1
|
Shah W, Gong Y, Qiao X, Lu Y, Ding Y, Zhang Z, Gao Y. Exploring Endothelial Cell Dysfunction's Impact on the Brain-Retina Microenvironment Connection: Molecular Mechanisms and Implications. Mol Neurobiol 2025; 62:7484-7505. [PMID: 39904964 DOI: 10.1007/s12035-025-04714-x] [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: 06/24/2024] [Accepted: 01/19/2025] [Indexed: 02/06/2025]
Abstract
The intricate linking between the health of blood vessels and the functioning of neurons has attracted growing attention in the context of disorders that affect the neurological environment. Endothelial cells, forming the blood-brain barrier and blood-retinal barrier, play a fundamental role in maintaining the integrity of the brain-retina microenvironment connection. This review explores the molecular foundations of endothelial cell dysfunction and its implications for the brain-retina interaction. A comprehensive analysis of the complex factors contributing to endothelial dysfunction is presented, including oxidative stress, inflammation, reduced nitric oxide signaling, and disrupted vascular autoregulation. The significance of endothelial dysfunction extends to neurovascular coupling, synaptic plasticity, and trophic support. To our knowledge, there is currently no existing literature review addressing endothelial microvascular dysfunction and its interplay with the brain-retina microenvironment. The review also explains bidirectional communication between the brain and retina, highlighting how compromised endothelial function can disrupt this vital crosstalk and inhibit normal physiological processes. As neurodegenerative diseases frequently exhibit vascular involvement, a deeper comprehension of the interaction between endothelial cells and neural tissue holds promise for innovative therapeutic strategies. By targeting endothelial dysfunction, we may enhance our ability to preserve the intricate dynamics of the brain-retina microenvironment connection and ameliorate the progression of neurological disorders.
Collapse
Affiliation(s)
- Wahid Shah
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, 030001, China
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, 030002, China
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, 030001, China
| | - Yuxing Gong
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, 030001, China
| | - Xin Qiao
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, 030002, China
| | - Yaling Lu
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, 030002, China
| | - Yufei Ding
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, 030002, China
| | - Ziting Zhang
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, 030002, China
| | - Yuan Gao
- Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, 030002, China.
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
2
|
Goel R, Mishra R, Kumar N, Imsong N, Singh N, Gaur P, Ansari MN, Madkhali HA. Hepatoprotective effect of diosmetin against thioacetamide-induced liver injury via modulating Bax/NF-κB/caspase-3/Nrf-2/PPAR ϒ signaling pathway in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04293-7. [PMID: 40418320 DOI: 10.1007/s00210-025-04293-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 05/12/2025] [Indexed: 05/27/2025]
Abstract
Thioacetamide (TAA), an established liver toxic chemical, is used to develop experimental models of liver injury by inducing inflammation, oxidative stress, and apoptosis. The current study investigates the hepatoprotective effect of diosmetin (DSM), a bioflavonoid molecule, with mechanistic investigations using an in vivo TAA-induced liver injury model because there is no precedence. In the current investigation, 30 Wistar rats were randomly assigned to five groups. The rats were given TAA (200 mg/kg) and DSM (50 and 100 mg/kg body weight) for 8 weeks. Liver function biomarkers (ALT, ASP, AST, bilirubin, GGT, CRP, albumin, globulin, and total protein) and inflammatory markers were analyzed on serum, whereas antioxidant levels, histological evaluation, and apoptotic markers were evaluated on liver tissue. The finding of this study indicated that DSM ameliorates TAA-induced increases in ALT, ASP, AST, bilirubin, GGT, CRP, MDA, albumin, globulin, total protein, and antioxidant enzyme activity such as SOD, CAT, and GSH levels. DSM reduced alterations in inflammatory biomarkers (IL-6, IL-1β, TNF-α) and apoptotic markers (Bax, NF-κB, caspase-3, Nrf-2, PPARϒ) in liver tissues. Furthermore, DSM therapy resulted in significant major changes in liver histology, fibrosis, and cell death. The current study's findings demonstrated that DSM significantly decreases hepatotoxicity by enhancing liver structure and function while suppressing oxidative stress, inflammation, and apoptosis.
Collapse
Affiliation(s)
- Radha Goel
- Department of Pharmacology, Lloyd Institute of Management & Technology, Plot No.-11, Knowledge Park-II, Greater Noida, Uttar Pradesh, 201306, India.
| | - Rosaline Mishra
- Department of Pharmacy, Metro College of Health Sciences and Research, Plot No- 41, Knowledge Park-III, Uttar Pradesh, 201306, Noida, India
| | - Nitin Kumar
- Department of Pharmacy, Meerut Institute of Technology, Meerut, India
| | - Nungsangtula Imsong
- Department of Pharmacology, Lloyd Institute of Management & Technology, Plot No.-11, Knowledge Park-II, Greater Noida, Uttar Pradesh, 201306, India
| | - Neelam Singh
- HR Institute of Pharmacy, HRIT University, 8th Km Stone, Delhi-Meerut Road, Ghaziabad, 201003, India
| | - Praveen Gaur
- Department of Pharmacy, Metro College of Health Sciences and Research, Plot No- 41, Knowledge Park-III, Uttar Pradesh, 201306, Noida, India
| | - Mohd Nazam Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Hassan A Madkhali
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| |
Collapse
|
3
|
Quarta S, Calabriso N, Carluccio MA, Albano C, Khalifa I, Wabitsch M, Blando F, Massaro M. Shielding Human Adipocytes From Inflammation: The Protective Potential of Polyphenol-Rich Opuntia ficus-indica Cladode Extract. Mol Nutr Food Res 2025:e70114. [PMID: 40377300 DOI: 10.1002/mnfr.70114] [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: 02/05/2025] [Revised: 03/28/2025] [Accepted: 04/29/2025] [Indexed: 05/18/2025]
Abstract
Opuntia ficus-indica (OFI) has attracted much attention as a source of antioxidant and antiinflammatory compounds. We hypothesize that the antioxidant content of OFI cladode extract may improve adipocyte dysfunction resulting from inflammatory stimulation of hypertrophic adipocytes. To this end, the properties of OFI cladode hydroalcoholic extract were evaluated in terms of antioxidant activity, regulation of adipocyte inflammation, and adipocyte/monocyte interaction in human adipocytes rendered dysfunctional by the proinflammatory cytokine tumor necrosis factor-α (TNF-α). The major phenolic compounds identified were isorhamnetin derivatives and phenolic acids, including piscidic and eucomic acids. Our results show that OFI cladode extract exhibits antiradical activities and reduces the adhesion and transmigration activity of monocytes to inflamed adipocytes by inhibiting various cytokines, chemokines, and adhesion molecules such as interleukin (IL)-6 and IL-8 by ∼80%, monocyte chemotactic protein (MCP)-1, C-X-C motif chemokine ligand (CXC-L)10, macrophage colony-stimulating factor (M-CSF) from 40% to 50%, and intercellular adhesion molecule-1 (ICAM-1) by 70% at the higher concentration. In structurally and mechanistically by protein-ligand docking profiling study, piscidic acid proved to be the best potential candidate for a regulatory interaction with the activities of nuclear factor erythroid 2-related factor 2 (NRF-2) and nuclear factor-κB (NF-κB). In summary, these data highlight the potential of OFI as a dietary supplement in nutritional treatments aimed at combating the inflammatory stigmata of obesity.
Collapse
Affiliation(s)
- Stefano Quarta
- National Research Council (CNR) Institute of Clinical Physiology, Lecce, Italy
| | - Nadia Calabriso
- National Research Council (CNR) Institute of Clinical Physiology, Lecce, Italy
| | | | - Clara Albano
- National Research Council (CNR) Institute of Sciences of Food Production (ISPA), CNR, Lecce, Italy
| | - Ibrahim Khalifa
- Food Technology Department, Faculty of Agriculture, Benha University, Moshtohor, Egypt
| | - Martin Wabitsch
- Division of Pediatric Endocrinology, Diabetes and Obesity, Department of Pediatrics and Adolescent Medicine, University of Ulm, Ulm, Germany
| | - Federica Blando
- National Research Council (CNR) Institute of Sciences of Food Production (ISPA), CNR, Lecce, Italy
| | - Marika Massaro
- National Research Council (CNR) Institute of Clinical Physiology, Lecce, Italy
| |
Collapse
|
4
|
Aslam H, Albaqami F, Rehman NU, Shah FA. Carvacrol attenuated myocardial infarction through NLRP3-mediated pyroptosis and mTOR/Nrf2/PPARγ-dependent autophagic signaling. Toxicol Appl Pharmacol 2025; 498:117281. [PMID: 40064378 DOI: 10.1016/j.taap.2025.117281] [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: 08/21/2024] [Revised: 02/03/2025] [Accepted: 02/24/2025] [Indexed: 03/22/2025]
Abstract
Myocardial ischemia, also known as myocardial infarction or heart attack, is a significant global health issue and a leading cause of mortality worldwide. The present study focuses on investigating the cardioprotective role of carvacrol on three specific pathways: nuclear factor E2-related factor (Nrf2) / peroxisome proliferator-activated receptor factor (PPARγ)-coupled anti-inflammatory response, inflammasome (NLRP3)-mediated pyroptosis, and mammalian target of rapamycin (mTOR)-dependent autophagic signaling. Male Sprague Dawley rats were divided into three experimental cohorts to determine the best dose for carvacrol (20 mg / kg, 50 mg / kg, and 80 mg/ kg) and the optimum treatment strategy. Our findings showed that isoproterenol raised the production of ROS, induced NLRP3-mediated pyroptosis, and modulated the mTOR-linked signaling cascade. Treatment with carvacrol activated the Nrf2 / HO-1 and PI3K / AKT signaling pathways that led to the reversal of NLRP3 inflammasome. Moreover, the Nrf2 inhibitor all-trans-retinoic acid (ATRA) antagonizes the protective effects of carvacrol and exacerbates myocardial infarction by inducing inflammatory mediators. Taken together, our findings suggest that carvacrol mitigated isoproterenol-induced myocardial ischemia, partially through the activation of Nrf2 and PPARγ and downregulation of NLRP3.
Collapse
Affiliation(s)
- Hina Aslam
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Faisal Albaqami
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Najeeb Ur Rehman
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia.
| | - Fawad Ali Shah
- Swat College of Pharmaceutical Sciences (SCPS), Gogdara, Swat, Khyber Pakhtunkhwa, Pakistan
| |
Collapse
|
5
|
Rabbaa S, Bouchab H, Laaziouez Y, Limami Y, Nasser B, Andreoletti P, Cherkaoui-Malki M, El Kebbaj R. Argan Oil: A Natural Bioactive Lipid Modulating Oxidative Stress and Inflammation. Antioxidants (Basel) 2025; 14:515. [PMID: 40427397 DOI: 10.3390/antiox14050515] [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: 03/23/2025] [Revised: 04/14/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025] Open
Abstract
Nutrition is a key determinant in modulating disease risk, with numerous studies highlighting the role of antioxidants and lipids, particularly the micronutrient and fatty acid composition of natural oils and their bioactive metabolites. In this context, argan oil-a vegetable oil extracted from the kernels of Argania spinosa-has gained significant attention due to its well-balanced fatty acid profile, rich in oleic and linoleic acids, and its high levels of antioxidant compounds, including tocopherols, polyphenols, and phytosterols, particularly schottenol and spinasterol. Thanks to its unique composition, argan oil exhibits protective properties against harmful biological processes, such as oxidative stress and inflammation, both of which play a significant role in various diseases. Preclinical studies, both in vitro and in vivo, have demonstrated that argan oil reduces oxidative stress by preventing DNA damage, protein carbonylation, and lipid peroxidation, while simultaneously increasing antioxidant defenses. Furthermore, it modulates inflammatory responses by decreasing pro-inflammatory biomarkers, increasing anti-inflammatory markers, and limiting immune cell infiltration across different tissues. These antioxidant and anti-inflammatory effects are thought to be linked to argan oil's ability to regulate key signaling pathways, such as Nrf-2 and NF-κB. Although direct evidence remains limited, studies suggest that its main bioactive components-fatty acid, γ-tocopherol, ferulic acid, and campesterol-can influence these pathways, either by activating Nrf2 to boost antioxidant defenses or by inhibiting NF-κB to suppress inflammation. This review explores the antioxidant and anti-inflammatory properties of argan oil, drawing insights from a range of scientific studies to better understand its therapeutic potential.
Collapse
Affiliation(s)
- Soufiane Rabbaa
- Sciences and Engineering of Biomedicals, Biophysics and Health Laboratory, Higher Institute of Health Sciences, Hassan First University, Settat 26000, Morocco
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne Europe, 21000 Dijon, France
| | - Habiba Bouchab
- Sciences and Engineering of Biomedicals, Biophysics and Health Laboratory, Higher Institute of Health Sciences, Hassan First University, Settat 26000, Morocco
- Higher Institute of Nursing Professions and Technical Health (ISPITS), Errachidia 52000, Morocco
| | - Yassir Laaziouez
- Sciences and Engineering of Biomedicals, Biophysics and Health Laboratory, Higher Institute of Health Sciences, Hassan First University, Settat 26000, Morocco
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne Europe, 21000 Dijon, France
| | - Youness Limami
- Sciences and Engineering of Biomedicals, Biophysics and Health Laboratory, Higher Institute of Health Sciences, Hassan First University, Settat 26000, Morocco
| | - Boubker Nasser
- Laboratory of Biochemistry, Neurosciences, Natural Resources and Environment, Faculty of Science and Technology, Hassan First University, Settat 26000, Morocco
| | - Pierre Andreoletti
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne Europe, 21000 Dijon, France
| | - Mustapha Cherkaoui-Malki
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne Europe, 21000 Dijon, France
| | - Riad El Kebbaj
- Sciences and Engineering of Biomedicals, Biophysics and Health Laboratory, Higher Institute of Health Sciences, Hassan First University, Settat 26000, Morocco
| |
Collapse
|
6
|
Bessetti RN, Cobb M, Lilley RM, Johnson NZ, Perez DA, Koonce VM, McCoy K, Litwa KA. Sulforaphane protects developing neural networks from VPA-induced synaptic alterations. Mol Psychiatry 2025:10.1038/s41380-025-02967-5. [PMID: 40175519 DOI: 10.1038/s41380-025-02967-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 02/13/2025] [Accepted: 03/20/2025] [Indexed: 04/04/2025]
Abstract
Prenatal brain development is particularly sensitive to chemicals that can disrupt synapse formation and cause neurodevelopmental disorders. In most cases, such chemicals increase cellular oxidative stress. For example, prenatal exposure to the anti-epileptic drug valproic acid (VPA), induces oxidative stress and synaptic alterations, promoting autism spectrum disorders (ASD) in humans and autism-like behaviors in rodents. Using VPA to model chemically induced ASD, we tested whether activation of cellular mechanisms that increase antioxidant gene expression would be sufficient to prevent VPA-induced synaptic alterations. As a master regulator of cellular defense pathways, the transcription factor nuclear factor erythroid 2-related factor 2 (NRF2) promotes expression of detoxification enzymes and antioxidant gene products. To increase NRF2 activity, we used the phytochemical and potent NRF2 activator, sulforaphane (SFN). In our models of human neurodevelopment, SFN activated NRF2, increasing expression of antioxidant genes and preventing oxidative stress. SFN also enhanced expression of genes associated with synapse formation. Consistent with these gene expression profiles, SFN protected developing neural networks from VPA-induced reductions in synapse formation. Furthermore, in mouse cortical neurons, SFN rescued VPA-induced reductions in neural activity. These results demonstrate the ability of SFN to protect developing neural networks during the vulnerable period of synapse formation, while also identifying molecular signatures of SFN-mediated neuroprotection that could be relevant for combatting other environmental toxicants.
Collapse
Affiliation(s)
- Riley N Bessetti
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University (ECU), Greenville, NC, USA
- East Carolina Diabetes and Obesity Institute at ECU, Greenville, NC, USA
| | - Michelle Cobb
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University (ECU), Greenville, NC, USA
- East Carolina Diabetes and Obesity Institute at ECU, Greenville, NC, USA
| | - Rosario M Lilley
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University (ECU), Greenville, NC, USA
- East Carolina Diabetes and Obesity Institute at ECU, Greenville, NC, USA
| | - Noah Z Johnson
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University (ECU), Greenville, NC, USA
- East Carolina Diabetes and Obesity Institute at ECU, Greenville, NC, USA
| | - Daisy A Perez
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University (ECU), Greenville, NC, USA
- East Carolina Diabetes and Obesity Institute at ECU, Greenville, NC, USA
| | - Virginia M Koonce
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University (ECU), Greenville, NC, USA
| | | | - Karen A Litwa
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University (ECU), Greenville, NC, USA.
- East Carolina Diabetes and Obesity Institute at ECU, Greenville, NC, USA.
| |
Collapse
|
7
|
Padhan P, Simran, Kumar N, Verma S. Glutathione S-transferase: A keystone in Parkinson's disease pathogenesis and therapy. Mol Cell Neurosci 2025; 132:103981. [PMID: 39644945 DOI: 10.1016/j.mcn.2024.103981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/01/2024] [Accepted: 11/30/2024] [Indexed: 12/09/2024] Open
Abstract
Parkinson's disease is a progressive neurodegenerative disorder that predominantly affects motor function due to the loss of dopaminergic neurons in the substantia nigra. It presents significant challenges, impacting millions worldwide with symptoms such as tremors, rigidity, bradykinesia, and postural instability, leading to decreased quality of life and increased morbidity. The pathogenesis of Parkinson's disease is multifaceted, involving complex interactions between genetic susceptibility, environmental factors, and aging, with oxidative stress playing a central role in neuronal degeneration. Glutathione S-Transferase enzymes are critical in the cellular defense mechanism against oxidative stress, catalysing the conjugation of the antioxidant glutathione to various toxic compounds, thereby facilitating their detoxification. Recent research underscores the importance of Glutathione S-Transferase in the pathophysiology of Parkinson's disease, revealing that genetic polymorphisms in Glutathione S-Transferase genes influence the risk and progression of the disease. These genetic variations can affect the enzymatic activity of Glutathione S-Transferase, thereby modulating an individual's capacity to detoxify reactive oxygen species and xenobiotics, which are implicated in Parkinson's disease neuropathological processes. Moreover, biochemical studies have elucidated the role of Glutathione S-Transferase in not only maintaining cellular redox balance but also in modulating various cellular signalling pathways, highlighting its neuroprotective potential. From a therapeutic perspective, targeting Glutathione S-Transferase pathways offers promising avenues for the development of novel treatments aimed at enhancing neuroprotection and mitigating disease progression. This review explores the evident and hypothesized roles of Glutathione S-Transferase in Parkinson's disease, providing a comprehensive overview of its importance and potential as a target for therapeutic intervention.
Collapse
Affiliation(s)
- Pratyush Padhan
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Simran
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Neeraj Kumar
- Department of Reproductive Biology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| | - Sonia Verma
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
8
|
Beigoli S, Kiani S, Asgharzadeh F, Memarzia A, Boskabady MH. Promising role of peroxisome proliferator-activated receptors in respiratory disorders, a review. Drug Metab Rev 2025; 57:26-50. [PMID: 39726246 DOI: 10.1080/03602532.2024.2442012] [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: 02/08/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024]
Abstract
Several studies indicate various pharmacological and therapeutic effects of peroxisome proliferator-activated receptors (PPARs) in different disorders. The current review describes the influences of PPARs on respiratory, allergic, and immunologic diseases. Various databases, including PubMed, Science Direct, and Scopus, were searched regarding the effect of PPARs on respiratory and allergic disorders from 1990 to 2024. The effects of PPARs stimulation on experimental animal models of respiratory diseases such as asthma, chronic obstructive pulmonary diseases (COPD), pulmonary fibrosis (PF), and lung infections were shown. Therapeutic potential mediated through PPARs has also been demonstrated in lung cancer, lung infections, and allergic and immunologic disorders. However, few clinical studies showed PPARs mediated therapeutic effects on asthma and COPD. The PPARs-mediated effects on various respiratory disorders were shown through antioxidant, immunomodulatory, anti-inflammatory, and other mechanisms. Therefore, this review indicated possible remedy effects mediated by these receptors in treating respiratory, allergic, and immunologic diseases. Moreover, this mechanistic review paves the way for researchers to consider further experimental and clinical studies.
Collapse
Affiliation(s)
- Sima Beigoli
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sahar Kiani
- Department of Stem Cell and Developmental Biology, Cell Science Research Center, ROYAN Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Brain and Cognitive Sciences, Cell Science Research Center, ROYAN Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Fereshteh Asgharzadeh
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arghavan Memarzia
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Hossein Boskabady
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
9
|
Collodel G, Moretti E, Marcucci C, Liguori L, Marchini D, Corsaro R, Centini G, Signorini C. PPARγ Expression in Human Spermatozoa and Its Relationship with Seminal F 2-Isoprostanes and Resolvin D1 in the Presence of Varicocele and Urogenital Infections. BIOLOGY 2025; 14:137. [PMID: 40001905 PMCID: PMC11851876 DOI: 10.3390/biology14020137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025]
Abstract
Peroxisome Proliferator-Activated Receptor Gamma (PPARγ) is a regulating agent in antioxidant response also involved in controlling inflammation. The impact of varicocele and urogenital infections on sperm PPARγ expression was studied. The PPARγ gene expression was investigated in spermatozoa of 26 normozoospermic men grouped according to their clinical conditions: normal semen parameters (N), normal semen parameters and varicocele (N + V), and normal semen parameters and urogenital infections (N + UI). Sperm PPARγ expression was correlated with F2-isoprostanes (F2-IsoPs), as markers of lipid peroxidation, and Resolvin D1 (RvD1), a pro-resolving mediator in inflammation. Sperm PPARγ expression was evaluated through comparative real-time PCR, and F2-IsoPs and RvD1 were quantified in the seminal plasma via GC/NCI-MS/MS and immunoassay, respectively. PPARγ expression correlates positively with sperm morphology and vitality and negatively with F2-IsoPs and RvD1. Sperm morphology positively correlates with vitality and negatively with F2-IsoP and RvD1 levels. Despite the normozoospermia in the three examined groups, sperm morphology and PPARγ expression were significantly reduced in N + V and N + UI groups compared to the N group. Additionally, F2-IsoP and RvD1 levels were elevated in N + V and N + UI patients. These data suggest that PPARγ expression is compromised by inflammation and lipoperoxidation, providing new insights to further explore new possibilities of targeted treatment of male infertility.
Collapse
Affiliation(s)
- Giulia Collodel
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (G.C.); (C.M.); (L.L.); (R.C.); (G.C.); (C.S.)
| | - Elena Moretti
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (G.C.); (C.M.); (L.L.); (R.C.); (G.C.); (C.S.)
| | - Caterina Marcucci
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (G.C.); (C.M.); (L.L.); (R.C.); (G.C.); (C.S.)
| | - Laura Liguori
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (G.C.); (C.M.); (L.L.); (R.C.); (G.C.); (C.S.)
| | - Daniela Marchini
- Department of Life Sciences, University of Siena, 53100 Siena, Italy;
| | - Roberta Corsaro
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (G.C.); (C.M.); (L.L.); (R.C.); (G.C.); (C.S.)
| | - Gabriele Centini
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (G.C.); (C.M.); (L.L.); (R.C.); (G.C.); (C.S.)
| | - Cinzia Signorini
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (G.C.); (C.M.); (L.L.); (R.C.); (G.C.); (C.S.)
| |
Collapse
|
10
|
Wei S, Li J, Zhang Y, Li Y, Wang Y. Ferroptosis in eye diseases: a systematic review. Eye (Lond) 2025; 39:18-27. [PMID: 39379520 PMCID: PMC11733247 DOI: 10.1038/s41433-024-03371-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] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 08/09/2024] [Accepted: 09/20/2024] [Indexed: 10/10/2024] Open
Abstract
Ferroptosis is a type of iron-dependent cell death that differs from apoptosis, necroptosis, autophagy, and other forms of cell death. It is mainly characterized by the accumulation of intracellular lipid peroxides, redox imbalance, and reduced levels of glutathione and glutathione peroxidase 4. Studies have demonstrated that ferroptosis plays an important regulatory role in the occurrence and development of neurodegenerative diseases, stroke, traumatic brain injury, and ischemia-reperfusion injuries. Multiple mechanisms, such as iron metabolism, ferritinophagy, p53, and p62/Keap1/Nrf2, as well as the combination of FSP1/CoQ/NADPH and hepcidin/FPN-1 can alter the vulnerability to ferroptosis. Nevertheless, there has been limited research on the development and management of ferroptosis in the realm of eye disorders, with most studies focusing on retinal conditions such as age-related macular degeneration and retinitis pigmentosa. This review offers a thorough examination of the disruption of iron homeostasis in eye disorders, investigating the underlying mechanisms. We anticipate that the occurrence of ferroptotic cell death will not only establish a fresh field of study in eye diseases, but also present a promising therapeutic target for treating these diseases.
Collapse
Affiliation(s)
- Shengsheng Wei
- Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital of Northwest University, Xi'an, China
| | - Jing Li
- Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital of Northwest University, Xi'an, China
| | - Yaohua Zhang
- Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital of Northwest University, Xi'an, China
| | - Yong Li
- Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital of Northwest University, Xi'an, China
| | - Yan Wang
- Tianjin Eye Institute, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Tianjin, China.
- Nankai University Eye Institute, Nankai University, Tianjin, China.
| |
Collapse
|
11
|
Guo J, Yang L, Song H, Bai L. Prevention of bleomycin-induced pulmonary fibrosis by vaccination with the Tocilizumab mimotope. Hum Vaccin Immunother 2024; 20:2319965. [PMID: 38408907 PMCID: PMC10900270 DOI: 10.1080/21645515.2024.2319965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 02/14/2024] [Indexed: 02/28/2024] Open
Abstract
Mimotope, a kind of peptide vaccine, is developed to bind natural receptor and inhibit the downstream signaling. We have demonstrated that the vaccination of Tocilizumab mimotopes could alleviate the renal fibrosis by interfering with both IL-6 and ferroptosis signaling. However, the effect of the vaccination of Tocilizumab mimotopes on the fibroblast was not investigated in previous study. Thus, we sought to explore the changes in the fibroblast induced by the Tocilizumab mimotopes vaccination. Bleomycin instillation was performed to construct the pulmonary fibrosis model after the immunization of Tocilizumab mimotopes. Lung histological analysis showed that the Tocilizumab mimotopes could significantly reduce the maladaptive repairment and abnormal remodeling. Immunoblotting assay and fluorescence staining showed that Immunization with the Tocilizumab mimotopes reduces the accumulation of fibrosis-related proteins. High level of lipid peroxidation product was observed in the animal model, while the Tocilizumab mimotopes vaccination could reduce the generation of lipid peroxidation product. Mechanism analysis further showed that Nrf-2 signaling, but not GPX-4 and FSP-1 signaling, was upregulated, and reduced the lipid peroxidation. Our results revealed that in the BLM-induced pulmonary fibrosis, high level of lipid peroxidation product was significantly accumulation in the lung tissues, which might lead to the occurrence of ferroptosis. The IL-6 pathway block therapy could inhibit lipid peroxidation product generation in the lung tissues by upregulating the Nrf-2 signaling, and further alleviate the pulmonary fibrosis.
Collapse
Affiliation(s)
- Jin Guo
- Department of Cardiorespiratory Rehabilitation, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Lin Yang
- Department of Nephrology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Haoming Song
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Li Bai
- The Central Lab, The First Affiliated Hospital of Baotou Medical College (Inner Mongolia Autoimmune Key Laboratory), Baotou, China
| |
Collapse
|
12
|
Rani N, Arya DS. Modulation of PPAR-γ/Nrf2 and AGE/RAGE signaling contributes to the chrysin cardioprotection against myocardial damage following ischemia/reperfusion in diabetic rats. J Pharm Pharmacol 2024:rgae140. [PMID: 39673242 DOI: 10.1093/jpp/rgae140] [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: 06/13/2024] [Accepted: 10/15/2024] [Indexed: 12/16/2024]
Abstract
OBJECTIVE Advanced glycation end products/receptor for AGEs (AGE/RAGE) signaling has a well-established role in the etiology of diabetic-related cardiovascular disorders. The purpose of the study was to elucidate the role of chrysin, a peroxisome proliferator-activated receptor-γ (PPAR-γ) agonist, against ischemia/reperfusion (IR) injury in diabetic rats and its functional interaction with the AGE/RAGE signaling pathway. METHODS A single intraperitoneal injection of streptozotocin (STZ, 70 mg/kg) was administered to rats for induction of diabetes. Rats having blood glucose levels more than 300 mg/dl following a 72 hr STZ injection were classified as diabetic. PPAR-γ antagonist GW9662 (1 mg/kg, i.p.), chrysin (60 mg/kg, p.o.), or both were administered to diabetic rats for 4 weeks. On the 29th day, rats were given ischemia for 45 min and then reperfusion for 1 hr to induce myocardial infarction (MI). KEY FINDINGS Pretreatment with chrysin significantly improved hemodynamic status, ventricular functions, and cardiac injury markers in diabetic myocardium. Increased PPAR-γ/Nrf2 and decreased RAGE protein expressions were linked to this improvement. Chrysin pretreatment resulted in the upregulation of endogenous antioxidants and reduced TBARS levels. Moreover, chrysin significantly decreased inflammation and apoptosis in diabetic myocardium. CONCLUSION PPAR-γ/Nrf2 co-activation by chrysin ameliorated IR-induced MI in diabetic rats, possibly via modulating AGE/RAGE signaling.
Collapse
Affiliation(s)
- Neha Rani
- Department of Pharmacology, Kalpana Chawla Government Medical College, Karnal, Haryana, 132001, India
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Dharamvir Singh Arya
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India
| |
Collapse
|
13
|
Zheng Y, Yan F, He S, Luo L. Targeting ferroptosis in autoimmune diseases: Mechanisms and therapeutic prospects. Autoimmun Rev 2024; 23:103640. [PMID: 39278299 DOI: 10.1016/j.autrev.2024.103640] [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: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024]
Abstract
Ferroptosis is a form of regulated cell death that relies on iron and exhibits unique characteristics, including disrupted iron balance, reduced antioxidant defenses, and abnormal lipid peroxidation. Recent research suggests that ferroptosis is associated with the onset and progression of autoimmune disorders such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), inflammatory bowel disease (IBD), and multiple sclerosis (MS). However, the precise effects and molecular mechanisms remain incompletely understood. This article presents an overview of how ferroptosis mechanisms contribute to the development and advancement of autoimmune diseases, as well as the involvement of various immune cells in linking ferroptosis to autoimmune conditions. It also explores potential drug targets within the ferroptosis pathway and recent advancements in therapeutic approaches aimed at preventing and treating autoimmune diseases by targeting ferroptosis. Lastly, the article discusses the challenges and opportunities in utilizing ferroptosis as a potential therapeutic avenue for autoimmune disorders.
Collapse
Affiliation(s)
- Yingzi Zheng
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| | - Fangfang Yan
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| | - Shasha He
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Chinese Medicine, Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing, China.
| | - Lianxiang Luo
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, China.
| |
Collapse
|
14
|
Baroi S, Czernik PJ, Khan MP, Letson J, Crowe E, Chougule A, Griffin PR, Rosen CJ, Lecka-Czernik B. PPARG in osteocytes controls cell bioenergetics and systemic energy metabolism independently of sclerostin levels in circulation. Mol Metab 2024; 88:102000. [PMID: 39074536 PMCID: PMC11367276 DOI: 10.1016/j.molmet.2024.102000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 07/31/2024] Open
Abstract
OBJECTIVE The skeleton is one of the largest organs in the body, wherein metabolism is integrated with systemic energy metabolism. However, the bioenergetic programming of osteocytes, the most abundant bone cells coordinating bone metabolism, is not well defined. Here, using a mouse model with partial penetration of an osteocyte-specific PPARG deletion, we demonstrate that PPARG controls osteocyte bioenergetics and their contribution to systemic energy metabolism independently of circulating sclerostin levels, which were previously correlated with metabolic status of extramedullary fat depots. METHODS In vivo and in vitro models of osteocyte-specific PPARG deletion, i.e. Dmp1CrePparγflfl male and female mice (γOTKO) and MLO-Y4 osteocyte-like cells with either siRNA-silenced or CRISPR/Cas9-edited Pparγ. As applicable, the models were analyzed for levels of energy metabolism, glucose metabolism, and metabolic profile of extramedullary adipose tissue, as well as the osteocyte transcriptome, mitochondrial function, bioenergetics, insulin signaling, and oxidative stress. RESULTS Circulating sclerostin levels of γOTKO male and female mice were not different from control mice. Male γOTKO mice exhibited a high energy phenotype characterized by increased respiration, heat production, locomotion and food intake. This high energy phenotype in males did not correlate with "beiging" of peripheral adipose depots. However, both sexes showed a trend for reduced fat mass and apparent insulin resistance without changes in glucose tolerance, which correlated with decreased osteocytic responsiveness to insulin measured by AKT activation. The transcriptome of osteocytes isolated from γOTKO males suggested profound changes in cellular metabolism, fuel transport, mitochondria dysfunction, insulin signaling and increased oxidative stress. In MLO-Y4 osteocytes, PPARG deficiency correlated with highly active mitochondria, increased ATP production, and accumulation of reactive oxygen species (ROS). CONCLUSIONS PPARG in male osteocytes acts as a molecular break on mitochondrial function, and protection against oxidative stress and ROS accumulation. It also regulates osteocyte insulin signaling and fuel usage to produce energy. These data provide insight into the connection between osteocyte bioenergetics and their sex-specific contribution to the balance of systemic energy metabolism. These findings support the concept that the skeleton controls systemic energy expenditure via osteocyte metabolism.
Collapse
Affiliation(s)
- Sudipta Baroi
- Department of Orthopaedic Surgery, University of Toledo, College of Medicine and Life Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA; Center for Diabetes and Endocrine Research, University of Toledo, College of Medicine and Life Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA.
| | - Piotr J Czernik
- Department of Orthopaedic Surgery, University of Toledo, College of Medicine and Life Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA; Center for Diabetes and Endocrine Research, University of Toledo, College of Medicine and Life Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA.
| | - Mohd Parvez Khan
- Department of Orthopaedic Surgery, University of Toledo, College of Medicine and Life Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA; Center for Diabetes and Endocrine Research, University of Toledo, College of Medicine and Life Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA.
| | - Joshua Letson
- Department of Orthopaedic Surgery, University of Toledo, College of Medicine and Life Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA; Center for Diabetes and Endocrine Research, University of Toledo, College of Medicine and Life Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA.
| | - Emily Crowe
- Department of Orthopaedic Surgery, University of Toledo, College of Medicine and Life Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA; Center for Diabetes and Endocrine Research, University of Toledo, College of Medicine and Life Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA.
| | - Amit Chougule
- Department of Orthopaedic Surgery, University of Toledo, College of Medicine and Life Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA; Center for Diabetes and Endocrine Research, University of Toledo, College of Medicine and Life Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA.
| | - Patrick R Griffin
- The Wertheim UF Scripps Institute, University of Florida, Jupiter, FL 33458, USA.
| | | | - Beata Lecka-Czernik
- Department of Orthopaedic Surgery, University of Toledo, College of Medicine and Life Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA; Center for Diabetes and Endocrine Research, University of Toledo, College of Medicine and Life Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA.
| |
Collapse
|
15
|
Senga SS, Bisson WH, Colacci A. Key characteristics of carcinogens meet hallmarks for prevention-cutting the Gordian knot. Front Oncol 2024; 14:1420687. [PMID: 39435286 PMCID: PMC11491790 DOI: 10.3389/fonc.2024.1420687] [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: 04/20/2024] [Accepted: 08/05/2024] [Indexed: 10/23/2024] Open
Abstract
The complexity of cancer requires a comprehensive approach to understand its diverse manifestations and underlying mechanisms. Initially outlined by Hanahan and Weinberg in 2000 and updated in 2010, the hallmarks of cancer provide a conceptual basis for understanding inherent variability in cancer biology. Recent expansions have further elucidated additional hallmarks, including phenotypic plasticity and senescent cells. The International Agency for Research on Cancer (IARC) has identified the key characteristics of carcinogens (KCCs) to evaluate their carcinogenic potential. We analyzed chemicals of concern for environmental exposure that interact with specific receptors to induce genomic instability, epigenetic alterations, immune suppression, and receptor-mediated effects, thereby contributing to chronic inflammation. Despite their varying degrees of carcinogenicity, these chemicals have similar KCC profiles. Our analysis highlights the pivotal role of receptor binding in activating most other KCCs, underscoring their significance in cancer initiation. Although KCCs are associated with early molecular or cellular events, they do not encompass processes directly linked to full cellular malignancy. Thus, there is a need to integrate clear endpoints that anchor KCCs to the acquisition of a complete malignant phenotype into chemical testing. From the perspective of toxicology and cancer research, an all-encompassing strategy that incorporates both existing and novel KCCs and cancer hallmarks is essential to enable the targeted identification of prevalent carcinogens and facilitate zone-specific prevention strategies. To achieve this goal, collaboration between the KCC and cancer hallmarks communities becomes essential.
Collapse
Affiliation(s)
- Sasi S. Senga
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - William H. Bisson
- Integrative Toxicology and Cancer Prevention, Durham, NC, United States
| | - Annamaria Colacci
- Agency for Prevention, Environment and Energy, Emilia-Romagna (Arpae), Bologna, Italy
- Alma Mater Institute on Healthy Planet – University of Bologna, Bologna, Italy
| |
Collapse
|
16
|
Li XC, Zhu XY, Wang YY, Tong SL, Chen ZL, Lu ZY, Zhang JH, Song LL, Wang XH, Zhang C, Sun YH, Zhong CY, Su LH, Wang LX, Huang XY. Canagliflozin alleviates pulmonary hypertension by activating PPARγ and inhibiting its S225 phosphorylation. Acta Pharmacol Sin 2024; 45:1861-1878. [PMID: 38719955 PMCID: PMC11335861 DOI: 10.1038/s41401-024-01286-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/07/2024] [Indexed: 08/22/2024]
Abstract
Pulmonary hypertension (PH) is a progressive fatal disease with no cure. Canagliflozin (CANA), a novel medication for diabetes, has been found to have remarkable cardiovascular benefits. However, few studies have addressed the effect and pharmacological mechanism of CANA in the treatment of PH. Therefore, our study aimed to investigate the effect and pharmacological mechanism of CANA in treating PH. First, CANA suppressed increased pulmonary artery pressure, right ventricular hypertrophy, and vascular remodeling in both mouse and rat PH models. Network pharmacology, transcriptomics, and biological results suggested that CANA could ameliorate PH by suppressing excessive oxidative stress and pulmonary artery smooth muscle cell proliferation partially through the activation of PPARγ. Further studies demonstrated that CANA inhibited phosphorylation of PPARγ at Ser225 (a novel serine phosphorylation site in PPARγ), thereby promoting the nuclear translocation of PPARγ and increasing its ability to resist oxidative stress and proliferation. Taken together, our study not only highlighted the potential pharmacological effect of CANA on PH but also revealed that CANA-induced inhibition of PPARγ Ser225 phosphorylation increases its capacity to counteract oxidative stress and inhibits proliferation. These findings may stimulate further research and encourage future clinical trials exploring the therapeutic potential of CANA in PH treatment.
Collapse
Affiliation(s)
- Xiu-Chun Li
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China
| | - Xia-Yan Zhu
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China
| | - Yang-Yue Wang
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China
| | | | - Zhi-Li Chen
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China
| | - Zi-Yi Lu
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China
| | | | - Lan-Lan Song
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China
| | - Xing-Hong Wang
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China
| | - Chi Zhang
- Wenzhou Medical University, Wenzhou, 325000, China
| | - Yi-Han Sun
- Wenzhou Medical University, Wenzhou, 325000, China
| | | | - Li-Huang Su
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China
| | - Liang-Xing Wang
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China
| | - Xiao-Ying Huang
- Division of Pulmonary Medicine, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China.
| |
Collapse
|
17
|
Huang HC, Shi YJ, Vo TLT, Hsu TH, Song TY. The Anti-Inflammatory Effects and Mechanism of the Submerged Culture of Ophiocordyceps sinensis and Its Possible Active Compounds. J Fungi (Basel) 2024; 10:523. [PMID: 39194849 DOI: 10.3390/jof10080523] [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: 07/04/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024] Open
Abstract
The pharmacological effects of the fruiting body of Ophiocordyceps sinensis (O. sinensis) such as antioxidant, anti-virus, and immunomodulatory activities have already been described, whereas the anti-inflammatory effects and active components of the submerged culture of O. sinesis (SCOS) still need to be further verified. This study aimed to investigate the active compounds in the fermented liquid (FLOS), hot water (WEOS), and 50-95% (EEOS-50, EEOS-95) ethanol extracts of SCOS and their anti-inflammatory effects and potential mechanisms in lipopolysaccharide (LPS)-stimulated microglial BV2 cells. The results demonstrated that all of the SCOS extracts could inhibit NO production in BV2 cells. EEOS-95 exhibited the strongest inhibitory effects (71% inhibitory ability at 500 µg/mL), and its ergosterol, γ-aminobutyric acid (GABA), total phenolic, and total flavonoid contents were significantly higher than those of the other extracts (18.60, 18.60, 2.28, and 2.14 mg/g, p < 0.05, respectively). EEOS-95 also has a strong inhibitory ability against IL-6, IL-1β, and TNF-α with an IC50 of 617, 277, and 507 µg/mL, respectively, which is higher than that of 1 mM melatonin. The anti-inflammatory mechanism of EEOS-95 seems to be associated with the up-regulation of PPAR-γ/Nrf-2/HO-1 antioxidant-related expression and the down-regulation of NF-κB/COX-2/iNOS pro-inflammatory expression signaling. In summary, we demonstrated that EEOS-95 exhibits neuroinflammation-mediated neurodegenerative disorder activities in LPS-induced inflammation in brain microglial cells.
Collapse
Affiliation(s)
- Hsien-Chi Huang
- PhD Program of Biotechnology and Bioindustry, College of Biotechnology and Bioresources, Da-Yeh University, Changhua 515, Taiwan
| | - Yu-Juan Shi
- Department of Medicinal Botanicals and Foods on Health Applications, Da-Yeh University, Changhua 515, Taiwan
| | - Thuy-Lan-Thi Vo
- Department of Medicinal Botanicals and Foods on Health Applications, Da-Yeh University, Changhua 515, Taiwan
| | - Tai-Hao Hsu
- Department of Medicinal Botanicals and Foods on Health Applications, Da-Yeh University, Changhua 515, Taiwan
| | - Tuzz-Ying Song
- Department of Medicinal Botanicals and Foods on Health Applications, Da-Yeh University, Changhua 515, Taiwan
| |
Collapse
|
18
|
Lu CL, Sha JJ, Ma RF, Dong XT, Su XR, Cong B, Wang SJ. Severe Hypothermia Induces Ferroptosis in Cerebral Cortical Nerve Cells. Int J Mol Sci 2024; 25:8086. [PMID: 39125656 PMCID: PMC11311695 DOI: 10.3390/ijms25158086] [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: 06/28/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Abnormal shifts in global climate, leading to extreme weather, significantly threaten the safety of individuals involved in outdoor activities. Hypothermia-induced coma or death frequently occurs in clinical and forensic settings. Despite this, the precise mechanism of central nervous system injury due to hypothermia remains unclear, hindering the development of targeted clinical treatments and specific forensic diagnostic indicators. The GEO database was searched to identify datasets related to hypothermia. Post-bioinformatics analyses, DEGs, and ferroptosis-related DEGs (FerrDEGs) were intersected. GSEA was then conducted to elucidate the functions of the Ferr-related genes. Animal experiments conducted in this study demonstrated that hypothermia, compared to the control treatment, can induce significant alterations in iron death-related genes such as PPARG, SCD, ADIPOQ, SAT1, EGR1, and HMOX1 in cerebral cortex nerve cells. These changes lead to iron ion accumulation, lipid peroxidation, and marked expression of iron death-related proteins. The application of the iron death inhibitor Ferrostatin-1 (Fer-1) effectively modulates the expression of these genes, reduces lipid peroxidation, and improves the expression of iron death-related proteins. Severe hypothermia disrupts the metabolism of cerebral cortex nerve cells, causing significant alterations in ferroptosis-related genes. These genetic changes promote ferroptosis through multiple pathways.
Collapse
Affiliation(s)
| | | | | | | | | | - Bin Cong
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, College of Forensic Medicine, Hebei Medical University, No. 361 Zhong Shan Road, Shijiazhuang 050017, China; (C.-L.L.); (J.-J.S.); (R.-F.M.); (X.-T.D.); (X.-R.S.)
| | - Song-Jun Wang
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, College of Forensic Medicine, Hebei Medical University, No. 361 Zhong Shan Road, Shijiazhuang 050017, China; (C.-L.L.); (J.-J.S.); (R.-F.M.); (X.-T.D.); (X.-R.S.)
| |
Collapse
|
19
|
Kuang S, Sheng W, Meng J, Liu W, Xiao Y, Tang H, Fu X, Kuang M, He Q, Gao S. Pyroptosis-related crosstalk in osteoarthritis: Macrophages, fibroblast-like synoviocytes and chondrocytes. J Orthop Translat 2024; 47:223-234. [PMID: 39040491 PMCID: PMC11262125 DOI: 10.1016/j.jot.2024.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 04/28/2024] [Accepted: 06/20/2024] [Indexed: 07/24/2024] Open
Abstract
The pathogenesis of osteoarthritis (OA) involves a multifaceted interplay of inflammatory processes. The initiation of pyroptosis involves the secretion of pro-inflammatory cytokines and has been identified as a critical factor in regulating the development of OA. Upon initiation of pyroptosis, a multitude of inflammatory mediators are released and can be disseminated throughout the synovial fluid within the joint cavity, thereby facilitating intercellular communication across the entire joint. The main cellular components of joints include chondrocytes (CC), fibroblast-like synoviocytes (FLS) and macrophages (MC). Investigating their interplay can enhance our understanding of OA pathogenesis. Therefore, we comprehensively examine the mechanisms underlying pyroptosis and specifically investigate the intercellular interactions associated with pyroptosis among these three cell types, thereby elucidating their collective contribution to the progression of OA. We propose the concept of ' CC-FLS-MC pyroptosis-related crosstalk', describe the various pathways of pyroptotic interactions among these three cell types, and focus on recent advances in intervening pyroptosis in these three cell types for treating OA. We hope this will provide a possible direction for diversification of treatment for OA. The Translational potential of this article. The present study introduces the concept of 'MC-FLS-CC pyroptosis-related crosstalk' and provides an overview of the mechanisms underlying pyroptosis, as well as the pathways through which it affects MC, FLS, and CC. In addition, the role of regulation of these three types of cellular pyroptosis in OA has also been concerned. This review offers novel insights into the interplay between these cell types, with the aim of providing a promising avenue for diversified management of OA.
Collapse
Affiliation(s)
- Shida Kuang
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- Andrology Laboratory, Hunan University of Chinese Medicine, Changsha, China
- Hunan University of Medicine, Huaihua, Hunan, China
| | - Wen Sheng
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- Andrology Laboratory, Hunan University of Chinese Medicine, Changsha, China
- Hunan University of Medicine, Huaihua, Hunan, China
| | - Jiahao Meng
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, Hunan, China
- Hunan Engineering Research Center of Osteoarthritis, Changsha, Hunan, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Weijie Liu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, Hunan, China
- Hunan Engineering Research Center of Osteoarthritis, Changsha, Hunan, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yifan Xiao
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, Hunan, China
- Hunan Engineering Research Center of Osteoarthritis, Changsha, Hunan, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hang Tang
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, Hunan, China
- Hunan Engineering Research Center of Osteoarthritis, Changsha, Hunan, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xinying Fu
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- Andrology Laboratory, Hunan University of Chinese Medicine, Changsha, China
- Hunan University of Medicine, Huaihua, Hunan, China
| | - Min Kuang
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- Andrology Laboratory, Hunan University of Chinese Medicine, Changsha, China
- Hunan University of Medicine, Huaihua, Hunan, China
| | - Qinghu He
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- Andrology Laboratory, Hunan University of Chinese Medicine, Changsha, China
- Hunan University of Medicine, Huaihua, Hunan, China
| | - Shuguang Gao
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, Hunan, China
- Hunan Engineering Research Center of Osteoarthritis, Changsha, Hunan, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
20
|
Artimovič P, Badovská Z, Toporcerová S, Špaková I, Smolko L, Sabolová G, Kriváková E, Rabajdová M. Oxidative Stress and the Nrf2/PPARγ Axis in the Endometrium: Insights into Female Fertility. Cells 2024; 13:1081. [PMID: 38994935 PMCID: PMC11240766 DOI: 10.3390/cells13131081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/13/2024] Open
Abstract
Successful pregnancy depends on precise molecular regulation of uterine physiology, especially during the menstrual cycle. Deregulated oxidative stress (OS), often influenced by inflammatory changes but also by environmental factors, represents a constant threat to this delicate balance. Oxidative stress induces a reciprocally regulated nuclear factor erythroid 2-related factor 2/peroxisome proliferator-activated receptor-gamma (Nrf2/PPARγ) pathway. However, increased PPARγ activity appears to be a double-edged sword in endometrial physiology. Activated PPARγ attenuates inflammation and attenuates OS to restore redox homeostasis. However, it also interferes with physiological processes during the menstrual cycle, such as hormonal signaling and angiogenesis. This review provides an elucidation of the molecular mechanisms that support the interplay between PPARγ and OS. Additionally, it offers fresh perspectives on the Nrf2/PPARγ pathway concerning endometrial receptivity and its potential implications for infertility.
Collapse
Affiliation(s)
- Peter Artimovič
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, 040 11 Košice, Slovakia; (P.A.); (I.Š.); (L.S.); (G.S.); (E.K.); (M.R.)
| | - Zuzana Badovská
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, 040 11 Košice, Slovakia; (P.A.); (I.Š.); (L.S.); (G.S.); (E.K.); (M.R.)
| | - Silvia Toporcerová
- Department of Gynaecology and Obstetrics, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, 040 11 Košice, Slovakia;
| | - Ivana Špaková
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, 040 11 Košice, Slovakia; (P.A.); (I.Š.); (L.S.); (G.S.); (E.K.); (M.R.)
| | - Lukáš Smolko
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, 040 11 Košice, Slovakia; (P.A.); (I.Š.); (L.S.); (G.S.); (E.K.); (M.R.)
| | - Gabriela Sabolová
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, 040 11 Košice, Slovakia; (P.A.); (I.Š.); (L.S.); (G.S.); (E.K.); (M.R.)
| | - Eva Kriváková
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, 040 11 Košice, Slovakia; (P.A.); (I.Š.); (L.S.); (G.S.); (E.K.); (M.R.)
| | - Miroslava Rabajdová
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, 040 11 Košice, Slovakia; (P.A.); (I.Š.); (L.S.); (G.S.); (E.K.); (M.R.)
| |
Collapse
|
21
|
Ezhilarasan D. Deciphering the molecular pathways of saroglitazar: A dual PPAR α/γ agonist for managing metabolic NAFLD. Metabolism 2024; 155:155912. [PMID: 38609038 DOI: 10.1016/j.metabol.2024.155912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/07/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024]
Abstract
Saroglitazar (SARO), a dual peroxisome proliferator activated receptor (PPAR)-α/γ agonist, has been used to treat metabolic diseases such as insulin resistance and diabetic dyslipidemia in patients with non-alcoholic fatty liver disease (NAFLD). SARO, administered at a dose of 4 mg/day, has been consistently studied in clinical trials with different time points ranging from 4 to 24 weeks with NAFLD patients. Due to its PPAR-γ agonistic action, SARO prevents adipose tissue-mediated fatty acid delivery to the liver by increasing insulin sensitivity and regulating adiponectin and leptin levels in adipose tissue. In hepatocytes, SARO induces fatty acid β-oxidation in mitochondria and transcriptionally activates lipid metabolizing genes in peroxisomes. SARO inhibits insulin resistance, thereby preventing the activation of sterol regulatory element-binding proteins -1c and carbohydrate response element binding protein in hepatocytes through its PPAR-α agonistic action. SARO treatment reduces lipotoxicity-mediated oxidative stress by activating the nuclear factor erythroid 2-related factor 2 and transcriptionally expressing the antioxidants from the antioxidant response element in the nucleus through its PPAR-γ agonistic action. SARO provides a PPAR-α/γ-mediated anti-inflammatory effect by preventing the phosphorylation of mitogen-activated protein kinases (JNK and ERK) and nuclear factor kappa B in hepatocytes. Additionally, SARO interferes with transforming growth factor-β/Smad downstream signaling, thereby reducing liver fibrosis progression through its PPAR-α/γ agonistic actions. Thus, SARO improves insulin resistance and dyslipidemia in NAFLD, reduces lipid accumulation in the liver, and thereby prevents mitochondrial toxicity, oxidative stress, inflammation, and fibrosis progression. This review summarizes the possible molecular mechanism of SARO in the NAFLD.
Collapse
Affiliation(s)
- Devaraj Ezhilarasan
- Department of Pharmacology, Hepatology and Molecular Medicine Lab, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamil Nadu 600 077, India.
| |
Collapse
|
22
|
Qian Z, Zhang Q, Li P, Li Y, Zhang Y, Li R, Zhao T, Xia M, Chen Y, Hong X. A Disintegrin and Metalloproteinase-8 Protects Against Erastin-Induced Neuronal Ferroptosis via Activating Nrf2/HO-1/FTH1 Signaling Pathway. Mol Neurobiol 2024; 61:3490-3502. [PMID: 37995078 DOI: 10.1007/s12035-023-03782-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/06/2023] [Indexed: 11/24/2023]
Abstract
Ferroptosis is a type of iron-dependent programmed cell death caused by the imbalance between oxidants and antioxidants. A disintegrin and metalloproteinase-8 (ADAM8) is a metalloproteinase that mediates cell adhesion, cell migration, and proteolytic activity. However, the molecular mechanism of ADAM8 regulating ferroptosis after neural disorder is unclear, especially in the neuron. In the present study, we identified the protective role of ADAM8 in Erastin-induced ferroptosis in vitro of the HT22 cells. It was found that overexpression of ADAM8 resulted in upregulated expression of GPX4 and FTH1 along with the decreased reactive oxygen species (ROS) production and reduced neuronal death; however, knockdown of ADAM8 resulted in an opposite. Mechanically, using the Nrf2 activator NK-252 and inhibitor nrf2-IN-1, we dmonstrated that ADAM8 regulates Erastin-mediated neuronal ferroptosis via activating the Nrf2/HO-1/FTH1 signaling pathway. In conclusion, the current study suggested that ADAM8 inhibited Erastin-induced neuronal ferroptosis through activating the Nrf2/HO-1/FTH1 signaling pathway, playing a protective role in vitro of the HT22 cell line. ADAM8 may be a promising and feasible target for neuronal survival in diseases of neural disorder.
Collapse
Affiliation(s)
- Zhanyang Qian
- Department of Orthopedics, Taizhou People Hospital, Nanjing Medical University, Taizhou, China
- Department of Orthopedics, Zhongda Hospital of Southeast University, Nanjing, China
| | - Qinyang Zhang
- Department of Orthopedics, Taizhou People Hospital, Nanjing Medical University, Taizhou, China
- Postgraduate School, Dalian Medical University, Dalian, China
| | - Pengfei Li
- Department of Orthopedics, Taizhou People Hospital, Nanjing Medical University, Taizhou, China
- Postgraduate School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Li
- Department of Orthopedics, Taizhou People Hospital, Nanjing Medical University, Taizhou, China
- Taizhou Clinical Medical School of Nanjing Medical University, Taizhou, China
| | - Yanan Zhang
- Department of Orthopedics, Taizhou People Hospital, Nanjing Medical University, Taizhou, China
| | - Rulin Li
- Department of Orthopedics, Taizhou People Hospital, Nanjing Medical University, Taizhou, China
- Postgraduate School, Dalian Medical University, Dalian, China
| | - Tianyu Zhao
- Department of Orthopedics, Taizhou People Hospital, Nanjing Medical University, Taizhou, China
- Postgraduate School, Dalian Medical University, Dalian, China
| | - Mingjie Xia
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong First People's Hospital, Nantong, China.
| | - Yongyi Chen
- Department of Anesthesiology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China.
| | - Xin Hong
- Department of Orthopedics, Zhongda Hospital of Southeast University, Nanjing, China.
| |
Collapse
|
23
|
Yi Y, Li T, lv C, He W, Li W, Zhou X, Qin S. Proanthocyanidins isolated from lotus seed skin mitigate glycolipid metabolism disorder through the p38/Nrf2/NF-κB signaling pathway. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1300-1310. [PMID: 38761010 PMCID: PMC11532215 DOI: 10.3724/abbs.2024042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 02/19/2024] [Indexed: 05/20/2024] Open
Abstract
Lotus seed skin extract is rich in flavonoids, making it a promising candidate for developing health products. In a previous study, we found that proanthocyanidins from lotus seed skin, particularly proanthocyanidin B1 (PB1), can indirectly activate the Nrf2 signaling pathway, exerting an antioxidant effect. In this study, we isolate proanthocyanidins from lotus seed skin (PLS) using ethanol extraction and RP-HPLC identification, and investigate its effects on glycolipid metabolism both in vivo and in vitro. Our results demonstrate that PLS reduces body weight in high-fat diet (HFD) mice by decreasing feed efficiency. PLS also normalizes serum glucose, insulin secretion, glycosylated hemoglobin (HbA1c), and intraperitoneal glucose tolerance (IPGTT). Furthermore, PLS significantly improves blood lipid parameters and inhibits the expressions of six proinflammatory factors, including IL-1α, IL-1β, IL-3, IL-6, IFN-γ and TNF-α in HFD mice. Additionally, analysis of fresh liver tissues reveals that PLS and PB1 induce the expressions of antioxidant proteins such as HO-1 and NQO1 by activating the p38-Nrf2 signaling pathway and inhibiting the NF-κB signaling pathway. In conclusion, proanthocyanidins from lotus seed skin regulate glycolipid metabolism disorders by targeting the p38/Nrf2/NF-κB signaling pathway. Our study offers a new approach for the high-value comprehensive utilization of lotus seed skin by-products and precise dietary intervention for metabolic syndrome.
Collapse
Affiliation(s)
- Yuhang Yi
- College of Food Science and TechnologyHunan Agricultural UniversityChangsha410128China
| | - Tao Li
- Hunan Agricultural Products Processing InstituteHunan Academy of Agricultural SciencesChangsha410125China
| | - Chenghao lv
- College of Biological Science and TechnologyHunan Agricultural UniversityChangsha410128China
| | - Wenjiang He
- College of Biological Science and TechnologyHunan Agricultural UniversityChangsha410128China
- Infinitus (China) Company Ltd.Guangzhou510665China
| | - Wenzhi Li
- Infinitus (China) Company Ltd.Guangzhou510665China
| | - Xixin Zhou
- College of Biological Science and TechnologyHunan Agricultural UniversityChangsha410128China
| | - Si Qin
- College of Food Science and TechnologyHunan Agricultural UniversityChangsha410128China
- College of Biological Science and TechnologyHunan Agricultural UniversityChangsha410128China
| |
Collapse
|
24
|
Greco G, Di Lorenzo R, Ricci L, Di Serio T, Vardaro E, Laneri S. Clinical Studies Using Topical Melatonin. Int J Mol Sci 2024; 25:5167. [PMID: 38791203 PMCID: PMC11121188 DOI: 10.3390/ijms25105167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/03/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Melatonin is ubiquitously present in all animals and plants, where it exerts a variety of physiological activities thanks to its antioxidant properties and its key role as the first messenger of extracellular signaling functions. Most of the clinical studies on melatonin refer to its widespread oral use as a dietary supplement to improve sleep. A far smaller number of articles describe the clinical applications of topical melatonin to treat or prevent skin disorders by exploiting its antioxidant and anti-inflammatory activities. This review focuses on the clinical studies in which melatonin was applied on the skin as a photoprotective, anti-aging, or hair growth-promoting agent. The methodologies and results of such studies are discussed to provide an overall picture of the state of the art in this intriguing field of research. The clinical studies in which melatonin was applied on the skin before exposure to radiation (UV, sunlight, and high-energy beams) were all characterized by an appropriate design (randomized, double-blind, and placebo-controlled) and strongly support its clinical efficacy in preventing or reducing skin damage such as dermatitis, erythema, and sunburn. Most of the studies examined in this review do not provide a clear demonstration of the efficacy of topical melatonin as a skin anti-aging or as a hair growth-promoting agent owing to limitations in their design and/or to the use of melatonin combined with extra active ingredients, except for one trial that suggests a possible beneficial role of melatonin in treating some forms of alopecia in women. Further research efforts are required to reach definitive conclusions concerning the actual benefits of topical melatonin to counteract skin aging and hair loss.
Collapse
Affiliation(s)
| | | | | | | | | | - Sonia Laneri
- Dipartimento di Farmacia, Università degli Studi di Napoli Federico II, Via Domenico Montesano, 49, 80131 Napoli, Italy; (G.G.); (R.D.L.); (L.R.); (T.D.S.); (E.V.)
| |
Collapse
|
25
|
Fan X, Yu W, Wang Q, Yang H, Tan D, Yu B, He J, Zheng P, Yu J, Luo J, Luo Y, Yan H, Wang J, Wang H, Wang Q, Mao X. Protective effect of Broussonetia papyrifera leaf polysaccharides on intestinal integrity in a rat model of diet-induced oxidative stress. Int J Biol Macromol 2024; 268:131589. [PMID: 38643924 DOI: 10.1016/j.ijbiomac.2024.131589] [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: 08/09/2023] [Revised: 03/28/2024] [Accepted: 04/12/2024] [Indexed: 04/23/2024]
Abstract
This study aimed to investigate the effect of Broussonetia papyrifera polysaccharides (BPP) on the jejunal intestinal integrity of rats ingesting oxidized fish oil (OFO) induced oxidative stress. Polysaccharides (Mw 16,956 Da) containing carboxyl groups were extracted from Broussonetia papyrifera leaves. In vitro antioxidant assays showed that this polysaccharide possessed antioxidant capabilities. Thirty-two male weaned rats were allocated into two groups orally infused BPP solution and PBS for 26 days, respectively. From day 9 to day 26, half of the rats in each group were fed food containing OFO, where the lipid peroxidation can induce intestinal oxidative stress. OFO administration resulted in diarrhea, decreased growth performance (p < 0.01), impaired jejunal morphology (p < 0.05) and antioxidant capacity (p < 0.01), increased the levels of ROS and its related products, IL-1β and IL-17 (p < 0.01) of jejunum, as well as down-regulated Bcl-2/Bax (p < 0.01) and Nrf2 signaling (p < 0.01) of jejunum in rats. BPP gavage effectively alleviated the negative effects of OFO on growth performance, morphology, enterocyte apoptosis, antioxidant capacity and inflammation of jejunum (p < 0.05) in rats. In the oxidative stress model cell assay, the use of receptor inhibitors inhibited the enhancement of antioxidant capacity by BPP. These results suggested that BPP protected intestinal morphology, thus improving growth performance and reducing diarrhea in rats ingesting OFO. This protective effect may be attributed to scavenging free radicals and activating the Nrf2 pathway, which enhances antioxidant capacity, consequently reducing inflammation and mitigating intestinal cell death.
Collapse
Affiliation(s)
- Xiangqi Fan
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Wei Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Qingxiang Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Heng Yang
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Dayan Tan
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Jun He
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Ping Zheng
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Jie Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Junqiu Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Yuheng Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Hui Yan
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Jianping Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Huifen Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Quyuan Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China
| | - Xiangbing Mao
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key laboratory of Animal Disease-resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key laboratory of Animal Disease-resistant Nutrition of Sichuan Province, Chengdu 611130, People's Republic of China.
| |
Collapse
|
26
|
Yi T, Zhang W, Hua Y, Xin X, Wu Z, Li Y, Wen C, Fan Y, Ji J, Xu L. Rutin alleviates lupus nephritis by inhibiting T cell oxidative stress through PPARγ. Chem Biol Interact 2024; 394:110972. [PMID: 38555047 DOI: 10.1016/j.cbi.2024.110972] [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: 01/27/2024] [Revised: 03/12/2024] [Accepted: 03/21/2024] [Indexed: 04/02/2024]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disorder characterized by complex clinical symptoms and multi-organ damage. One of the most prevalent complications of SLE is lupus nephritis (LN). Rutin, a natural flavonoid compound found in various plants used in traditional Chinese medicine, has shown promising anti-inflammatory, antioxidant, and renal protective effects. In our study, we treated MRL/lpr mice, a model known for spontaneously developing LN, with Rutin. Our findings reveal that Rutin markedly reduced serum cytokine and autoantibody levels and decreased inflammatory cell infiltration in renal tissues, thereby ameliorating kidney pathology. In vitro experiments indicated that Rutin's therapeutic effect on LN is linked to its significant reduction of oxidative stress in T cells. Further investigations suggest that Rutin enhances oxidative stress management through the modulation of Peroxisome proliferator-activated receptor gamma (PPARγ). We observed that Rutin modulates PPARγ activity, leading to reduced transcriptional activity of NF-κB and STAT3, which in turn inhibits the secretion of inflammatory cytokines such as IL-6, TNF-α, and IL-17. In summary, Rutin can exert an antioxidant effect by regulating PPARγ and shows therapeutic action against LN.
Collapse
Affiliation(s)
- Tongtong Yi
- College of Basic Medical, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, 548 Binwen Road, Hangzhou, 310051, China
| | - Wei Zhang
- College of Basic Medical, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, 548 Binwen Road, Hangzhou, 310051, China
| | - Ying Hua
- College of Basic Medical, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, 548 Binwen Road, Hangzhou, 310051, China
| | - Xingpan Xin
- College of Basic Medical, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, 548 Binwen Road, Hangzhou, 310051, China
| | - Zhenyu Wu
- College of Basic Medical, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, 548 Binwen Road, Hangzhou, 310051, China
| | - Ying Li
- College of Basic Medical, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, 548 Binwen Road, Hangzhou, 310051, China
| | - Chengping Wen
- College of Basic Medical, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, 548 Binwen Road, Hangzhou, 310051, China
| | - Yongsheng Fan
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, 548 Binwen Road, Hangzhou, 310051, China; Department of Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jinjun Ji
- College of Basic Medical, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, 548 Binwen Road, Hangzhou, 310051, China.
| | - Li Xu
- College of Basic Medical, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, 548 Binwen Road, Hangzhou, 310051, China.
| |
Collapse
|
27
|
Baroi S, Czernik PJ, Khan MP, Letson J, Crowe E, Chougule A, Griffin PR, Rosen CJ, Lecka-Czernik B. PPARG in osteocytes controls cell bioenergetics and systemic energy metabolism independently of sclerostin levels in circulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.04.588029. [PMID: 38645043 PMCID: PMC11030235 DOI: 10.1101/2024.04.04.588029] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Objective The skeleton is one of the largest organs in the body, wherein metabolism is integrated with systemic energy metabolism. However, the bioenergetic programming of osteocytes, the most abundant bone cells coordinating bone metabolism, is not well defined. Here, using a mouse model with partial penetration of an osteocyte-specific PPARG deletion, we demonstrate that PPARG controls osteocyte bioenergetics and their contribution to systemic energy metabolism independently of circulating sclerostin levels. Methods In vivo and in vitro models of osteocyte-specific PPARG deletion, i.e. Dmp 1 Cre Pparγ flfl male and female mice (γOT KO ) and MLO-Y4 osteocyte-like cells with either siRNA-silenced or CRISPR/Cas9-edited Pparγ . As applicable, the models were analyzed for levels of energy metabolism, glucose metabolism, and metabolic profile of extramedullary adipose tissue, as well as the osteocyte transcriptome, mitochondrial function, bioenergetics, insulin signaling, and oxidative stress. Results Circulating sclerostin levels of γOT KO male and female mice were not different from control mice. Male γOT KO mice exhibited a high energy phenotype characterized by increased respiration, heat production, locomotion and food intake. This high energy phenotype in males did not correlate with "beiging" of peripheral adipose depots. However, both sexes showed a trend for reduced fat mass and apparent insulin resistance without changes in glucose tolerance, which correlated with decreased osteocytic responsiveness to insulin measured by AKT activation. The transcriptome of osteocytes isolated from γOT KO males suggested profound changes in cellular metabolism, fuel transport and usage, mitochondria dysfunction, insulin signaling and increased oxidative stress. In MLO-Y4 osteocytes, PPARG deficiency correlated with highly active mitochondria, increased ATP production, shifts in fuel utilization, and accumulation of reactive oxygen species (ROS). Conclusions PPARG in male osteocytes acts as a molecular break on mitochondrial function, and protection against oxidative stress and ROS accumulation. It also regulates osteocyte insulin signaling and fuel usage to produce energy. These data provide insight into the connection between osteocyte bioenergetics and their sex-specific contribution to the balance of systemic energy metabolism. These findings support the concept that the skeleton controls systemic energy expenditure via osteocyte metabolism. Highlights Osteocytes function as a body energostat via their bioenergeticsPPARG protein acts as a "molecular break" of osteocyte mitochondrial activityPPARG deficiency activates TCA cycle, oxidative stress and ROS accumulationPPARG controls osteocyte insulin signaling and fuel utilization.
Collapse
|
28
|
Zhou B, Wang L, Yang S, Liang Y, Zhang Y, Liu X, Pan X, Li J. Pyrogallol protects against influenza A virus-triggered lethal lung injury by activating the Nrf2-PPAR-γ-HO-1 signaling axis. MedComm (Beijing) 2024; 5:e531. [PMID: 38617435 PMCID: PMC11014464 DOI: 10.1002/mco2.531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 02/04/2024] [Accepted: 02/26/2024] [Indexed: 04/16/2024] Open
Abstract
Pyrogallol, a natural polyphenol compound (1,2,3-trihydroxybenzene), has shown efficacy in the therapeutic treatment of disorders associated with inflammation. Nevertheless, the mechanisms underlying the protective properties of pyrogallol against influenza A virus infection are not yet established. We established in this study that pyrogallol effectively alleviated H1N1 influenza A virus-induced lung injury and reduced mortality. Treatment with pyrogallol was found to promote the expression and nuclear translocation of nuclear factor erythroid-2-related factor 2 (Nrf2) and peroxisome proliferator-activated receptor gamma (PPAR-γ). Notably, the activation of Nrf2 by pyrogallol was involved in elevating the expression of PPAR-γ, both of which act synergistically to enhance heme oxygenase-1 (HO-1) synthesis. Blocking HO-1 by zinc protoporphyrin (ZnPP) reduced the suppressive impact of pyrogallol on H1N1 virus-mediated aberrant retinoic acid-inducible gene-I-nuclear factor kappa B (RIG-I-NF-κB) signaling, which thus abolished the dampening effects of pyrogallol on excessive proinflammatory mediators and cell death (including apoptosis, necrosis, and ferroptosis). Furthermore, the HO-1-independent inactivation of janus kinase 1/signal transducers and activators of transcription (JAK1/STATs) and the HO-1-dependent RIG-I-augmented STAT1/2 activation were both abrogated by pyrogallol, resulting in suppression of the enhanced transcriptional activity of interferon-stimulated gene factor 3 (ISGF3) complexes, thus prominently inhibiting the amplification of the H1N1 virus-induced proinflammatory reaction and apoptosis in interferon-beta (IFN-β)-sensitized cells. The study provides evidence that pyrogallol alleviates excessive proinflammatory responses and abnormal cell death via HO-1 induction, suggesting it could be a potential agent for treating influenza.
Collapse
Affiliation(s)
- Beixian Zhou
- The People's Hospital of GaozhouGaozhouChina
- Cancer Center, Integrated Hospital of Traditional Chinese MedicineSouthern Medical UniversityGuangzhouChina
| | | | - Sushan Yang
- The People's Hospital of GaozhouGaozhouChina
| | | | | | - Xuanyu Liu
- The People's Hospital of GaozhouGaozhouChina
| | | | - Jing Li
- State Key Laboratory of Respiratory DiseaseNational Clinical Research Center of Respiratory DiseaseGuangzhou Institute of Respiratory HealthInstitute of Chinese Integrative MedicineGuangdong‐Hongkong‐Macao Joint Laboratory of Infectious Respiratory Diseasethe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou Medical UniversityGuangzhouChina
| |
Collapse
|
29
|
Monsalvo-Maraver LA, Ovalle-Noguez EA, Nava-Osorio J, Maya-López M, Rangel-López E, Túnez I, Tinkov AA, Tizabi Y, Aschner M, Santamaría A. Interactions Between the Ubiquitin-Proteasome System, Nrf2, and the Cannabinoidome as Protective Strategies to Combat Neurodegeneration: Review on Experimental Evidence. Neurotox Res 2024; 42:18. [PMID: 38393521 PMCID: PMC10891226 DOI: 10.1007/s12640-024-00694-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 01/13/2024] [Accepted: 02/04/2024] [Indexed: 02/25/2024]
Abstract
Neurodegenerative disorders are chronic brain diseases that affect humans worldwide. Although many different factors are thought to be involved in the pathogenesis of these disorders, alterations in several key elements such as the ubiquitin-proteasome system (UPS), the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway, and the endocannabinoid system (ECS or endocannabinoidome) have been implicated in their etiology. Impairment of these elements has been linked to the origin and progression of neurodegenerative disorders, while their potentiation is thought to promote neuronal survival and overall neuroprotection, as proved with several experimental models. These key neuroprotective pathways can interact and indirectly activate each other. In this review, we summarize the neuroprotective potential of the UPS, ECS, and Nrf2 signaling, both separately and combined, pinpointing their role as a potential therapeutic approach against several hallmarks of neurodegeneration.
Collapse
Affiliation(s)
- Luis Angel Monsalvo-Maraver
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Av. Universidad 3000, C.U. Coyoacán, 04510, Mexico City, Mexico.
| | - Enid A Ovalle-Noguez
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Av. Universidad 3000, C.U. Coyoacán, 04510, Mexico City, Mexico
| | - Jade Nava-Osorio
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Av. Universidad 3000, C.U. Coyoacán, 04510, Mexico City, Mexico
| | - Marisol Maya-López
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Av. Universidad 3000, C.U. Coyoacán, 04510, Mexico City, Mexico
- Doctorado en Ciencias Biológicas y de La Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| | - Edgar Rangel-López
- Instituto Nacional de Neurología y Neurocirugía, S.S.A., Mexico City, Mexico
| | - Isaac Túnez
- Instituto de Investigaciones Biomédicas Maimonides de Córdoba (IMIBIC), Departamento de Bioquímica y Biología Molecular, Facultad de Medicina y Enfermería, Universidad de Córdoba, Red Española de Excelencia en Estimulación Cerebral (REDESTIM), Córdoba, Spain
| | - Alexey A Tinkov
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
- Yaroslavl State University, Yaroslavl, Russia
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Abel Santamaría
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Av. Universidad 3000, C.U. Coyoacán, 04510, Mexico City, Mexico.
| |
Collapse
|
30
|
Díaz M, Valdés-Baizabal C, de Pablo DP, Marin R. Age-Dependent Changes in Nrf2/Keap1 and Target Antioxidant Protein Expression Correlate to Lipoxidative Adducts, and Are Modulated by Dietary N-3 LCPUFA in the Hippocampus of Mice. Antioxidants (Basel) 2024; 13:206. [PMID: 38397804 PMCID: PMC10886099 DOI: 10.3390/antiox13020206] [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: 10/22/2023] [Revised: 01/26/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
The brain has a high metabolism rate that may generate reactive oxygen and nitrogen species. Consequently, nerve cells require highly efficient antioxidant defenses in order to prevent a condition of deleterious oxidative stress. This is particularly relevant in the hippocampus, a highly complex cerebral area involved in processing superior cognitive functions. Most current evidence points to hippocampal oxidative damage as a causal effect for neurodegenerative disorders, especially Alzheimer's disease. Nuclear factor erythroid-2-related factor 2/Kelch-like ECH-associated protein 1 (Nrf2/Keap1) is a master key for the transcriptional regulation of antioxidant and detoxifying systems. It is ubiquitously expressed in brain areas, mainly supporting glial cells. In the present study, we have analyzed the relationships between Nrf2 and Keap1 isoforms in hippocampal tissue in response to aging and dietary long-chain polyunsaturated fatty acids (LCPUFA) supplementation. The possible involvement of lipoxidative and nitrosative by-products in the dynamics of the Nrf2/Keap1 complex was examined though determination of protein adducts, namely malondialdehyde (MDA), 4-hydroxynonenal (HNE), and 3-nitro-tyrosine (NTyr) under basal conditions. The results were correlated to the expression of target proteins heme-oxygenase-1 (HO-1) and glutathione peroxidase 4 (GPx4), whose expressions are known to be regulated by Nrf2/Keap1 signaling activation. All variables in this study were obtained simultaneously from the same preparations, allowing multivariate approaches. The results demonstrate a complex modification of the protein expression patterns together with the formation of adducts in response to aging and diet supplementation. Both parameters exhibited a strong interaction. Noticeably, LCPUFA supplementation to aged animals restored the Nrf2/Keap1/target protein patterns to the status observed in young animals, therefore driving a "rejuvenation" of hippocampal antioxidant defense.
Collapse
Affiliation(s)
- Mario Díaz
- Department of Physics, Faculty of Sciences, University of La Laguna, 38200 Tenerife, Spain
- Instituto Universitario de Neurociencias (IUNE), University of La Laguna, 38320 Tenerife, Spain; (C.V.-B.); (D.P.d.P.); (R.M.)
| | - Catalina Valdés-Baizabal
- Instituto Universitario de Neurociencias (IUNE), University of La Laguna, 38320 Tenerife, Spain; (C.V.-B.); (D.P.d.P.); (R.M.)
- Laboratory of Cellular Neurobiology, Department of Basic Medical Sciences, Faculty of Health Sciences, University of La Laguna, 38200 Tenerife, Spain
| | - Daniel Pereda de Pablo
- Instituto Universitario de Neurociencias (IUNE), University of La Laguna, 38320 Tenerife, Spain; (C.V.-B.); (D.P.d.P.); (R.M.)
- Laboratory of Cellular Neurobiology, Department of Basic Medical Sciences, Faculty of Health Sciences, University of La Laguna, 38200 Tenerife, Spain
| | - Raquel Marin
- Instituto Universitario de Neurociencias (IUNE), University of La Laguna, 38320 Tenerife, Spain; (C.V.-B.); (D.P.d.P.); (R.M.)
- Laboratory of Cellular Neurobiology, Department of Basic Medical Sciences, Faculty of Health Sciences, University of La Laguna, 38200 Tenerife, Spain
- Associate Research Unit ULL-CSIC “Membrane Physiology and Biophysics in Neurodegenerative and Cancer Diseases”, 38200 Tenerife, Spain
| |
Collapse
|
31
|
Pfefferlé M, Vallelian F. Transcription Factor NRF2 in Shaping Myeloid Cell Differentiation and Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1459:159-195. [PMID: 39017844 DOI: 10.1007/978-3-031-62731-6_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
NFE2-related factor 2 (NRF2) is a master transcription factor (TF) that coordinates key cellular homeostatic processes including antioxidative responses, autophagy, proteostasis, and metabolism. The emerging evidence underscores its significant role in modulating inflammatory and immune processes. This chapter delves into the role of NRF2 in myeloid cell differentiation and function and its implication in myeloid cell-driven diseases. In macrophages, NRF2 modulates cytokine production, phagocytosis, pathogen clearance, and metabolic adaptations. In dendritic cells (DCs), it affects maturation, cytokine production, and antigen presentation capabilities, while in neutrophils, NRF2 is involved in activation, migration, cytokine production, and NETosis. The discussion extends to how NRF2's regulatory actions pertain to a wide array of diseases, such as sepsis, various infectious diseases, cancer, wound healing, atherosclerosis, hemolytic conditions, pulmonary disorders, hemorrhagic events, and autoimmune diseases. The activation of NRF2 typically reduces inflammation, thereby modifying disease outcomes. This highlights the therapeutic potential of NRF2 modulation in treating myeloid cell-driven pathologies.
Collapse
Affiliation(s)
- Marc Pfefferlé
- Department of Internal Medicine, Spital Limmattal, Schlieren, Switzerland
| | - Florence Vallelian
- Department of Internal Medicine, University of Zurich and University Hospital of Zurich, Zurich, Switzerland.
| |
Collapse
|
32
|
Chang CY, Armstrong D, Corry DB, Kheradmand F. Alveolar macrophages in lung cancer: opportunities challenges. Front Immunol 2023; 14:1268939. [PMID: 37822933 PMCID: PMC10562548 DOI: 10.3389/fimmu.2023.1268939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 09/12/2023] [Indexed: 10/13/2023] Open
Abstract
Alveolar macrophages (AMs) are critical components of the innate defense mechanism in the lung. Nestled tightly within the alveoli, AMs, derived from the yolk-sac or bone marrow, can phagocytose foreign particles, defend the host against pathogens, recycle surfactant, and promptly respond to inhaled noxious stimuli. The behavior of AMs is tightly dependent on the environmental cues whereby infection, chronic inflammation, and associated metabolic changes can repolarize their effector functions in the lungs. Several factors within the tumor microenvironment can re-educate AMs, resulting in tumor growth, and reducing immune checkpoint inhibitors (ICIs) efficacy in patients treated for non-small cell lung cancer (NSCLC). The plasticity of AMs and their critical function in altering tumor responses to ICIs make them a desirable target in lung cancer treatment. New strategies have been developed to target AMs in solid tumors reprograming their suppressive function and boosting the efficacy of ICIs. Here, we review the phenotypic and functional changes in AMs in response to sterile inflammation and in NSCLC that could be critical in tumor growth and metastasis. Opportunities in altering AMs' function include harnessing their potential function in trained immunity, a concept borrowed from memory response to infections, which could be explored therapeutically in managing lung cancer treatment.
Collapse
Affiliation(s)
- Cheng-Yen Chang
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Dominique Armstrong
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - David B. Corry
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
- Biology of Inflammation Center, Baylor College of Medicine, Houston, TX, United States
- Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, TX, United States
| | - Farrah Kheradmand
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
- Biology of Inflammation Center, Baylor College of Medicine, Houston, TX, United States
- Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, TX, United States
| |
Collapse
|
33
|
Tian HY, Huang BY, Nie HF, Chen XY, Zhou Y, Yang T, Cheng SW, Mei ZG, Ge JW. The Interplay between Mitochondrial Dysfunction and Ferroptosis during Ischemia-Associated Central Nervous System Diseases. Brain Sci 2023; 13:1367. [PMID: 37891735 PMCID: PMC10605666 DOI: 10.3390/brainsci13101367] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/12/2023] [Accepted: 09/22/2023] [Indexed: 10/29/2023] Open
Abstract
Cerebral ischemia, a leading cause of disability and mortality worldwide, triggers a cascade of molecular and cellular pathologies linked to several central nervous system (CNS) disorders. These disorders primarily encompass ischemic stroke, Alzheimer's disease (AD), Parkinson's disease (PD), epilepsy, and other CNS conditions. Despite substantial progress in understanding and treating the underlying pathological processes in various neurological diseases, there is still a notable absence of effective therapeutic approaches aimed specifically at mitigating the damage caused by these illnesses. Remarkably, ischemia causes severe damage to cells in ischemia-associated CNS diseases. Cerebral ischemia initiates oxygen and glucose deprivation, which subsequently promotes mitochondrial dysfunction, including mitochondrial permeability transition pore (MPTP) opening, mitophagy dysfunction, and excessive mitochondrial fission, triggering various forms of cell death such as autophagy, apoptosis, as well as ferroptosis. Ferroptosis, a novel type of regulated cell death (RCD), is characterized by iron-dependent accumulation of lethal reactive oxygen species (ROS) and lipid peroxidation. Mitochondrial dysfunction and ferroptosis both play critical roles in the pathogenic progression of ischemia-associated CNS diseases. In recent years, growing evidence has indicated that mitochondrial dysfunction interplays with ferroptosis to aggravate cerebral ischemia injury. However, the potential connections between mitochondrial dysfunction and ferroptosis in cerebral ischemia have not yet been clarified. Thus, we analyzed the underlying mechanism between mitochondrial dysfunction and ferroptosis in ischemia-associated CNS diseases. We also discovered that GSH depletion and GPX4 inactivation cause lipoxygenase activation and calcium influx following cerebral ischemia injury, resulting in MPTP opening and mitochondrial dysfunction. Additionally, dysfunction in mitochondrial electron transport and an imbalanced fusion-to-fission ratio can lead to the accumulation of ROS and iron overload, which further contribute to the occurrence of ferroptosis. This creates a vicious cycle that continuously worsens cerebral ischemia injury. In this study, our focus is on exploring the interplay between mitochondrial dysfunction and ferroptosis, which may offer new insights into potential therapeutic approaches for the treatment of ischemia-associated CNS diseases.
Collapse
Affiliation(s)
- He-Yan Tian
- School of Medical Technology and Nursing, Shenzhen Polytechnic University, Xili Lake, Nanshan District, Shenzhen 518000, China;
| | - Bo-Yang Huang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Hui-Fang Nie
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Xiang-Yu Chen
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Yue Zhou
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Tong Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Shao-Wu Cheng
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Zhi-Gang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Jin-Wen Ge
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
- Hunan Academy of Traditional Chinese Medicine, Changsha 410208, China
| |
Collapse
|
34
|
Vázquez-González D, Corona JC. Pioglitazone enhances brain mitochondrial biogenesis and phase II detoxification capacity in neonatal rats with 6-OHDA-induced unilateral striatal lesions. Front Neurosci 2023; 17:1186520. [PMID: 37575308 PMCID: PMC10416244 DOI: 10.3389/fnins.2023.1186520] [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: 03/16/2023] [Accepted: 07/17/2023] [Indexed: 08/15/2023] Open
Abstract
The psychostimulant methylphenidate (MPH) is the first-line pharmacological treatment for attention-deficit/hyperactivity disorder (ADHD), but has numerous adverse side effects. The PPARγ receptor agonist pioglitazone (PIO) is known to improve mitochondrial bioenergetics and antioxidant capacity, both of which may be deficient in ADHD, suggesting utility as an adjunct therapy. Here, we assessed the effects of PIO on ADHD-like symptoms, mitochondrial biogenesis and antioxidant pathways in multiple brain regions of neonate rats with unilateral striatal lesions induced by 6-hydroxydopamine (6-OHDA) as an experimental ADHD model. Unilateral striatal injection of 6-OHDA reduced ipsilateral dopaminergic innervation by 33% and increased locomotor activity. This locomotor hyperactivity was not altered by PIO treatment for 14 days. However, PIO increased the expression of proteins contributing to mitochondrial biogenesis in the striatum, hippocampus, cerebellum and prefrontal cortex of 6-OHDA-lesioned rats. In addition, PIO treatment enhanced the expression of the phase II transcription factor Nrf2 in the striatum, prefrontal cortex and cerebellum. In contrast, no change in the antioxidant enzyme catalase was observed in any of the brain regions analyzed. Thus, PIO may improve mitochondrial biogenesis and phase 2 detoxification in the ADHD brain. Further studies are required to determine if different dose regimens can exert more comprehensive therapeutic effects against ADHD neuropathology and behavior.
Collapse
Affiliation(s)
| | - Juan Carlos Corona
- Laboratory of Neurosciences, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| |
Collapse
|
35
|
Kogami M, Abe S, Nakamura H, Aoshiba K. Fenofibrate attenuates the cytotoxic effect of cisplatin on lung cancer cells by enhancing the antioxidant defense system in vitro. Oncol Lett 2023; 26:313. [PMID: 37332337 PMCID: PMC10272955 DOI: 10.3892/ol.2023.13899] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 05/11/2023] [Indexed: 06/20/2023] Open
Abstract
Fenofibrate (FF) is a peroxisome proliferator- activated receptor (PPAR)-α agonist that is widely used for the treatment of hyperlipidemia. It has been shown to have pleiotropic actions beyond its hypolipidemic effect. FF has been shown to exert a cytotoxic effect on some cancer cells when used at higher than clinically relevant concentrations; on the other hand, its cytoprotective effect on normal cells has also been reported. The present study assessed the effect of FF on cisplatin (CDDP) cytotoxicity to lung cancer cells in vitro. The results demonstrated that the effect of FF on lung cancer cells depends on its concentration. FF at ≤50 µM, which is a clinically achievable blood concentration, attenuated CDDP cytotoxicity to lung cancer cells, whereas FF at ≥100 µM, albeit clinically unachievable, had an anticancer effect. The mechanism of FF attenuation of CDDP cytotoxicity involved PPAR-α-dependent aryl hydrocarbon receptor (AhR) expression, which in turn stimulated nuclear factor erythroid 2-related factor 2 (Nrf2) expression and antioxidant production, resulting in lung cancer cell protection from CDDP-evoked oxidative damage. In conclusion, the present study revealed that FF, at clinically relevant concentrations, attenuated CDDP cytotoxicity to lung cancer cells by enhancing the antioxidant defense system through activation of a pathway that involves the PPAR-α-PPAR response element-AhR xenobiotic response element-Nrf2-antioxidant response element. These findings suggested that concomitant use of FF with CDDP may compromise the efficacy of chemotherapy. Although the anticancer property of FF has recently attracted much attention, concentrations that exceed clinically relevant concentrations are required.
Collapse
Affiliation(s)
- Mariko Kogami
- Department of Respiratory Medicine, Tokyo Medical University Ibaraki Medical Center, Ami, Ibaraki 300-0395, Japan
- Department of Respiratory Medicine, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Shinji Abe
- Department of Respiratory Medicine, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Hiroyuki Nakamura
- Department of Respiratory Medicine, Tokyo Medical University Ibaraki Medical Center, Ami, Ibaraki 300-0395, Japan
| | - Kazutetsu Aoshiba
- Department of Respiratory Medicine, Tokyo Medical University Ibaraki Medical Center, Ami, Ibaraki 300-0395, Japan
| |
Collapse
|
36
|
Borgonovi SM, Iametti S, Di Nunzio M. Docosahexaenoic Acid as Master Regulator of Cellular Antioxidant Defenses: A Systematic Review. Antioxidants (Basel) 2023; 12:1283. [PMID: 37372014 DOI: 10.3390/antiox12061283] [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: 05/18/2023] [Revised: 06/08/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Docosahexaenoic acid (DHA) is a polyunsaturated fatty acid that benefits the prevention of chronic diseases. Due to its high unsaturation, DHA is vulnerable to free radical oxidation, resulting in several unfavorable effects, including producing hazardous metabolites. However, in vitro and in vivo investigations suggest that the relationship between the chemical structure of DHA and its susceptibility to oxidation may not be as clear-cut as previously thought. Organisms have developed a balanced system of antioxidants to counteract the overproduction of oxidants, and the nuclear factor erythroid 2-related factor 2 (Nrf2) is the key transcription factor identified for transmitting the inducer signal to the antioxidant response element. Thus, DHA might preserve the cellular redox status promoting the transcriptional regulation of cellular antioxidants through Nrf2 activation. Here, we systematically summarize the research on the possible role of DHA in controlling cellular antioxidant enzymes. After the screening process, 43 records were selected and included in this review. Specifically, 29 studies related to the effects of DHA in cell cultures and 15 studies concerned the effects of consumption or treatment with DHA in animal. Despite DHA's promising and encouraging effects at modulating the cellular antioxidant response in vitro/in vivo, some differences observed among the reviewed studies may be accounted for by the different experimental conditions adopted, including the time of supplementation/treatment, DHA concentration, and cell culture/tissue model. Moreover, this review offers potential molecular explanations for how DHA controls cellular antioxidant defenses, including involvement of transcription factors and the redox signaling pathway.
Collapse
Affiliation(s)
- Sara Margherita Borgonovi
- Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Via Celoria 2, 20133 Milan, Italy
| | - Stefania Iametti
- Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Via Celoria 2, 20133 Milan, Italy
| | - Mattia Di Nunzio
- Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Via Celoria 2, 20133 Milan, Italy
| |
Collapse
|
37
|
Nandanpawar P, Sahoo L, Sahoo B, Murmu K, Chaudhari A, Pavan kumar A, Das P. Identification of differentially expressed genes and SNPs linked to harvest body weight of genetically improved rohu carp, Labeo rohita. Front Genet 2023; 14:1153911. [PMID: 37359361 PMCID: PMC10285081 DOI: 10.3389/fgene.2023.1153911] [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: 01/30/2023] [Accepted: 05/25/2023] [Indexed: 06/28/2023] Open
Abstract
In most of the aquaculture selection programs, harvest body weight has been a preferred performance trait for improvement. Molecular interplay of genes linked to higher body weight is not elucidated in major carp species. The genetically improved rohu carp with 18% average genetic gain per generation with respect to harvest body weight is a promising candidate for studying genes' underlying performance traits. In the present study, muscle transcriptome sequencing of two groups of individuals, with significant difference in breeding value, belonging to the tenth generation of rohu carp was performed using the Illumina HiSeq 2000 platform. A total of 178 million paired-end raw reads were generated to give rise to 173 million reads after quality control and trimming. The genome-guided transcriptome assembly and differential gene expression produced 11,86,119 transcripts and 451 upregulated and 181 downregulated differentially expressed genes (DEGs) between high-breeding value and low-breeding value (HB & LB) groups, respectively. Similarly, 39,158 high-quality coding SNPs were identified with the Ts/Tv ratio of 1.23. Out of a total of 17 qPCR-validated transcripts, eight were associated with cellular growth and proliferation and harbored 13 SNPs. The gene expression pattern was observed to be positively correlated with RNA-seq data for genes such as myogenic factor 6, titin isoform X11, IGF-1 like, acetyl-CoA, and thyroid receptor hormone beta. A total of 26 miRNA target interactions were also identified to be associated with significant DETs (p-value < 0.05). Genes such as Myo6, IGF-1-like, and acetyl-CoA linked to higher harvest body weight may serve as candidate genes in marker-assisted breeding and SNP array construction for genome-wide association studies and genomic selection.
Collapse
Affiliation(s)
- P. Nandanpawar
- ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar, Odisha, India
| | - L. Sahoo
- ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar, Odisha, India
| | - B. Sahoo
- ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar, Odisha, India
| | - K. Murmu
- ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar, Odisha, India
| | - A. Chaudhari
- ICAR-Central Institute of Fisheries Education, Mumbai, Maharashtra, India
| | - A. Pavan kumar
- ICAR-Central Institute of Fisheries Education, Mumbai, Maharashtra, India
| | - P. Das
- ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar, Odisha, India
| |
Collapse
|
38
|
Lu P, Li X, Li B, Li X, Wang C, Liu Z, Ji Y, Wang X, Wen Z, Fan J, Yi C, Song M, Wang X. The mitochondrial-derived peptide MOTS-c suppresses ferroptosis and alleviates acute lung injury induced by myocardial ischemia reperfusion via PPARγ signaling pathway. Eur J Pharmacol 2023:175835. [PMID: 37290680 DOI: 10.1016/j.ejphar.2023.175835] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/16/2023] [Accepted: 06/06/2023] [Indexed: 06/10/2023]
Abstract
Acute lung injury (ALI) is a life-threatening complication of cardiac surgery that has a high rate of morbidity and mortality. Epithelial ferroptosis is believed to be involved in the pathogenesis of ALI. MOTS-c has been reported to play a role in regulating inflammation and sepsis-associated ALI. The purpose of this study is to observe the effect of MOTS-c on myocardial ischemia reperfusion (MIR)-induced ALI and ferroptosis. In humans, we used ELISA kits to investigate MOTS-c and malondialdehyde (MDA) levels in patients undergoing off-pump coronary artery bypass grafting (CABG). In vivo, we pretreated Sprague-Dawley rats with MOTS-c, Ferrostatin-1 and Fe-citrate(Ⅲ). We conducted Hematoxylin and Eosin (H&E) staining and detection of ferroptosis-related genes in MIR-induced ALI rats. In vitro, we evaluated the effect of MOTS-c on hypoxia regeneration (HR)-induced mouse lung epithelial-12 (MLE-12) ferroptosis and analyzed the expression of PPARγ through western blotting. We found that circulating MOTS-c levels were decreased in postoperative ALI patients after off-pump CABG, and that ferroptosis contributed to ALI induced by MIR in rats. MOTS-c suppressed ferroptosis and alleviated ALI induced by MIR, and the protective effect of MOTS-c- was dependent on PPARγ signaling pathway. Additionally, HR promoted ferroptosis in MLE-12 cells, and MOTS-c inhibited ferroptosis against HR through the PPARγ signaling pathway. These findings highlight the therapeutic potential of MOTS-c for improving postoperative ALI induced by cardiac surgery.
Collapse
Affiliation(s)
- Peng Lu
- Department of Cardiovascular Surgery, PR China
| | - Xiaopei Li
- Department of Cardiovascular Surgery, PR China
| | - Ben Li
- Department of Cardiovascular Surgery, PR China
| | - Xiangyu Li
- Department of Cardiovascular Surgery, PR China
| | - Chufan Wang
- Department of Cardiovascular Surgery, PR China
| | | | - Yumeng Ji
- Department of Cardiovascular Surgery, PR China
| | - Xufeng Wang
- Department of Cardiovascular Surgery, PR China
| | - Ziang Wen
- Department of Cardiovascular Surgery, PR China
| | - Jidan Fan
- Department of Cardiovascular Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, PR China
| | - Chenlong Yi
- Department of Cardiovascular Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, PR China
| | - Meijuan Song
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China.
| | - Xiaowei Wang
- Department of Cardiovascular Surgery, PR China; Department of Cardiovascular Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, PR China.
| |
Collapse
|
39
|
Li J, Jia YC, Ding YX, Bai J, Cao F, Li F. The crosstalk between ferroptosis and mitochondrial dynamic regulatory networks. Int J Biol Sci 2023; 19:2756-2771. [PMID: 37324946 PMCID: PMC10266069 DOI: 10.7150/ijbs.83348] [Citation(s) in RCA: 100] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 05/14/2023] [Indexed: 06/17/2023] Open
Abstract
Ferroptosis is an iron-driven cell death modality characterized by iron accumulation and excessive lipid peroxidation. Ferroptosis is closely related to mitochondrial function, as indicated by studies showing that mitochondrial dysfunction and damage promote oxidative stress, which in turn induces ferroptosis. Mitochondria play crucial roles in cellular homeostasis, and abnormalities in their morphology and function are closely associated with the development of many diseases. Mitochondria are highly dynamic organelles, and their stability is maintained through a series of regulatory pathways. Mitochondrial homeostasis is dynamically regulated, mainly via key processes such as mitochondrial fission, mitochondrial fusion and mitophagy; however, mitochondrial processes are prone to dysregulation. Mitochondrial fission and fusion and mitophagy are intimately related to ferroptosis. Therefore, investigations into the dynamic regulation of mitochondrial processes during ferroptosis are important to provide a better understanding of the development of disease. In this paper, we systematically summarized changes in ferroptosis, mitochondrial fission and fusion and mitophagy to promote an in-depth understanding of the mechanism underlying ferroptosis and provide a corresponding reference for the treatment of related diseases.
Collapse
Affiliation(s)
- Jie Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China
| | - Yu-chen Jia
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China
| | - Yi-xuan Ding
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China
| | - Jian Bai
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China
| | - Feng Cao
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China
| | - Fei Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China
| |
Collapse
|
40
|
Araki W. Aβ Oligomer Toxicity-Reducing Therapy for the Prevention of Alzheimer's Disease: Importance of the Nrf2 and PPARγ Pathways. Cells 2023; 12:1386. [PMID: 37408220 DOI: 10.3390/cells12101386] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/30/2023] [Accepted: 05/09/2023] [Indexed: 07/07/2023] Open
Abstract
Recent studies have revealed that soluble amyloid-β oligomers (AβOs) play a pathogenetic role in Alzheimer's disease (AD). Indeed, AβOs induce neurotoxic and synaptotoxic effects and are also critically involved in neuroinflammation. Oxidative stress appears to be a crucial event underlying these pathological effects of AβOs. From a therapeutic standpoint, new drugs for AD designed to remove AβOs or inhibit the formation of AβOs are currently being developed. However, it is also worth considering strategies for preventing AβO toxicity itself. In particular, small molecules with AβO toxicity-reducing activity have potential as drug candidates. Among such small molecules, those that can enhance Nrf2 and/or PPARγ activity can effectively inhibit AβO toxicity. In this review, I summarize studies on the small molecules that counteract AβO toxicity and are capable of activating Nrf2 and/or PPARγ. I also discuss how these interrelated pathways are involved in the mechanisms by which these small molecules prevent AβO-induced neurotoxicity and neuroinflammation. I propose that AβO toxicity-reducing therapy, designated ATR-T, could be a beneficial, complementary strategy for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Wataru Araki
- Department of Neurology and Neurological Science, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
- Memory Clinic Ochanomizu, Bunkyo-ku, Tokyo 113-8510, Japan
| |
Collapse
|
41
|
Felipe Souza E Silva L, Siena Dos Santos A, Mayumi Yuzawa J, Luiz de Barros Torresi J, Ziroldo A, Rosado Rosenstock T. SIRTUINS MODULATORS COUNTERACT MITOCHONDRIAL DYSFUNCTION IN CELLULAR MODELS OF HYPOXIA: RELEVANCE TO SCHIZOPHRENIA. Neuroscience 2023:S0306-4522(23)00200-2. [PMID: 37169164 DOI: 10.1016/j.neuroscience.2023.04.027] [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: 10/24/2022] [Revised: 04/16/2023] [Accepted: 04/27/2023] [Indexed: 05/13/2023]
Abstract
Schizophrenia (SZ) is a neurodevelopmental-associated disorder strongly related to environmental factors, such as hypoxia. Because there is no cure for SZ or any pharmacological approach that could revert hypoxia-induced cellular damages, we evaluated whether modulators of sirtuins could abrogate hypoxia-induced mitochondrial deregulation as a neuroprotective strategy. Firstly, astrocytes from control (Wistar) and Spontaneously Hypertensive Rats (SHR), a model of both SZ and neonatal hypoxia, were submitted to chemical hypoxia. Then, cells were exposed to different concentrations of Nicotinamide (NAM), Resveratrol (Resv), and Sirtinol (Sir) for 48hrs. Our data indicate that sirtuins modulation reduces cell death increasing the acetylation of histone 3. This outcome is related to the rescue of loss of mitochondrial membrane potential, changes in mitochondrial calcium buffering capacity, decreased O2-• levels and increased expression of metabolic regulators (Nrf-1 and Nfe2l2) and mitochondrial content. Such findings are relevant not only for hypoxia-associated conditions, named pre-eclampsia but also for SZ since prenatal hypoxia is a relevant environmental factor related to this burdensome neuropsychiatric disorder.
Collapse
Affiliation(s)
- Luiz Felipe Souza E Silva
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Amanda Siena Dos Santos
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Jessica Mayumi Yuzawa
- Department of Physiological Science, Santa Casa de São Paulo School of Medical Science, São Paulo, Brazil
| | | | - Alan Ziroldo
- Department of Physiological Science, Santa Casa de São Paulo School of Medical Science, São Paulo, Brazil
| | - Tatiana Rosado Rosenstock
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil; Dept. of Bioscience, In-vitro Neuroscience, Sygnature Discovery, Nottingham, United Kingdom.
| |
Collapse
|
42
|
Li Q, Zhang W, Cheng N, Zhu Y, Li H, Zhang S, Guo W, Ge G. Pectolinarigenin ameliorates acetaminophen-induced acute liver injury via attenuating oxidative stress and inflammatory response in Nrf2 and PPARa dependent manners. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 113:154726. [PMID: 36863308 DOI: 10.1016/j.phymed.2023.154726] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 01/30/2023] [Accepted: 02/20/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Cirsii Japonici Herba Carbonisata (Dajitan in Chinese) has been used to treat liver disorders in Asian countries. Pectolinarigenin (PEC), an abundant constituent in Dajitan, has been found to possess a wide range of biological benefits, including hepatoprotective effects. However, the effects of PEC on acetaminophen (APAP)-induced liver injury (AILI) and the underlying mechanisms have not been studied. PURPOSES To explore the role and mechanisms of PEC in protecting against AILI. STUDY DESIGN AND METHODS The hepatoprotective benefits of PEC were studied using a mouse model and HepG2 cells. PEC was tested for its effects by injecting it intraperitoneally before APAP administration. To assess liver damage, histological and biochemical tests were performed. The levels of inflammatory factors in the liver were measured using RT-PCR and ELISA. Western blotting was used to measure the expression of a panel of key proteins involved in APAP metabolism, as well as Nrf2 and PPARα. PEC mechanisms on AILI were investigated using HepG2 cells, while the Nrf2 inhibitor (ML385) and PPARα inhibitor (GW6471) were used to validate the importance of either Nrf2 and PPARα in the hepatoprotective effects of PEC. RESULTS PEC treatment decreased serum aspartate aminotransferase (AST), alanine aminotransferase (ALT), tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and interleukin-1β (IL-1β) levels in the liver. PEC pretreatment increased the activity of superoxide dismutase (SOD) and glutathione (GSH) while decreasing malondialdehyde production (MDA). PEC could also up-regulate two important APAP detoxification enzymes (UGT1A1 and SULT1A1). Further research revealed that PEC reduced hepatic oxidative damage and inflammation, and up-regulated APAP detoxification enzymes in hepatocytes by activating the Nrf2 and PPARα signaling pathways. CONCLUSIONS PEC ameliorates AILI by decreasing hepatic oxidative stress and inflammation while increasing phase Ⅱ detoxification enzymes related to APAP harmless metabolism through activation of Nrf2 and PPARα signaling. Hence, PEC may serve as a promising therapeutic drug against AILI.
Collapse
Affiliation(s)
- Qian Li
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| | - Wen Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China; Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University; Henan Engineering Technology Research Center of Organ Transplantation; Henan Research Centre for Organ Transplantation, No. 1, East Jianshe Road, Zhengzhou 450001, China
| | - Nuo Cheng
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University; Henan Engineering Technology Research Center of Organ Transplantation; Henan Research Centre for Organ Transplantation, No. 1, East Jianshe Road, Zhengzhou 450001, China
| | - Yadi Zhu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| | - Hao Li
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University; Henan Engineering Technology Research Center of Organ Transplantation; Henan Research Centre for Organ Transplantation, No. 1, East Jianshe Road, Zhengzhou 450001, China
| | - Shuijun Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University; Henan Engineering Technology Research Center of Organ Transplantation; Henan Research Centre for Organ Transplantation, No. 1, East Jianshe Road, Zhengzhou 450001, China
| | - Wenzhi Guo
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University; Henan Engineering Technology Research Center of Organ Transplantation; Henan Research Centre for Organ Transplantation, No. 1, East Jianshe Road, Zhengzhou 450001, China.
| | - Guangbo Ge
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China.
| |
Collapse
|
43
|
Abstract
ABSTRACT Ferroptosis is an iron-dependent cell death pathway that is different from apoptosis, pyroptosis, and necrosis. The main characteristics of ferroptosis are the Fenton reaction mediated by intracellular free divalent iron ions, lipid peroxidation of cell membrane lipids, and inhibition of the anti-lipid peroxidation activity of intracellular glutathione peroxidase 4 (GPX4). Recent studies have shown that ferroptosis can be involved in the pathological processes of many disorders, such as ischemia-reperfusion injury, nervous system diseases, and blood diseases. However, the specific mechanisms by which ferroptosis participates in the occurrence and development of acute leukemia still need to be more fully and deeply studied. This article reviews the characteristics of ferroptosis and the regulatory mechanisms promoting or inhibiting ferroptosis. More importantly, it further discusses the role of ferroptosis in acute leukemia and predicts a change in treatment strategy brought about by increased knowledge of the role of ferroptosis in acute leukemia.
Collapse
Affiliation(s)
- Tianxin Lyu
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Xudong Li
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan 450008, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yongping Song
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| |
Collapse
|
44
|
Xu M, Che L, Gao K, Wang L, Yang X, Wen X, Li M, Jiang Z. Taurine alleviates oxidative stress in porcine mammary epithelial cells by stimulating the Nrf2-MAPK signaling pathway. Food Sci Nutr 2023; 11:1736-1746. [PMID: 37051345 PMCID: PMC10084955 DOI: 10.1002/fsn3.3203] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 12/04/2022] [Accepted: 12/13/2022] [Indexed: 01/23/2023] Open
Abstract
The high incidence of oxidative stress in sows during late gestation and lactation affects mammary gland health, milk yield, and milk quality. Recently, we found that supplementing maternal diets with 1% taurine improved antioxidant capability and enhanced growth performance in offspring; however, the mechanisms underlying these are unknown. This study aimed to investigate the cytoprotective effects and the mechanism of taurine in mitigating oxidative stress in porcine mammary epithelial cells (PMECs). PMECs were pretreated with 0-2.0 mM taurine for 12 h and then subjected to oxidative injury with 500 μM hydrogen peroxide (H2O2). Pretreatment with taurine attenuated decreased cell viability, enhanced superoxide dismutase, and reduced the intracellular reactive oxygen species accumulation after H2O2 exposure. Taurine also prevented H2O2-induced endoplasmic reticulum stress. Nuclear factor erythroid 2-related factor 2 (Nrf2) was essential to the cytoprotective effects of taurine on PMECs, as Nrf2 knockdown significantly inhibited taurine-induced cytoprotection against oxidative stress. Moreover, we confirmed that Nrf2 induction by taurine was mediated through the inactivation of the p38/MAPK pathway. Overall, taurine supplementation has beneficial effects on redox balance regulation and may protect against oxidative stress in lactating animals.
Collapse
Affiliation(s)
- Mengmeng Xu
- College of Animal Science and TechnologyHenan University of Animal Husbandry and EconomyZhengzhouChina
- State Key Laboratory of Livestock and poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangdong public Laboratory of Animal Breeding and Nutrition, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal ScienceGuangdong Academy of Agricultural SciencesGuangzhouChina
| | - Long Che
- College of Animal Science and TechnologyHenan University of Animal Husbandry and EconomyZhengzhouChina
- State Key Laboratory of Livestock and poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangdong public Laboratory of Animal Breeding and Nutrition, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal ScienceGuangdong Academy of Agricultural SciencesGuangzhouChina
| | - Kaiguo Gao
- State Key Laboratory of Livestock and poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangdong public Laboratory of Animal Breeding and Nutrition, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal ScienceGuangdong Academy of Agricultural SciencesGuangzhouChina
| | - Li Wang
- State Key Laboratory of Livestock and poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangdong public Laboratory of Animal Breeding and Nutrition, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal ScienceGuangdong Academy of Agricultural SciencesGuangzhouChina
| | - Xuefen Yang
- State Key Laboratory of Livestock and poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangdong public Laboratory of Animal Breeding and Nutrition, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal ScienceGuangdong Academy of Agricultural SciencesGuangzhouChina
| | - Xiaolu Wen
- State Key Laboratory of Livestock and poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangdong public Laboratory of Animal Breeding and Nutrition, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal ScienceGuangdong Academy of Agricultural SciencesGuangzhouChina
| | - Mengyun Li
- College of Animal Science and TechnologyHenan University of Animal Husbandry and EconomyZhengzhouChina
| | - Zongyong Jiang
- State Key Laboratory of Livestock and poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangdong public Laboratory of Animal Breeding and Nutrition, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal ScienceGuangdong Academy of Agricultural SciencesGuangzhouChina
| |
Collapse
|
45
|
Kuret T, Kreft ME, Romih R, Veranič P. Cannabidiol as a Promising Therapeutic Option in IC/BPS: In Vitro Evaluation of Its Protective Effects against Inflammation and Oxidative Stress. Int J Mol Sci 2023; 24:ijms24055055. [PMID: 36902479 PMCID: PMC10003465 DOI: 10.3390/ijms24055055] [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: 01/24/2023] [Revised: 02/25/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023] Open
Abstract
Several animal studies have described the potential effect of cannabidiol (CBD) in alleviating the symptoms of interstitial cystitis/bladder pain syndrome (IC/BPS), a chronic inflammatory disease of the urinary bladder. However, the effects of CBD, its mechanism of action, and modulation of downstream signaling pathways in urothelial cells, the main effector cells in IC/BPS, have not been fully elucidated yet. Here, we investigated the effect of CBD against inflammation and oxidative stress in an in vitro model of IC/BPS comprised of TNFα-stimulated human urothelial cells SV-HUC1. Our results show that CBD treatment of urothelial cells significantly decreased TNFα-upregulated mRNA and protein expression of IL1α, IL8, CXCL1, and CXCL10, as well as attenuated NFκB phosphorylation. In addition, CBD treatment also diminished TNFα-driven cellular reactive oxygen species generation (ROS), by increasing the expression of the redox-sensitive transcription factor Nrf2, the antioxidant enzymes superoxide dismutase 1 and 2, and hem oxygenase 1. CBD-mediated effects in urothelial cells may occur by the activation of the PPARγ receptor since inhibition of PPARγ resulted in significantly diminished anti-inflammatory and antioxidant effects of CBD. Our observations provide new insights into the therapeutic potential of CBD through modulation of PPARγ/Nrf2/NFκB signaling pathways, which could be further exploited in the treatment of IC/BPS.
Collapse
|
46
|
El-Beheiry KM, El-Sayed El-Sayad M, El-Masry TA, Elsisi AE. Combination of metformin and hesperidin mitigates cyclophosphamide-induced hepatotoxicity. Emerging role of PPAR-γ/Nrf-2/NF-κB signaling pathway. Int Immunopharmacol 2023; 117:109891. [PMID: 36812672 DOI: 10.1016/j.intimp.2023.109891] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/02/2023] [Accepted: 02/11/2023] [Indexed: 02/22/2023]
Abstract
Cyclophosphamide (CP) is widely used as an immunosuppressive and chemotherapeutic drug. However, its therapeutic application is restricted by its adverse effects, particularly hepatotoxicity. Both metformin (MET) and hesperidin (HES) have promising antioxidant, anti-inflammatory, and anti-apoptotic effects. Therefore, the principal aim of the current study is to investigate the hepatoprotective effects of MET, HES, and their combinations on the CP-induced hepatotoxicity model. Hepatotoxicity was evoked by a single (I.P) injection of CP (200 mg/kg) on day 7. For this study, 64 albino rats were randomly categorized into eight equal groups; naïve, control vehicle, untreated CP (200 mg/kg, IP), and CP 200 groups treated with MET 200, HES 50, HES 100 or a combination of MET 200 with HES 50 and HES 100 respectively orally daily for 12 days. At the end of the study, the liver function biomarkers, oxidative stress, inflammatory parameters, histopathological and immunohistochemical analysis of PPAR-γ, Nrf-2, NF-κB, Bcl-2, and caspase3 were assessed. CP significantly increased serum ALT, AST, total bilirubin, hepatic MDA, NO content, NF-κB, and TNF-α. Otherwise, albumin, hepatic GSH content, Nrf-2, and PPAR-γ expression decreased considerably compared to the control vehicle group. The combinations of MET200 with HES50 or HES100 induced pronounced hepatoprotective, anti-oxidative, anti-inflammatory, and anti-apoptotic effects on CP-treated rats. The possible explanation of such hepatoprotective effects may be mediated via upregulation of Nrf-2, PPAR-γ, Bcl-2 expression, hepatic GSH content, and marked suppression of TNF-α and NF-κB expression. In conclusion, the current study showed that combining MET and HES revealed a remarkable hepatoprotective effect against CP-induced hepatotoxicity.
Collapse
Affiliation(s)
- Kareman M El-Beheiry
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt.
| | - Magda El-Sayed El-Sayad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Thanaa A El-Masry
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Alaa E Elsisi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
47
|
Jîtcă G, Ősz BE, Vari CE, Rusz CM, Tero-Vescan A, Pușcaș A. Cannabidiol: Bridge between Antioxidant Effect, Cellular Protection, and Cognitive and Physical Performance. Antioxidants (Basel) 2023; 12:antiox12020485. [PMID: 36830042 PMCID: PMC9952814 DOI: 10.3390/antiox12020485] [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: 01/13/2023] [Revised: 02/08/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
The literature provides scientific evidence for the beneficial effects of cannabidiol (CBD), and these effects extend beyond epilepsy treatment (e.g., Lennox-Gastaut and Dravet syndromes), notably the influence on oxidative status, neurodegeneration, cellular protection, cognitive function, and physical performance. However, products containing CBD are not allowed to be marketed everywhere in the world, which may ultimately have a negative effect on health as a result of the uncontrolled CBD market. After the isolation of CBD follows the discovery of CB1 and CB2 receptors and the main enzymatic components (diacylglycerol lipase (DAG lipase), monoacyl glycerol lipase (MAGL), fatty acid amino hydrolase (FAAH)). At the same time, the antioxidant potential of CBD is due not only to the molecular structure but also to the fact that this compound increases the expression of the main endogenous antioxidant systems, superoxide dismutase (SOD), and glutathione peroxidase (GPx), through the nuclear complex erythroid 2-related factor (Nrf2)/Keep1. Regarding the role in the control of inflammation, this function is exercised by inhibiting (nuclear factor kappa B) NF-κB, and also the genes that encode the expression of molecules with a pro-inflammatory role (cytokines and metalloproteinases). The other effects of CBD on cognitive function and physical performance should not be excluded. In conclusion, the CBD market needs to be regulated more thoroughly, given the previously listed properties, with the mention that the safety profile is a very good one.
Collapse
Affiliation(s)
- George Jîtcă
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540139 Târgu Mureș, Romania
| | - Bianca E. Ősz
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540139 Târgu Mureș, Romania
- Correspondence:
| | - Camil E. Vari
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540139 Târgu Mureș, Romania
| | - Carmen-Maria Rusz
- Doctoral School of Medicine and Pharmacy, I.O.S.U.D, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540139 Târgu Mureș, Romania
| | - Amelia Tero-Vescan
- Department of Biochemistry, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540139 Târgu Mureș, Romania
| | - Amalia Pușcaș
- Department of Biochemistry, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540139 Târgu Mureș, Romania
| |
Collapse
|
48
|
Diversified Effects of COVID-19 as a Consequence of the Differential Metabolism of Phospholipids and Lipid Peroxidation Evaluated in the Plasma of Survivors and Deceased Patients upon Admission to the Hospital. Int J Mol Sci 2022; 23:ijms231911810. [PMID: 36233111 PMCID: PMC9570244 DOI: 10.3390/ijms231911810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/29/2022] [Accepted: 10/03/2022] [Indexed: 12/03/2022] Open
Abstract
As a result of SARS-CoV-2 infection, inflammation develops, which promotes oxidative stress, leading to modification of phospholipid metabolism. Therefore, the aim of this study is to compare the effects of COVID-19 on the levels of phospholipid and free polyunsaturated fatty acids (PUFAs) and their metabolites produced in response to reactions with reactive oxygen species (ROS) and enzymes (cyclooxygenases-(COXs) and lipoxygenase-(LOX)) in the plasma of patients who either recovered or passed away within a week of hospitalization. In the plasma of COVID-19 patients, especially of the survivors, the actions of ROS and phospholipase A2 (PLA2) cause a decrease in phospholipid fatty acids level and an increase in free fatty acids (especially arachidonic acid) despite increased COXs and LOX activity. This is accompanied by an increased level in lipid peroxidation products (malondialdehyde and 8-isoprostaglandin F2α) and lipid mediators generated by enzymes. There is also an increase in eicosanoids, both pro-inflammatory as follows: thromboxane B2 and prostaglandin E2, and anti-inflammatory as follows: 15-deoxy-Δ-12,14-prostaglandin J2 and 12-hydroxyeicosatetraenoic acid, as well as endocannabinoids (anandamide-(AEA) and 2-arachidonylglycerol-(2-AG)) observed in the plasma of patients who recovered. Moreover, the expression of tumor necrosis factor α and interleukins (IL-6 and IL-10) is increased in patients who recovered. However, in the group of patients who died, elevated levels of N-oleoylethanolamine and N-palmitoylethanolamine are found. Since lipid mediators may have different functions depending on the onset of pathophysiological processes, a stronger pro-inflammatory response in patients who have recovered may be the result of the defensive response to SARS-CoV-2 in survivors associated with specific changes in the phospholipid metabolism, which could also be considered a prognostic factor.
Collapse
|
49
|
Meher A. Role of Transcription Factors in the Management of Preterm Birth: Impact on Future Treatment Strategies. Reprod Sci 2022; 30:1408-1420. [PMID: 36131222 DOI: 10.1007/s43032-022-01087-7] [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: 06/01/2022] [Accepted: 09/15/2022] [Indexed: 10/14/2022]
Abstract
Preterm birth is defined as the birth of a neonate before 37 weeks of gestation and is considered as a leading cause of the under five deaths of neonates. Neonates born preterm are known to have higher perinatal mortality and morbidity with associated risks of low birth weight, respiratory distress syndrome, gastrointestinal, immunologic, central nervous system, hearing, and vision problems, cerebral palsy, and delayed development. India leads the list of countries with the greatest number of preterm births. The studies focusing on the molecular mechanisms related to the etiology of preterm birth have described the role of different transcription factors. With respect to this, transcription factors like peroxisome proliferator activated receptors (PPAR), nuclear factor kappa β (NF-kβ), nuclear erythroid 2-related factor 2 (Nrf2), and progesterone receptor (PR) are known to be associated with preterm labor. All these transcription factors are linked together with a common cascade involving inflammatory processes. Thus, the current review describes the possible cross-talk between these transcription factors and their therapeutic potential to prevent or manage preterm labor.
Collapse
Affiliation(s)
- Akshaya Meher
- Central Research Laboratory, Dr. Vasantrao Pawar Medical College, Hospital and Research Centre, Nashik, Maharashtra, India, 422003.
| |
Collapse
|
50
|
Enayati A, Ghojoghnejad M, Roufogalis BD, Maollem SA, Sahebkar A. Impact of Phytochemicals on PPAR Receptors: Implications for Disease Treatments. PPAR Res 2022; 2022:4714914. [PMID: 36092543 PMCID: PMC9453090 DOI: 10.1155/2022/4714914] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 07/10/2022] [Indexed: 11/17/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are members of the ligand-dependent nuclear receptor family. PPARs have attracted wide attention as pharmacologic mediators to manage multiple diseases and their underlying signaling targets. They mediate a broad range of specific biological activities and multiple organ toxicity, including cellular differentiation, metabolic syndrome, cancer, atherosclerosis, neurodegeneration, cardiovascular diseases, and inflammation related to their up/downstream signaling pathways. Consequently, several types of selective PPAR ligands, such as fibrates and thiazolidinediones (TZDs), have been approved as their pharmacological agonists. Despite these advances, the use of PPAR agonists is known to cause adverse effects in various systems. Conversely, some naturally occurring PPAR agonists, including polyunsaturated fatty acids and natural endogenous PPAR agonists curcumin and resveratrol, have been introduced as safe agonists as a result of their clinical evidence or preclinical experiments. This review focuses on research on plant-derived active ingredients (natural phytochemicals) as potential safe and promising PPAR agonists. Moreover, it provides a comprehensive review and critique of the role of phytochemicals in PPARs-related diseases and provides an understanding of phytochemical-mediated PPAR-dependent and -independent cascades. The findings of this research will help to define the functions of phytochemicals as potent PPAR pharmacological agonists in underlying disease mechanisms and their related complications.
Collapse
Affiliation(s)
- Ayesheh Enayati
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mobina Ghojoghnejad
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Basil D. Roufogalis
- Discipline of Pharmacology, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
- NICM Health Research Institute, Western Sydney University, Penrith, NSW, Australia
| | - Seyed Adel Maollem
- Department of Pharmacology and Toxicology, College of Pharmacy, Al-Zahraa University for Women, Karbala, Iraq
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - 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
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|