14040471] [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: 02/18/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
Heat stress (HS) is one of the most important stressors in chickens, and its adverse effects are primarily caused by disturbing the redox homeostasis. An increase in electron leakage from the mitochondrial electron transport chain is the major source of free radical production under HS, which triggers other enzymatic systems to generate more radicals. As a defense mechanism, cells have enzymatic and non-enzymatic antioxidant systems that work cooperatively against free radicals. The generation of free radicals, particularly the reactive oxygen species (ROS) and reactive nitrogen species (RNS), under HS condition outweighs the cellular antioxidant capacity, resulting in oxidative damage to macromolecules, including lipids, carbohydrates, proteins, and DNA. Understanding these detrimental oxidative processes and protective defense mechanisms is important in developing mitigation strategies against HS. This review summarizes the current understanding of major oxidative and antioxidant systems and their molecular mechanisms in generating or neutralizing the ROS/RNS. Importantly, this review explores the potential mechanisms that lead to the development of oxidative stress in heat-stressed chickens, highlighting their unique behavioral and physiological responses against thermal stress. Further, we summarize the major findings associated with these oxidative and antioxidant mechanisms in chickens.
Collapse
Affiliation(s)
- Bikash Aryal
- NutriGenomics Laboratory, Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA or (B.A.); (J.K.); (O.W.A.); (A.F.A.G.); (M.C.M.); (A.L.F.)
- Department of Animal Sciences, The Ohio State University, Wooster, OH 44691, USA;
| | - Josephine Kwakye
- NutriGenomics Laboratory, Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA or (B.A.); (J.K.); (O.W.A.); (A.F.A.G.); (M.C.M.); (A.L.F.)
| | - Oluwatomide W. Ariyo
- NutriGenomics Laboratory, Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA or (B.A.); (J.K.); (O.W.A.); (A.F.A.G.); (M.C.M.); (A.L.F.)
| | - Ahmed F. A. Ghareeb
- NutriGenomics Laboratory, Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA or (B.A.); (J.K.); (O.W.A.); (A.F.A.G.); (M.C.M.); (A.L.F.)
- Boehringer Ingelheim Animal Health (BIAH), Gainesville, GA 30501, USA
| | - Marie C. Milfort
- NutriGenomics Laboratory, Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA or (B.A.); (J.K.); (O.W.A.); (A.F.A.G.); (M.C.M.); (A.L.F.)
| | - Alberta L. Fuller
- NutriGenomics Laboratory, Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA or (B.A.); (J.K.); (O.W.A.); (A.F.A.G.); (M.C.M.); (A.L.F.)
| | - Saroj Khatiwada
- Department of Animal Sciences, The Ohio State University, Wooster, OH 44691, USA;
| | - Romdhane Rekaya
- Department of Animal and Dairy Science, The University of Georgia, Athens, GA 30602, USA;
| | - Samuel E. Aggrey
- NutriGenomics Laboratory, Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA or (B.A.); (J.K.); (O.W.A.); (A.F.A.G.); (M.C.M.); (A.L.F.)
| |
Collapse
|
4
|
Jayasinghe T, Jenkins J, Medara N, Choowong P, Dharmarathne G, Kong F, Cho H, Kim SH, Zhang Y, Franco-Duarte R, Eberhard J, Spahr A. Dietary Fibre Modulates Body Composition, Blood Glucose, Inflammation, Microbiome, and Metabolome in a Murine Model of Periodontitis. Nutrients 2025; 17:1146. [PMID: 40218904 PMCID: PMC11990244 DOI: 10.3390/nu17071146] [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: 02/24/2025] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 04/14/2025] Open
Abstract
Background: Dietary fibre plays a crucial role in metabolic regulation, inflammation, and microbiome composition. However, its impact on systemic and oral health, particularly in periodontitis, remains unclear. This study investigated the effects of high- and low-fibre diets on body composition, glycaemic control, inflammation, microbiome, and metabolome in a murine model of experimental periodontitis. Methods: Thirty-six male C57BL/6 mice were randomised to a high-fibre (40% fibre) or low-fibre (5% fibre) diet for eight weeks. Body weight, fat mass, lean mass, fasting blood glucose, serum inflammatory markers, alveolar bone loss, and root length were assessed. Oral and faecal microbiome composition was analysed using 16S rRNA sequencing. Metabolomic and short-chain fatty acid (SCFA) profiling was conducted using liquid chromatography-mass spectrometry (LC-MS). Results: Mice on the high-fibre diet exhibited significantly lower body weight (p < 0.0001), fat mass (p = 0.0007), and lean mass (p < 0.0001) compared to the low-fibre group. Fasting blood glucose levels were significantly lower in the high-fibre group (p = 0.0013). TNF-α and IFN-γ levels were significantly elevated in the low-fibre group (p < 0.0001), suggesting a heightened pro-inflammatory state. While alveolar bone loss and root length did not differ significantly, microbiome analysis revealed distinct bacterial compositions (PERMANOVA, p < 0.05), with fibre-fermenting taxa enriched in high-fibre-fed mice. Metabolomic analysis identified 19 significantly altered metabolites, indicating dietary adaptations. Conclusions: A high-fibre diet improves glycaemic control, reduces systemic inflammation, and alters microbial and metabolic profiles in experimental periodontitis. These findings highlight dietary fibre's role in modulating metabolic and inflammatory pathways relevant to periodontal and systemic diseases.
Collapse
Affiliation(s)
- Thilini Jayasinghe
- The Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050, Australia; (J.J.); (P.C.); (J.E.)
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| | - Josie Jenkins
- The Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050, Australia; (J.J.); (P.C.); (J.E.)
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| | - Nidhi Medara
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| | - Phannaphat Choowong
- The Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050, Australia; (J.J.); (P.C.); (J.E.)
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| | - Gangani Dharmarathne
- Australian Laboratory Services Global, Water and Hydrographic, Hume, ACT 2620, Australia;
| | - Fay Kong
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| | - Hanna Cho
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| | - Se Hun Kim
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| | - Yuchen Zhang
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| | - Ricardo Franco-Duarte
- Centre of Molecular and Environmental Biology, Department of Biology, University of Minho, 4710-057 Braga, Portugal;
| | - Joerg Eberhard
- The Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050, Australia; (J.J.); (P.C.); (J.E.)
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| | - Axel Spahr
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| |
Collapse
|
5
|
Ujie Y, Saito S, Banno T, Yaguchi T, Arai MA. Co-culture of Aspergillus niger IFM 59706 and RAW264 cells enhances the production of aurasperone A with nitric oxide inhibitory activity. Biosci Biotechnol Biochem 2025; 89:541-547. [PMID: 39737716 DOI: 10.1093/bbb/zbae211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 12/23/2024] [Indexed: 01/01/2025]
Abstract
Most actinomycetes and fungi have a multitude of silent biosynthetic genes whose activation could lead to the production of new natural products. Our group recently designed and used a co-culture method to isolate new natural products, based on the idea that pathogens might produce immune suppressors to avoid attack by immune cells. Here, we searched for compounds produced by the co-culture of immune cells with pathogenic fungi isolated from clinical specimens. The production of dimeric naphtho-γ-pyrone aurasperone A (1) was enhanced by the co-culture of pathogenic fungus Aspergillus niger IFM 59706 and RAW264 mouse macrophage-like cells. The absolute configuration of 1 was confirmed by comparison with the reported electronic circular dichroism spectrum. This is the first report of the inhibitory activity of 1 on nitric oxide production, an inflammatory mediator.
Collapse
Affiliation(s)
- Yukiko Ujie
- Department of Biosciences & Informatics, Faculty of Science and Technology, Keio University, Yokohama, Japan
| | - Shun Saito
- Department of Biosciences & Informatics, Faculty of Science and Technology, Keio University, Yokohama, Japan
| | - Tomoya Banno
- Department of Biosciences & Informatics, Faculty of Science and Technology, Keio University, Yokohama, Japan
| | - Takashi Yaguchi
- Medical Mycology Research Center, Chiba University, Chiba, Japan
| | - Midori A Arai
- Department of Biosciences & Informatics, Faculty of Science and Technology, Keio University, Yokohama, Japan
| |
Collapse
|
6
|
Lu X, Liu F, Chen H, Cai H, Zhang L, Li J. Effects of WN1703 on Cardiovascular Function in Chronic Hyperuricemia Rats and Myocardial Injury Mechanism Exploration in H9C2 Cells. J Appl Toxicol 2025; 45:418-431. [PMID: 39435646 DOI: 10.1002/jat.4710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/09/2024] [Accepted: 09/25/2024] [Indexed: 10/23/2024]
Abstract
Hyperuricemia, a prevalent condition, is typically preceded by disturbances in purine metabolism and is frequently associated with hyperlipidemia and other dysfunctions of metabolism. WN1703 demonstrated an inhibitory activity against xanthine oxidoreductase (XOR) that was comparable to febuxostat in our prior investigation. In this study, we assessed the cardiovascular safety of WN1703 in a chronic hyperuricemia rat model induced by potassium oxonate in combination with hypoxanthine. We investigated the changes in cardiovascular biomarkers in chronic hyperuricemia rats treated with febuxostat and WN1703, including creatine kinase (CK), CK-MB, B type natriuretic peptide (BNP), Corin protein (CRN), Neprilysin (NEP), myeloperoxidase (MPO), 8-hydroxy-2-deoxyguanosine (8-OHdG), tumor necrosis factor (TNF-α), interleukin-1β (IL-1β), and interleukin-8 (IL-8). Additionally, we validated the potential mechanism of cardiac injury induced by WN1703 in H9C2 cells, guided by cardiotoxicity predictions from the cardioToxCSM database and network pharmacology. We observed that excessively rapid urate-lowering, oxidative stress, and inflammation could disrupt myocardial functional homeostasis and increase the risk of cardiovascular injury in hyperuricemia rats, and WN1703 treatment effectively reduced the levels oxidative stress marker 8-OHdG and inflammatory factor TNF-α. Despite the absence of organic damage to the heart with prolonged treatment of febuxostat and WN1703, potential hazard of cardiovascular injury could be associated with the modulation of the TGFβ and RHO/ROCK signaling pathways by febuxostat and WN1703. This could offer new insights into the mechanisms underlying the adverse effects caused by XOR inhibitors.
Collapse
Affiliation(s)
- Xiaodan Lu
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Fuyao Liu
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Hongming Chen
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Haojie Cai
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Lei Zhang
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Jing Li
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| |
Collapse
|
7
|
Yagi H, Akazawa H, Liu Q, Yamamoto K, Nawata K, Saga-Kamo A, Umei M, Kadowaki H, Matsuoka R, Shindo A, Okamura S, Toko H, Takeda N, Ando M, Yamauchi H, Takeda N, Fini MA, Ono M, Komuro I. XOR-Derived ROS in Tie2-Lineage Cells Including Endothelial Cells Promotes Aortic Aneurysm Progression in Marfan Syndrome. Arterioscler Thromb Vasc Biol 2025; 45:e63-e77. [PMID: 39882602 DOI: 10.1161/atvbaha.124.321527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 12/24/2024] [Accepted: 01/09/2025] [Indexed: 01/31/2025]
Abstract
BACKGROUND Marfan syndrome (MFS) is an inherited disorder caused by mutations in the FBN1 gene encoding fibrillin-1, a matrix component of extracellular microfibrils. The main cause of morbidity and mortality in MFS is thoracic aortic aneurysm and dissection, but the underlying mechanisms remain undetermined. METHODS To elucidate the role of endothelial XOR (xanthine oxidoreductase)-derived reactive oxygen species in aortic aneurysm progression, we inhibited in vivo function of XOR either by endothelial cell (EC)-specific disruption of the Xdh gene or by systemic administration of an XOR inhibitor febuxostat in MFS mice harboring the Fbn1 missense mutation p.(Cys1041Gly). We assessed the aberrant activation of mechanosensitive signaling in the ascending aorta of Fbn1C1041G/+ mice. Further analysis of human aortic ECs investigated the mechanisms by which mechanical stress upregulates XOR expression. RESULTS We found a significant increase in reactive oxygen species generation in the ascending aorta of patients with MFS and Fbn1C1041G/+ mice, which was associated with a significant increase in protein expression and enzymatic activity of XOR protein in aortic ECs. Genetic disruption of Xdh in ECs or treatment with febuxostat significantly suppressed aortic aneurysm progression and improved perivascular infiltration of macrophages. Mechanistically, mechanosensitive signaling involving FAK (focal adhesion kinase)-p38 MAPK (p38 mitogen-activated protein kinase) and Egr-1 (early growth response-1) was aberrantly activated in the ascending aorta of Fbn1C1041G/+ mice, and mechanical stress on human aortic ECs upregulated XOR expression through Egr-1 upregulation. Consistently, EC-specific knockout of XOR or systemic administration of febuxostat in Fbn1C1041G/+ mice suppressed reactive oxygen species generation, FAK-p38 MAPK activation, and Egr-1 upregulation. CONCLUSIONS Aberrant activation of mechanosensitive signaling in vascular ECs triggered endothelial XOR activation and reactive oxygen species generation, which contributes to the progression of aortic aneurysms in MFS. These findings highlight a drug repositioning approach using a uric acid-lowering drug febuxostat as a potential therapy for MFS.
Collapse
MESH Headings
- Animals
- Marfan Syndrome/genetics
- Marfan Syndrome/complications
- Marfan Syndrome/enzymology
- Marfan Syndrome/pathology
- Reactive Oxygen Species/metabolism
- Humans
- Fibrillin-1/genetics
- Fibrillin-1/metabolism
- Disease Progression
- Endothelial Cells/enzymology
- Endothelial Cells/pathology
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Disease Models, Animal
- Mechanotransduction, Cellular
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/enzymology
- Aortic Aneurysm, Thoracic/etiology
- Cells, Cultured
- Mice
- Male
- Early Growth Response Protein 1/metabolism
- Mice, Inbred C57BL
- Aortic Aneurysm/pathology
- Aortic Aneurysm/genetics
- Aortic Aneurysm/enzymology
- Aortic Aneurysm/etiology
- Focal Adhesion Kinase 1/metabolism
- Mice, Knockout
- Female
- Aorta/pathology
- Aorta/enzymology
- p38 Mitogen-Activated Protein Kinases/metabolism
- Stress, Mechanical
- Adipokines
Collapse
Affiliation(s)
- Hiroki Yagi
- Department of Cardiovascular Medicine (H. Yagi, H.A., Q.L., A.S.-K., M.U., H.K., R.M., A.S., S.O., H.T., Norifumi Takeda, I.K.), The University of Tokyo, Bunkyo-ku, Japan
- Marfan Syndrome Center, The University of Tokyo Hospital, Bunkyo-ku, Japan (H. Yagi, Norifumi Takeda, M.A., H. Yamauchi)
| | - Hiroshi Akazawa
- Department of Cardiovascular Medicine (H. Yagi, H.A., Q.L., A.S.-K., M.U., H.K., R.M., A.S., S.O., H.T., Norifumi Takeda, I.K.), The University of Tokyo, Bunkyo-ku, Japan
| | - Qing Liu
- Department of Cardiovascular Medicine (H. Yagi, H.A., Q.L., A.S.-K., M.U., H.K., R.M., A.S., S.O., H.T., Norifumi Takeda, I.K.), The University of Tokyo, Bunkyo-ku, Japan
| | - Kimiko Yamamoto
- Laboratory of System Physiology, Department of Biomedical Engineering, Graduate School of Medicine (K.Y.)
| | - Kan Nawata
- Department of Cardiovascular Surgery, St. Marianna University School of Medicine, Kawasaki, Japan (K.N.)
| | - Akiko Saga-Kamo
- Department of Cardiovascular Medicine (H. Yagi, H.A., Q.L., A.S.-K., M.U., H.K., R.M., A.S., S.O., H.T., Norifumi Takeda, I.K.), The University of Tokyo, Bunkyo-ku, Japan
| | - Masahiko Umei
- Department of Cardiovascular Medicine (H. Yagi, H.A., Q.L., A.S.-K., M.U., H.K., R.M., A.S., S.O., H.T., Norifumi Takeda, I.K.), The University of Tokyo, Bunkyo-ku, Japan
| | - Hiroshi Kadowaki
- Department of Cardiovascular Medicine (H. Yagi, H.A., Q.L., A.S.-K., M.U., H.K., R.M., A.S., S.O., H.T., Norifumi Takeda, I.K.), The University of Tokyo, Bunkyo-ku, Japan
| | - Ryo Matsuoka
- Department of Cardiovascular Medicine (H. Yagi, H.A., Q.L., A.S.-K., M.U., H.K., R.M., A.S., S.O., H.T., Norifumi Takeda, I.K.), The University of Tokyo, Bunkyo-ku, Japan
| | - Akito Shindo
- Department of Cardiovascular Medicine (H. Yagi, H.A., Q.L., A.S.-K., M.U., H.K., R.M., A.S., S.O., H.T., Norifumi Takeda, I.K.), The University of Tokyo, Bunkyo-ku, Japan
| | - Shun Okamura
- Department of Cardiovascular Medicine (H. Yagi, H.A., Q.L., A.S.-K., M.U., H.K., R.M., A.S., S.O., H.T., Norifumi Takeda, I.K.), The University of Tokyo, Bunkyo-ku, Japan
| | - Haruhiro Toko
- Department of Cardiovascular Medicine (H. Yagi, H.A., Q.L., A.S.-K., M.U., H.K., R.M., A.S., S.O., H.T., Norifumi Takeda, I.K.), The University of Tokyo, Bunkyo-ku, Japan
| | - Norifumi Takeda
- Department of Cardiovascular Medicine (H. Yagi, H.A., Q.L., A.S.-K., M.U., H.K., R.M., A.S., S.O., H.T., Norifumi Takeda, I.K.), The University of Tokyo, Bunkyo-ku, Japan
- Marfan Syndrome Center, The University of Tokyo Hospital, Bunkyo-ku, Japan (H. Yagi, Norifumi Takeda, M.A., H. Yamauchi)
| | - Masahiko Ando
- Department of Cardiac Surgery, Graduate School of Medicine (M.A., H. Yamauchi, M.O.), The University of Tokyo, Bunkyo-ku, Japan
- Marfan Syndrome Center, The University of Tokyo Hospital, Bunkyo-ku, Japan (H. Yagi, Norifumi Takeda, M.A., H. Yamauchi)
| | - Haruo Yamauchi
- Department of Cardiac Surgery, Graduate School of Medicine (M.A., H. Yamauchi, M.O.), The University of Tokyo, Bunkyo-ku, Japan
- Marfan Syndrome Center, The University of Tokyo Hospital, Bunkyo-ku, Japan (H. Yagi, Norifumi Takeda, M.A., H. Yamauchi)
| | - Norihiko Takeda
- Division of Cardiology and Metabolism, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan (Norihiko Takeda)
| | - Mehdi A Fini
- Division of Pulmonary and Critical Care, Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora (M.A.F.)
| | - Minoru Ono
- Department of Cardiac Surgery, Graduate School of Medicine (M.A., H. Yamauchi, M.O.), The University of Tokyo, Bunkyo-ku, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine (H. Yagi, H.A., Q.L., A.S.-K., M.U., H.K., R.M., A.S., S.O., H.T., Norifumi Takeda, I.K.), The University of Tokyo, Bunkyo-ku, Japan
| |
Collapse
|
8
|
Yang CC, Yue Y, Chen YL, Chiang JY, Wang YT, Huang CR, Cheng BC, Hsu TW, Yip HK. Febuxostat therapy improved the outcomes of cardiorenal syndrome rodent through alleviating xanthine oxidase-induced oxidative stress and mitochondrial dysfunction. Int J Biol Sci 2025; 21:1749-1766. [PMID: 39990671 PMCID: PMC11844282 DOI: 10.7150/ijbs.99194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 11/20/2024] [Indexed: 02/25/2025] Open
Abstract
Background: We tested the hypothesis that febuxostat (Feb) therapy effectively protected cardiorenal syndrome (CRS) rats via repressing the xanthine-oxidase (XO)-caused oxidative stress. Methods and Results: Cellular levels of apoptosis/oxidative stress/mitochondrial-membrane potential were higher in p-Cresol treated-NRK-52E cells than in control group that were reversed by Feb treatment or silencing XO gene (all P<0.001). Pilot study demonstrated that: XO activity was significantly increased in CRS than in SC group; a significant negative correlation between XO activity and left ventricular ejection fraction (LVEF) (%); a significant positive correlation between XO activity and BNP/BUN/creatinine/proteinuria levels (all P<0.01). Male-adult SD-rats were classified into groups 1(sham-control)/2 (CRS)/3 [CRS+Feb (10mg/kg/day)]/4 [CRS+Feb (30mg/kg/day)]. By day-63, the survival rate was significantly lower in group 2 than in other groups (P=0.029), and circulatory levels of FGF23/BNP/XO-activity BUN/creatinine/proteinuria and renal-artery resistance were highest in group 2/lowest in group 1/significantly lower in group 4 than in group 3, whereas the LVEF exhibited an opposite pattern of XO among the groups (all P<0.0001). Cellular levels of fibrosis/XO/H2DCFDA/CD68/CHAC1, and protein expressions of oxidative-stress (NOX-2/NOX-4/XO)/inflammatory (NF-κB/IL-1β)/fibrotic (Smad3/TFG-β)/apoptotic (CHAC1/2)/mitochondrial-damaged (p-DRP1) biomarkers in kidney/heart tissues displayed a similar pattern of XO (all P<0.0001). Conclusion: Feb therapy improved cardiorenal function and prognostic outcome in CRS rats.
Collapse
Affiliation(s)
- Chih-Chao Yang
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan, R.O.C
| | - Ya Yue
- The First Hospital of Guangzhou Medical University, Guangzhou, 510120, China, P. R. C
| | - Yi-Ling Chen
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital Kaohsiung 833401, Taiwan, R. O. C
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan, R. O. C
| | - John Y. Chiang
- Department of Computer Science and Engineering, National Sun Yat-Sen University, Kaohsiung 804201, Taiwan, R. O. C
- Department of Healthcare Administration and Medical Informatics, Kaohsiung Medical University, Kaohsiung 807378, Taiwan, R. O. C
| | - Yi-Ting Wang
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital Kaohsiung 833401, Taiwan, R. O. C
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan, R. O. C
| | - Chi-Ruei Huang
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan, R. O. C
- Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital Kaohsiung 833401, Taiwan, R. O. C
| | - Ben-Chung Cheng
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan, R.O.C
| | - Tsuen-Wei Hsu
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan, R.O.C
| | - Hon-Kan Yip
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital Kaohsiung 833401, Taiwan, R. O. C
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan, R. O. C
- Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital Kaohsiung 833401, Taiwan, R. O. C
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404333, Taiwan, R. O. C
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan, R. O. C
| |
Collapse
|
9
|
Zhang F, Zhang T, Dong H, Jiang J, Yang G, Seim I, Tian R. Comparative Genomics Uncovers Molecular Adaptations for Cetacean Deep-Sea Diving. Mol Ecol 2025:e17678. [PMID: 39898416 DOI: 10.1111/mec.17678] [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: 05/26/2024] [Revised: 12/20/2024] [Accepted: 01/23/2025] [Indexed: 02/04/2025]
Abstract
Cetaceans show remarkable diversity in diving capability, implying a range of adaptive strategies to hazards such as hydrostatic pressure and oxidative stress, but few studies have considered the evolution of extreme diving. Here, we first examined the relationship between morphological and physiological factors and diving capability and then considered the molecular evolution of candidate deep-sea diving traits in a genomic dataset of cetaceans. Our dataset included six super-divers, sperm whales (families Physeteridae and Kogiidae) and beaked whales (Ziphiidae), species that can dive deeper than 1000 m for about an hour or longer. We found a positive association between diving capability and oxygen-linked globins, and super-diver myoglobin (MB) is under positive selection and harbours a reported functional amino acid change. Blubber thickness was positively associated, likely to provide thermal insulation and hydrostatic pressure resistance. Super-divers have gene changes that may contribute to differences in the composition of outer blubber neutral lipids (triacylglycerols and wax esters), fatty acids and cholesterol. Total lung capacity relative to body mass showed a negative association, ostensibly to limit gas bubbles that can cause decompression sickness. A functional assay suggests that an ATP8B1 amino acid substitution may reduce lung injury in super-divers. Super-diver XDH has two unique amino acids and a decreased ability to produce uric acid under hypoxia when its ROS-generating XO isoform is prevalent, suggesting that it reduces cell damage from oxidative stress and uric acid accumulation in species with prolonged dives. Our study deepens the understanding of how deep-sea diving emerged in the cetacean lineage.
Collapse
Affiliation(s)
- Fan Zhang
- Jiangsu Key Laboratory for the Biodiversity Conservation and Sustainable Utilization in the Middle and Lower Reaches of the Yangtze River Basin, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Tong Zhang
- Jiangsu Key Laboratory for the Biodiversity Conservation and Sustainable Utilization in the Middle and Lower Reaches of the Yangtze River Basin, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Hao Dong
- Jiangsu Key Laboratory for the Biodiversity Conservation and Sustainable Utilization in the Middle and Lower Reaches of the Yangtze River Basin, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Jie Jiang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Guang Yang
- Jiangsu Key Laboratory for the Biodiversity Conservation and Sustainable Utilization in the Middle and Lower Reaches of the Yangtze River Basin, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Inge Seim
- Marine Mammal and Marine Bioacoustics Laboratory, Institute of Deep-Sea Science and Engineering, Chinese Academy of Sciences, Sanya, China
| | - Ran Tian
- Jiangsu Key Laboratory for the Biodiversity Conservation and Sustainable Utilization in the Middle and Lower Reaches of the Yangtze River Basin, College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|
10
|
Castillo-Lopez E, Biber P, Sener-Aydemir A, Hummel K, Razzazi-Fazeli E, Reisinger N, Zebeli Q, Kreuzer-Redmer S, Hartinger T. Characterization of the colostrum proteome of primiparous Holstein cows and its association with colostrum immunoglobulin G concentrations. J Anim Sci Biotechnol 2025; 16:10. [PMID: 39833978 PMCID: PMC11748342 DOI: 10.1186/s40104-024-01144-y] [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: 08/09/2024] [Accepted: 12/17/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND The objective was to characterize the colostrum proteome of primiparous Holstein cows in association with immunoglobulin G (IgG) content. Immediately after calving, colostrum samples were collected from 18 cows to measure IgG concentration. Based on colostrum IgG content, samples were classified through cluster analysis and were identified as poor, average, and excellent quality. The proteome was assessed with quantitative shotgun proteomics; abundance data were compared among the colostrum types; enrichment analysis of metabolic processes and proteins classes was performed as well. We also tested correlations between this proteome and blood globulin level of cows and passive immunity level of calves. RESULTS On average, 428 proteins were identified per sample, which belonged mainly to cellular process, biological regulation, response to stimulus, metabolic process, and immune system process. Most abundant proteins were complement C3 (Q2UVX4), alpha-S1-casein (P02662), Ig-like domain-containing protein (A0A3Q1M032), albumin (A0A140T897), polymeric immunoglobulin receptor (P81265), lactotransferrrin (P24627), and IGHG1*01 (X16701_4). Colostrum of excellent quality had greater (P < 0.05) abundance of serpin A3-7 (A2I7N3), complement factor I (A0A3Q1MIF4), lipocalin/cytosolic fatty-acid binding domain-containing protein (A0A3Q1MRQ2), complement C3 (E1B805), complement component 4 binding protein alpha (A0AAF6ZHP5), and complement component C6 (F1MM86). However, colostrum of excellent quality had lower (P < 0.05) abundance of HGF activator (E1BCW0), alpha-S1-casein (P02662), and xanthine dehydrogenase/oxidase (P80457). This resulted in enrichment of the biological processes predominantly for complement activation alternative pathway, complement activation, complement activation classical pathway, humoral immune response, leukocyte mediated immunity, and negative regulation of endopeptidase activity in excellent-quality colostrum. Additionally, some colostrum proteins were found to be correlated with the blood globulin level of cows and with the passive immunity level of calves (P < 0.05; r ≥ 0.57). CONCLUSIONS This study provides new insights into the bovine colostrum proteome, demonstrating associations between IgG levels and the abundance of other proteins, as well as the enrichment of metabolic processes related to innate immune response. Thus, results suggest that the colostrum proteomic profile is associated with the content of IgG. Future research should deeply explore the association of these findings with pre-calving nutrition status and blood composition of the cow, and with passive immunity transfer to the calf.
Collapse
Affiliation(s)
- Ezequias Castillo-Lopez
- Center for Animal Nutrition and Welfare, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210, Vienna, Austria.
- Christian Doppler Laboratory for Innovative Gut Health Concepts of Livestock, Vienna, Austria.
| | - Patrick Biber
- Center for Animal Nutrition and Welfare, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210, Vienna, Austria
- Christian Doppler Laboratory for Innovative Gut Health Concepts of Livestock, Vienna, Austria
| | - Arife Sener-Aydemir
- Center for Animal Nutrition and Welfare, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210, Vienna, Austria
- Christian Doppler Laboratory for Innovative Gut Health Concepts of Livestock, Vienna, Austria
| | - Karin Hummel
- University of Veterinary Medicine Vienna, VetCore Facility (Mass Spectrometry), Vienna, Austria
| | - Ebrahim Razzazi-Fazeli
- University of Veterinary Medicine Vienna, VetCore Facility (Mass Spectrometry), Vienna, Austria
| | - Nicole Reisinger
- Dsm-Firmenich, Animal Nutrition & Health R&D Center, Tulln, Austria
| | - Qendrim Zebeli
- Center for Animal Nutrition and Welfare, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210, Vienna, Austria
- Christian Doppler Laboratory for Innovative Gut Health Concepts of Livestock, Vienna, Austria
| | - Susanne Kreuzer-Redmer
- Center for Animal Nutrition and Welfare, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210, Vienna, Austria
| | - Thomas Hartinger
- Center for Animal Nutrition and Welfare, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210, Vienna, Austria
- Christian Doppler Laboratory for Innovative Gut Health Concepts of Livestock, Vienna, Austria
| |
Collapse
|
11
|
Alharbi O, Al-Mutairi KA, Ibrahim MM, Ramu R, Al-Ghorbani M. New Pyranopyrazole-Based Indolin-2,3-Dione Hybrid as Effective Inhibitors of Xanthine Oxidase: Synthesis, In Vitro, and Molecular Modeling Approaches. Chem Biodivers 2025:e202402104. [PMID: 39777976 DOI: 10.1002/cbdv.202402104] [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: 08/25/2024] [Revised: 12/29/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025]
Abstract
In the current study, new pyranopyrazole analogs (9a-d and 10a-d) were synthesized through a one-pot condensation reaction of 2-arylacetohydrazide. The inhibitory abilities were investigated against the xanthine oxidase (XO) enzyme through experimental and molecular docking analyses. The synthesis studies were based on ultrasound-mediated condensation reactions of four-component containing 2-arylacetohydrazide, ethyl acetoacetate, indoline-2,3-dione, and ethyl 2-cyanoacetate/malononitrile in various solvents and catalysts to yield pyranopyrazole analogs (9a-d and 10a-d) in a short reaction time and remarkably favorable yields ranging from 79% to 92%. On the basis of the XO inhibition study of compounds 9a-d and 10a-d, compound 10d was the most potent (IC50 = 0.09 ± 0.22 µM), followed by 9c (0.12 ± 0.11 µM). With IC50 values of 0.20 ± 0.27 and 0.17 ± 0.11 µM respectively, compounds 10a and 10c exhibited moderate activity. The other compounds have shown less activity compared to the allopurinol control (IC50 = 0.14 ± 0.10 µM). Furthermore, in the molecular docking analysis, compound 10d was predicted to have the highest binding affinity against the target XO enzyme.
Collapse
Affiliation(s)
- Osama Alharbi
- Department of Chemistry, Faculty of Science, Taibah University, Medina Manora, Saudi Arabia
| | | | - Munjed M Ibrahim
- Department of Pharmacy, Faculty of Pharmacy, Nursing, and Health Professions, Birzeit University, West Bank, Palestine
| | - Ramith Ramu
- Department of Biotechnology and Bioinformatics, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Mohammed Al-Ghorbani
- Department of Chemistry, Faculty of Science, Taibah University, Medina Manora, Saudi Arabia
- Department of Chemistry, College of Education, University of Thamar, Dhamar, Yemen
| |
Collapse
|
12
|
Gwozdzinski L, Pieniazek A, Gwozdzinski K. The Roles of Oxidative Stress and Red Blood Cells in the Pathology of the Varicose Vein. Int J Mol Sci 2024; 25:13400. [PMID: 39769165 PMCID: PMC11678264 DOI: 10.3390/ijms252413400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 11/25/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
This review discusses sources of reactive oxygen species, enzymatic antioxidant systems, and low molecular weight antioxidants. We present the pathology of varicose veins (VVs), including factors such as hypoxia, inflammation, dysfunctional endothelial cells, risk factors in varicose veins, the role of RBCs in venous thrombus formation, the influence of reactive oxygen species (ROS) and RBCs on VV pathology, and the role of hemoglobin in the damage of particles and macromolecules in VVs. This review discusses the production of ROS, enzymatic and nonenzymatic antioxidants, the pathogenesis of varicose veins as a pathology based on hypoxia, inflammation, and oxidative stress, as well as the participation of red blood cells in the pathology of varicose veins.
Collapse
Affiliation(s)
- Lukasz Gwozdzinski
- Department of Pharmacology and Toxicology, Medical University of Lodz, 90-752 Lodz, Poland
| | - Anna Pieniazek
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland; (A.P.); (K.G.)
| | - Krzysztof Gwozdzinski
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland; (A.P.); (K.G.)
| |
Collapse
|
13
|
Jorge-Rosas F, Díaz-Godínez C, García-Aguirre S, Martínez-Calvillo S, Carrero JC. Entamoeba histolytica-induced NETs are highly cytotoxic on hepatic and colonic cells due to serine proteases and myeloperoxidase activities. Front Immunol 2024; 15:1493946. [PMID: 39687618 PMCID: PMC11646992 DOI: 10.3389/fimmu.2024.1493946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/14/2024] [Indexed: 12/18/2024] Open
Abstract
During intestinal and liver invasion by the protozoan parasite Entamoeba histolytica, extensive tissue destruction linked to large neutrophil infiltrates is observed. It has been proposed that microbicidal components of neutrophils are responsible for the damage, however, the mechanism by which they are released and act in the extracellular space remains unknown. In previous studies, we have shown that E. histolytica trophozoites induce NET formation, leading to the release of neutrophil granule content into extruded DNA. In this work, we evaluate the possible participation of NETs in the development of amoeba-associated pathology and analyze the contribution of anti-microbial components of the associated granules. E. histolytica-induced NETs were isolated and their effect on the viability and integrity of HCT 116 colonic and Hep G2 liver cultures were evaluated. The results showed that simple incubation of cell monolayers with purified NETs for 24 h resulted in cell detachment and death in a dose-dependent manner. The effect was thermolabile and correlated with the amount of DNA and protein present in NETs. Pretreatment of NETs with specific inhibitors of some microbicidal components suggested that serine proteases, are mostly responsible for the damage caused by NETs on HCT 116 cells, while the MPO activity was the most related to Hep G2 cells damage. Our study also points to a very important role of DNA as a scaffold for the activity of these proteins. We show evidence of the development of NETs in amoebic liver abscesses in hamsters as a preamble to evaluate their participation in tissue damage. In conclusion, these studies demonstrate that amoebic-induced NETs have potent cytotoxic effects on target cells and, therefore, may be responsible for the intense damage associated with tissue invasion by this parasite.
Collapse
Affiliation(s)
- Fabian Jorge-Rosas
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - César Díaz-Godínez
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - Samuel García-Aguirre
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - Santiago Martínez-Calvillo
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, EM, Mexico
| | - Julio César Carrero
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| |
Collapse
|
14
|
Smith PP, Chicca IJ, Heaney JLJ, Muchova M, Khanim FL, Shields AM, Drayson MT, Chapple ILC, Hirschfeld J. Paracetamol suppresses neutrophilic oxygen radicals through competitive inhibition and scavenging. Chem Biol Interact 2024; 404:111283. [PMID: 39428054 DOI: 10.1016/j.cbi.2024.111283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/07/2024] [Accepted: 10/18/2024] [Indexed: 10/22/2024]
Abstract
Neutrophils, pivotal cells of innate and adaptive immune responses, employ reactive oxygen species (ROS) to combat pathogens and control gene expression. Paracetamol (acetaminophen) is widely used as an analgesic and antipyretic medication, yet its precise mechanisms of action are not yet fully understood. Here, we investigate the impact of both ingested and in-vitro paracetamol on neutrophil ROS activity, using flow cytometry and antioxidant assays. Our studies reveal that paracetamol significantly suppresses ROS activity ex-vivo in the short term. Additionally, both paracetamol and its metabolite N-acetyl-p-benzoquinone imine exhibited direct in vitro antioxidant effects, and paracetamol suppressed neutrophil extracellular trap formation ex vivo. These findings suggest a connection between paracetamol use and altered neutrophil responses, with potential implications for use in some patient groups, such as immunocompromised individuals. Further investigation into paracetamol's effects on neutrophil antimicrobial functions is warranted to elucidate possible risks, particularly when taken frequently or in conjunction with other treatments such as vaccinations.
Collapse
Affiliation(s)
- Peter P Smith
- College of Medicine and Health, School of Health Sciences, Dentistry, Periodontal Research Group, University of Birmingham, Birmingham, UK
| | - Ilaria J Chicca
- College of Medical and Health, School of Infection, Inflammation and Immunology, Clinical Immunology Service, University of Birmingham, Birmingham, UK
| | - Jennifer L J Heaney
- College of Medical and Health, School of Infection, Inflammation and Immunology, Clinical Immunology Service, University of Birmingham, Birmingham, UK
| | - Maria Muchova
- College of Medicine and Health, School of Health Sciences, Dentistry, Periodontal Research Group, University of Birmingham, Birmingham, UK
| | - Farhat L Khanim
- College of Medical and Health, School of Infection, Inflammation and Immunology, Clinical Immunology Service, University of Birmingham, Birmingham, UK
| | - Adrian M Shields
- College of Medical and Health, School of Infection, Inflammation and Immunology, Clinical Immunology Service, University of Birmingham, Birmingham, UK
| | - Mark T Drayson
- College of Medical and Health, School of Infection, Inflammation and Immunology, Clinical Immunology Service, University of Birmingham, Birmingham, UK
| | - Iain L C Chapple
- College of Medicine and Health, School of Health Sciences, Dentistry, Periodontal Research Group, University of Birmingham, Birmingham, UK; Birmingham NIHR Biomedical Research Centre in Inflammation, University of Birmingham, Birmingham, UK.
| | - Josefine Hirschfeld
- College of Medicine and Health, School of Health Sciences, Dentistry, Periodontal Research Group, University of Birmingham, Birmingham, UK; Birmingham NIHR Biomedical Research Centre in Inflammation, University of Birmingham, Birmingham, UK.
| |
Collapse
|
15
|
Trinh PTN, Truc NC, Danh TT, Trang NTT, Le Hang DT, Vi LNT, Hung QT, Dung LT. A study on the antioxidant, anti-inflammatory, and xanthine oxidase inhibitory activity of the Artemisia vulgaris L. extract and its fractions. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118519. [PMID: 38971340 DOI: 10.1016/j.jep.2024.118519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/25/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Vietnamese people use mugwort (Artemisia vulgaris L.) to treat arthritis and gout. Our previous research shows that mugwort contains flavonoids, and its extract possesses antibacterial and anti-inflammatory activities. However, no publications have been on the xanthine oxidase inhibitory activity of mugwort and acute anti-inflammatory activity in vivo. AIM OF THE STUDY The study aimed to verify the antioxidant, xanthine oxidase inhibitory, and anti-inflammatory capabilities of mugwort extract in vitro and in vivo, isolate phyto-compounds from potential bioactive fractions, and then evaluate their potential in inhibiting xanthine oxidase. METHODS According to established methods, the extract and the active flavonoids were obtained using different chromatographic techniques. DPPH, ABTS, reducing power, and H2O2 elimination were used to evaluate antioxidant activity. The model of LPS-induced RAW264.7 cells was used to measure the inhibition of NO production. The carrageenan-induced paw oedema model was used to assess acute inflammation in mice. In vitro, xanthine oxidase inhibition assay was applied to investigate the effects of extract/compounds on uric acid production. Chemical structures were identified by spectral analysis. RESULTS The assessment of the acute inflammatory model in mice revealed that both the 96% ethanol and the 50% ethanol extracts significantly decreased oedema in the mice's feet following carrageenan-induced inflammation. 96% ethanol extract exhibited a better reduction in oedema at the low dose. The analysis revealed that the ethyl acetate fraction had the highest levels of total polyphenols and flavonoids. Additionally, this fraction demonstrated significant antioxidant activity in various assays, such as DPPH, ABTS, reducing power, and H2O2 removal. Furthermore, it displayed the most potent inhibition of xanthine oxidase, an anti-inflammatory activity. Five phytochemicals were isolated and determined from the active fraction such as luteolin (1), rutin (2), apigenin (3), myricetin (4), and quercetin (5). Except for rutin, the other compounds demonstrated the ability to inhibit effective xanthine oxidase compared to standard (allopurinol). Moreover, quercetin (5) inhibited NO production (IC50 21.87 μM). CONCLUSION The results indicate that extracts from A. vulgaris effectively suppressed the activity of xanthine oxidase and exhibited antioxidant and anti-inflammatory properties, potentially leading to a reduction in the production of uric acid in the body and eliminating ROS. The study identified mugwort extract and bioactive compounds derived from Artemisia vulgaris, specifically luteolin, apigenin, and quercetin, as promising xanthine oxidase inhibitors. These findings suggest that further development of these compounds is warranted. At the same time, the above results also strengthen the use of mugwort to treat gout disease in Vietnam.
Collapse
Affiliation(s)
- Pham Thi Nhat Trinh
- Department of Natural Science, Tien Giang University, 119 Ap Bac, My Tho, Tien Giang, Viet Nam
| | - Nguyen Cong Truc
- Department of Organic Chemistry, Faculty of Chemical Engineering, Ho Chi Minh City University of Technology (HCMUT), 268 Ly Thuong Kiet, Ho Chi Minh, Viet Nam; Institute of Applied Materials Science, Vietnam Academy of Science and Technology (VAST), 1B TL29, Ho Chi Minh, Viet Nam
| | - Tong Thanh Danh
- Department of Organic Chemistry, Faculty of Chemical Engineering, Ho Chi Minh City University of Technology (HCMUT), 268 Ly Thuong Kiet, Ho Chi Minh, Viet Nam
| | | | - Dang Thi Le Hang
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology (VAST), 1B TL29, Ho Chi Minh, Viet Nam; Graduate University of Science and Technology (VAST), 18 Hoang Quoc Viet, Cau Giay, Hanoi, Viet Nam.
| | - Le Nguyen Tuong Vi
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology (VAST), 1B TL29, Ho Chi Minh, Viet Nam
| | - Quach Tong Hung
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology (VAST), 1B TL29, Ho Chi Minh, Viet Nam
| | - Le Tien Dung
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology (VAST), 1B TL29, Ho Chi Minh, Viet Nam; Graduate University of Science and Technology (VAST), 18 Hoang Quoc Viet, Cau Giay, Hanoi, Viet Nam.
| |
Collapse
|
16
|
Janakiraman V, Sudhan M, Ahmad SF, Attia SM, Emran TB, Ahmed SSSJ. Molecular Docking, Quantum Mechanics and Molecular Dynamics Simulation of Anti-CAD Drugs Against High-Risk Xanthine Dehydrogenase Variants Associated with Oxidative Stress Pathways. JOURNAL OF COMPUTATIONAL BIOPHYSICS AND CHEMISTRY 2024; 23:1109-1128. [DOI: 10.1142/s2737416524500315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Xanthine dehydrogenase (XDH) contributes significantly to generating reactive oxygen species in coronary artery disease (CAD). XDH has been proposed as a therapeutic target, but its genetic variants could affect protein structure and drug response. We aimed to assess protein structure modification occur due to genetic variants and to screen 215 CAD drugs for their utility in personalized CAD treatment against the XDH variants. A series of computational methods were implemented to identify pathogenic variants that cause XDH structure instability localized at the con served regions contributing to functional significance. Then, the XDH structures with the pathogenic variants were modeled using two different approaches to select the best models for docking with the CAD drugs. Finally, the stability of the docked complexes and their ability to transfer electrons were evaluated using molecular dynamics (MD) simulations and quantum mechanics/molecular mechanics (QM/MM) calculation. Among 751 variants examined; R149C and Q919R showed high pathogenicity, localized in conserved regions could alter protein structure and function. Further, docking of CAD drugs against XDH (native, R149C and Q919R) showed vericiguat with higher affinity, ranging from −7.95 kcal/mol to −10.41 kcal/mol, than the well-known XDH inhibitor (febuxostat, −5.73 kcal/mol to −8.35 kcal/mol). This indicates that vericiguat will be effective in CAD treatment, regardless of the XDH variants. Additionally, MD simulation and QM/MM confirmed vericiguat stability and electron transfer ability to form hydrogen bonds with the XDH protein. In conclusion, vericiguat will be beneficial for the personalized treatment of CAD by inhibiting XDH variants. Additional clinical studies are necessary to confirm our findings.
Collapse
Affiliation(s)
- V. Janakiraman
- Drug Discovery and Multi-omics Laboratory, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Kelambakkam 603103, Tamil Nadu, India
| | - M. Sudhan
- Drug Discovery and Multi-omics Laboratory, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Kelambakkam 603103, Tamil Nadu, India
| | - Sheikh F. Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sabry M. Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Talha Bin Emran
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Legorreta Cancer Center, Brown University, Providence, RI 02912, USA
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Shiek S. S. J. Ahmed
- Drug Discovery and Multi-omics Laboratory, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Kelambakkam 603103, Tamil Nadu, India
| |
Collapse
|
17
|
Nwokocha GC, Ghosh A, Grove A. Regulation of bacterial virulence genes by PecS family transcription factors. J Bacteriol 2024; 206:e0030224. [PMID: 39287432 PMCID: PMC11500572 DOI: 10.1128/jb.00302-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024] Open
Abstract
Bacterial plant pathogens adjust their gene expression programs in response to environmental signals and host-derived compounds. This ensures that virulence genes or genes encoding proteins, which promote bacterial fitness in a host environment, are expressed only when needed. Such regulation is in the purview of transcription factors, many of which belong to the ubiquitous multiple antibiotic resistance regulator (MarR) protein family. PecS proteins constitute a subset of this large protein family. PecS has likely been distributed by horizontal gene transfer, along with the divergently encoded efflux pump PecM, suggesting its integration into existing gene regulatory networks. Here, we discuss the roles of PecS in the regulation of genes associated with virulence and fitness of bacterial plant pathogens. A comparison of phenotypes and differential gene expression associated with the disruption of pecS shows that functional consequences of PecS integration into existing transcriptional networks are highly variable, resulting in distinct PecS regulons. Although PecS universally binds to the pecS-pecM intergenic region to repress the expression of both genes, binding modes differ. A particularly relaxed sequence preference appears to apply for Dickeya dadantii PecS, perhaps to optimize its integration as a global regulator and regulate genes ancestral to the acquisition of pecS-pecM. Even inducing ligands for PecS are not universally conserved. It appears that PecS function has been optimized to match the unique regulatory needs of individual bacterial species and that its roles must be appreciated in the context of the regulatory networks into which it was recruited.
Collapse
Affiliation(s)
| | - Arpita Ghosh
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Anne Grove
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, USA
| |
Collapse
|
18
|
Annesi L, Tossetta G, Borghi C, Piani F. The Role of Xanthine Oxidase in Pregnancy Complications: A Systematic Review. Antioxidants (Basel) 2024; 13:1234. [PMID: 39456486 PMCID: PMC11505381 DOI: 10.3390/antiox13101234] [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: 08/22/2024] [Revised: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
Xanthine oxidoreductase (XOR) is an enzyme involved in the oxidation of hypoxanthine and xanthine to uric acid. XOR has two isoforms: xanthine dehydrogenase and xanthine oxidase (XO). XO plays a major role in oxidative stress, causing the formation of reactive oxygen species. In the present study, we aimed to summarize the evidence on the association between XO and pregnancy complications. The PRISMA checklist guided the reporting of the data. We conducted systematic searches in the PubMed and Web of Science databases to identify all human studies investigating XO in pregnancy diseases up to June 2024. A total of 195 references have been identified and 14 studies were included. Most studies focused on women with PE and GD. Overall, all the included studies found a statistically significant increase in maternal, placental, and/or fetal XO levels, activity, or tissue expression in women with pregnancy complications, compared to those with uncomplicated pregnancies. Although promising, the quality and dimension of the included studies do not allow for a definitive answer to the question of whether XO may play a crucial role in pregnancy complications. Future studies are warranted to confirm if XO could represent a prognostic and therapeutic marker in pregnancy complications and their impact on long-term maternal and offspring cardiovascular health.
Collapse
Affiliation(s)
- Lorenzo Annesi
- Hypertension and Cardiovascular Risk Research Center, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; (L.A.); (C.B.)
| | - Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy;
| | - Claudio Borghi
- Hypertension and Cardiovascular Risk Research Center, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; (L.A.); (C.B.)
| | - Federica Piani
- Hypertension and Cardiovascular Risk Research Center, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; (L.A.); (C.B.)
| |
Collapse
|
19
|
van Drie RWA, van de Wouw J, Zandbergen LM, Dehairs J, Swinnen JV, Mulder MT, Verhaar MC, MaassenVanDenBrink A, Duncker DJ, Sorop O, Merkus D. Vasodilator reactive oxygen species ameliorate perturbed myocardial oxygen delivery in exercising swine with multiple comorbidities. Basic Res Cardiol 2024; 119:869-887. [PMID: 38796544 PMCID: PMC11461570 DOI: 10.1007/s00395-024-01055-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/28/2024]
Abstract
Multiple common cardiovascular comorbidities produce coronary microvascular dysfunction. We previously observed in swine that a combination of diabetes mellitus (DM), high fat diet (HFD) and chronic kidney disease (CKD) induced systemic inflammation, increased oxidative stress and produced coronary endothelial dysfunction, altering control of coronary microvascular tone via loss of NO bioavailability, which was associated with an increase in circulating endothelin (ET). In the present study, we tested the hypotheses that (1) ROS scavenging and (2) ETA+B-receptor blockade improve myocardial oxygen delivery in the same female swine model. Healthy female swine on normal pig chow served as controls (Normal). Five months after induction of DM (streptozotocin, 3 × 50 mg kg-1 i.v.), hypercholesterolemia (HFD) and CKD (renal embolization), swine were chronically instrumented and studied at rest and during exercise. Sustained hyperglycemia, hypercholesterolemia and renal dysfunction were accompanied by systemic inflammation and oxidative stress. In vivo ROS scavenging (TEMPOL + MPG) reduced myocardial oxygen delivery in DM + HFD + CKD swine, suggestive of a vasodilator influence of endogenous ROS, while it had no effect in Normal swine. In vitro wire myography revealed a vasodilator role for hydrogen peroxide (H2O2) in isolated small coronary artery segments from DM + HFD + CKD, but not Normal swine. Increased catalase activity and ceramide production in left ventricular myocardial tissue of DM + HFD + CKD swine further suggest that increased H2O2 acts as vasodilator ROS in the coronary microvasculature. Despite elevated ET-1 plasma levels in DM + HFD + CKD swine, ETA+B blockade did not affect myocardial oxygen delivery in Normal or DM + HFD + CKD swine. In conclusion, loss of NO bioavailability due to 5 months exposure to multiple comorbidities is partially compensated by increased H2O2-mediated coronary vasodilation.
Collapse
Affiliation(s)
- R W A van Drie
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
- Laboratory of Vascular Medicine, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - J van de Wouw
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - L M Zandbergen
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
- Walter Brendel Center of Experimental Medicine (WBex), University Clinic Munich, 81377 LMU, Munich, Germany
| | - J Dehairs
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven-University of Leuven, Leuven, Belgium
| | - J V Swinnen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven-University of Leuven, Leuven, Belgium
| | - M T Mulder
- Laboratory of Vascular Medicine, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - M C Verhaar
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - A MaassenVanDenBrink
- Laboratory of Vascular Medicine, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - D J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - O Sorop
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - D Merkus
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.
- Walter Brendel Center of Experimental Medicine (WBex), University Clinic Munich, 81377 LMU, Munich, Germany.
- Center for Cardiovascular Research (DZHK), Munich Heart Alliance (MHA), Partner Site Munich, 81377, Munich, Germany.
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), University Clinic Munich, LMU, Munich, Germany.
| |
Collapse
|
20
|
Owumi S, Agbarogi H, Oluwawibe BJ, Otunla MT, Anifowose MM, Arunsi UO. Modulation of the Nrf-2 and HO-1 signalling axis is associated with Betaine's abatement of fluoride-induced hepatorenal toxicities in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7725-7745. [PMID: 38713257 DOI: 10.1007/s00210-024-03133-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 04/29/2024] [Indexed: 05/08/2024]
Abstract
Sodium fluoride (NaF) ingestion has several detrimental effects in humans and rodents. NaF mechanisms of toxicity include perturbation of intracellular redox homeostasis and apoptosis. Betaine (BET) is a modified amino acid with anti-inflammatory, antioxidant, and anti-apoptotic properties. This study investigates BET's effect on NaF-induced hepatorenal toxicities in rats. Experimental rats (n = 30) were randomly assigned to groups (n = 6) and treated by gavage for 28 days. Group I (2 mL of distilled water), Group II (NaF: 9 mg/kg) alone, Group III: (BET: 100 mg/kg), Group IV: (NaF: 9 mg/kg and BET 1: 50 mg/kg), and Group V: (NaF: 9 mg/kg and BET 2: 100 mg/kg). Our findings revealed significantly (p < 0.05) increased hepatic transaminase activities alongside creatinine and urea levels following NaF-alone treatment in addition to increased oxidative status, lipid peroxidation, reactive oxygen and nitrogen species, decreased superoxide dismutase, catalase, glutathione-s-transferase, glutathione peroxidase, glutathione, and total sulfhydryl groups. The reduced levels of nuclear factor erythroid 2-related factor-2 and the activities of heme oxygenase-1, thioredoxin, and thioredoxin reductase in NaF-alone treated rats equally compromised cellular molecular responses to oxidative stress. Also, NaF increased (p < 0.05) hepatorenal inflammatory biomarkers-nitric oxide, interleukin-10, myeloperoxidase, and xanthine oxidase. Furthermore, caspase-3 and caspase-9 were increased (p < 0.05) in rats treated with NaF alone. Contrastingly, BET was observed to alleviate the harmful effects of NaF. Treatment with BET mitigated NaF-induced oxido-inflammatory responses and apoptosis in the experimental rat's hepatorenal system. The study demonstrates the potential of BET to abate NaF-induced hepatorenal toxicity.
Collapse
Affiliation(s)
- Solomon Owumi
- ChangeLab-changing lives; Room New Building 302, Cancer Research and Molecular Biology Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, 200005, Oyo State, Nigeria.
| | - Harieme Agbarogi
- ChangeLab-changing lives; Room New Building 302, Cancer Research and Molecular Biology Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, 200005, Oyo State, Nigeria
| | - Bayode J Oluwawibe
- ChangeLab-changing lives; Room New Building 302, Cancer Research and Molecular Biology Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, 200005, Oyo State, Nigeria
| | - Moses T Otunla
- ChangeLab-changing lives; Room New Building 302, Cancer Research and Molecular Biology Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, 200005, Oyo State, Nigeria
| | - Mayowa M Anifowose
- School of Chemistry & Biochemistry, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332-0400, USA
| | - Uche O Arunsi
- School of Chemistry & Biochemistry, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332-0400, USA
| |
Collapse
|
21
|
Suganuma T, Hassan H, Gogol M, Workman JL. C G composition in transposon-derived genes is increased in FXD with perturbed immune system. NAR MOLECULAR MEDICINE 2024; 1:ugae015. [PMID: 39465205 PMCID: PMC11500580 DOI: 10.1093/narmme/ugae015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/01/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024]
Abstract
Increasing incidence of Fragile X disorders (FXD) and of immune-mediated disorders in FXD suggests that additional factors besides FMR1 mutations contribute to the pathogenesis. Here, we discovered that the expression levels or splicing of specific transposon element (TE)-derived genes, regulating purine metabolism and immune responses against viral infections are altered in FXD. These genes include HLA genes clustered in chr6p21.3 and viral responsive genes in chr5q15. Remarkably, these TE-derived genes contain a low A T/C G suggesting base substitutions of A T to C G. The TE-derived genes with changed expression levels contained a higher content of 5'-CG-3' dinucleotides in FXD compared to healthy donors. This resembles the genomes of some RNA viruses, which maintain high contents of CG dinucleotides to sustain their latent infection exploiting antiviral responses. Thus, past viral infections may have persisted as TEs, provoking immune-mediated disorders in FXD.
Collapse
Affiliation(s)
- Tamaki Suganuma
- Stowers Institute for Medical Research, 1000 E. 50 Street, Kansas City, MO 64110, USA
| | - Huzaifa Hassan
- Stowers Institute for Medical Research, 1000 E. 50 Street, Kansas City, MO 64110, USA
| | - Madelaine Gogol
- Stowers Institute for Medical Research, 1000 E. 50 Street, Kansas City, MO 64110, USA
| | - Jerry L Workman
- Stowers Institute for Medical Research, 1000 E. 50 Street, Kansas City, MO 64110, USA
| |
Collapse
|
22
|
Pawluk H, Tafelska-Kaczmarek A, Sopońska M, Porzych M, Modrzejewska M, Pawluk M, Kurhaluk N, Tkaczenko H, Kołodziejska R. The Influence of Oxidative Stress Markers in Patients with Ischemic Stroke. Biomolecules 2024; 14:1130. [PMID: 39334896 PMCID: PMC11430825 DOI: 10.3390/biom14091130] [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: 06/25/2024] [Revised: 07/27/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
Stroke is the second leading cause of death worldwide, and its incidence is rising rapidly. Acute ischemic stroke is a subtype of stroke that accounts for the majority of stroke cases and has a high mortality rate. An effective treatment for stroke is to minimize damage to the brain's neural tissue by restoring blood flow to decreased perfusion areas of the brain. Many reports have concluded that both oxidative stress and excitotoxicity are the main pathological processes associated with ischemic stroke. Current measures to protect the brain against serious damage caused by stroke are insufficient. For this reason, it is important to investigate oxidative and antioxidant strategies to reduce oxidative damage. This review focuses on studies assessing the concentration of oxidative stress biomarkers and the level of antioxidants (enzymatic and non-enzymatic) and their impact on the clinical prognosis of patients after stroke. Mechanisms related to the production of ROS/RNS and the role of oxidative stress in the pathogenesis of ischemic stroke are presented, as well as new therapeutic strategies aimed at reducing the effects of ischemia and reperfusion.
Collapse
Affiliation(s)
- Hanna Pawluk
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Karlowicza 24, 85-092 Bydgoszcz, Poland
| | - Agnieszka Tafelska-Kaczmarek
- Department of Organic Chemistry, Faculty of Chemistry, Nicolaus Copernicus University, Gagarina 7, 87-100 Torun, Poland
| | - Małgorzata Sopońska
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Karlowicza 24, 85-092 Bydgoszcz, Poland
| | - Marta Porzych
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Karlowicza 24, 85-092 Bydgoszcz, Poland
| | - Martyna Modrzejewska
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Karlowicza 24, 85-092 Bydgoszcz, Poland
| | - Mateusz Pawluk
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Karlowicza 24, 85-092 Bydgoszcz, Poland
| | - Natalia Kurhaluk
- Institute of Biology, Pomeranian University in Slupsk, Arciszewski 22B, 76-200 Slupsk, Poland
| | - Halina Tkaczenko
- Institute of Biology, Pomeranian University in Slupsk, Arciszewski 22B, 76-200 Slupsk, Poland
| | - Renata Kołodziejska
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Karlowicza 24, 85-092 Bydgoszcz, Poland
| |
Collapse
|
23
|
Chien HT, Lin YW, Shen LJ, Hsieh SC, Lin LY, Chen YA, Lin FJ. Sex-specific associations between prolonged serum uric acid levels and risk of major adverse cardiovascular events. INTERNATIONAL JOURNAL OF CARDIOLOGY. CARDIOVASCULAR RISK AND PREVENTION 2024; 22:200302. [PMID: 39035111 PMCID: PMC11259864 DOI: 10.1016/j.ijcrp.2024.200302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/01/2024] [Accepted: 06/19/2024] [Indexed: 07/23/2024]
Abstract
Background While hyperuricemia has been correlated with cardiovascular (CV) diseases, further evidence is required to evaluate the implications of stable serum uric acid (sUA) levels, especially concerning low sUA. This study aimed to investigate prolonged stable sUA levels and CV events/mortality. Methods We conducted a retrospective cohort study at a medical center using electronic medical records linked with the national claims database. Patients with at least two sUA measurements, with intervals ranging from 6 months to 4 years, were included. The mean of the first two eligible sUA measurements were analyzed, stratified by sex. Outcomes of interest comprised major adverse cardiovascular events (MACE), heart failure hospitalization, CV and all-cause mortality. Results This study included 33,096 patients (follow-up: men 6.6 years, women 6.4 years). After multivariable adjustment, cubic spline models showed that long-term high sUA levels were consistently associated with a higher risk of MACE, heart failure hospitalization, CV and all-cause mortality. A U-shaped association was observed between sUA levels and all-cause mortality in both sexes and between sUA levels and CV mortality in women. The impact of sUA, especially lower levels, on CV events and mortality was more pronounced in women than in men. Conclusion Long-term high sUA levels are consistently associated with increased risk of CV events and mortality. A U-shaped association between sUA levels and all-cause mortality was observed in both men and women and was pronounced in women. The findings underscore the importance of considering sUA levels, especially in women, when assessing CV risk.
Collapse
Affiliation(s)
- Hsiu-Ting Chien
- Graduate Institute of Clinical Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Wen Lin
- Graduate Institute of Clinical Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Li-Jiuan Shen
- Graduate Institute of Clinical Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Pharmacy, National Taiwan University Hospital, Taipei, Taiwan
| | - Song-Chou Hsieh
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Division of Allergy, Immunology & Rheumatology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Lian-Yu Lin
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yi-An Chen
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Fang-Ju Lin
- Graduate Institute of Clinical Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Pharmacy, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
24
|
Kumar N, Yang ML, Sun P, Hunker KL, Li J, Jia J, Fan F, Wang J, Ning X, Gao W, Xu M, Zhang J, Chang L, Chen YE, Huo Y, Zhang Y, Ganesh SK. Genetic variation in CCDC93 is associated with elevated central systolic blood pressure, impaired arterial relaxation, and mitochondrial dysfunction. PLoS Genet 2024; 20:e1011151. [PMID: 39250516 PMCID: PMC11421807 DOI: 10.1371/journal.pgen.1011151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 09/24/2024] [Accepted: 01/23/2024] [Indexed: 09/11/2024] Open
Abstract
Genetic studies of blood pressure (BP) traits to date have been performed on conventional measures by brachial cuff sphygmomanometer for systolic BP (SBP) and diastolic BP, integrating several physiologic occurrences. Genetic associations with central SBP (cSBP) have not been well-studied. Genetic discovery studies of BP have been most often performed in European-ancestry samples. Here, we investigated genetic associations with cSBP in a Chinese population and functionally validated the impact of a novel associated coiled-coil domain containing 93 (CCDC93) gene on BP regulation. An exome-wide association study (EWAS) was performed using a mixed linear model of non-invasive cSBP and peripheral BP traits in a Han Chinese population (N = 5,954) from Beijing, China genotyped with a customized Illumina ExomeChip array. We identified four SNP-trait associations with three SNPs, including two novel associations (rs2165468-SBP and rs33975708-cSBP). rs33975708 is a coding variant in the CCDC93 gene, c.535C>T, p.Arg179Cys (MAF = 0.15%), and was associated with increased cSBP (β = 29.3 mmHg, P = 1.23x10-7). CRISPR/Cas9 genome editing was used to model the effect of Ccdc93 loss in mice. Homozygous Ccdc93 deletion was lethal prior to day 10.5 of embryonic development. Ccdc93+/- heterozygous mice were viable and morphologically normal, with 1.3-fold lower aortic Ccdc93 protein expression (P = 0.0041) and elevated SBP as compared to littermate Ccdc93+/+ controls (110±8 mmHg vs 125±10 mmHg, P = 0.016). Wire myography of Ccdc93+/- aortae showed impaired acetylcholine-induced relaxation and enhanced phenylephrine-induced contraction. RNA-Seq transcriptome analysis of Ccdc93+/- mouse thoracic aortae identified significantly enriched pathways altered in fatty acid metabolism and mitochondrial metabolism. Plasma free fatty acid levels were elevated in Ccdc93+/- mice (96±7mM vs 124±13mM, P = 0.0031) and aortic mitochondrial dysfunction was observed through aberrant Parkin and Nix protein expression. Together, our genetic and functional studies support a novel role of CCDC93 in the regulation of BP through its effects on vascular mitochondrial function and endothelial function.
Collapse
Affiliation(s)
- Nitin Kumar
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Min-Lee Yang
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Pengfei Sun
- Department of Cardiology, Peking University First hospital, Beijing, China
- Department of Cardiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Kristina L. Hunker
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Jianping Li
- Department of Cardiology, Peking University First hospital, Beijing, China
| | - Jia Jia
- Department of Cardiology, Peking University First hospital, Beijing, China
| | - Fangfang Fan
- Department of Cardiology, Peking University First hospital, Beijing, China
| | - Jinghua Wang
- Laboratory of Epidemiology, Tianjin Neurological Institute, Tianjin, China
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xianjia Ning
- Laboratory of Epidemiology, Tianjin Neurological Institute, Tianjin, China
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Wei Gao
- Department of Cardiology, Peking University Third hospital, Beijing, China
| | - Ming Xu
- Department of Cardiology, Peking University Third hospital, Beijing, China
| | - Jifeng Zhang
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Lin Chang
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Y. Eugene Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Yong Huo
- Department of Cardiology, Peking University First hospital, Beijing, China
| | - Yan Zhang
- Department of Cardiology, Peking University First hospital, Beijing, China
- Institute of Cardiovascular Disease, Peking University First Hospital, Beijing, China
- Hypertension Precision Diagnosis and Treatment Research Center, Peking University First Hospital, Beijing, China
| | - Santhi K. Ganesh
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| |
Collapse
|
25
|
Rameshbabu S, Alehaideb Z, Alghamdi SS, Suliman RS, Almourfi F, Yacoob SAM, Venkataraman A, Messaoudi S, Matou-Nasri S. Identification of Anastatica hierochuntica L. Methanolic-Leaf-Extract-Derived Metabolites Exhibiting Xanthine Oxidase Inhibitory Activities: In Vitro and In Silico Approaches. Metabolites 2024; 14:368. [PMID: 39057691 PMCID: PMC11278686 DOI: 10.3390/metabo14070368] [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: 05/22/2024] [Revised: 06/06/2024] [Accepted: 06/12/2024] [Indexed: 07/28/2024] Open
Abstract
There is a growing interest in the discovery of novel xanthine oxidase inhibitors for gout prevention and treatment with fewer side effects. This study aimed to identify the xanthine oxidase (XO) inhibitory potential and drug-likeness of the metabolites present in the methanolic leaf extract of Anastatica (A.) hierochuntica L. using in vitro and in silico models. The extract-derived metabolites were identified by liquid-chromatography-quadrupole-time-of-flight-mass-spectrometry (LC-QTOF-MS). Molecular docking predicted the XO inhibitory activity of the identified metabolites and validated the best scored in vitro XO inhibitory activities for experimental verification, as well as predictions of their anticancer, pharmacokinetic, and toxic properties; oral bioavailability; and endocrine disruption using SwissADMET, PASS, ProTox-II, and Endocrine Disruptome web servers. A total of 12 metabolites, with a majority of flavonoids, were identified. Rutin, quercetin, and luteolin flavonoids demonstrated the highest ranked docking scores of -12.39, -11.15, and -10.43, respectively, while the half-maximal inhibitory concentration (IC50) values of these metabolites against XO activity were 11.35 µM, 11.1 µM, and 21.58 µM, respectively. In addition, SwissADMET generated data related to the physicochemical properties and drug-likeness of the metabolites. Similarly, the PASS, ProTox-II, and Endocrine Disruptome prediction models stated the safe and potential use of these natural compounds. However, in vivo studies are necessary to support the development of the prominent and promising therapeutic use of A. hierochuntica methanolic-leaf-extract-derived metabolites as XO inhibitors for the prevention and treatment of hyperuricemic and gout patients. Furthermore, the predicted findings of the present study open a new paradigm for these extract-derived metabolites by revealing novel oncogenic targets for the potential treatment of human malignancies.
Collapse
Affiliation(s)
- Saranya Rameshbabu
- PG & Research Department of Biotechnology, Mohamed Sathak College of Arts and Science, Chennai 600119, India; (S.R.); (S.A.M.Y.)
| | - Zeyad Alehaideb
- Department of Core Medical Research Facility and Platform, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11481, Saudi Arabia; (Z.A.); (F.A.)
| | - Sahar S. Alghamdi
- Department of Pharmaceutical Sciences, College of Pharmacy, KSAU-HS, MNGHA, Riyadh 11481, Saudi Arabia;
| | - Rasha S. Suliman
- Department of Pharmacy, Fatima College of Health Sciences, Abu Dhabi P.O. Box 3798, United Arab Emirates;
| | - Feras Almourfi
- Department of Core Medical Research Facility and Platform, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11481, Saudi Arabia; (Z.A.); (F.A.)
| | - Syed Ali Mohamed Yacoob
- PG & Research Department of Biotechnology, Mohamed Sathak College of Arts and Science, Chennai 600119, India; (S.R.); (S.A.M.Y.)
| | - Anuradha Venkataraman
- PG & Research Department of Biochemistry, Mohamed Sathak College of Arts and Science, Chennai 600119, India;
| | - Safia Messaoudi
- Department of Forensic Science, College of Criminal Justice, Naif Arab University for Security Sciences, Riyadh 11452, Saudi Arabia;
| | - Sabine Matou-Nasri
- Department of Blood and Cancer Research, KAIMRC, KSAU-HS, MNGHA, Riyadh 11481, Saudi Arabia
- Department of Biosciences, Faculty of the School of Systems Biology, George Mason University, Manassas, VA 22030, USA
| |
Collapse
|
26
|
Korsmo HW, Ekperikpe US, Daehn IS. Emerging Roles of Xanthine Oxidoreductase in Chronic Kidney Disease. Antioxidants (Basel) 2024; 13:712. [PMID: 38929151 PMCID: PMC11200862 DOI: 10.3390/antiox13060712] [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: 05/03/2024] [Revised: 06/09/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Xanthine Oxidoreductase (XOR) is a ubiquitous, essential enzyme responsible for the terminal steps of purine catabolism, ultimately producing uric acid that is eliminated by the kidneys. XOR is also a physiological source of superoxide ion, hydrogen peroxide, and nitric oxide, which can function as second messengers in the activation of various physiological pathways, as well as contribute to the development and the progression of chronic conditions including kidney diseases, which are increasing in prevalence worldwide. XOR activity can promote oxidative distress, endothelial dysfunction, and inflammation through the biological effects of reactive oxygen species; nitric oxide and uric acid are the major products of XOR activity. However, the complex relationship of these reactions in disease settings has long been debated, and the environmental influences and genetics remain largely unknown. In this review, we give an overview of the biochemistry, biology, environmental, and current clinical impact of XOR in the kidney. Finally, we highlight recent genetic studies linking XOR and risk for kidney disease, igniting enthusiasm for future biomarker development and novel therapeutic approaches targeting XOR.
Collapse
Affiliation(s)
| | | | - Ilse S. Daehn
- Department of Medicine, Division of Nephrology, The Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, Box 1243, New York, NY 10029, USA
| |
Collapse
|
27
|
Olatunji LA, Badmus OO, Abdullahi KO, Usman TO, ologe M, Adejare A. Depletion of hepatic glutathione and adenosine by glucocorticoid exposure in Wistar rats is pregnancy-independent. Toxicol Rep 2024; 12:485-491. [PMID: 38741615 PMCID: PMC11090063 DOI: 10.1016/j.toxrep.2024.04.011] [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: 06/03/2023] [Revised: 02/24/2024] [Accepted: 04/29/2024] [Indexed: 05/16/2024] Open
Abstract
Liver diseases have gained increasing attention due to their substantial impact on health, independently as well as in association with cardio-metabolic disorders. Studies have suggested that glutathione and adenosine assist in providing protection against oxidative stress and inflammation while glucocorticoid (GC) therapy has been associated with chronic inflammatory disorders, even in pregnancy. The implications of Glucocorticoid exposure on maternal health and fetal growth is a concern, however, the possible role of glutathione and adenosine has not been thoroughly investigated. The study therefore hypothesize that exposure to glucocorticoids leads to depletion of hepatic glutathione and adenosine levels, contributing to oxidative stress and tissue injury. Additionally, we aim to investigate whether the effects of glucocorticoids on hepatic health are pregnancy dependent in female rats. Twelve Pregnant and twelve age-matched non-pregnant rats were used for this study; an exogenous administration of glucocorticoid (Dex: 0.2 mg/kg) or vehicle (po) was administered to six pregnant and six non-pregnant rats from gestational day 14 to 19 or for a period of 6 days respectively. Data obtained showed that GC exposure led to a decrease in hepatic glucose-6-phosphate dehydrogenase, glutathione peroxidase, GSH/GSSG ratio and adenosine content in both pregnant and non-pregnant rats. In addition, increased activities of adenosine deaminase and xanthine oxidase, along with increased production of uric acid and increased levels of lactate dehydrogenase, aspartate aminotransferase, alanine transferase, alkaline phosphatase and gamma-glutamyl transferase were observed. In summary, the study indicates that GC-induced liver damage is underlined by depleted hepatic adenosine and glutathione levels as well as elevated markers of tissue inflammation and/or injury. Furthermore, the findings suggest that the effects of GC exposure on hepatic health are pregnancy independent.
Collapse
Affiliation(s)
- Lawrence A. Olatunji
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Olufunto O. Badmus
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
- Department of Physiology and Biophysics, Cardiorenal, and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Kamaldeen O. Abdullahi
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Taofeek O. Usman
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
- Division of Endocrinology and Diabetes, Department of Pediatrics, Children’s Hospital of Pittsburgh of University of Pittsburgh School of Medicine, Pittsburg, PA, USA
| | - Mary ologe
- Department of Pharmacology and Therapeutics, University of Ilorin, Ilorin, Nigeria
| | | |
Collapse
|
28
|
Bouyahya A, Bakrim S, Aboulaghras S, El Kadri K, Aanniz T, Khalid A, Abdalla AN, Abdallah AA, Ardianto C, Ming LC, El Omari N. Bioactive compounds from nature: Antioxidants targeting cellular transformation in response to epigenetic perturbations induced by oxidative stress. Biomed Pharmacother 2024; 174:116432. [PMID: 38520868 DOI: 10.1016/j.biopha.2024.116432] [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: 12/17/2023] [Revised: 03/09/2024] [Accepted: 03/15/2024] [Indexed: 03/25/2024] Open
Abstract
Oxidative stress results from a persistent imbalance in oxidation levels that promotes oxidants, playing a crucial role in the early and sustained phases of DNA damage and genomic and epigenetic instability, both of which are intricately linked to the development of tumors. The molecular pathways contributing to carcinogenesis in this context, particularly those related to double-strand and single-strand breaks in DNA, serve as indicators of DNA damage due to oxidation in cancer cases, as well as factors contributing to epigenetic instability through ectopic expressions. Oxidative stress has been considered a therapeutic target for many years, and an increasing number of studies have highlighted the promising effectiveness of natural products in cancer treatment. In this regard, we present significant research on the therapeutic targeting of oxidative stress using natural molecules and underscore the essential role of oxidative stress in cancer. The consequences of stress, especially epigenetic instability, also offer significant therapeutic prospects. In this context, the use of natural epi-drugs capable of modulating and reorganizing the epigenetic network is beginning to emerge remarkably. In this review, we emphasize the close connections between oxidative stress, epigenetic instability, and tumor transformation, while highlighting the role of natural substances as antioxidants and epi-drugs in the anti-tumoral context.
Collapse
Affiliation(s)
- Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco.
| | - Saad Bakrim
- Geo-Bio-Environment Engineering and Innovation Laboratory, Molecular Engineering, Biotechnology and Innovation Team, Polydisciplinary Faculty of Taroudant, Ibn Zohr University, Agadir 80000, Morocco
| | - Sara Aboulaghras
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco
| | - Kawtar El Kadri
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco
| | - Tarik Aanniz
- Biotechnology Lab (MedBiotech), Bioinova Research Center, Rabat Medical & Pharmacy School, Mohammed V University in Rabat, Morocco
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Center, Jazan University, Jazan PO Box: 114, Saudi Arabia.
| | - Ashraf N Abdalla
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Ahmed A Abdallah
- Department of Anatomy, Faculty of Medicine, Umm Alqura University, Makkah 21955, Saudi Arabia
| | - Chrismawan Ardianto
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia.
| | - Long Chiau Ming
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia; School of Medical and Life Sciences, Sunway University, Sunway City, Malaysia; Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei Darussalam.
| | - Nasreddine El Omari
- High Institute of Nursing Professions and Health Techniques of Tetouan, Tetouan, Morocco
| |
Collapse
|
29
|
Jing Q, Zhou C, Zhang J, Zhang P, Wu Y, Zhou J, Tong X, Li Y, Du J, Wang Y. Role of reactive oxygen species in myelodysplastic syndromes. Cell Mol Biol Lett 2024; 29:53. [PMID: 38616283 PMCID: PMC11017617 DOI: 10.1186/s11658-024-00570-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 03/27/2024] [Indexed: 04/16/2024] Open
Abstract
Reactive oxygen species (ROS) serve as typical metabolic byproducts of aerobic life and play a pivotal role in redox reactions and signal transduction pathways. Contingent upon their concentration, ROS production not only initiates or stimulates tumorigenesis but also causes oxidative stress (OS) and triggers cellular apoptosis. Mounting literature supports the view that ROS are closely interwoven with the pathogenesis of a cluster of diseases, particularly those involving cell proliferation and differentiation, such as myelodysplastic syndromes (MDS) and chronic/acute myeloid leukemia (CML/AML). OS caused by excessive ROS at physiological levels is likely to affect the functions of hematopoietic stem cells, such as cell growth and self-renewal, which may contribute to defective hematopoiesis. We review herein the eminent role of ROS in the hematological niche and their profound influence on the progress of MDS. We also highlight that targeting ROS is a practical and reliable tactic for MDS therapy.
Collapse
Affiliation(s)
- Qiangan Jing
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- HEALTH BioMed Research & Development Center, Health BioMed Co., Ltd, Ningbo, 315803, Zhejiang, China
| | - Chaoting Zhou
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Junyu Zhang
- Department of Hematology, Lishui Central Hospital, Lishui, 323000, Zhejiang, China
| | - Ping Zhang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Yunyi Wu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Junyu Zhou
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Xiangmin Tong
- Department of Central Laboratory, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, Zhejiang, China
| | - Yanchun Li
- Department of Central Laboratory, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, Zhejiang, China.
| | - Jing Du
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
| | - Ying Wang
- Department of Central Laboratory, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, Zhejiang, China.
| |
Collapse
|
30
|
Ramos-González EJ, Bitzer-Quintero OK, Ortiz G, Hernández-Cruz JJ, Ramírez-Jirano LJ. Relationship between inflammation and oxidative stress and its effect on multiple sclerosis. Neurologia 2024; 39:292-301. [PMID: 38553104 DOI: 10.1016/j.nrleng.2021.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 10/28/2021] [Indexed: 04/02/2024] Open
Abstract
INTRODUCTION This paper highlights the relationship of inflammation and oxidative stress as damage mechanisms of Multiple Sclerosis (MS), considered an inflammatory and autoimmune disease. DEVELOPMENT The oxidative stress concept has been defined by an imbalance between oxidants and antioxidants in favor of the oxidants. There is necessary to do physiological functions, like the respiration chain, but in certain conditions, the production of reactive species overpassed the antioxidant systems, which could cause tissue damage. On the other hand, it is well established that inflammation is a complex reaction in the vascularized connective tissue in response to diverse stimuli. However, an unregulated prolonged inflammatory process also can induce tissue damage. CONCLUSION Both inflammation and oxidative stress are interrelated since one could promote the other, leading to a toxic feedback system, which contributes to the inflammatory and demyelination process in MS.
Collapse
Affiliation(s)
- E J Ramos-González
- Unidad de Investigacion Biomedica de Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas, Zacatecas, Mexico
| | - O K Bitzer-Quintero
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, Mexico
| | - G Ortiz
- Departamento de Diciplinas Metodológicas y Filosóficas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - J J Hernández-Cruz
- Departamento de Diciplinas Metodológicas y Filosóficas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - L J Ramírez-Jirano
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, Mexico.
| |
Collapse
|
31
|
Eker F, Akdaşçi E, Duman H, Yalçıntaş YM, Canbolat AA, Kalkan AE, Karav S, Šamec D. Antimicrobial Properties of Colostrum and Milk. Antibiotics (Basel) 2024; 13:251. [PMID: 38534686 DOI: 10.3390/antibiotics13030251] [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: 01/31/2024] [Revised: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 03/28/2024] Open
Abstract
The growing number of antibiotic resistance genes is putting a strain on the ecosystem and harming human health. In addition, consumers have developed a cautious attitude towards chemical preservatives. Colostrum and milk are excellent sources of antibacterial components that help to strengthen the immunity of the offspring and accelerate the maturation of the immune system. It is possible to study these important defenses of milk and colostrum, such as lactoferrin, lysozyme, immunoglobulins, oligosaccharides, etc., as biotherapeutic agents for the prevention and treatment of numerous infections caused by microbes. Each of these components has different mechanisms and interactions in various places. The compound's mechanisms of action determine where the antibacterial activity appears. The activation of the antibacterial activity of milk and colostrum compounds can start in the infant's mouth during lactation and continue in the gastrointestinal regions. These antibacterial properties possess potential for therapeutic uses. In order to discover new perspectives and methods for the treatment of bacterial infections, additional investigations of the mechanisms of action and potential complexes are required.
Collapse
Affiliation(s)
- Furkan Eker
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Turkey
| | - Emir Akdaşçi
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Turkey
| | - Hatice Duman
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Turkey
| | - Yalçın Mert Yalçıntaş
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Turkey
| | - Ahmet Alperen Canbolat
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Turkey
| | - Arda Erkan Kalkan
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Turkey
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Turkey
| | - Dunja Šamec
- Department of Food Technology, University North, Trg Dr. Žarka Dolinara 1, 48000 Koprivnica, Croatia
| |
Collapse
|
32
|
Morin CR, Baeva ME, Hollenberg MD, Brain MC. Milk and multiple sclerosis: A possible link? Mult Scler Relat Disord 2024; 83:105477. [PMID: 38308914 DOI: 10.1016/j.msard.2024.105477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 01/07/2024] [Accepted: 01/26/2024] [Indexed: 02/05/2024]
Abstract
Despite having been formally defined over 150 years ago, the etiology of multiple sclerosis (MS) is still relatively unknown. However, it is now recognized as a multifactorial disease in which genetics, infection, immune function, and environment play a role. We propose an additional piece to the puzzle: milk. In this review, milk is highlighted as a potential risk factor for MS. We examine the overall correlation between bovine milk consumption and the incidence of MS. We then discuss possible mechanisms that may explain the positive association between milk consumption and the development of MS. For instance, butyrophilin (BTN), a milk glycoprotein, can provide molecular mimicry of myelin oligodendrocyte glycoprotein and induce an autoinflammatory response against myelin. Other milk components such as casein, gangliosides, xanthine oxidase, and saturated fats are also analyzed for their potential involvement in the pathophysiology of MS. Finally, we fit milk alongside other well known risk factors of MS: vitamin D levels, Epstein Barr virus infection, and gut dysbiosis. In conclusion, this review summarizes potential mechanisms linking milk as an underappreciated potential risk factor for the development of MS.
Collapse
Affiliation(s)
- Caleb R Morin
- University of Calgary Cumming School of Medicine, Calgary, AB T2N 4N1, Canada
| | | | - Morley D Hollenberg
- Department of Physiology & Pharmacology, University of Calgary Cumming School of Medicine, Calgary, AB T2N 4N1, Canada
| | - Michael C Brain
- Department of Biochemistry and Molecular Biology, University of Calgary Cumming School of Medicine, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
33
|
Mugundhan V, Arthanari A, Parthasarathy PR. Protective Effect of Ferulic Acid on Acetylcholinesterase and Amyloid Beta Peptide Plaque Formation in Alzheimer's Disease: An In Vitro Study. Cureus 2024; 16:e54103. [PMID: 38487137 PMCID: PMC10938272 DOI: 10.7759/cureus.54103] [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: 10/06/2023] [Accepted: 02/13/2024] [Indexed: 03/17/2024] Open
Abstract
Aim This study aims to comprehensively evaluate the effects of ferulic acid (FA) on acetylcholinesterase (AChE) enzyme activity and amyloid beta (Aβ) peptide plaque formation in an in vitro model of Alzheimer's disease (AD). Background AD is a progressive neurological condition marked by disrupted cholinergic signaling, accumulation of Aβ peptide, and tau protein hyperphosphorylation. Currently, no direct anti-Alzheimer drug that effectively prevents the cognitive decline from AD has been reported. To combat this, a multi-target drug addressing several molecular aspects would be ideal for AD. Natural compounds are preferred over synthetic drugs due to their accessibility, cost-efficiency, and lower toxicity The proven association between polyphenol consumption and the prevention of AD has led to the investigation of the effect of FA, a polyphenolic compound, on acetylcholinesterase enzyme activity and Aβ peptide formation, the key targets of AD. Materials and method The free radical scavenging ability of FA was assessed by xanthine oxidase inhibitory activity. Furthermore, FA was also evaluated for its inhibitory activity against AChE enzyme and amyloid beta peptide formation to evaluate the neuroprotective potential of FA. Results The results showed that FA has the potential to be an AChE inhibitor, thus helping in blocking the activity of AChE and also reducing the incidence of amyloid beta plaque formation. Furthermore, the compound also exhibited a significant antioxidant property which was demonstrated by the xanthine oxidase enzyme inhibitory effect. Conclusion From the observed results, FA has significant antioxidant and neuroprotective effects which are compared with those of their respective standards. More research is required to determine the efficacy and safety of this compound as a treatment for neurodegenerative diseases like AD because the precise mechanism and degree of its AChE inhibitory effects in the brain are still elusive. A potent, selective, and effective drug is desperately needed to treat patients with AD and those at risk of developing the disease.
Collapse
Affiliation(s)
- Varsha Mugundhan
- Department of Forensic Odontology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Abirami Arthanari
- Department of Forensic Odontology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Parameswari R Parthasarathy
- Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| |
Collapse
|
34
|
Najjar RS, Roy RK, Stern JE, Feresin RG. Raspberry polyphenols target molecular pathways of heart failure. J Nutr Biochem 2024; 124:109535. [PMID: 37984734 DOI: 10.1016/j.jnutbio.2023.109535] [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: 06/01/2023] [Revised: 11/03/2023] [Accepted: 11/15/2023] [Indexed: 11/22/2023]
Abstract
Approximately 650,000 new cases of heart failure (HF) are diagnosed annually with a 50% five-year mortality rate. HF is characterized by reduced left ventricular (LV) ejection fraction and hypertrophy of the LV wall. The pathophysiological remodeling of the heart is mediated by increased oxidative stress and inflammation. Raspberries are rich in polyphenols which may favorably impact enzymes involved in redox homeostasis while also targeting inflammatory signaling. Thus, the objective of this study was to investigate whether raspberry polyphenols could attenuate HF. Sprague Dawley rats consumed a 10% (w/w) raspberry diet for 7 weeks. At week 3, HF was surgically induced via coronary artery ligation. Hemodynamics and morphology of the heart were assessed. Expression of cardiac proteins involved in oxidative stress, inflammation, apoptosis, and remodeling were examined, and histological analysis was conducted. Additionally, human cardiomyocytes were treated with raspberry polyphenol extract (RBPE) followed by CoCl2 to chemically induce hypoxia. Redox status, apoptosis, and mitochondrial dysfunction were measured. Raspberries attenuated reductions in cardiac function and reduced morphological changes which coincided with reduced toll-like receptor (TLR)4 signaling. Reductions in oxidative stress, apoptosis, and remodeling occurred in vivo. Incubation of cardiomyocytes with RBPE attenuated CoCl2-induced oxidative stress and apoptosis despite pronounced hypoxia-inducible factor (HIF)-1α expression. These data indicate that consumption of raspberries can reduce the underlying molecular drivers of HF; thus, leading to the observed improvements in cardiac functional capacity and morphology. This dietary strategy may be an effective alternative strategy for treating HF. However, further investigation into alternative models of HF is warranted.
Collapse
Affiliation(s)
- Rami S Najjar
- Department of Nutrition, Georgia State University, Atlanta, Georgia, USA; Department of Chemistry, Georgia State University, Atlanta, Georgia, USA
| | - Ranjan K Roy
- Center for Neuroinflammation and Cardiometabolic Diseases, Department of Neuroscience, Georgia State University, Atlanta, Georgia, USA
| | - Javier E Stern
- Center for Neuroinflammation and Cardiometabolic Diseases, Department of Neuroscience, Georgia State University, Atlanta, Georgia, USA
| | - Rafaela G Feresin
- Department of Nutrition, Georgia State University, Atlanta, Georgia, USA; Department of Chemistry, Georgia State University, Atlanta, Georgia, USA; Center for Neuroinflammation and Cardiometabolic Diseases, Department of Neuroscience, Georgia State University, Atlanta, Georgia, USA.
| |
Collapse
|
35
|
Gwozdzinski L, Pieniazek A, Gwozdzinski K. Factors Influencing Venous Remodeling in the Development of Varicose Veins of the Lower Limbs. Int J Mol Sci 2024; 25:1560. [PMID: 38338837 PMCID: PMC10855638 DOI: 10.3390/ijms25031560] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
One of the early symptoms of chronic venous disease (CVD) is varicose veins (VV) of the lower limbs. There are many etiological environmental factors influencing the development of chronic venous insufficiency (CVI), although genetic factors and family history of the disease play a key role. All these factors induce changes in the hemodynamic in the venous system of the lower limbs leading to blood stasis, hypoxia, inflammation, oxidative stress, proteolytic activity of matrix metalloproteinases (MMPs), changes in microcirculation and, consequently, the remodeling of the venous wall. The aim of this review is to present current knowledge on CVD, including the pathophysiology and mechanisms related to vein wall remodeling. Particular emphasis has been placed on describing the role of inflammation and oxidative stress and the involvement of extracellular hemoglobin as pathogenetic factors of VV. Additionally, active substances used in the treatment of VV were discussed.
Collapse
Affiliation(s)
- Lukasz Gwozdzinski
- Department of Pharmacology and Toxicology, Medical University of Lodz, 90-752 Lodz, Poland;
| | - Anna Pieniazek
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland;
| | - Krzysztof Gwozdzinski
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland;
| |
Collapse
|
36
|
Mudgal R, Singh S. Xanthine Oxidoreductase in the Pathogenesis of Endothelial Dysfunction: An Update. Curr Hypertens Rev 2024; 20:10-22. [PMID: 38318826 DOI: 10.2174/0115734021277772240124075120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 12/04/2023] [Accepted: 12/19/2023] [Indexed: 02/07/2024]
Abstract
Xanthine oxidoreductase (XOR) is a rate-limiting enzyme in the formation of uric acid (UA) and is involved in the generation of reactive oxygen species (ROS). Overproduction of ROS has been linked to the pathogenesis of hypertension, atherosclerosis, and cardiovascular disease, with multiple studies over the last 30 years demonstrating that XOR inhibition is beneficial. The involvement of XOR and its constituents in the advancement of chronic inflammation and ROS, which are responsible for endothelial dysfunction, is the focus of this evidence-based review. An overabundance of XOR products and ROS appears to drive the inflammatory response, resulting in significant endothelium damage. It has also been demonstrated that XOR activity and ED are connected. Diabetes, hypertension, and cardiovascular disease are all associated with endothelial dysfunction. ROS mainly modifies the activity of vascular cells and can be important in normal vascular physiology as well as the development of vascular disease. Suppressing XOR activity appears to decrease endothelial dysfunction, probably because it lessens the generation of reactive oxygen species and the oxidative stress brought on by XOR. Although there has long been a link between higher vascular XOR activity and worse clinical outcomes, new research suggests a different picture in which positive results are mediated by XOR enzymatic activity. Here in this study, we aimed to review the association between XOR and vascular endothelial dysfunction. The prevention and treatment approaches against vascular endothelial dysfunction in atherosclerotic disease.
Collapse
Affiliation(s)
- Rajat Mudgal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Bihar, India
| | - Sanjiv Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Bihar, India
| |
Collapse
|
37
|
Amir-Ata JS, Mohammad-Reza V, Malekinejad H. The Benzene-induced Hepatic Cytochrome P450 2E1 Expression and Activity are Reduced by Quercetin Administration in Mice. Curr Pharm Des 2024; 30:676-682. [PMID: 38424425 DOI: 10.2174/0113816128285832240216120053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/20/2024] [Accepted: 01/26/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Benzene as an environmental and industrial agent induces adverse effects that are mainly metabolism-dependent. OBJECTIVES Effects of Quercetin (QCN) on Benzene (BNZ)-induced changes in the hepatic Cytochrome P450 2E1 expression and activity were investigated. METHODS Thirty-six adult male mice were divided into 6 groups (n = 6) and nominated as control, BNZ (exposed to BNZ: 30 ppm), QCN (received QCN: 50 mg/kg, orally), and the fourth, fifth and sixth groups were exposed to 30 ppm BNZ and received 10, 50 and 100 mg/kg QCN respectively, for 28 days. The microsomal subcellular fraction was isolated from the liver samples and the activity of CYP 2E1 was measured based on the hydroxylation rate of 4-nitrophenol. The hepatic activity of myeloperoxidase also was assessed. Total antioxidant capacity and nitric oxide contents of the liver were determined. Expression changes of CYP 2E1 at the mRNA level were examined by qPCR technique. RESULTS QCN lowered significantly (p < 0.05) the BNZ-increased hepatic nitric oxide levels and restored the BNZ-reduced antioxidant capacity. The BNZ-elevated activity of myeloperoxidase was declined in QCN-received mice. QCN downregulated the expression and activity of hepatic CYP 2E1 in BNZ-exposed animals. CONCLUSION Our results suggest that QCN could be a novel hepatoprotective compound for BNZ-induced hepatotoxicities, which is attributed to its capability in the down-regulation of CYP 2E1 expression and activity.
Collapse
Affiliation(s)
- Jambour-Shabestary Amir-Ata
- Department of Pharmacology & Toxicology, Experimental and Applied Pharmaceutical Sciences Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Vardast Mohammad-Reza
- Department of Medicinal Chemistry, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Hassan Malekinejad
- Department of Pharmacology & Toxicology, Experimental and Applied Pharmaceutical Sciences Research Center, Urmia University of Medical Sciences, Urmia, Iran
- Department of Pharmacology & Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
38
|
Singh D, Malhotra P, Agarwal P, Kumar R. N-acetyl-l-tryptophan (NAT) ameliorates radiation-induced cell death in murine macrophages J774A.1 via regulating redox homeostasis and mitochondrial dysfunction. J Biochem Mol Toxicol 2024; 38:e23529. [PMID: 37702290 DOI: 10.1002/jbt.23529] [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: 02/21/2023] [Revised: 08/02/2023] [Accepted: 08/25/2023] [Indexed: 09/14/2023]
Abstract
Ionizing radiation interacts with the immune system and induces molecular damage in the cellular milieu by generating reactive oxygen species (ROS) leading to cell death. The present study was performed to investigate the protective efficacy of N-acetyl-L-tryptophan (NAT) against gamma-radiation-induced cell death in murine macrophage J774A.1 cells. The radioprotective efficacy of NAT was evaluated in terms of cell survivability, effect on antioxidant enzyme activity, and free radicals inhibition. Radioprotective efficacy of NAT pretreatment to irradiated cells was assessed via cell cycle progression, mitochondrial membrane potential (MMP) perturbation, and apoptosis regulation using flow cytometry. Results of the study demonstrated significant radioprotective efficacy (>80%) of NAT in irradiated cells as estimated by sulforhodamine B (SRB), MTT, and clonogenic assay. Significant (p < 0.001) reduction in ROS, xanthine oxidase, and mitochondrial superoxide levels along with increment in catalase, glutathione-s-transferase, glutathione, and ATPase activities in NAT pretreated plus irradiated cells was observed as compared to the gamma-irradiated cells. Further, significant (p < 0.001) stabilization of MMP and reduction in apoptosis was also observed in NAT pretreated plus irradiated cells as compared to irradiated cells that not pretreated with NAT. The current study demonstrates that NAT pretreatment to irradiated cells protects against gamma radiation-induced cell death by reducing oxidative stress, stabilizing MMP, and inhibiting apoptosis. These observations conclusively highlight the potential of developing NAT as a prospective radioprotective agent upon further validation using in-depth preclinical assessment in cellular and animal models.
Collapse
Affiliation(s)
- Darshana Singh
- Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Poonam Malhotra
- Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Prerna Agarwal
- Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Raj Kumar
- Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| |
Collapse
|
39
|
Bellanti F, Mangieri D, Vendemiale G. Redox Biology and Liver Fibrosis. Int J Mol Sci 2023; 25:410. [PMID: 38203581 PMCID: PMC10778611 DOI: 10.3390/ijms25010410] [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: 11/24/2023] [Revised: 12/21/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Hepatic fibrosis is a complex process that develops in chronic liver diseases. Even though the initiation and progression of fibrosis rely on the underlying etiology, mutual mechanisms can be recognized and targeted for therapeutic purposes. Irrespective of the primary cause of liver disease, persistent damage to parenchymal cells triggers the overproduction of reactive species, with the consequent disruption of redox balance. Reactive species are important mediators for the homeostasis of both hepatocytes and non-parenchymal liver cells. Indeed, other than acting as cytotoxic agents, reactive species are able to modulate specific signaling pathways that may be relevant to hepatic fibrogenesis. After a brief introduction to redox biology and the mechanisms of fibrogenesis, this review aims to summarize the current evidence of the involvement of redox-dependent pathways in liver fibrosis and focuses on possible therapeutic targets.
Collapse
Affiliation(s)
- Francesco Bellanti
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy;
| | - Domenica Mangieri
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy;
| | - Gianluigi Vendemiale
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy;
| |
Collapse
|
40
|
Lim EY, Lee SY, Shin HS, Kim GD. Reactive Oxygen Species and Strategies for Antioxidant Intervention in Acute Respiratory Distress Syndrome. Antioxidants (Basel) 2023; 12:2016. [PMID: 38001869 PMCID: PMC10669909 DOI: 10.3390/antiox12112016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threatening pulmonary condition characterized by the sudden onset of respiratory failure, pulmonary edema, dysfunction of endothelial and epithelial barriers, and the activation of inflammatory cascades. Despite the increasing number of deaths attributed to ARDS, a comprehensive therapeutic approach for managing patients with ARDS remains elusive. To elucidate the pathological mechanisms underlying ARDS, numerous studies have employed various preclinical models, often utilizing lipopolysaccharide as the ARDS inducer. Accumulating evidence emphasizes the pivotal role of reactive oxygen species (ROS) in the pathophysiology of ARDS. Both preclinical and clinical investigations have asserted the potential of antioxidants in ameliorating ARDS. This review focuses on various sources of ROS, including NADPH oxidase, uncoupled endothelial nitric oxide synthase, cytochrome P450, and xanthine oxidase, and provides a comprehensive overview of their roles in ARDS. Additionally, we discuss the potential of using antioxidants as a strategy for treating ARDS.
Collapse
Affiliation(s)
- Eun Yeong Lim
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea; (E.Y.L.); (S.-Y.L.); (H.S.S.)
| | - So-Young Lee
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea; (E.Y.L.); (S.-Y.L.); (H.S.S.)
- Department of Food Biotechnology, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Hee Soon Shin
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea; (E.Y.L.); (S.-Y.L.); (H.S.S.)
- Department of Food Biotechnology, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Gun-Dong Kim
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea; (E.Y.L.); (S.-Y.L.); (H.S.S.)
| |
Collapse
|
41
|
Siani A, Infante-Teixeira L, d'Arcy R, Roberts IV, El Mohtadi F, Donno R, Tirelli N. Polysulfide nanoparticles inhibit fibroblast-to-myofibroblast transition via extracellular ROS scavenging and have potential anti-fibrotic properties. BIOMATERIALS ADVANCES 2023; 153:213537. [PMID: 37406516 DOI: 10.1016/j.bioadv.2023.213537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 06/06/2023] [Accepted: 06/22/2023] [Indexed: 07/07/2023]
Abstract
This paper is about the effects of reactive oxygen species (ROS) - and of their nanoparticle-mediated extracellular removal - in the TGF-β1-induced differentiation of fibroblasts (human dermal fibroblasts - HDFa) to more contractile myofibroblasts, and in the maintenance of this phenotype. Here, poly(propylene sulfide) (PPS) nanoparticles have been employed on 2D and 3D in vitro models, showing extremely low toxicity and undergoing negligible internalization, thereby ensuring an extracellular-only action. Firstly, PPS nanoparticles abrogated ROS-mediated downstream molecular events such as glutathione oxidation, NF-κB activation, and heme oxidase-1 (HMOX) overexpression. Secondly, PPS nanoparticles were also capable to inhibit, prevent and reverse the TGF-β1-induced upregulation of key biomechanical elements, such as ED-a fibronectin (EF-A FN) and alpha-smooth muscle actin (α-SMA), respectively markers of protomyofibroblastic and of myofibroblastic differentiation. We also confirmed that ROS alone are ineffective promoters of the myofibroblastic transition, although their presence contributes to its stabilization. Finally, the particles also countered TGF-β1-induced matrix- and tissue-level phenomena, e.g., the upregulation of collagen type 1, the development of aberrant collagen type 1/3 ratios and the contracture of HDFa 3D-seeded fibrin constructs. In short, experimental data at molecular, cellular and tissue levels show a significant potential in the use of PPS nanoparticles as anti-fibrotic agents.
Collapse
Affiliation(s)
- Alessandro Siani
- Division of Pharmacy and Optometry, School of Health Sciences, Stopford Building, The University of Manchester, Manchester M13 9PL, UK
| | - Lorena Infante-Teixeira
- Laboratory of Polymers and Biomaterials, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Richard d'Arcy
- Division of Pharmacy and Optometry, School of Health Sciences, Stopford Building, The University of Manchester, Manchester M13 9PL, UK; Laboratory of Polymers and Biomaterials, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy.
| | - Iwan V Roberts
- Division of Pharmacy and Optometry, School of Health Sciences, Stopford Building, The University of Manchester, Manchester M13 9PL, UK
| | - Farah El Mohtadi
- Division of Pharmacy and Optometry, School of Health Sciences, Stopford Building, The University of Manchester, Manchester M13 9PL, UK
| | - Roberto Donno
- Laboratory of Polymers and Biomaterials, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Nicola Tirelli
- Division of Pharmacy and Optometry, School of Health Sciences, Stopford Building, The University of Manchester, Manchester M13 9PL, UK; Laboratory of Polymers and Biomaterials, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy.
| |
Collapse
|
42
|
Seo YS, Park JM, Kim JH, Lee MY. Cigarette Smoke-Induced Reactive Oxygen Species Formation: A Concise Review. Antioxidants (Basel) 2023; 12:1732. [PMID: 37760035 PMCID: PMC10525535 DOI: 10.3390/antiox12091732] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 08/28/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Smoking is recognized as a significant risk factor for numerous disorders, including cardiovascular diseases, respiratory conditions, and various forms of cancer. While the exact pathogenic mechanisms continue to be explored, the induction of oxidative stress via the production of excess reactive oxygen species (ROS) is widely accepted as a primary molecular event that predisposes individuals to these smoking-related ailments. This review focused on how cigarette smoke (CS) promotes ROS formation rather than the pathophysiological repercussions of ROS and oxidative stress. A comprehensive analysis of existing studies revealed the following key ways through which CS imposes ROS burden on biological systems: (1) ROS, as well as radicals, are intrinsically present in CS, (2) CS constituents generate ROS through chemical reactions with biomolecules, (3) CS stimulates cellular ROS sources to enhance production, and (4) CS disrupts the antioxidant system, aggravating the ROS generation and its functions. While the evidence supporting these mechanisms is chiefly based on in vitro and animal studies, the direct clinical relevance remains to be fully elucidated. Nevertheless, this understanding is fundamental for deciphering molecular events leading to oxidative stress and for developing intervention strategies to counter CS-induced oxidative stress.
Collapse
Affiliation(s)
| | | | | | - Moo-Yeol Lee
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Goyang-si 10326, Gyeonggi-do, Republic of Korea; (Y.-S.S.); (J.-M.P.); (J.-H.K.)
| |
Collapse
|
43
|
An Y, Xu BT, Wan SR, Ma XM, Long Y, Xu Y, Jiang ZZ. The role of oxidative stress in diabetes mellitus-induced vascular endothelial dysfunction. Cardiovasc Diabetol 2023; 22:237. [PMID: 37660030 PMCID: PMC10475205 DOI: 10.1186/s12933-023-01965-7] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 08/14/2023] [Indexed: 09/04/2023] Open
Abstract
Diabetes mellitus is a metabolic disease characterized by long-term hyperglycaemia, which leads to microangiopathy and macroangiopathy and ultimately increases the mortality of diabetic patients. Endothelial dysfunction, which has been recognized as a key factor in the pathogenesis of diabetic microangiopathy and macroangiopathy, is characterized by a reduction in NO bioavailability. Oxidative stress, which is the main pathogenic factor in diabetes, is one of the major triggers of endothelial dysfunction through the reduction in NO. In this review, we summarize the four sources of ROS in the diabetic vasculature and the underlying molecular mechanisms by which the pathogenic factors hyperglycaemia, hyperlipidaemia, adipokines and insulin resistance induce oxidative stress in endothelial cells in the context of diabetes. In addition, we discuss oxidative stress-targeted interventions, including hypoglycaemic drugs, antioxidants and lifestyle interventions, and their effects on diabetes-induced endothelial dysfunction. In summary, our review provides comprehensive insight into the roles of oxidative stress in diabetes-induced endothelial dysfunction.
Collapse
Affiliation(s)
- Ying An
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, 646000, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, 646000, China
| | - Bu-Tuo Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, 646000, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, 646000, China
| | - Sheng-Rong Wan
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, 646000, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, 646000, China
| | - Xiu-Mei Ma
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, 646000, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, 646000, China
- Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau, China
| | - Yang Long
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, 646000, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, 646000, China
| | - Yong Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China.
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, 646000, China.
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, 646000, China.
| | - Zong-Zhe Jiang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China.
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, 646000, China.
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, 646000, China.
| |
Collapse
|
44
|
Soliman E, Elshazly SM, Shewaikh SM, El-Shaarawy F. Reno- and hepato-protective effect of allopurinol after renal ischemia/reperfusion injury: Crosstalk between xanthine oxidase and peroxisome proliferator-activated receptor gamma signaling. Food Chem Toxicol 2023; 178:113868. [PMID: 37269893 DOI: 10.1016/j.fct.2023.113868] [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: 01/16/2023] [Revised: 05/21/2023] [Accepted: 05/31/2023] [Indexed: 06/05/2023]
Abstract
Renal ischemia/reperfusion (I/R) is a common cause of acute kidney injury and remote liver damage is an ultimate negative outcome. Current treatments for renal I/R typically involve the use of antioxidants and anti-inflammatory to protect against oxidative stress and inflammation. Xanthine oxidase (XO) and PPAR-γ contribute to renal I/R-induced oxidative stress; however, the crosstalk between the two pathways remains unexplored. In the present study, we report that XO inhibitor, allopurinol (ALP), protects kidney and liver after renal I/R by PPAR-γ activation. Rats with renal I/R showed reduced kidney and liver functions, increased XO, and decreased PPAR-γ. ALP increased PPAR-γ expression and improved liver and kidney functions. ALP also reduced inflammation and nitrosative stress indicated by reduction in TNF-α, iNOS, nitric oxide (NO), and peroxynitrite formation. Interestingly, rats co-treated with PPAR-γ inhibitor, BADGE, and ALP showed diminished beneficial effect on renal and kidney functions, inflammation, and nitrosative stress. This data suggests that downregulation of PPAR-γ contributes to nitrosative stress and inflammation in renal I/R and the use of ALP reverses this effect by increasing PPAR-γ expression. In conclusion, this study highlights the potential therapeutic value of ALP and suggests targeting XO-PPAR-γ pathway as a promising strategy for preventing renal I/R injury.
Collapse
Affiliation(s)
- Eman Soliman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| | - Shimaa Mustafa Elshazly
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| | - Samar M Shewaikh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| | - Fatma El-Shaarawy
- Department of Biochemistry, Faculty of Pharmacy, Sinai University, Arish, 45511, Egypt.
| |
Collapse
|
45
|
Gwozdzinski L, Bernasinska-Slomczewska J, Hikisz P, Wiktorowska-Owczarek A, Kowalczyk E, Pieniazek A. The Effect of Diosmin, Escin, and Bromelain on Human Endothelial Cells Derived from the Umbilical Vein and the Varicose Vein-A Preliminary Study. Biomedicines 2023; 11:1702. [PMID: 37371797 DOI: 10.3390/biomedicines11061702] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 05/29/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
In this study, we investigated the properties of human varicose vein (VV) endothelial cells (HVVEC) in comparison to the human umbilical vein endothelial cells (HUVEC). The cells were treated with three bioactive compounds with proven beneficial effects in the therapy of patients with VV, diosmin, escin, and bromelain. Two concentrations of tested drugs were used (1, 10 mg/mL), which did not affect the viability of either cell type. Escin led to a slight generation of reactive oxygen species in HUVEC cells. We observed a slight release of superoxide in HVVEC cells upon treatment with diosmin and escin. Diosmin and bromelain showed a tendency to release nitric oxide in HUVEC. Using membrane fluorescent probes, we demonstrated a reduced fluidity of HVVEC, which may lead to their increased adhesion, and, consequently, a much more frequent occurrence of venous thrombosis. For the first time, we show the mechanism of action of drugs used in VV therapy on endothelial cells derived from a VV. Studies with HVVEC have shown that tested drugs may lead to a reduction in the adhesive properties of these cells, and thus to a lower risk of thrombosis.
Collapse
Affiliation(s)
- Lukasz Gwozdzinski
- Department of Pharmacology and Toxicology, Medical University of Lodz, 90-752 Lodz, Poland
| | - Joanna Bernasinska-Slomczewska
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland
| | - Pawel Hikisz
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland
| | | | - Edward Kowalczyk
- Department of Pharmacology and Toxicology, Medical University of Lodz, 90-752 Lodz, Poland
| | - Anna Pieniazek
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland
| |
Collapse
|
46
|
Aboukhater D, Morad B, Nasrallah N, Nasser SA, Sahebkar A, Kobeissy F, Boudaka A, Eid AH. Inflammation and hypertension: Underlying mechanisms and emerging understandings. J Cell Physiol 2023; 238:1148-1159. [PMID: 37039489 DOI: 10.1002/jcp.31019] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/24/2023] [Indexed: 04/12/2023]
Abstract
Hypertension remains a major contributor to cardiovascular disease (CVD), a leading cause of global death. One of the major insults that drive increased blood pressure is inflammation. While it is the body's defensive response against some homeostatic imbalances, inflammation, when dysregulated, can be very deleterious. In this review, we highlight and discuss the causative relationship between inflammation and hypertension. We critically discuss how the interplay between inflammation and reactive oxygen species evokes endothelial damage and dysfunction, ultimately leading to narrowing and stiffness of blood vessels. This, along with phenotypic switching of the vascular smooth muscle cells and the abnormal increase in extracellular matrix deposition further exacerbates arterial stiffness and noncompliance. We also discuss how hyperhomocysteinemia and microRNA act as links between inflammation and hypertension. The premises we discuss suggest that the blue-sky scenarios for targeting the underlying mechanisms of hypertension necessitate further research.
Collapse
Affiliation(s)
- Diana Aboukhater
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Bassel Morad
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Nadim Nasrallah
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | | | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Firas Kobeissy
- Department of Neurobiology and Neuroscience, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Ammar Boudaka
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
47
|
Nunes KZ, Scorza FA, Cavalheiro EA, Vassallo DV. Reduction of vascular reactivity in rat aortas following pilocarpine-induced status epilepticus. Clinics (Sao Paulo) 2023; 78:100195. [PMID: 37099815 PMCID: PMC10149400 DOI: 10.1016/j.clinsp.2023.100195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 03/20/2023] [Accepted: 03/28/2023] [Indexed: 04/28/2023] Open
Abstract
OBJECTIVE The authors investigated changes in vascular reactivity in rats following pilocarpine-induced status epilepticus. METHOD Male Wistar rats weighing between 250g and 300g were used. Status epilepticus was induced using 385 mg/kg i.p. pilocarpine. After 40 days the thoracic aorta was dissected and divided into 4 mm rings and the vascular smooth muscle reactivity to phenylephrine was evaluated. RESULTS Epilepsy decreased the contractile responses of the aortic rings to phenylephrine (0.1 nM-300 mM). To investigate if this reduction was induced by increasing NO production with/or hydrogen peroxide L-NAME and Catalase were used. L-NAME (N-nitro-L arginine methyl ester) increased vascular reactivity but the contractile response to phenylephrine increased in the epileptic group. Catalase administration decreased the contractile responses only in the rings of rats with epilepsy. CONCLUSIONS Our findings demonstrated for the first time that epilepsy is capable of causing a reduction of vascular reactivity in rat aortas. These results suggest that vascular reactivity reduction is associated with increased production of Nitric Oxide (NO) as an organic attempt to avoid hypertension produced by excessive sympathetic activation.
Collapse
Affiliation(s)
- Karolini Zuqui Nunes
- Postgraduate Program in Nutrition and Health, Universidade Federal do Espírito Santo, Vitória, ES, Brazil.
| | - Fulvio Alexandre Scorza
- Discipline of Neuroscience, Universidade Federal de São Paulo/Escola Paulista de Medicina, São PauloSP, Brazil
| | - Esper Abrão Cavalheiro
- Discipline of Neuroscience, Universidade Federal de São Paulo/Escola Paulista de Medicina, São PauloSP, Brazil
| | - Dalton Valentim Vassallo
- Graduate Program in Physiological Sciences, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| |
Collapse
|
48
|
Ivanova ON, Krasnov GS, Snezhkina AV, Kudryavtseva AV, Fedorov VS, Zakirova NF, Golikov MV, Kochetkov SN, Bartosch B, Valuev-Elliston VT, Ivanov AV. Transcriptome Analysis of Redox Systems and Polyamine Metabolic Pathway in Hepatoma and Non-Tumor Hepatocyte-like Cells. Biomolecules 2023; 13:714. [PMID: 37189460 PMCID: PMC10136275 DOI: 10.3390/biom13040714] [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: 01/27/2023] [Revised: 04/10/2023] [Accepted: 04/19/2023] [Indexed: 05/17/2023] Open
Abstract
Reactive oxygen species (ROS) play a major role in the regulation of various processes in the cell. The increase in their production is a factor contributing to the development of numerous pathologies, including inflammation, fibrosis, and cancer. Accordingly, the study of ROS production and neutralization, as well as redox-dependent processes and the post-translational modifications of proteins, is warranted. Here, we present a transcriptomic analysis of the gene expression of various redox systems and related metabolic processes, such as polyamine and proline metabolism and the urea cycle in Huh7.5 hepatoma cells and the HepaRG liver progenitor cell line, that are widely used in hepatitis research. In addition, changes in response to the activation of polyamine catabolism that contribute to oxidative stress were studied. In particular, differences in the gene expression of various ROS-producing and ROS-neutralizing proteins, the enzymes of polyamine metabolisms and proline and urea cycles, as well as calcium ion transporters between cell lines, are shown. The data obtained are important for understanding the redox biology of viral hepatitis and elucidating the influence of the laboratory models used.
Collapse
Affiliation(s)
- Olga N. Ivanova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - George S. Krasnov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anastasiya V. Snezhkina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anna V. Kudryavtseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Vyacheslav S. Fedorov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Natalia F. Zakirova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Michail V. Golikov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Sergey N. Kochetkov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Birke Bartosch
- Lyon Cancer Research Center, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, 69008 Lyon, France
| | | | - Alexander V. Ivanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
49
|
Sarkar S, Karmakar S, Basu M, Ghosh P, Ghosh MK. Neurological damages in COVID-19 patients: Mechanisms and preventive interventions. MedComm (Beijing) 2023; 4:e247. [PMID: 37035134 PMCID: PMC10080216 DOI: 10.1002/mco2.247] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 02/14/2023] [Accepted: 03/01/2023] [Indexed: 04/11/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a novel coronavirus, causes coronavirus disease 2019 (COVID-19) which led to neurological damage and increased mortality worldwide in its second and third waves. It is associated with systemic inflammation, myocardial infarction, neurological illness including ischemic strokes (e.g., cardiac and cerebral ischemia), and even death through multi-organ failure. At the early stage, the virus infects the lung epithelial cells and is slowly transmitted to the other organs including the gastrointestinal tract, blood vessels, kidneys, heart, and brain. The neurological effect of the virus is mainly due to hypoxia-driven reactive oxygen species (ROS) and generated cytokine storm. Internalization of SARS-CoV-2 triggers ROS production and modulation of the immunological cascade which ultimately initiates the hypercoagulable state and vascular thrombosis. Suppression of immunological machinery and inhibition of ROS play an important role in neurological disturbances. So, COVID-19 associated damage to the central nervous system, patients need special care to prevent multi-organ failure at later stages of disease progression. Here in this review, we are selectively discussing these issues and possible antioxidant-based prevention therapies for COVID-19-associated neurological damage that leads to multi-organ failure.
Collapse
Affiliation(s)
- Sibani Sarkar
- Division of Cancer Biology and Inflammatory DisorderSignal Transduction in Cancer and Stem Cells LaboratoryCouncil of Scientific and Industrial Research‐Indian Institute of Chemical Biology (CSIR‐IICB)KolkataIndia
| | - Subhajit Karmakar
- Division of Cancer Biology and Inflammatory DisorderSignal Transduction in Cancer and Stem Cells LaboratoryCouncil of Scientific and Industrial Research‐Indian Institute of Chemical Biology (CSIR‐IICB)KolkataIndia
| | - Malini Basu
- Department of MicrobiologyDhruba Chand Halder College, University of CalcuttaDakshin BarasatWBIndia
| | - Pratyasha Ghosh
- Department of EconomicsBethune CollegeUniversity of CalcuttaKolkataIndia
| | - Mrinal K Ghosh
- Division of Cancer Biology and Inflammatory DisorderSignal Transduction in Cancer and Stem Cells LaboratoryCouncil of Scientific and Industrial Research‐Indian Institute of Chemical Biology (CSIR‐IICB)KolkataIndia
| |
Collapse
|
50
|
PPARβ/δ Ligands Regulate Oxidative Status and Inflammatory Response in Inflamed Corpus Luteum-An In Vitro Study. Int J Mol Sci 2023; 24:ijms24054993. [PMID: 36902426 PMCID: PMC10003567 DOI: 10.3390/ijms24054993] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/19/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
Inflammation in the female reproductive system causes serious health problems including infertility. The aim of this study was to determine the in vitro effects of peroxisome proliferator-activated receptor-beta/delta (PPARβ/δ) ligands on the transcriptomic profile of the lipopolysaccharide (LPS)-stimulated pig corpus luteum (CL) in the mid-luteal phase of the estrous cycle using RNA-seq technology. The CL slices were incubated in the presence of LPS or in combination with LPS and the PPARβ/δ agonist-GW0724 (1 μmol/L or 10 μmol/L) or the antagonist-GSK3787 (25 μmol/L). We identified 117 differentially expressed genes after treatment with LPS; 102 and 97 differentially expressed genes after treatment, respectively, with the PPARβ/δ agonist at a concentration of 1 μmol/L or 10 μmol/L, as well as 88 after the treatment with the PPARβ/δ antagonist. In addition, biochemical analyses of oxidative status were performed (total antioxidant capacity and activity of peroxidase, catalase, superoxide dismutase, and glutathione S-transferase). This study revealed that PPARβ/δ agonists regulate genes involved in the inflammatory response in a dose-dependent manner. The results indicate that the lower dose of GW0724 showed an anti-inflammatory character, while the higher dose seems to be pro-inflammatory. We propose that GW0724 should be considered for further research to alleviate chronic inflammation (at the lower dose) or to support the natural immune response against pathogens (at the higher dose) in the inflamed corpus luteum.
Collapse
|