1
|
Kamata S, Honda A, Yashiro S, Kaneko C, Komori Y, Shimamura A, Masuda R, Oyama T, Ishii I. Competitive Ligand-Induced Recruitment of Coactivators to Specific PPARα/δ/γ Ligand-Binding Domains Revealed by Dual-Emission FRET and X-Ray Diffraction of Cocrystals. Antioxidants (Basel) 2025; 14:494. [PMID: 40298866 PMCID: PMC12024163 DOI: 10.3390/antiox14040494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/18/2025] [Accepted: 04/18/2025] [Indexed: 04/30/2025] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs), composed of the α/δ/γ subtypes, are ligand-activated nuclear receptors/transcription factors that sense endogenous fatty acids or therapeutic drugs to regulate lipid/glucose metabolism and oxidative stress. PPAR forms a multiprotein complex with a retinoid X receptor and corepressor complex in an unliganded/inactive state, and ligand binding induces the replacement of the corepressor complex with the coactivator complex to initiate the transcription of various genes, including the metabolic and antioxidant ones. We investigated the processes by which the corepressor is replaced with the coactivator or in which two coactivators compete for the PPARα/δ/γ-ligand-binding domains (LBDs) using single- and dual-emission fluorescence resonance energy transfer (FRET) assays. Single-FRET revealed that the respective PPARα/δ/γ-selective agonists (pemafibrate, seladelpar, and pioglitazone) induced the dissociation of the two corepressor peptides, NCoR1 and NCoR2, from the PPARα/δ/γ-LBDs and the recruitment of the two coactivator peptides, CBP and TRAP220. Meanwhile, dual-FRET demonstrated that these processes are simultaneous and that the four coactivator peptides, CBP, TRAP220, PGC1α, and SRC1, were competitively recruited to the PPARα/δ/γ-LBDs with different preferences upon ligand activation. Furthermore, the five newly obtained cocrystal structures using X-ray diffraction, PPARα-LBDs-NCoR2/CBP/TRAP220/PGC1α and PPARγ-LBD-NCoR2, were co-analyzed with those from our previous studies. This illustrates that these coactivators bound to the same PPARα-LBD loci via their consensus LXXLL motifs in the liganded state; that NCoR1/NCoR2 corepressors bound to the same loci via the IXXXL sequences within their consensus LXXXIXXXL motifs in the unliganded state; and that ligand activation induced AF-2 helix 12 formation that interfered with corepressor binding and created a binding space for the coactivator. These PPARα/γ-related biochemical and physicochemical findings highlight the coregulator dynamics on limited PPARα/δ/γ-LBDs loci.
Collapse
Affiliation(s)
- Shotaro Kamata
- Department of Health Chemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Akihiro Honda
- Department of Health Chemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Sayaka Yashiro
- Department of Health Chemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Chihiro Kaneko
- Department of Health Chemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Yuna Komori
- Department of Health Chemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Ayumi Shimamura
- Department of Health Chemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Risa Masuda
- Department of Health Chemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Takuji Oyama
- Faculty of Life and Environmental Sciences, University of Yamanashi, Kofu, Yamanashi 400-8510, Japan
| | - Isao Ishii
- Department of Health Chemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| |
Collapse
|
2
|
Arafa ESA, Hassanein EHM, Hussein RM, Mohamed WR. Impact of lisinopril on cisplatin-induced inflammation, oxidative stress, apoptosis, and impaired steroidogenesis in rat testis: involvement of Nrf2/Keap1/HO-1 and PPARγ signaling. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03924-3. [PMID: 40009171 DOI: 10.1007/s00210-025-03924-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 02/12/2025] [Indexed: 02/27/2025]
Abstract
Testicular dysfunction is a significant long-term side effect of the chemotherapeutic drug cisplatin (CDDP), primarily due to DNA damage and oxidative stress. Lisinopril (LSP), an angiotensin-converting enzyme (ACE) inhibitor commonly used for hypertension treatment, has a debated impact on reproductive function. This study investigates, for the first time, the ability of LSP to counteract CDDP-induced inflammation, oxidative stress, apoptosis, and steroidogenic disturbances in rat testis. In addition, LSP's effect on testicular Nrf2/Keap1/HO-1 and PPARγ signaling is examined. Rats were divided into Control, LSP, CDDP, and LSP + CDDP groups. Rats were treated with 10 mg/kg of LSP orally for 10 days, and blood and testis samples were collected after sacrifice for histopathological, biochemical, and genetic analysis. Our results revealed that LSP administration with CDDP effectively increased luteinizing, follicle-stimulating, and testosterone hormone levels (effect size f = 2.56, 2.32, and 3.02; respectively, and power = 1.00) and upregulated testicular expression of CYP11a1, HSD17B3, and StAR genes. LSP counteracted the histopathological aberrations induced by CDDP. The LSP + CDDP group also showed increased levels of reduced glutathione and superoxide dismutase (effect size f = 1.72 and power = 0.99) and decreased levels of malondialdehyde (effect size f = 3.07 and power = 1), interleukin-1β, tumor necrosis factor-α, interleukin-6, nuclear factor kappa B, cyclooxygenase-2, and cleaved caspase 3 (effect size f = 4.61 and power = 1). On the molecular level, the LSP + CDDP group showed a reduction in Keap1 protein level but an increase in Nrf2 (effect size f = 5.50 and power = 1), HO-1 (effect size f = 3.66 and power = 1), and PPARγ protein levels, compared to the CDDP group. In conclusion, LSP revealed prominent anti-oxidant, anti-apoptotic, and anti-inflammatory effects protecting against CDDP-induced testicular damage. Moreover, it preserved the steroidogenic process and testicular tissue characteristics. LSP modulated the expression of Nrf2/Keap1/HO-1 and PPARγ signaling. Therefore, our data presents LSP as a promising candidate for enhancing reproductive health in patients undergoing CDDP treatment.
Collapse
Affiliation(s)
- El-Shaimaa A Arafa
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman, United Arab Emirates.
- Centre of Medical and Bio-Allied Health Sciences Research (CMBAHSR), Ajman University, Ajman, United Arab Emirates.
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt.
| | - Emad H M Hassanein
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Rasha M Hussein
- Department of Biochemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt
- Department of Clinical Pharmacy, Faculty of Pharmacy, Mutah University, Al-Karak, 61710, Jordan
| | - Wafaa R Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt.
| |
Collapse
|
3
|
Alfwuaires MA. Rosmarinic acid protects against cyclophosphamide-induced hepatotoxicity via inhibition of oxidative stress, inflammation, and apoptosis and upregulation of Nrf2 in mice. J Mol Histol 2024; 56:49. [PMID: 39702535 DOI: 10.1007/s10735-024-10290-6] [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: 09/17/2024] [Accepted: 10/29/2024] [Indexed: 12/21/2024]
Abstract
Cyclophosphamide (CP) is widely used in chemotherapy to treat various types of cancer. However, it is toxic to the liver and other organs. Rosmarinic acid (RA) possesses anti-inflammatory, antioxidant, and cytoprotective properties. This study investigated the protective effects of RA against CP-induced liver injury in mice. Mice were treated with RA (25, 50, and 100 mg/kg) for 15 days and followed by a single injection of CP on day 16th. CP injection resulted in an elevation in serum AST, ALT, and ALP, along with multiple histopathological alterations in the liver. CP also induced increased levels of MDA and NO, associated with declined GSH, SOD and CAT. RA pretreatment prevented liver injury, alleviated the enhanced levels of MDA and NO, and restored antioxidants defenses, hence avoiding the oxidative injury in the liver. Moreover, RA pretreatment attenuated NF-κB p65 and proinflammatory cytokines levels. Liver of CP-injected mice also showed a decrease in Bcl2, accompanied with elevated BAX and caspase-3 expression, an effect that RA pretreatment alleviated. In addition, pretreatment of CP-administrated mice with RA restored the Nrf2 expression in the liver. Taken together, this study suggests a potential application value of RA in preventing CP hepatotoxicity and sheds light on the possible mechanism.
Collapse
Affiliation(s)
- Manal A Alfwuaires
- Department of Biological Sciences, Faculty of Science, King Faisal University, 31982, Al Hofuf, Al-Ahsa, Saudi Arabia.
| |
Collapse
|
4
|
Alruhaimi RS, Hassanein EHM, Ahmeda AF, Alnasser SM, Atwa AM, Sabry M, Alzoghaibi MA, Mahmoud AM. Attenuation of inflammation, oxidative stress and TGF-β1/Smad3 signaling and upregulation of Nrf2/HO-1 signaling mediate the protective effect of diallyl disulfide against cadmium nephrotoxicity. Tissue Cell 2024; 91:102576. [PMID: 39353227 DOI: 10.1016/j.tice.2024.102576] [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: 08/20/2024] [Revised: 09/25/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Abstract
Heavy metals are toxic environmental pollutants with serious health effects on humans and animals. Cadmium (Cd) is known for its serious nephrotoxic effect and its toxicity involves oxidative stress (OS) and inflammation. Diallyl disulfide (DADS), a main constituent of garlic, exhibites cytoprotective and antioxidant activities. This study investigated the effect of DADS on OS, inflammation, and fibrosis induced by Cd in rat kidney, pointing to the involvement of transforming growth factor-β (TGF-β)/Smad3 and nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) signaling, and peroxisome proliferator-activated receptor gamma (PPARγ). Rats received DADS for 14 days and Cd on day 7 and blood and kidney samples were collected. Cd elevated serum creatinine, urea and uric acid, provoked kidney histopathological alterations and collagen deposition, increased kidney malondialdehyde (MDA) level, and decreased glutathione (GSH) and antioxidant enzymes. Nuclear factor-kappaB (NF-κB) p65, interleukin (IL)-6, tumor necrosis factor (TNF)-α, IL-1β, and CD68 were upregulated in Cd-administered rat kidney. DADS prevented kidney injury, mitigated OS, suppressed NF-κB, CD68 and pro-inflammatory mediators, and boosted antioxidants. DADS downregulated TGF-β1, Smad3 phosphorylation and Kelch-like ECH-associated protein-1 (Keap1), and increased Nrf2, HO-1, cytoglobin, and PPARγ. In conclusion, DADS protects the kidney against Cd toxicity by attenuating OS, inflammation, and TGF-β1/Smad3 signaling, and enhancement of Nrf2/HO-1 signaling, antioxidants, and PPARγ.
Collapse
Affiliation(s)
- Reem S Alruhaimi
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia
| | - Emad H M Hassanein
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University-Assiut Branch, Assiut 71524, Egypt
| | - Ahmad F Ahmeda
- Department of Basic Medical Sciences, College of Medicine, Ajman University, Ajman 346, United Arab Emirates; Center of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman 346, United Arab Emirates
| | - Sulaiman M Alnasser
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia
| | - Ahmed M Atwa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo 11829, Egypt
| | - Mostafa Sabry
- Department of Biochemistry, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
| | - Mohammed A Alzoghaibi
- Physiology Department, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
| | - Ayman M Mahmoud
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester M1 5GD, UK; Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef 62514, Egypt.
| |
Collapse
|
5
|
Liu Z, Zheng J, Ding T, Chen H, Wan R, Zhang X, Zhang W. HIF-1α protects nucleus pulposus cells from oxidative stress-induced mitochondrial impairment through PDK-1. Free Radic Biol Med 2024; 224:39-49. [PMID: 39128487 DOI: 10.1016/j.freeradbiomed.2024.08.007] [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: 07/07/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
The pathogenesis of intervertebral disc degeneration (IVDD) involves complex signaling networks and various effector molecules, and our understanding of the pathogenesis of IVDD is limited. Hypoxia inducible factor-1α (HIF-1α) is closely related to IVDD, and there is excessive oxidative stress concurrent with IVDD. In this study, we found that HIF-1α could protect nucleus pulposus cells from excessive oxidative stress by reversing the imbalance between oxidants and antioxidants and thus mitigating the oxidative stress-induced mitochondrial impairment. With further exploration, we found that pyruvate dehydrogenase kinase 1 (PDK-1) was involved in the protective effect of HIF-1α on nucleus pulposus cells under oxidative stress. We suggested that HIF-1α could preserve the mitochondrial integrity and activate glycolysis in nucleus pulposus cells via PDK-1, and the addition of DCA, a PDK-1 inhibitor, could blunt the protective effect of HIF-1α. In addition, the HIF-1α/PDK-1 regulatory axis was also confirmed in vivo through HIF-1α knockout mice model. Therefore, we propose that HIF-1α protects nucleus pulposus cells from excessive oxidative stress by maintaining the mitochondrial integrity and glycolysis via PDK-1, thus enriching the insight into the protective mechanism of HIF-1α against IVDD, and providing a novel therapeutic target for the treatment of IVDD.
Collapse
Affiliation(s)
- Zhuochao Liu
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Jiancheng Zheng
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Tao Ding
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Haoyi Chen
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Rong Wan
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| | - Xingkai Zhang
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; Department of Orthopedics, Ruijin Hospital Wuxi Branch, Shanghai Jiao Tong University School of Medicine, Jiangsu, PR China.
| | - Weibin Zhang
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| |
Collapse
|
6
|
Abou Taha MA, Ali FEM, Saleh IG, Akool ES. Sorafenib and edaravone protect against renal fibrosis induced by unilateral ureteral obstruction via inhibition of oxidative stress, inflammation, and RIPK-3/MLKL pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8961-8977. [PMID: 38874805 PMCID: PMC11522075 DOI: 10.1007/s00210-024-03146-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/05/2024] [Indexed: 06/15/2024]
Abstract
Renal fibrosis is the common endpoint of nearly all chronic and progressive nephropathies. Cell death and sterile inflammation are the main characteristics of renal fibrosis, which can lead to end-stage renal failure. The inflammatory reaction triggered by tissue damage is strongly related to necroptosis, a type of caspase-independent, regulated cell death. Using an animal model of unilateral ureteral obstruction (UUO), the anti-fibrotic effects of sorafenib (SOF), a multi-kinase inhibitor, and edaravone (EDV), a potent antioxidant and free radical scavenger, were examined in rats with obstructive nephropathy. Experimentally, animals were divided randomly into five groups: sham; UUO; UUO + SOF (5 mg/kg/day, P.O.); UUO + EDV (20 mg/kg/day, P.O.); and UUO + SOF + EDV groups. The kidney function biomarkers, oxidant/antioxidant status, renal mRNA expressions of TNF-α, collagen-1α, protein expressions of RIPK-1, RIPK-3, MLKL, caspase-8, HYP, MPO, and TNF-α were all significantly modulated by UUO. Administration of either SOF or EDV significantly attenuated cellular and molecular changes induced by UUO. Also, histopathological changes were improved. Moreover, SOF in combination with EDV, significantly improved UUO-induced renal fibrosis compared with each drug alone. Collectively, administration of either SOF or EDV or both of them significantly attenuated the rats with obstructive nephropathy, possibly by blocking the RIPK-3/MLKL necroptotic pathway and suppressing renal oxidative stress and inflammation.
Collapse
Affiliation(s)
- Mohamed A Abou Taha
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University Assiut Branch, Assiut, 71524, Egypt
| | - Fares E M Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University Assiut Branch, Assiut, 71524, Egypt.
| | - Ibrahim G Saleh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
- Department of Pharmacy Practice, Faculty of Pharmacy, Sinai University, Kantara, Ismailia, Egypt
| | - El-Sayed Akool
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
7
|
Alruhaimi RS, Hassanein EHM, Ahmeda AF, Atwa AM, Alnasser SM, Sayed GA, Alotaibi M, Alzoghaibi MA, Mahmoud AM. Farnesol attenuates cadmium-induced kidney injury by mitigating oxidative stress, inflammation and necroptosis and upregulating cytoglobin and PPARγ in rats. Tissue Cell 2024; 90:102526. [PMID: 39181090 DOI: 10.1016/j.tice.2024.102526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/11/2024] [Accepted: 08/16/2024] [Indexed: 08/27/2024]
Abstract
Heavy metals are environmental pollutants that can harm animals and humans even at low concentrations. Cadmium (Cd) is known for its serious health effects on different organs and its toxicity is associated with oxidative stress (OS) and inflammation. Farnesol (FAR), a sesquiterpene alcohol found in many vegetables and fruits, possesses promising anti-inflammatory and antioxidant activities. This study evaluated the effect of FAR on Cd-induced kidney injury, pinpointing its effect of the redox status, inflammation, fibrosis and necroptosis. Rats in this study received FAR for 14 days and Cd on day 7. Elevated serum creatinine, urea and uric acid, and several kidney histopathological alterations were observed in Cd-administered rats. Cd increased MDA, decreased antioxidants, downregulated PPARγ and upregulated NF-κB p65, IL-6, TNF-α, and IL-1β. Necroptosis mediators (RIP1, RIP3, MLKL, and caspase-8) and α-SMA were upregulated, and collagen deposition was increased in Cd-administered rats. FAR ameliorated kidney injury markers and tissue damage, attenuated OS, suppressed NF-κB and inflammatory mediators, and enhanced antioxidants. In addition, FAR suppressed RIP1, RIP3, MLKL, caspase-8, and α-SMA, and enhanced kidney cytoglobin and PPARγ. In conclusion, FAR protects against Cd nephrotoxicity by suppressing OS, inflammatory response and necroptosis, effects associated with enhanced antioxidants, cytoglobin, and PPARγ.
Collapse
Affiliation(s)
- Reem S Alruhaimi
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia
| | - Emad H M Hassanein
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University-Assiut Branch, Egypt
| | - Ahmad F Ahmeda
- Department of Basic Medical Sciences, College of Medicine, Ajman University, Ajman 346, United Arab Emirates; Center of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman 346, United Arab Emirates
| | - Ahmed M Atwa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo 11829, Egypt
| | - Sulaiman M Alnasser
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia
| | - Ghadir A Sayed
- Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Cairo 11829, Egypt
| | - Meshal Alotaibi
- Department of Pharmacy Practice, College of Pharmacy, University of Hafr Albatin, Hafar Al Batin 39524, Saudi Arabia
| | - Mohammed A Alzoghaibi
- Physiology Department, College of Medicine, King Saud University, Riyadh, 11461, Saudi Arabia
| | - Ayman M Mahmoud
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester M1 5GD, UK; Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef 62514, Egypt.
| |
Collapse
|
8
|
Paoli S, Eidelman DH, Mann KK, Baglole C. Sex-specific alterations in pulmonary metabolic, xenobiotic and lipid signalling pathways after e-cigarette aerosol exposure during adolescence in mice. BMJ Open Respir Res 2024; 11:e002423. [PMID: 39299769 PMCID: PMC11418501 DOI: 10.1136/bmjresp-2024-002423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 08/19/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND E-cigarette use is now prevalent among adolescents and young adults, raising concerns over potential adverse long-term health effects. Although it is hypothesised that e-cigarettes promote inflammation, studies have yielded conflicting evidence. Our previous work showed that JUUL, a popular e-cigarette brand, elicited minimal lung inflammation but induced significant molecular changes in adult C57BL/6 mice. METHODS Now, we have profiled immunological and proteomic changes in the lungs of adolescent male and female BALB/c and C57BL/6 mice exposed to a flavoured JUUL aerosol containing 18 mg/mL of nicotine for 14 consecutive days. We evaluated changes in the immune composition by flow cytometry, gene expression levels by reverse transcription-quantitative PCR and assessed the proteomic profile of the lungs and bronchoalveolar lavage (BAL) by tandem mass tag-labelled mass spectroscopy. RESULTS While there were few significant changes in the immune composition of the lungs, proteomic analysis revealed that JUUL exposure caused significant sex-dependent and strain-dependent differences in lung and BAL proteins that are implicated in metabolic pathways, including those related to lipids and atherosclerosis, as well as pathways related to immune function and response to xenobiotics. Notably, these changes were more pronounced in male mice. CONCLUSIONS These findings raise the possibility that vaping dysregulates numerous biological responses in lungs that may affect disease risk, disproportionally impacting males and raising significant concerns for the future health of male youth who currently vape.
Collapse
Affiliation(s)
- Sofia Paoli
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - David H Eidelman
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Koren K Mann
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Lady Davis Institute for Medical Research, Montreal, Quebec, Canada
| | - Carolyn Baglole
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Department of Medicine, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
9
|
Sultan WS, Mahmoud AM, Ahmed SA, Alruhaimi RS, Alzoghaibi MA, El-Bassuony AA, Hasona NA, Kamel EM. Phytochemical Analysis and Anti-dyslipidemia and Antioxidant Activities of Pluchea dioscoridis: In Vitro, In Silico and In Vivo Studies. Chem Biodivers 2024; 21:e202400842. [PMID: 38884416 DOI: 10.1002/cbdv.202400842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/15/2024] [Accepted: 06/17/2024] [Indexed: 06/18/2024]
Abstract
Pluchea dioscoridis (L.) DC. is a flowering wild plant used traditionally in the treatment of rhematic disorders. This study investigated the phytochemical and in vitro radical scavenging activity (RSA), and in vivo anti-hyperlipidemic, antioxidant and anti-inflammatory properties of P. dioscoridis. The antihyperlipidemic efficacy was determined in a rat model of dyslipidemia. The extract and fractions of P. dioscoridis showed RSA with the ethyl acetate (EA) fraction exhibiting the most potent activity. The phytochemical analysis of P. dioscoridis EA fraction (PDEAF) led to the isolation of five compounds (lupeol, quercetin, lupeol acetate, stigmasterol, and syringic acid). To evaluate its anti-hyperlipidemic effect, three doses of PDEAF were supplemented to rats for 14 days and poloxamer-407 was administered on day 15 to induce dyslipidemia. All doses of PDEAF decreased plasma triglycerides, cholesterol, low-density lipoprotein-cholesterol (LDL-C) and very low-density lipoprotein-cholesterol (vLDL-C), and increased plasma lipoprotein lipase (LPL). PDEAF upregulated hepatic LDL receptor and suppressed 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase, decreased lipid peroxidation and tumor necrosis factor (TNF)-α and enhanced reduced glutathione (GSH) and enzymatic antioxidants in dyslipidmeic rats. In silico findings revealed the binding affinity of the isolated compounds towards LPL, HMG-CoA reductase, and LDL receptor. In conclusion, P. dioscoridis is rich in phytoconstituents, exhibited RSA and its EA fraction effectively prevented acute dyslipidemia and its associated oxidative stress and inflammatory response.
Collapse
Affiliation(s)
- Wageha S Sultan
- Department of Chemistry, Research Institute of Medicinal and Aromatic Plants, Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Ayman M Mahmoud
- Department of Life Sciences, Faculty of Science & Engineering, Manchester Metropolitan University, Manchester, M1 5GD, UK
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Shimaa A Ahmed
- Chemistry Department, Faculty of Science, Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Reem S Alruhaimi
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, 11671, Saudi Arabia
| | - Mohammed A Alzoghaibi
- Physiology Department, College of Medicine, King Saud University, Riyadh, 11461, Saudi Arabia
| | - Ashraf A El-Bassuony
- Chemistry Department, Faculty of Science, Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Nabil A Hasona
- Biochemistry Department, Faculty of Science, Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Emadeldin M Kamel
- Chemistry Department, Faculty of Science, Beni-Suef University, Beni-Suef, 62514, Egypt
| |
Collapse
|
10
|
Ahmed SA, Kamel EM, Mahmoud AM, Nasr HMD, Hassan HM, Alanazi MM, Rateb ME, Hozayen WG, Ahmed SA. Phytochemical Analysis, and Antioxidant and Hepatoprotective Activities of Chamaerops humilis L. Leaves; A Focus on Xanthine Oxidase. Chem Biodivers 2024; 21:e202400865. [PMID: 38867399 DOI: 10.1002/cbdv.202400865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/14/2024]
Abstract
Chamaerops humilis L. is clumping palm of the family Arecaceae with promising health-promoting effects. Parts of this species are utilized as food and employed in folk medicine to treat several disorders. This study investigated the phytochemical constituents of C. humilis leaves and their antioxidant and xanthine oxidase (XO) inhibitory activities in vitro and in vivo in acetaminophen (APAP)-induced hepatotoxicity in rats. The chemical structure of the isolated phytochemicals was determined using data obtained from UV, MS, IR, and 1H-, 13C-NMR spectroscopic tools as well as comparison with authentic markers. Eleven compounds, including tricin 7-O-β-rutinoside, vicenin, tricin, astragalin, borassoside D, pregnane-3,5,6,16-tetrol, oleanolic acid, β-sitosterol and campesterol were isolated from C. humilis ethanolic extract (CHEE). CHEE and the butanol, n-hexane, and dichloromethane fractions exhibited in vitro radical scavenging and XO inhibitory efficacies. The computational findings revealed the tendency of the isolated compounds towards the active site of XO. In vivo, CHEE ameliorated liver function markers and prevented tissue injury induced by APAP in rats. CHEE suppressed hepatic XO, decreased serum uric acid and liver malondialdehyde (MDA), and enhanced reduced glutathione (GSH), superoxide dismutase (SOD), and catalase in APAP-treated rats. CHEE ameliorated serum tumor necrosis factor alpha (TNF-α) and interleukin (IL)-1β in APAP-treated rats. Thus, C. humilis is rich in beneficial phytochemicals that possess binding affinity towards XO. C. humilis exhibited potent in vitro antioxidant and XO inhibitory activities, and prevented APAP hepatotoxicity by attenuating tissue injury, oxidative stress and inflammation.
Collapse
Affiliation(s)
- Shimaa A Ahmed
- Department of Chemistry, Faculty of Science, Beni-Suef University, Beni-Seuf, 62514, Egypt
| | - Emadeldin M Kamel
- Department of Chemistry, Faculty of Science, Beni-Suef University, Beni-Seuf, 62514, Egypt
| | - Ayman M Mahmoud
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, M1 5GD, UK
| | - Hamdi M D Nasr
- Department of Chemistry, Faculty of Science, Al-Azhar University (Assiut), Assiut, 71524, Egypt
| | - Hossam M Hassan
- Department of Pharmacognosy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Mohammed M Alanazi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mostafa E Rateb
- Department of Pharmacognosy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Walaa G Hozayen
- Department of Biochemistry, Faculty of Science, Beni-Suef University, Beni-Seuf, 62514, Egypt
| | - Sayed A Ahmed
- Department of Chemistry, Faculty of Science, Beni-Suef University, Beni-Seuf, 62514, Egypt
| |
Collapse
|
11
|
Mohammadi T. Effect of quercetin and mirtazapine on spermatogenesis and testis structure in phenylhydrazine-induced hemolytic anemia mice: An experimental study. Food Chem Toxicol 2024; 189:114732. [PMID: 38740240 DOI: 10.1016/j.fct.2024.114732] [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: 03/18/2024] [Revised: 04/30/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024]
Abstract
Anemia poses a significant healthcare challenge across different socioeconomic groups and can result in reproductive system damage through the generation of free radicals and lipid peroxidation. This study examines the protective effects of quercetin (QUE) and mirtazapine (MIR) against the reproductive damage caused by phenylhydrazine (PHZ) in mice. Fifty NMRI mice, aged 8-10 weeks with an average weight of 27.0 ± 2.0 g, were randomly divided into five groups. The control group (Group 1) received oral administration of 10 mL/kg/day of normal saline. Group 2 (PHZ group) received an initial intraperitoneal dose of 8 mg/100 g body weight of PHZ, followed by subsequent doses of 6 mg/100 g every 48 h. Group 3 received PHZ along with oral QUE at a dosage of 50 mg/kg/day. Group 4 received PHZ along with oral MIR at a dosage of 30 mg/kg/day. Group 5 received PHZ along with oral QUE at a dosage of 50 mg/kg/day and MIR at a dosage of 30 mg/kg/day. The treatment duration was 35 days. Sperm samples were collected from the caudal region of the epididymis post-euthanasia to assess the total mean sperm count, sperm viability, motility, DNA damage, and morphology. Testicular tissue was employed to quantify total antioxidant capacity (TAC), superoxide dismutase (SOD), glutathione peroxidase (GPx), and malondialdehyde (MDA) concentrations, while serum levels of testosterone, luteinizing hormone (LH), and follicle-stimulating hormone (FSH) were analyzed. Additionally, various aspects, including testicular histopathology, oxidative enzyme levels, gene expression related to apoptosis and antiapoptotic pathways, and in vivo fertility index, were evaluated after 35 days. The QUE, MIR, and QUE + MIR groups showed less abnormal morphology and DNA damage, as well as better total and progressive sperm motility, motility characteristics, viability, and plasma membrane function compared to the PHZ group. QUE, MIR, and QUE + MIR administration increased TAC, SOD, and GPx activities in testicular tissue, while reducing MDA levels compared to the PHZ group. Furthermore, QUE, MIR, and QUE + MIR significantly reduced Bax, and caspase-3 expression levels, and increased Bcl-2 expression levels, compared to the PHZ group. Mice treated with QUE, MIR, and QUE + MIR exhibited an increased in vivo fertility index and plasma sex hormone levels compared to the PHZ group. These results show that QUE, MIR, and QUE + MIR might be able to improve the fertility index, boost the testicular antioxidant defense system, and control the death of germ cells. This could mean that they could be used to treat mice with PHZ-induced testicular damage.
Collapse
Affiliation(s)
- Tohid Mohammadi
- Department of Basic Science, Faculty of Veterinary Medicine, Urmia Branch, Islamic Azad University, Urmia, Iran.
| |
Collapse
|
12
|
Hassanein EHM, Althagafy HS, Baraka MA, Abd-Alhameed EK, Ibrahim IM. Pharmacological update of mirtazapine: a narrative literature review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:2603-2619. [PMID: 37943296 PMCID: PMC11074035 DOI: 10.1007/s00210-023-02818-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 10/26/2023] [Indexed: 11/10/2023]
Abstract
Mirtazapine (MTZ) is an antidepressant drug with an exceptional pharmacological profile. It also has an excellent safety and tolerability profile. The present review provides a pharmacological update on MTZ and summarizes the research findings of MTZ's effects on different diseases. MTZ is hypothesized to have antidepressant effects because of the synergy between noradrenergic and serotonergic actions and is effective in treating major depressive disorder and depression associated with epilepsy, Alzheimer's disease, stroke, cardiovascular disease, and respiratory disease. In cancer patients, MTZ significantly reduced sadness, nausea, sleep disruption, and pain and improved quality of life. Also, it has promising effects on Parkinson's disease, schizophrenia, dysthymia, social anxiety disorder, alcohol dependency, posttraumatic stress disorder, panic disorder, pain syndromes, obsessive-compulsive disorder, and sleep disorders. Additionally, MTZ is potentially therapeutic in different situations associated with depression, such as liver, kidney, cardiovascular, respiratory, infertility, heavy metal-induced neurotoxicity, and pruritus. Potent antioxidative, anti-inflammatory, and anti-apoptotic bioactivities mediate these promising effects. These positive outcomes of the scientific investigations motivate more and more clinical trials for a golden exceptional antidepressant in different conditions.
Collapse
Affiliation(s)
- Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt.
| | - Hanan S Althagafy
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Mohammad A Baraka
- Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Esraa K Abd-Alhameed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Islam M Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
13
|
Hasan IH, Shaheen SY, Alhusaini AM, Mahmoud AM. Simvastatin mitigates diabetic nephropathy by upregulating farnesoid X receptor and Nrf2/HO-1 signaling and attenuating oxidative stress and inflammation in rats. Life Sci 2024; 340:122445. [PMID: 38278349 DOI: 10.1016/j.lfs.2024.122445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/03/2024] [Accepted: 01/16/2024] [Indexed: 01/28/2024]
Abstract
Diabetic nephropathy is one of the complications of diabetes that affects the kidney and can result in renal failure. The cholesterol-lowering drug simvastatin (SIM) has shown promising effects against diabetic nephropathy (DN). This study evaluated the protective role of SIM on DN, pointing to the involvement of farnesoid X receptor (FXR) and Nrf2/HO-1 signaling in attenuating inflammatory response, oxidative injury, and tissue damage in streptozotocin-induced diabetic rats. SIM was supplemented orally for 8 weeks, and samples were collected for analysis. SIM effectively ameliorated hyperglycemia, kidney hypertrophy, body weight loss, and tissue injury and fibrosis in diabetic animals. SIM mitigated oxidative stress (OS), inflammatory response, and cell death, as evidenced by the suppressed malondialdehyde, nitric oxide, myeloperoxidase, NF-kB, TNF-α, IL-1β, CD68, Bax, and caspase-3 in the diabetic kidney. These effects were linked to suppressed Keap1, upregulated FXR, Nrf2, and HO-1, and enhanced antioxidant defenses and Bcl-2. The in silico findings revealed the binding affinity of SIM with NF-kB, caspase-3, Keap1, HO-1, and FXR. In conclusion, SIM protects against DN by attenuating hyperglycemia, kidney injury, fibrosis, inflammation, and OS, and upregulating antioxidants, FXR, and Nrf2/HO-1 signaling.
Collapse
Affiliation(s)
- Iman H Hasan
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11459, Saudi Arabia..
| | - Sameerah Y Shaheen
- Department of Anatomy, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
| | - Ahlam M Alhusaini
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11459, Saudi Arabia
| | - Ayman M Mahmoud
- Department of Life Sciences, Faculty of Science & Engineering, Manchester Metropolitan University, Manchester M1 5GD, UK..
| |
Collapse
|
14
|
Ghodrat M, Separham A, Banisefid E, Alamdari NM, Akbarzadeh M, Alipour S, Yahyapoor T, Roshanravan N, Ghaffari S. The expression levels of PPAR-α/γ and UCP1/2 on the slow coronary flow phenomenon; results from a case-control study. Mol Biol Rep 2023; 50:7527-7533. [PMID: 37501045 DOI: 10.1007/s11033-023-08668-y] [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: 05/16/2023] [Accepted: 07/05/2023] [Indexed: 07/29/2023]
Abstract
PURPOSE The slow coronary flow (SCF) phenomenon is considered a coronary artery disorder. Because of the critical function of peroxisome proliferator-activated receptors (PPARs) in regulating the oxidative stress and inflammatory reactions in cardiovascular disease, The aim of the current study was to investigate the expression of the genes for uncoupling proteins 1 and 2 (UCP1 and UCP2), peroxisome proliferator-activated receptors and (PPAR- PPAR-), and PPAR- in SCF patients. METHODS In this case-control study, coronary angiography examination was used to analyze 35 individuals with SCF and 35 subjects with normal coronary flow (NCF). SCF was diagnosed using the TIMI (thrombolysis in myocardial infarction frame count) method. The SCF phenomenon is thought to be the TIMI > 27. In the peripheral blood mononuclear cells (PBMCs), the messenger ribonucleic acid (mRNA) expression levels of the PPAR-, PPAR-, UCP1, and UCP2 genes were evaluated. RESULTS UCP1 and UCP2 expression levels were significantly higher in the SCF group compared to the NCF group (P = 0.034 and P0.001, respectively). The PPAR- and PPAR- levels were found to be significantly lower in the SCF group compared to the NCF group (P = 0.015, P0.001, respectively). According to the results of the logistic regression analysis, high UCP1 and UCP2 levels and low PPAR- and PPAR- levels are each independent predictors of the SCF phenomenon. CONCLUSION This research provided evidence about the potential role of PPAR-α, PPAR-γ, UCP1, and UCP2 as biomarkers in SCF. More investigations are suggested to assess the functions of these factors in SCF patients mechanistically.
Collapse
Affiliation(s)
- Mahshid Ghodrat
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, 5166615573, Iran
| | - Ahmad Separham
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, 5166615573, Iran
| | - Erfan Banisefid
- Students Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Moloud Akbarzadeh
- Centre de Recherche de L'Institut Universitaire de Cardiologie Et de Pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Shahriar Alipour
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Clinical Biochemistry and Applied Cell Sciences, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Tohid Yahyapoor
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, 5166615573, Iran
| | - Neda Roshanravan
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, 5166615573, Iran.
| | - Samad Ghaffari
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, 5166615573, Iran.
| |
Collapse
|
15
|
Le NT. The significance of ERK5 catalytic-independent functions in disease pathways. Front Cell Dev Biol 2023; 11:1235217. [PMID: 37601096 PMCID: PMC10436230 DOI: 10.3389/fcell.2023.1235217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 07/26/2023] [Indexed: 08/22/2023] Open
Abstract
Extracellular signal-regulated kinase 5 (ERK5), also known as BMK1 or MAPK7, represents a recent addition to the classical mitogen-activated protein kinase (MAPK) family. This family includes well-known members such as ERK1/2, c-Jun N-terminal kinase (JNK), and p38 mitogen-activated protein kinase (p38 MAPK), as well as atypical MAPKs such as ERK3, ERK4, ERK7 (ERK8), and Nemo-like kinase (NLK). Comprehensive reviews available elsewhere provide detailed insights into ERK5, which interested readers can refer to for in-depth knowledge (Nithianandarajah-Jones et al., 2012; Monti et al., Cancers (Basel), 2022, 14). The primary aim of this review is to emphasize the essential characteristics of ERK5 and shed light on the intricate nature of its activation, with particular attention to the catalytic-independent functions in disease pathways.
Collapse
Affiliation(s)
- Nhat-Tu Le
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
16
|
Gao S, Chen X, Yu Z, Du R, Chen B, Wang Y, Cai X, Xu J, Chen J, Duan H, Cai Y, Zheng G. Progress of research on the role of active ingredients of Citri Reticulatae Pericarpium in liver injury. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 115:154836. [PMID: 37119760 DOI: 10.1016/j.phymed.2023.154836] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/01/2023] [Accepted: 04/18/2023] [Indexed: 05/21/2023]
Abstract
BACKGROUND Liver is a vital organ responsible for metabolizing and detoxifying both endogenous and exogenous substances in the body. However, it is susceptible to damage from chemical and natural toxins. The high incidence and mortality rates of liver disease and its associated complications impose a significant economic burden and survival pressure on patients and their families. Various liver diseases exist, including cholestasis, viral and non-viral hepatitis, fatty liver disease, drug-induced liver injury, alcoholic liver injury, and severe end-stage liver diseases such as cirrhosis, hepatocellular carcinoma (HCC), and cholangiocellular carcinoma (CCA). Recent research has shown that flavonoids found in Citri Reticulatae Pericarpium (CRP) have the potential to normalize blood glucose, cholesterol levels, and liver lipid levels. Additionally, these flavonoids exhibit anti-inflammatory properties, prevent oxidation and lipid peroxidation, and reduce liver toxicity, thereby preventing liver injury. Given these promising findings, it is essential to explore the potential of active components in CRP for developing new drugs to treat liver diseases. OBJECTIVE Recent studies have revealed that flavonoids, including hesperidin (HD), hesperetin (HT), naringenin (NIN), nobiletin (NOB), naringin (NRG), tangerine (TN), and erodcyol (ED), are the primary bioactive components in CRP. These flavonoids exhibit various therapeutic effects on liver injury, including anti-oxidative stress, anti-cytotoxicity, anti-inflammatory, anti-fibrosis, and anti-tumor mechanisms. In this review, we have summarized the research progress on the hepatoprotective effects of HD, HT, NIN, NOB, NRG, TN, ED and limonene (LIM), highlighting their underlying molecular mechanisms. Despite their promising effects, the current clinical application of these active ingredients in CRP has some limitations. Therefore, further studies are needed to explore the full potential of these flavonoids and develop new therapeutic strategies for liver diseases. METHODS For this review, we conducted a systematic search of three databases (ScienceNet, PubMed, and Science Direct) up to July 2022, using the search terms "CRP active ingredient," "liver injury," and "flavonoids." The search data followed the PRISMA standard. RESULTS Our findings indicate that flavonoids found in CRP can effectively reduce drug-induced liver injury, alcoholic liver injury, and non-alcoholic liver injury. These therapeutic effects are mainly attributed to the ability of flavonoids to improve liver resistance to oxidative stress and inflammation while normalizing cholesterol and liver lipid levels by exhibiting anti-free radical and anti-lipid peroxidation properties. CONCLUSION Our review provides new insights into the potential of active components in CRP for preventing and treating liver injury by regulating various molecular targets within different cell signaling pathways. This information can aid in the development of novel therapeutic strategies for liver disease.
Collapse
Affiliation(s)
- Shuhan Gao
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xiaojing Chen
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhiqian Yu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Rong Du
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Baizhong Chen
- Guangdong Xinbaotang Biological Technology Co., Ltd, Guangdong Jiangmen, 529000, China
| | - Yuxin Wang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xiaoting Cai
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jiepei Xu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jiamin Chen
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Huiying Duan
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yi Cai
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Guodong Zheng
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
17
|
Punicalagin Protects against the Development of Methotrexate-Induced Hepatotoxicity in Mice via Activating Nrf2 Signaling and Decreasing Oxidative Stress, Inflammation, and Cell Death. Int J Mol Sci 2022; 23:ijms232012334. [PMID: 36293191 PMCID: PMC9604463 DOI: 10.3390/ijms232012334] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/11/2022] [Accepted: 10/13/2022] [Indexed: 11/06/2022] Open
Abstract
Despite its effectiveness in treating inflammatory diseases and various malignancies, methotrexate (MTX) is well known to cause hepatotoxicity, which involves increased oxidative stress and inflammation, limiting its clinical use. Herein, we looked into the effect of punicalagin (PU), a polyphenolic molecule having a variety of health-promoting attributes, on MTX-induced hepatotoxicity in mice. PU (25 and 50 mg/kg/day) was given orally to the mice for 10 days, while a single dose of MTX (20 mg/kg) was injected intraperitoneally (i.p.) at day 7. The MTX-induced liver damage was demonstrated by remarkably higher transaminases (ALT and AST), ALP, and LDH, as well as significant histological alterations in hepatic tissues. MTX-injected mice also demonstrated increases in hepatic oxidative stress markers, including malondialdehyde (MDA) and nitric oxide (NO), with a concordant drop in glutathione (GSH) content and superoxide dismutase (SOD) and catalase (CAT) activities. PU significantly attenuated the MTX-induced serum transaminases, ALP and LDH elevations, and hepatic oxidative stress measures and boosted antioxidant defenses in the liver. Moreover, the liver of MTX-treated mice showed increases in NF-κB p65 expression, pro-inflammatory cytokine (IL-6 and TNF-α) levels, and pro-apoptotic protein (caspase-3 and Bax) expression, whereas Bcl-2 and Nrf2 expressions were reduced, which were all attenuated by PU treatment. Collectively, PU inhibits oxidative damage, inflammation, and apoptosis and upregulates Nrf2 in the liver of MTX-induced mice. Thus, these findings suggest that PU may have great therapeutic potential for the prevention of MTX-induced hepatotoxicity, pending further exploration in upcoming studies.
Collapse
|
18
|
Essawy AA, Abdel-Farid IB. Hybrid solvothermal/sonochemical-mediated synthesis of ZnO NPs generative of OH radicals: Photoluminescent approach to evaluate OH scavenging activity of Egyptian and Yemeni Punica granatum arils extract. ULTRASONICS SONOCHEMISTRY 2022; 89:106152. [PMID: 36055013 PMCID: PMC9445396 DOI: 10.1016/j.ultsonch.2022.106152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 08/25/2022] [Accepted: 08/28/2022] [Indexed: 06/15/2023]
Abstract
Zinc oxide NPs were synthesized solvothermally within sonochemical mediation and characterized by XRD, FTIR, SEM, EDX, elemental mapping, TEM and UV-vis. spectrophotometry. To evaluate the hydroxyl radicals (OH) scavenging activity of arils extract of Egyptian (EGY-PAM) and Yemeni Punica granatum (YEM-PAM), the developed zinc oxide nano particles (ZnO NPs) as a highly productive source of hydroxyl radicals (under Solar-illumination) was used. The yield of OH was trapped and probed via fluorimetric monitoring. This suits the first sensitive/selective photoluminescent avenue to evaluate the OH scavenging activity. The high percentage of DPPH radical scavenging reflected higher contents of phenolics, flavonoids, and anthocyanins that were found in EGY-PAM and YEM-PAM. Although, some secondary metabolites contents were significantly different in EGY-PAM and YEM-PAM, the traditional DPPH radical scavenging methodology revealed insignificant IC50. Unlike, the developed fluorimetric probing, sensitively discriminated the OH scavenging activity with IC50 (105.7 µg/mL) and lower rate of OH productivity (k = 0.031 min-1) in case of EGY-PAM in comparison to IC50 (153.4 µg/mL) and higher rate of OH productivity (k = 0.053 min-1) for YEM-PAM. Our findings are interestingly superior to the TBHQ that is synthetic antioxidant. Moreover, our developed methodology for fluorimetric probing of OH radicals scavenging, recommends EGY-PAM as OH radicals scavenger for diabetic patients while YEM-PAM exhibited a better OH radicals scavenging appropriate for high blood pressure patients. More interestingly, EGY-PAM and YEM-PAM exhibited high anticancer potentiality. The aforementioned OH and DPPH scavenging activities as well as the anticancer potentiality present EGY-PAM and YEM-PAM as promising sources of natural antioxidants, that may have crucial roles in some chronic diseases such as diabetics and hypertension in addition to cancer therapeutic protocols.
Collapse
Affiliation(s)
- Amr A Essawy
- Chemistry Department, College of Science, Jouf University, Sakaka P.O. Box 2014, Saudi Arabia; Chemistry Department, Faculty of Science, Fayoum University, 63514 Fayoum, Egypt.
| | - Ibrahim B Abdel-Farid
- Biology Department, College of Science, Jouf University, Sakaka P.O. Box 2014, Saudi Arabia; Botany Department, Faculty of Science, Aswan University, Aswan, Egypt
| |
Collapse
|
19
|
Soliman MM, Aldhahrani A, Elshazly SA, Shukry M, Abouzed TK. Borate Ameliorates Sodium Nitrite-Induced Oxidative Stress Through Regulation of Oxidant/Antioxidant Status: Involvement of the Nrf2/HO-1 and NF-κB Pathways. Biol Trace Elem Res 2022; 200:197-205. [PMID: 33559025 DOI: 10.1007/s12011-021-02613-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/24/2021] [Indexed: 10/22/2022]
Abstract
The widespread industrial use of nitrite in preservatives, colorants, and manufacturing rubber products and dyes increases the possibilities of organ toxicity. Lithium borate (LB) is known as an antioxidant and an oxidative stress reliever. Therefore, this study is aimed at examining the effect of LB on nitrite-induced hepatorenal dysfunction. Twenty-eight male Swiss mice were divided into four equal groups. Group 1, the control group, received saline. Group 2 received LB orally for 5 consecutive days at a dose of 15 mg/kg bw. Group 3, the nitrite group, received sodium nitrite (NaNO2) on Day 5 (60 mg/kg bw intraperitoneally). Group 4, the protective group (LB + NaNO2 group), received LB for 5 days and then a single dose of NaNO2 intraperitoneally on Day 5, the same as in Groups 2 and 3, respectively. Samples of blood and kidney were taken for serum analysis of hepatorenal biomarkers, levels of antioxidants and cytokines, and the expression of genes associated with oxidative stress and inflammation. NaNO2 intoxication increased markers of liver and kidney functions yet decreased reduced glutathione (GSH), superoxide dismutase (SOD), and catalase activities in blood. NaNO2 also increased the expression of tumor necrosis factor (TNF-α), interleukin-1β and interleukin-6 (IL-1β and IL-6). Pre-administration of LB protected mice from oxidative stress, lipid peroxidation, and the decrease in antioxidant enzyme activity. Moreover, LB protected mice from cytokine changes, which remained within normal levels. LB ameliorated the changes induced by NaNO2 on the mRNA of nuclear factor erythroid 2-related factor 2 (Nfr2), heme oxygenase-1 (HO-1), nuclear factor-kappa B (NF-κB), transforming growth factor-beta 2 (TGF-β2), and glutathione-S-transferase (GST) as determined using quantitative real-time PCR (qRT-PCR). These results collectively demonstrate that LB ameliorated NaNO2-induced oxidative stress by controlling the oxidative stress biomarkers and the oxidant/antioxidant state through the involvement of the Nrf2/HO-1 and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Mohamed Mohamed Soliman
- Clinical Laboratory Sciences Department, Turabah University College, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia.
- Biochemistry Department, Faculty of Veterinary Medicine, Benha University, Benha, Egypt.
| | - Adil Aldhahrani
- Clinical Laboratory Sciences Department, Turabah University College, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
| | - Samir Ahmed Elshazly
- Biochemistry Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Mustafa Shukry
- Physiology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Tarek Kamal Abouzed
- Biochemistry Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, Egypt
| |
Collapse
|
20
|
Galangin Attenuates Liver Injury, Oxidative Stress and Inflammation, and Upregulates Nrf2/HO-1 Signaling in Streptozotocin-Induced Diabetic Rats. Processes (Basel) 2021. [DOI: 10.3390/pr9091562] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Chronic hyperglycemia increases the risk of liver damage. Oxidative stress and aberrant inflammatory response are entangled in diabetes-associated liver injury. This study evaluated the protective effect of the flavonoid galangin (Gal) on glucose intolerance, liver injury, oxidative stress, inflammatory response, and Nrf2/HO-1 signaling in diabetic rats. Diabetes was induced by streptozotocin (STZ), and the rats received Gal for six weeks. STZ-induced rats showed glucose intolerance, hypoinsulinemia, elevated glycated hemoglobin (HbA1c), and decreased liver glycogen. Gal ameliorated glucose intolerance, reduced HbA1c%, increased serum insulin and liver glycogen and hexokinase activity, and suppressed glycogen phosphorylase, glucose-6-phosphatase and fructose-1,6-biphosphatase in diabetic rats. Circulating transaminases, ALP and LDH, and liver ROS, MDA, TNF-α, IL-1β, and IL-6 were increased and GSH, SOD, and CAT were diminished in diabetic rats. In addition, diabetic rats exhibited multiple histopathological alterations and marked collagen deposition. Treatment with Gal mitigated liver injury, prevented histopathological alterations, decreased ROS, MDA, pro-inflammatory cytokines, Bax and caspase-3, and enhanced cellular antioxidants and Bcl-2. Gal downregulated hepatic Keap1 in diabetic rats and upregulated Nrf2 and HO-1 mRNA as well as HO-1 activity. Molecular modeling studies revealed the ability of Gal to bind to and inhibit NF-κB and Keap1, and also showed its binding pattern with HO-1. In conclusion, Gal ameliorates hyperglycemia, glucose intolerance, oxidative stress, inflammation, and apoptosis in diabetic rats. Gal improved carbohydrate metabolizing enzymes and upregulated Nrf2/HO-1 signaling.
Collapse
|
21
|
Phloretin suppresses neuroinflammation by autophagy-mediated Nrf2 activation in macrophages. J Neuroinflammation 2021; 18:148. [PMID: 34218792 PMCID: PMC8254976 DOI: 10.1186/s12974-021-02194-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 06/16/2021] [Indexed: 12/31/2022] Open
Abstract
Background Macrophages play a dual role in neuroinflammatory disorders such as multiple sclerosis (MS). They are involved in lesion onset and progression but can also promote the resolution of inflammation and repair of damaged tissue. In this study, we investigate if and how phloretin, a flavonoid abundantly present in apples and strawberries, lowers the inflammatory phenotype of macrophages and suppresses neuroinflammation. Methods Transcriptional changes in mouse bone marrow-derived macrophages upon phloretin exposure were assessed by bulk RNA sequencing. Underlying pathways related to inflammation, oxidative stress response and autophagy were validated by quantitative PCR, fluorescent and absorbance assays, nuclear factor erythroid 2–related factor 2 (Nrf2) knockout mice, western blot, and immunofluorescence. The experimental autoimmune encephalomyelitis (EAE) model was used to study the impact of phloretin on neuroinflammation in vivo and confirm underlying mechanisms. Results We show that phloretin reduces the inflammatory phenotype of macrophages and markedly suppresses neuroinflammation in EAE. Phloretin mediates its effect by activating the Nrf2 signaling pathway. Nrf2 activation was attributed to 5′ AMP-activated protein kinase (AMPK)-dependent activation of autophagy and subsequent kelch-like ECH-associated protein 1 (Keap1) degradation. Conclusions This study opens future perspectives for phloretin as a therapeutic strategy for neuroinflammatory disorders such as MS. Trial registration Not applicable. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02194-z.
Collapse
|
22
|
Seol D, Coleman MC, Martin JA, Song I, Jaidev LR, Salem AK, Lim TH. Targeting oxidative stress with amobarbital to prevent intervertebral disc degeneration: Part I. in vitro and ex vivo studies. Spine J 2021; 21:1021-1030. [PMID: 33610806 PMCID: PMC11789274 DOI: 10.1016/j.spinee.2021.02.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 02/08/2021] [Accepted: 02/13/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND Mounting evidence that oxidative stress contributes to the pathogenesis of intervertebral disc (IVD) degeneration (IDD) suggests that therapies targeting oxidative stress may slow or prevent disease progression. PURPOSE The objective of this study was to investigate the inhibitory effects of amobarbital (Amo) on the mitochondria of nucleus pulposus (NP) cells under tert-butyl hydrogen peroxide (tBHP)-induced oxidative stress or in NP tissues under oxidative stress from tissue injury as a means of identifying therapeutic targets for IDD. STUDY DESIGN/SETTING We tested the effects inhibiting mitochondria, a major source of oxidants, with Amo in NP cells subjected to two different forms of insult: exposure to tBHP, and physical injury induced by disc transection. N-acetylcysteine (NAC), an antioxidant known to protect NP cells, was compared to the complex I inhibitor, Amo. METHODS NP cells were pre-treated for 2 hours with Amo, NAC, or both, and then exposed to tBHP for 1 hour. Apoptosis, necrosis, and reactive oxygen species (ROS) production were assessed using confocal microscopy and fluorescent probes (Annexin V, propidium iodide, and MitoSox Red, respectively). The activation of mitogen-activated protein kinases (MAPKs) involved in oxidative stress responses were interrogated by confocal imaging of immunofluorescence stains using phospho-specific antibodies to extracellular signal-regulated kinase (ERK), c-JUN N-terminal kinase (JNK), and p38. Mitochondrial function was assessed by imaging JC-1 staining, a probe for membrane potential. RESULTS Amo was modestly more protective than NAC by some measures, while both agents improved mitochondrial function and lowered tBHP-induced apoptosis, necrosis, and ROS production. Activation of MAPK by tBHP was significantly suppressed by both drugs. Physically injured IVDs were treated immediately after transection with Amo or NAC for 24 hours, and then stained with dihydroethidium (DHE), a fluorescent probe for ROS production. Immunofluorescence was used to track the expression of nuclear factor (erythroid-derived 2)-like 2 (Nrf2), a transcription factor that induces the expression of antioxidant genes. Amo and NAC significantly reduced ROS production and increased Nrf2 expression. CONCLUSION These findings suggest that the progression of IDD may be forestalled by Amo via protection of NP cells from oxidative stress following IVD injury. CLINICAL SIGNIFICANCE This study will define the extent to which a novel, minimally invasive procedure targeting oxidative stress in NP cells can augment surgical interventions intended to retard IVD degeneration.
Collapse
Affiliation(s)
- Dongrim Seol
- Departments of Orthopedics and Rehabilitation, University of Iowa, Iowa City, IA 52242, USA.
| | - Mitchell C Coleman
- Departments of Orthopedics and Rehabilitation, University of Iowa, Iowa City, IA 52242, USA; Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA
| | - James A Martin
- Departments of Orthopedics and Rehabilitation, University of Iowa, Iowa City, IA 52242, USA; Roy J. Carver Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA; Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa, Iowa City, IA 52242, USA
| | - Ino Song
- Departments of Orthopedics and Rehabilitation, University of Iowa, Iowa City, IA 52242, USA; Roy J. Carver Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - L R Jaidev
- Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa, Iowa City, IA 52242, USA
| | - Aliasger K Salem
- Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa, Iowa City, IA 52242, USA
| | - Tae-Hong Lim
- Roy J. Carver Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
23
|
Kim SY, Zhang X, Schiattarella GG, Altamirano F, Ramos TAR, French KM, Jiang N, Szweda PA, Evers BM, May HI, Luo X, Li H, Szweda LI, Maracaja-Coutinho V, Lavandero S, Gillette TG, Hill JA. Epigenetic Reader BRD4 (Bromodomain-Containing Protein 4) Governs Nucleus-Encoded Mitochondrial Transcriptome to Regulate Cardiac Function. Circulation 2020; 142:2356-2370. [PMID: 33113340 PMCID: PMC7736324 DOI: 10.1161/circulationaha.120.047239] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 10/23/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND BET (bromodomain and extraterminal) epigenetic reader proteins, in particular BRD4 (bromodomain-containing protein 4), have emerged as potential therapeutic targets in a number of pathological conditions, including cancer and cardiovascular disease. Small-molecule BET protein inhibitors such as JQ1 have demonstrated efficacy in reversing cardiac hypertrophy and heart failure in preclinical models. Yet, genetic studies elucidating the biology of BET proteins in the heart have not been conducted to validate pharmacological findings and to unveil potential pharmacological side effects. METHODS By engineering a cardiomyocyte-specific BRD4 knockout mouse, we investigated the role of BRD4 in cardiac pathophysiology. We performed functional, transcriptomic, and mitochondrial analyses to evaluate BRD4 function in developing and mature hearts. RESULTS Unlike pharmacological inhibition, loss of BRD4 protein triggered progressive declines in myocardial function, culminating in dilated cardiomyopathy. Transcriptome analysis of BRD4 knockout mouse heart tissue identified early and specific disruption of genes essential to mitochondrial energy production and homeostasis. Functional analysis of isolated mitochondria from these hearts confirmed that BRD4 ablation triggered significant changes in mitochondrial electron transport chain protein expression and activity. Computational analysis identified candidate transcription factors participating in the BRD4-regulated transcriptome. In particular, estrogen-related receptor α, a key nuclear receptor in metabolic gene regulation, was enriched in promoters of BRD4-regulated mitochondrial genes. CONCLUSIONS In aggregate, we describe a previously unrecognized role for BRD4 in regulating cardiomyocyte mitochondrial homeostasis, observing that its function is indispensable to the maintenance of normal cardiac function.
Collapse
MESH Headings
- Animals
- Cardiomyopathy, Dilated/genetics
- Cardiomyopathy, Dilated/metabolism
- Cardiomyopathy, Dilated/pathology
- Cardiomyopathy, Dilated/physiopathology
- Cell Nucleus/genetics
- Cell Nucleus/metabolism
- Cell Nucleus/pathology
- Electron Transport Chain Complex Proteins/genetics
- Electron Transport Chain Complex Proteins/metabolism
- Energy Metabolism/genetics
- Epigenesis, Genetic
- Estrogen Receptor alpha/genetics
- Estrogen Receptor alpha/metabolism
- Gene Expression Profiling
- Heart Failure/genetics
- Heart Failure/metabolism
- Heart Failure/pathology
- Heart Failure/physiopathology
- Mice, Knockout
- Mitochondria, Heart/genetics
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcriptome
- Ventricular Dysfunction, Left/genetics
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/pathology
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Function, Left/genetics
Collapse
Affiliation(s)
- Soo Young Kim
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX, USA
| | - Xin Zhang
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Gabriele G. Schiattarella
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX, USA
| | - Francisco Altamirano
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX, USA
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX
| | - Thais A. R. Ramos
- Advanced Center for Chronic Disease, Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago, Chile
- Bioinformatics Multidisciplinary Environment, Digital Metropolis Institute, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Kristin M. French
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX, USA
| | - Nan Jiang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX, USA
| | - Pamela A. Szweda
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX, USA
| | - Bret M. Evers
- Department of Pathology, University of Texas Southwestern, Dallas, TX, USA
| | - Herman I. May
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX, USA
| | - Xiang Luo
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX, USA
| | - Hongliang Li
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Luke I. Szweda
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX, USA
| | - Vinicius Maracaja-Coutinho
- Advanced Center for Chronic Disease, Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago, Chile
- Bioinformatics Multidisciplinary Environment, Digital Metropolis Institute, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Sergio Lavandero
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX, USA
- Advanced Center for Chronic Disease, Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago, Chile
| | - Thomas G. Gillette
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX, USA
| | - Joseph A. Hill
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX, USA
- Department of Molecular Biology, University of Texas Southwestern, Dallas, TX, USA
| |
Collapse
|
24
|
Bhatt S, Nagappa AN, Patil CR. Role of oxidative stress in depression. Drug Discov Today 2020; 25:1270-1276. [DOI: 10.1016/j.drudis.2020.05.001] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 04/25/2020] [Accepted: 05/04/2020] [Indexed: 12/15/2022]
|
25
|
Coumarins as Modulators of the Keap1/Nrf2/ARE Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1675957. [PMID: 32377290 PMCID: PMC7196981 DOI: 10.1155/2020/1675957] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/14/2020] [Accepted: 04/06/2020] [Indexed: 12/30/2022]
Abstract
The Keap1/Nrf2/ARE system is a central defensive mechanism against oxidative stress which plays a key role in the pathogenesis and progression of many diseases. Nrf2 is a redox-sensitive transcription factor controlling a variety of downstream antioxidant and cytodefensive genes. Nrf2 has a powerful anti-inflammatory activity mediated via modulating NF-κB. Therefore, pharmacological activation of Nrf2 is a promising therapeutic strategy for the treatment/prevention of several diseases that are underlined by both oxidative stress and inflammation. Coumarins are natural products with promising pharmacological activities, including antioxidant, anticancer, antimicrobial, and anti-inflammatory efficacies. Coumarins are found in many plants, fungi, and bacteria and have been widely used as complementary and alternative medicines. Some coumarins have shown an ability to activate Nrf2 signaling in different cells and animal models. The present review compiles the research findings of seventeen coumarin derivatives of plant origin (imperatorin, visnagin, urolithin B, urolithin A, scopoletin, esculin, esculetin, umbelliferone, fraxetin, fraxin, daphnetin, anomalin, wedelolactone, glycycoumarin, osthole, hydrangenol, and isoimperatorin) as antioxidant and anti-inflammatory agents, emphasizing the role of Nrf2 activation in their pharmacological activities. Additionally, molecular docking simulations were utilized to investigate the potential binding mode of these coumarins with Keap1 as a strategy to disrupt Keap1/Nrf2 protein-protein interaction and activate Nrf2 signaling.
Collapse
|
26
|
Metabolomic Profiling and Antioxidant, Anticancer and Antimicrobial Activities of Hyphaene thebaica. Processes (Basel) 2020. [DOI: 10.3390/pr8030266] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
This study investigated the metabolic content and biological activities of Hyphaene thebaica leaves, male parts and fruits methanolic extracts. The phytochemical constituents were determined, and multivariate data analysis discriminated the evaluated three parts into three groups according to the content of phenolics, flavonoids, flavonols, saponins, anthocyanins and tannins. High-performance liquid chromatography (HPLC) profiling of polyphenols revealed the dominance of catechins, rutin and apigenin-7-glucosides in leaves, protocatechuin, p-hydroxybenzoic, syringic, vanillic, rosmarinic, p-coumaric acids and chrysin in male parts, and chlorogenic acid in fruits. Leaves and male parts showed stronger free radical scavenging activity than the fruits. Positive correlations between total antioxidant capacity and carbohydrates, phenolics and flavonols were observed. The three extracts exhibited potent anti-cancer activity against liver and lung carcinoma cell lines. All extracts exhibited antibacterial activity, while only fruits showed antifungal efficacy. In conclusion, H. thebaica leaves, male parts and fruits contain a variety of phytochemicals with antioxidant, anticancer and antimicrobial activities.
Collapse
|
27
|
Tabassum R, Jeong NY, Jung J. Protective effect of hydrogen sulfide on oxidative stress-induced neurodegenerative diseases. Neural Regen Res 2020; 15:232-241. [PMID: 31552888 PMCID: PMC6905340 DOI: 10.4103/1673-5374.265543] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 06/06/2019] [Indexed: 12/11/2022] Open
Abstract
Hydrogen sulfide is an antioxidant molecule that has a wide range of biological effects against oxidative stress. Balanced oxidative stress is also vital for maintaining cellular function in biological system, where reactive oxygen species are the main source of oxidative stress. When the normal redox balance is disturbed, deoxyribonucleic acid, lipid, and protein molecules are oxidized under pathological conditions, like diabetes mellitus that leads to diabetic peripheral neuropathy. In diabetes mellitus-induced diabetic peripheral neuropathy, due to hyperglycemia, pancreatic beta cell (β cell) shows resistance to insulin secretion. As a consequence, glucose metabolism is disturbed in neuronal cells which are distracted from providing proper cell signaling pathway. Not only diabetic peripheral neuropathy but also other central damages occur in brain neuropathy. Neurological studies regarding type 1 diabetes mellitus patients with Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis have shown changes in the central nervous system because high blood glucose levels (HbA1c) appeared with poor cognitive function. Oxidative stress plays a role in inhibiting insulin signaling that is necessary for brain function. Hydrogen sulfide exhibits antioxidant effects against oxidative stress, where cystathionine β synthase, cystathionine γ lyase, and 3-mercaptopyruvate sulfurtransferase are the endogenous sources of hydrogen sulfide. This review is to explore the pathogenesis of diabetes mellitus-induced diabetic peripheral neuropathy and other neurological comorbid disorders under the oxidative stress condition and the anti-oxidative effects of hydrogen sulfide.
Collapse
Affiliation(s)
- Rubaiya Tabassum
- Department of Anatomy and Cell Biology, College of Medicine, Dong-A University, Seo-gu, Busan, Korea
- Department of Medicine, Graduate School, Dong-A University, Seo-gu, Busan, Korea
| | - Na Young Jeong
- Department of Anatomy and Cell Biology, College of Medicine, Dong-A University, Seo-gu, Busan, Korea
- Department of Medicine, Graduate School, Dong-A University, Seo-gu, Busan, Korea
| | - Junyang Jung
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Dongdaemun-gu, Seoul, Korea
| |
Collapse
|
28
|
Beneficial Effects of Citrus Flavonoids on Cardiovascular and Metabolic Health. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:5484138. [PMID: 30962863 PMCID: PMC6431442 DOI: 10.1155/2019/5484138] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 01/06/2019] [Accepted: 01/30/2019] [Indexed: 12/20/2022]
Abstract
The prevalence of cardiovascular disease (CVD) is increasing over time. CVD is a comorbidity in diabetes and contributes to premature death. Citrus flavonoids possess several biological activities and have emerged as efficient therapeutics for the treatment of CVD. Citrus flavonoids scavenge free radicals, improve glucose tolerance and insulin sensitivity, modulate lipid metabolism and adipocyte differentiation, suppress inflammation and apoptosis, and improve endothelial dysfunction. The intake of citrus flavonoids has been associated with improved cardiovascular outcomes. Although citrus flavonoids exerted multiple beneficial effects, their mechanisms of action are not completely established. In this review, we summarized recent findings and advances in understanding the mechanisms underlying the protective effects of citrus flavonoids against oxidative stress, inflammation, diabetes, dyslipidemia, endothelial dysfunction, and atherosclerosis. Further studies and clinical trials to assess the efficacy and to explore the underlying mechanism(s) of action of citrus flavonoids are recommended.
Collapse
|