1
|
Ali MA, Michel HE, Menze ET, Tadros MG, Wahdan SA. The potential neuroprotective effect of empagliflozin against depressive-like behavior induced by chronic unpredictable mild stress in rats: Involvement of NLRP3 inflammasome. Eur J Pharmacol 2025; 998:177525. [PMID: 40107336 DOI: 10.1016/j.ejphar.2025.177525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 03/15/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025]
Abstract
Depression is a prevalent and debilitating condition that has a severe negative impact on a person's life. Chronic stress exposure plays a substantial role in the development of depression. In the present study, rats were exposed to chronic unpredictable mild stress (CUMS) for four weeks. Empagliflozin (EMPA), a Sodium-Glucose Cotransporter-2 (SGLT-2) inhibitor, is an oral antidiabetic agent exhibiting antioxidant, anti-inflammatory, and antiapoptotic effects. This study aimed to examine the antidepressant effect of EMPA in an experimental animal model of depression induced by CUMS in rats and explore the probable underlying mechanisms. Rats were treated with EMPA, per-orally, at a dose of 10 mg/kg/day for four weeks. EMPA treatment counteracted CUMS-induced histopathological, biochemical and behavioral alterations. EMPA suppressed the CUMS-induced increase in the oxidative stress, inflammatory, and apoptotic markers, where levels of MDA, IL-1β, TNF-α, NF-κB, NLRP3 and active caspase 3 were reduced by 29.6 %, 24.8 %, 17.9 %, 36.6 %, 24.5 % and 41.5 %, respectively, compared to the disease group. Furthermore, EMPA decreased the level of the microglial activation marker, iba-1 by 24 % in comparison to the disease group. In addition, EMPA treatment decreased blood glucose levels by 39 %, decreased serum insulin levels by 60.6 %, decreased HOMA-IR by 76.5 % and increased GLUT 4 expression, compared to the CUMS group, all which proves that EMPA has an effect insulin signaling and alleviates insulin resistance. Our results conclude that modulating key factors involved in depression, such as inflammation, oxidative stress, and NLRP3 inflammasome pathway, accounts for the anti-depressant effect of EMPA.
Collapse
Affiliation(s)
- Marwa A Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Haidy E Michel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Esther T Menze
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Marianne G Tadros
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Sara A Wahdan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt.
| |
Collapse
|
2
|
Alzokaky AA, Saber SK, Zaki MO. The reno-protective effect of Empagliflozin against carbon tetrachloride (CCl4)-induced nephrotoxicity in mice halting JNK/MKK4/NRF2/NF-KB pathway. Food Chem Toxicol 2025; 201:115439. [PMID: 40204264 DOI: 10.1016/j.fct.2025.115439] [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: 01/17/2025] [Revised: 03/12/2025] [Accepted: 04/07/2025] [Indexed: 04/11/2025]
Abstract
AIM This study designed to evaluate the reno-protective effects of Empagliflozin (EMPA), a sodium-glucose co-transporter 2 (SGLT2) inhibitor, against carbon tetrachloride (CCl4)-induced nephrotoxicity in mice targeting JNK/MKK4/NRF2/NF-KB pathway. METHODS Male albino mice were given EMPA (10 mg/kg, orally) for 4 weeks prior to a single i.p. injection of 10 % CCl4 (20 ml/kg). Mice were sacrificed 48 h post CCl4 injection. KEY FINDINGS EMPA attenuated CCl4-induced renal injury, as reflected by a decrease in serum urea and creatinine levels, also preserved the histological integrity of kidney tissue. Theses reno-protective effects of EMPA can be mainly due to its 1. Antioxidant, (↑CAT, ↑SOD, ↑Nrf-2 and ↑ARE), 2. Anti-inflammatory (↓NF-κB and ↓TNF-α) and 3. Anti-apoptotic (↓Caspase-3) proprieties. EMPA also inhibited JNK/MKK4 signaling pathway, which plays a critical role in kidney damage. CONCLUSION These finding confirm the reno-protective effect of EMPA with a modulatory impact on JNK/MKK4/Nrf2/NF-κB signaling network; suggesting its therapeutic utility to minimize acute kidney injury (AKI) in clinical setting in the future.
Collapse
Affiliation(s)
- Amany A Alzokaky
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11651, Egypt; Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta, 34518, Egypt
| | - Shimaa K Saber
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| | - Mennatallah O Zaki
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta, 34518, Egypt
| |
Collapse
|
3
|
Qusty NF, Bokhari BT, Taha M, Alobaidy MA, Al-Kushi AG, Sembawa HA, Abdelbagi O, Baokbah TAS, Obaid R, Albar HT, Babateen O, Dahran N. Empagliflozin Inhibits Cadmium-Induced Hepatic Cell Apoptosis Through Endoplasmic Reticulum Stress and Autophagy Pathways. Biol Trace Elem Res 2025:10.1007/s12011-025-04631-z. [PMID: 40372601 DOI: 10.1007/s12011-025-04631-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 04/16/2025] [Indexed: 05/16/2025]
Abstract
Cadmium (Cd), a well-known toxic heavy metal, adversely affects multiple organs. The SGLT-2 inhibitor empagliflozin (EMPA) exhibits significant antioxidant properties and hypoglycemic potential. This study aimed to investigate the hepatoprotective effect of EMPA against Cd-induced liver injury and elucidate its molecular mechanisms. Thirty-two male rats were allocated into four groups of eight rats each: group I (control group), group II (EMPA group), group III (Cd group), and group IV (Cd + EMPA group). Cd intake disrupted liver enzymes (ALT, AST, and ALP) and impaired hepatic histological architecture. Cd induced hepatic oxidative stress, as evidenced by increased MDA levels and reduced antioxidant enzymes, including SOD, GPx, and CAT. It downregulated the Nrf2/HO-1 pathway and elevated proinflammatory mediators IL-1β, IL-6, and TNF-α. Furthermore, Cd increased ER stress markers GRP78 and CHOP, along with apoptotic markers Bax and caspase-3 while decreasing anti-apoptotic Bcl-2 and reducing the autophagic indicator Beclin-1. Interestingly, EMPA administration in the Cd + EMPA group attenuated Cd-induced hepatic deterioration, improving hepatocyte structure. This beneficial effect was driven by the downregulation of hepatic oxidative stress, inflammation, ER stress, and apoptosis, alongside the upregulation of the autophagy process. In conclusion, this study highlights the hepatoprotective effect of EMPA against Cd-induced liver injury, elucidating its underlying molecular mechanisms.
Collapse
Affiliation(s)
- Naeem F Qusty
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al‒Qura University, Makkah, 21955, Saudi Arabia
| | - Bayan T Bokhari
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al‒Qura University, Makkah, 21955, Saudi Arabia
| | - Medhat Taha
- Department of Anatomy, Al-Qunfudah Medical College, Umm Al-Qura University, Al-Qunfudhah, Saudi Arabia.
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | - Mohammad Ahmad Alobaidy
- Department of Anatomy, Faculty of Medicine, Umm Al-Qura University, Makkah, P.O. Box 7607, Saudi Arabia
| | - Abdullah G Al-Kushi
- Department of Anatomy, Faculty of Medicine, Umm Al-Qura University, Makkah, P.O. Box 7607, Saudi Arabia
| | - Hatem A Sembawa
- Department of Surgery, Faculty of Medicine, Umm Al-Qura University, Holy Makkah, Saudi Arabia
| | - Omer Abdelbagi
- Department of Pathology, Qunfudah Faculty of Medicine, Umm-Al-Qura University, Makkah, Kingdom of Saudi Arabia
| | - Tourki A S Baokbah
- Department of Medical Emergency Services, College of Health Sciences-AlQunfudah, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Rami Obaid
- Department of Medical Genetics, Faculty of Medicine, -Qunfudah, Umm Al-Qura University, Al-Qunfudhah, Saudi Arabia
| | - Halah Tariq Albar
- Department of Physiology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Omar Babateen
- Department of Physiology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Naief Dahran
- Department of Basic Medical Sciences, College of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| |
Collapse
|
4
|
Deniz E, Topcu A, Ozturk A, Ozturk SD, Akyildiz K. Hedera helix folium extract attenuates methotrexate-induced hepatotoxicity by modulating oxidative stress and inflammatory mediators. Tissue Cell 2025; 96:102967. [PMID: 40403377 DOI: 10.1016/j.tice.2025.102967] [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: 09/30/2024] [Revised: 04/02/2025] [Accepted: 05/06/2025] [Indexed: 05/24/2025]
Abstract
Methotrexate (MTX)-induced hepatotoxicity is linked to oxidative damage and inflammatory processes. Hedera helix folium (HHF) extract protects cells against oxidative damage. We investigated the role of HHF extract in tumor necrosis factor alpha (TNF-α) and interleukin-10 (IL-10)-associated inflammation and oxidative stress in the pathology of MTX-associated liver injury in rats. Forty male rats were divided into one of five equal groups: Control, HHF, MTX, H100+MTX and H200+MTX. HHF extract was administered via the oral route at 100 mg/kg or 200 mg/kg once daily for seven days, while MTX was administered as a single dose of 20 mg/kg intraperitoneally. Intracardiac blood samples and liver tissue samples were collected at the conclusion of the experiment. Serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels increased due to MTX. Increased ALT levels were significantly reduced by low-dose HHF and increased AST levels were significantly reduced by high-dose HHF administration. The application of MTX significantly increased malondialdehyde (MDA) and TNF-α levels, while significantly reducing those of glutathione (GSH) and IL-10. High-dose HHF also significantly lowered MDA and TNF-α levels, while significantly increasing those of GSH and IL-10. Histopathological damage findings observed due to MTX were significantly attenuated with high-dose HHF. In addition, the increased caspase-3, p53, and Bcl2 levels caused by MTX decreased with high-dose HHF administration. HHF extract can alleviate liver damage induced by MTX. This extract, which has the ability to reduce damage due to oxidative stress and inflammation, may represent an alternative approach to preventing MTX-induced liver damage.
Collapse
Affiliation(s)
- Esra Deniz
- Department of Pharmacology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize 53100, Turkey.
| | - Atilla Topcu
- Department of Pharmacology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize 53100, Turkey
| | - Aykut Ozturk
- Department of Pharmacology, Ministry of Health, Derince Training and Research Hospital, Kocaeli 41100, Turkey
| | - Seda Duman Ozturk
- Department of Pathology, Faculty of Medicine, Kocaeli University, Kocaeli 41100, Turkey
| | - Kerimali Akyildiz
- Department of Medical Services and Techniques, Health Care Services Vocational School, Recep Tayyip Erdogan University, Rize 53100, Turkey
| |
Collapse
|
5
|
Alsaab J, Sarawi WS, Alhusaini AM, Hasan IH, Alturaif S, Ali RA, Alrasheed NM, Mohammad R, Algarzae NK. Procyanidin B2 mitigates methotrexate-induced hepatic pyroptosis by suppressing TLR4/NF-κB and caspase-3/GSDME pathways. Food Chem Toxicol 2025; 199:115341. [PMID: 39988050 DOI: 10.1016/j.fct.2025.115341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 02/08/2025] [Accepted: 02/19/2025] [Indexed: 02/25/2025]
Abstract
Methotrexate (MTX), a potent chemotherapeutic and immunosuppressive agent, is widely used for cancer and autoimmune diseases. MTX-induced hepatotoxicity is a well-recognized adverse response, even at relatively low doses. This study investigates the possible protective effects of procyanidin B2 (PCB2) on MTX-induced hepatotoxicity. Rats were orally treated with PCB2 (40 mg/kg) for 10 days, followed by a single intraperitoneal MTX injection (20 mg/kg) on day 8. The study also included a positive control group treated with quercetin (20 mg/kg), a known antioxidant, alongside MTX. The results revealed that MTX-induced hepatic injury was evidenced by elevation in serum transaminases. This elevation was accompanied by hepatic oxidative stress due to an imbalance in oxidative/antioxidant markers, specifically elevated malondialdehyde (MDA) and decreased glutathione (GSH) levels and superoxide dismutase (SOD) activity. The inflammatory cytokines, including tumor necrosis factor-alpha (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6), were markedly upregulated in the liver of MTX-intoxicated rats. Additionally, the expressions of nuclear factor kappa B (NF-κB), toll-like receptor 4 (TLR4), caspase-3 and gasdermin E (GSDME) were significantly increased in MTX rats. The use of PCB2 significantly ameliorated the deleterious effect of MTX on previous parameters by restoring oxidant/antioxidant balance, decreasing the inflammatory markers, and normalizing the expression of NF-κB, TLR4, caspase-3 and GSDME. In conclusion, this study uncovered the potential role of PCB2 on MTX-induced hepatotoxicity, confirming its antioxidant, anti-inflammatory, and anti-pyroptosis effects yet, further studies are needed to support its use as a protective therapy against such toxicity.
Collapse
Affiliation(s)
- Juman Alsaab
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh, 11495, Saudi Arabia.
| | - Wedad S Sarawi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh, 11495, Saudi Arabia.
| | - Ahlam M Alhusaini
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh, 11495, Saudi Arabia.
| | - Iman H Hasan
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh, 11495, Saudi Arabia.
| | - Sumayya Alturaif
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh, 11495, Saudi Arabia.
| | - Rehab A Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh, 11495, Saudi Arabia.
| | - Nouf M Alrasheed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh, 11495, Saudi Arabia.
| | - Raeesa Mohammad
- Department of Histology, College of Medicine, King Saud University, P.O. Box 2925, Riyadh, 11461, Saudi Arabia.
| | - Norah K Algarzae
- Department of Physiology, College of Medicine, King Saud University, P.O. Box 2925, Riyadh, 11461, Saudi Arabia.
| |
Collapse
|
6
|
Kamel GAM, Hussein S. Vinpocetine Mitigates Methotrexate-Induced Liver Injury in Rats Through Modulating Intercellular Communication. J Biochem Mol Toxicol 2025; 39:e70300. [PMID: 40342225 DOI: 10.1002/jbt.70300] [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: 04/02/2025] [Revised: 04/05/2025] [Accepted: 04/29/2025] [Indexed: 05/11/2025]
Abstract
Methotrexate (MTX) has been widely implemented in managing several malignancies, inflammatory conditions such as rheumatic arthritis, and autoimmune illnesses. Hepatotoxicity is a significant side effect of MTX, characterized by increased oxidative stress (OS) and inflammation. Vinpocetine (Vinpo) is a prescription medication with a favorable safety profile. It exerts anti-inflammatory and oxidant implications that might be novel candidates for protecting against MTX-induced hepatotoxicity. This study investigates the therapeutic impact of Vinpo against MTX-stimulated liver damage via the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathways. Rats are allocated into three groups: (1) the Control (saline); (2) the MTX-control (20 mg/kg; injected once i.p.), and (3) the Vinpo + MTX groups. Vinpo was administered orally for 7 days, during which MTX was given intraperitoneally once at the end of Day 3. The liver functions, OS markers, inflammatory mediators, Nrf2, HO-1, NF-κB, and apoptotic signals were estimated. Vinpo lead to enhancement in superoxide dismutase (SOD) enzyme activity, elevation in glutathione (GSH), and a hindrance in malondialdehyde (MDA). It also enhances Nrf2 and HO-1, inhibiting NF-κB (p65) expression and apoptotic markers. Moreover, Vinpo therapy, in conjunction with MTX, restores the normal histological structure of hepatic tissues. Our data suggested that Vinpo exerts a preventive effect against MTX-induced toxicity through anti-oxidative, anti-inflammatory, and apoptotic activities, mediated via Nrf2/HO-1/Nf-κB and caspase-3/Bax/Bcl-2 pathways.
Collapse
Affiliation(s)
- Gellan Alaa Mohamed Kamel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
- Department of Pharmacology & Toxicology, College of Pharmacy, Uruk University, Baghdad, Iraq
| | - Shaimaa Hussein
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Aljouf, Saudi Arabia
| |
Collapse
|
7
|
El-Dessouki AM, Alzokaky AA, Raslan NA, Ibrahim S, Selim HMRM, Al-Karmalawy AA. Dabigatran attenuates methotrexate-induced hepatotoxicity by regulating coagulation, endothelial dysfunction, and the NF-kB/IL-1β/MCP-1 and TLR4/NLRP3 signaling pathways. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:5129-5145. [PMID: 39527308 DOI: 10.1007/s00210-024-03567-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
This study examines Dabigatran's (Dab) capacity to mitigate methotrexate (MTX)-induced coagulation disorders and endothelial dysfunction, while exploring its effects on oxidative stress and inflammatory pathways (NF-kB/IL-1β/MCP-1, TLR4/NLRP3) in reducing hepatotoxicity. Rats were assigned to four groups: a control group receiving saline intraperitoneally (i.p.); an MTX group with a single MTX dose (20 mg/kg, i.p.) to induce hepatotoxicity; and two pretreatment groups receiving Dab orally at 15 mg/kg and 25 mg/kg for seven days before and 4 days after MTX administration. MTX-treated rats showed significant increases in liver enzymes (ALT, AST, ALP) and reductions in antioxidant enzymes (SOD, GSH), along with elevated coagulation parameters (tissue factor (TF), thrombin, fibrin, plasminogen activator inhibitor-1 (PAI-1)), leading to coagulation disorders. Endothelial dysfunction was evident with reduced eNOS expression, while inflammation increased through elevated iNOS, ICAM-1, and pro-inflammatory cytokines (MPO, NF-kB, TNF-α, IL-1β, MCP-1), alongside activation of the TLR4/NLRP3 inflammasome pathway and decreased IL-10 (p < 0.05). Immunohistochemistry revealed increased cytochrome c and caspase-3 expression, with histopathological damage. Dabigatran mitigated these effects, downregulating liver enzymes, modulating coagulation factors, restoring eNOS levels, and reducing histopathological and inflammatory markers. Dabigatran demonstrates significant therapeutic potential in alleviating methotrexate-induced hepatotoxicity through its antioxidant, anti-inflammatory, anticoagulant, and anti-apoptotic effects. Its regulation of coagulation parameters and endothelial function suggests a protective role against tissue damage, warranting further investigation.
Collapse
Affiliation(s)
- Ahmed M El-Dessouki
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Ahram Canadian University (ACU), 6th of October City, 12566, Giza, Egypt.
| | - Amany A Alzokaky
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, 11651, Egypt
- Pharmacology and Biochemistry Department, Faculty of Pharmacy, Horus University, New Damietta, 34518, Egypt
| | - Nahed A Raslan
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, 11651, Egypt
- Clinical Pharmacy Department, College of Health Sciences and Nursing, Al-Rayan Colleges, AL-Madinah AL-Munawarah, Saudi Arabia
| | - Samar Ibrahim
- Pharmacy Practice and Clinical Pharmacy Department, Faculty of Pharmacy, Galala University-Ataka, Suez, Egypt
| | - Heba Mohammed Refat M Selim
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, 11597, Riyadh, Saudi Arabia
| | - Ahmed A Al-Karmalawy
- Department of Pharmaceutical Chemistry, College of Pharmacy, The University of Mashreq, Baghdad, 10023, Iraq.
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University, New Damietta, 34518, Egypt.
| |
Collapse
|
8
|
Ezhilarasan D, Karthikeyan S, Najimi M, Vijayalakshmi P, Bhavani G, Jansi Rani M. Preclinical liver toxicity models: Advantages, limitations and recommendations. Toxicology 2025; 511:154020. [PMID: 39637935 DOI: 10.1016/j.tox.2024.154020] [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: 10/21/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
Experimental animal models are crucial for elucidating the pathophysiology of liver injuries and for assessing new hepatoprotective agents. Drugs and chemicals such as acetaminophen, isoniazid, valproic acid, ethanol, carbon tetrachloride (CCl4), dimethylnitrosamine (DMN), and thioacetamide (TAA) are metabolized by the CYP2E1 enzyme, producing hepatotoxic metabolites that lead to both acute and chronic liver injuries. In experimental settings, acetaminophen (centrilobular necrosis), carbamazepine (centrilobular necrosis and inflammation), sodium valproate (necrosis, hydropic degeneration and mild inflammation), methotrexate (sinusoidal congestion and inflammation), and TAA (centrilobular necrosis and inflammation) are commonly used to induce various types of acute liver injuries. Repeated and intermittent low-dose administration of CCl4, TAA, and DMN activates quiescent hepatic stellate cells, transdifferentiating them into myofibroblasts, which results in abnormal extracellular matrix production and fibrosis induction, more rapidly with DMN and CCL4 than TAA (DMN > CCl4 > TAA). Regarding toxicity and mortality, CCl4 is more toxic than DMN and TAA (CCl4 > DMN > TAA). Models used to induce metabolic dysfunction-associated liver disease (MAFLD) vary, but MAFLD's multifactorial nature driven by factors like obesity, fatty liver, dyslipidaemia, type II diabetes, hypertension, and cardiovascular disease makes it challenging to replicate human metabolic dysfunction-associated steatohepatitis accurately. From an experimental point of view, the degree and pattern of liver injury are influenced by various factors, including the type of hepatotoxic agent, exposure duration, route of exposure, dosage, frequency of administration, and the animal model utilized. Therefore, there is a pressing need for standardized protocols and regulatory guidelines to streamline the selection of animal models in preclinical studies.
Collapse
Affiliation(s)
- Devaraj Ezhilarasan
- Department of Pharmacology, Hepatology and Molecular Medicine Lab, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India.
| | - Sivanesan Karthikeyan
- Department of Pharmacology and Environmental Toxicology, Dr. A.L.M. Postgraduate Institute of Basic Medical Sciences, University of Madras, Chennai, India
| | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
| | - Paramasivan Vijayalakshmi
- Department of Pharmacology and Environmental Toxicology, Dr. A.L.M. Postgraduate Institute of Basic Medical Sciences, University of Madras, Chennai, India; Department of Pharmacology, Asan Memorial Dental College and Hospital, Chengalpattu, Tamil Nadu, India
| | - Ganapathy Bhavani
- Department of Pharmacology and Environmental Toxicology, Dr. A.L.M. Postgraduate Institute of Basic Medical Sciences, University of Madras, Chennai, India; Department of Pharmacology, Meenakshi Ammal Dental College and Hospital, Meenakshi Academy of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Muthukrishnan Jansi Rani
- Department of Pharmacology and Environmental Toxicology, Dr. A.L.M. Postgraduate Institute of Basic Medical Sciences, University of Madras, Chennai, India
| |
Collapse
|
9
|
El-Sayed NM, Menze ET, Tadros MG, Hanna DMF. Mangiferin mitigates methotrexate-induced liver injury and suppresses hepatic stellate cells activation in rats: Imperative role of Nrf2/NF-κB/NLRP3 signaling axis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119296. [PMID: 39732297 DOI: 10.1016/j.jep.2024.119296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 11/16/2024] [Accepted: 12/26/2024] [Indexed: 12/30/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Mangifera indica (family Anacardiaceae), often acknowledged as mango and renowned for being a plant of diverse ethnopharmacological background since ancient times, harbors the polyphenolic bioactive constituent, mangiferin (MNG). MNG is a major phytochemical of Mangifera indica and other plants with a wide range of reported pharmacological activities, including antioxidant, anti-inflammatory, neuroprotective and hepatoprotective effects. MNG has also been utilized in traditional medicine; it is reportedly a major bioactive element in over 40 polyherbal products in traditional Chinese medicine (TCM), and two prominent anti-inflammatory, immunomodulatory and antiviral Cuban formulations. Despite the availability of evidence in support of MNG hepatoprotective properties, its hepatoprotective potential against MTX-induced liver injury and fibrosis has not been explored yet. AIM To unravel the hepatoprotective potential of MNG against MTX-induced hepatic injury and fibrosis and elucidate the possible underlying molecular mechanisms. MATERIALS AND METHODS Male Sprague-Dawley rats were, randomly, distributed into five groups; two of which were administered MNG 50 mg/kg and MNG 100 mg/kg intraperitoneally (i.p.) for ten days, and a single i.p. injection of MTX 40 mg/kg on the seventh day to establish hepatotoxicity. Blood and liver tissue samples were retrieved from all study groups and analyzed for liver functions, histopathological alterations, and oxidative stress, inflammatory, and fibrotic biomarkers. RESULTS MNG restored the MTX-induced degenerations in hepatic architecture and function. Moreover, it combated the MTX-elicited oxidative stress evidently by the significantly attenuated hepatic tissue levels of malondialdehyde, and the significantly elevated reduced glutathione and Nrf2 levels. MNG also halted inflammation depicted by the downregulation of the NF-κB/NLRP3 inflammasome axis. It further demonstrated anti-fibrogenic potential as evidenced by the significant reduction in fibrous tissue deposition and hepatic expression of α-SMA. CONCLUSION The current study proved the hepatoprotective, and anti-fibrogenic effects of MNG against MTX-induced hepatotoxicity via the downregulation of NF-κB/NLRP3 inflammasome signaling axis, preceded by the amelioration of oxidative stress and Nrf2 signaling upregulation.
Collapse
Affiliation(s)
- Nada M El-Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566, Cairo, Egypt; School of Life and Medical Sciences, University of Hertfordshire Hosted By Global Academic Foundation, New Administrative Capital, Cairo, Egypt
| | - Esther T Menze
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566, Cairo, Egypt
| | - Mariane G Tadros
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566, Cairo, Egypt
| | - Diana M F Hanna
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566, Cairo, Egypt.
| |
Collapse
|
10
|
Famurewa AC, George MY, Ukwubile CA, Kumar S, Kamal MV, Belle VS, Othman EM, Pai SRK. Trace elements and metal nanoparticles: mechanistic approaches to mitigating chemotherapy-induced toxicity-a review of literature evidence. Biometals 2024; 37:1325-1378. [PMID: 39347848 DOI: 10.1007/s10534-024-00637-7] [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: 04/15/2024] [Accepted: 08/30/2024] [Indexed: 10/01/2024]
Abstract
Anticancer chemotherapy (ACT) remains a cornerstone in cancer treatment, despite significant advances in pharmacology over recent decades. However, its associated side effect toxicity continues to pose a major concern for both oncology clinicians and patients, significantly impacting treatment protocols and patient quality of life. Current clinical strategies to mitigate ACT-induced toxicity have proven largely unsatisfactory, leaving a critical unmet need to block toxicity mechanisms without diminishing ACT's therapeutic efficacy. This review aims to document the molecular mechanisms underlying ACT toxicity and highlight research efforts exploring the protective effects of trace elements (TEs) and their nanoparticles (NPs) against these mechanisms. Our literature review reveals that the primary driver of ACT toxicity is redox imbalance, which triggers oxidative inflammation, apoptosis, endoplasmic reticulum stress, mitochondrial dysfunction, autophagy, and dysregulation of signaling pathways such as PI3K/mTOR/Akt. Studies suggest that TEs, including zinc, selenium, boron, manganese, and molybdenum, and their NPs, can potentially counteract ACT-induced toxicity by inhibiting oxidative stress-mediated pathways, including NF-κB/TLR4/MAPK/NLRP3, STAT-3/NLRP3, Bcl-2/Bid/p53/caspases, and LC3/Beclin-1/CHOP/ATG6, while also upregulating protective signaling pathways like Sirt1/PPAR-γ/PGC-1α/FOXO-3 and Nrf2/HO-1/ARE. However, evidence regarding the roles of lncRNA and the Wnt/β-catenin pathway in ACT toxicity remains inconsistent, and the impact of TEs and NPs on ACT efficacy is not fully understood. Further research is needed to confirm the protective effects of TEs and their NPs against ACT toxicity in cancer patients. In summary, TEs and their NPs present a promising avenue as adjuvant agents for preventing non-target organ toxicity induced by ACT.
Collapse
Affiliation(s)
- Ademola C Famurewa
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medical Sciences, Alex Ekwueme Federal University Ndufu-Alike Ikwo, Abakaliki, Ebonyi, Nigeria.
- Centre for Natural Products Discovery, School of P harmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF, UK.
- Department of Pharmacology, Manipal College of Pharmaceutical Science, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| | - Mina Y George
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Cletus A Ukwubile
- Department of Pharmacognosy, Faculty of Pharmacy, University of Maiduguri, Bama Road, Maiduguri, Borno, Nigeria
| | - Sachindra Kumar
- Department of Pharmacology, Manipal College of Pharmaceutical Science, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Mehta V Kamal
- Department of Biochemistry, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Vijetha S Belle
- Department of Biochemistry, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Eman M Othman
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
- Cancer Therapy Research Center, Department of Biochemistry-I, Biocenter, University of Würzburg, Am Hubland, 97074, Würzburg, Germany
- Department of Bioinformatics, University of Würzburg, Am Hubland, 97074, BiocenterWürzburg, Germany
| | - Sreedhara Ranganath K Pai
- Department of Pharmacology, Manipal College of Pharmaceutical Science, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| |
Collapse
|
11
|
Chen Q, Zhao X, Xu Z, Liu Y. Endoplasmic reticulum stress mechanisms and exercise intervention in type 2 diabetes mellitus. Biomed Pharmacother 2024; 177:117122. [PMID: 38991302 DOI: 10.1016/j.biopha.2024.117122] [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: 04/02/2024] [Revised: 07/08/2024] [Accepted: 07/08/2024] [Indexed: 07/13/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a metabolic disease primarily characterized by insulin resistance (IR) and insufficient insulin secretion. The unfolded protein response (UPR) overactivation induced by endoplasmic reticulum stress (ERS) appears to play a key role in this process, although the exact pathogenesis of T2DM is not fully understood. Studies have demonstrated that appropriate exercise can regulate ERS in the heart, liver, pancreas, skeletal muscle, and other body tissues leading to an improvement in diabetes and its complications. However, the exact mechanism remains unclear. By analyzing the relationship between ERS, T2DM pathology, and exercise intervention, this review concludes that exercise can increase insulin sensitivity, inhibit IR, promote insulin secretion and alleviate T2DM by regulating ERS. This paper specifically reviews the signaling pathways by which ERS induces diabetes, the mechanisms of exercise regulation of ERS in diabetes, and the varying effects of different types of exercise on diabetes improvement through ERS mechanisms. Physical exercise is an effective non-pharmacological intervention for T2DM. Thus, further exploration of how exercise regulates ERS in diabetes could refine "precision exercise medicine" for diabetes and identify new drug targets.
Collapse
Affiliation(s)
- Qianyu Chen
- College of Physical Education, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China.
| | - Xiaoqin Zhao
- College of Physical Education, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China.
| | - Zujie Xu
- College of Physical Education, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China.
| | - Yiyao Liu
- College of Physical Education, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China.
| |
Collapse
|
12
|
El-Gohary RM, Abdeen A, Ibrahim HA, Taher ES, Ghabrial MM, Younis RL, Khattab H, Seleem MA, Alwutayed KM, Mihaela O, Ioan BD, El-Nablaway M, Aldarmahi AA, Ibrahim AM, Al-Serwi RH, Ghalwash AA. Empagliflozin impact on experimentally induced acetaminophen toxicity: Imprint of mitochondrial dynamics, biogenesis, and cGAS/STING signal in amending liver insult. FASEB J 2024; 38:e23816. [PMID: 39072779 DOI: 10.1096/fj.202400254rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 06/25/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024]
Abstract
Acetaminophen (APAP) is one of the most clinically relevant medications associated with acute liver damage. A prolific deal of research validated the hepatoprotective effect of empagliflozin (EMPA); however, its effect on APAP-induced hepatotoxicity has still not been investigated. In this study, the prospective hepatoprotective impact of EMPA against APAP-induced hepatotoxicity was investigated. Twenty-eight Balb-C mice were assigned to four groups: control, APAP, EMPA10/APAP, and EMPA25/APAP. At the end of the experiment, serum hepatotoxicity biomarkers, MDA level, and GSH content were estimated. Hepatic mitofusin-2 (MFN2), optic atrophy 1 (OPA1), dynamin-related protein 1 (Drp1), and mitochondrial fission 1 protein (FIS1) were immunoassayed. PGC-1α, cGAS, and STING mRNA expression were assessed by real-time PCR. Histopathological changes and immunohistochemistry of INF-β, p-NF-κB, and iNOS were evaluated. APAP treatment caused significant hepatic functional impairment and increased hepatic MDA levels, as well as a concomitant decrease in GSH content. Marked elevation in Drp1 and FIS1 levels, INF-ß, p-NF-κB, and iNOS immunoreactivity, and reduction in MFN2 and OPA1 levels in the APAP-injected group, PGC-1α downregulation, and high expression of cGAS and STING were also documented. EMPA effectively ameliorated APAP-generated structural and functional changes in the liver, restored redox homeostasis and mitochondrial dynamics balance, and enhanced mitochondrial biogenesis, remarkably diminished hepatic expression of cGAS and STING, and elicited a reduction in hepatic inflammation. Moreover, the computational modeling data support the interaction of APAP with antioxidant system-related proteins as well as the interactions of EMPA against Drp1, cGAS, IKKA, and iNOS proteins. Our findings demonstrated for the first time that EMPA has an ameliorative impact against APAP-induced hepatotoxicity in mice via modulation of mitochondrial dynamics, biogenesis, and cGAS/STING-dependent inflammation. Thus, this study concluded that EMPA could be a promising therapeutic modality for acute liver toxicity.
Collapse
Affiliation(s)
- Rehab M El-Gohary
- Department of Medical Biochemistry, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Ahmed Abdeen
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Benha University, Toukh, Egypt
| | - Hoda A Ibrahim
- Department of Medical Biochemistry, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Ehab S Taher
- Department of Basic Medical and Dental Sciences, Faculty of Dentistry, Zarqa University, Zarqa, Jordan
| | - Maram M Ghabrial
- Department of Anatomy and Embryology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Reham L Younis
- Department of Physiology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Haidy Khattab
- Department of Physiology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Monira A Seleem
- Department of Medical Pharmacology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Khairiah M Alwutayed
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Ostan Mihaela
- Department of Biology, Faculty of Agriculture, University of Life Sciences "King Michael I" from Timisoara, Timisoara, Romania
| | - Banatean-Dunea Ioan
- Department of Biology, Faculty of Agriculture, University of Life Sciences "King Michael I" from Timisoara, Timisoara, Romania
| | - Mohammad El-Nablaway
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia
| | - Ahmed A Aldarmahi
- Department of Basic Science, College of Science and Health Professions, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- National Guard- Health Affairs, King Abdullah International Medical Research Centre, Jeddah, Saudi Arabia
| | - Ateya M Ibrahim
- Department of Administration and Nursing Education, College of Nursing, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
- Department of Family and Community Health Nursing, Faculty of Nursing, Port-Said University, Port Said, Egypt
| | - Rasha H Al-Serwi
- Department of Basic Sciences, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Asmaa A Ghalwash
- Department of Medical Biochemistry, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
13
|
Li Y, Yang P, Ye J, Xu Q, Wu J, Wang Y. Updated mechanisms of MASLD pathogenesis. Lipids Health Dis 2024; 23:117. [PMID: 38649999 PMCID: PMC11034170 DOI: 10.1186/s12944-024-02108-x] [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: 11/20/2023] [Accepted: 04/11/2024] [Indexed: 04/25/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) has garnered considerable attention globally. Changing lifestyles, over-nutrition, and physical inactivity have promoted its development. MASLD is typically accompanied by obesity and is strongly linked to metabolic syndromes. Given that MASLD prevalence is on the rise, there is an urgent need to elucidate its pathogenesis. Hepatic lipid accumulation generally triggers lipotoxicity and induces MASLD or progress to metabolic dysfunction-associated steatohepatitis (MASH) by mediating endoplasmic reticulum stress, oxidative stress, organelle dysfunction, and ferroptosis. Recently, significant attention has been directed towards exploring the role of gut microbial dysbiosis in the development of MASLD, offering a novel therapeutic target for MASLD. Considering that there are no recognized pharmacological therapies due to the diversity of mechanisms involved in MASLD and the difficulty associated with undertaking clinical trials, potential targets in MASLD remain elusive. Thus, this article aimed to summarize and evaluate the prominent roles of lipotoxicity, ferroptosis, and gut microbes in the development of MASLD and the mechanisms underlying their effects. Furthermore, existing advances and challenges in the treatment of MASLD were outlined.
Collapse
Affiliation(s)
- Yuxuan Li
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Translational Medicine Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Peipei Yang
- Translational Medicine Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Jialu Ye
- Translational Medicine Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Qiyuan Xu
- Wenzhou Medical University, Wenzhou, China
| | - Jiaqi Wu
- Translational Medicine Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China.
- Department of Gastroenterology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China.
| | - Yidong Wang
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|