1
|
Rofaeil RR, Sharata EE, Attya ME, Abo-Youssef AM, Hemeida RA, Khalaf MM. Repurposing levomilnacipran to attenuate premature ovarian insufficiency induced by cyclophosphamide in female Wistar albino rats through modulation of TLR4/p38-MAPK/NF-κB p65, caspase-3-driven apoptosis, and Klotho protein expression. Food Chem Toxicol 2025; 200:115406. [PMID: 40154831 DOI: 10.1016/j.fct.2025.115406] [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: 03/06/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
AIM This study aims to explore the mitigative impact of levomilnacipran (LVM) against cyclophosphamide (CPA)-induced premature ovarian insufficiency by targeting TLR4/p38 MAPK/NF-κB p65, and klotho expression. METHODS Rats were allocated into five groups as follows: control, LVM, CPA, CPA + LVM, and CPA + TRI. Serum hormones and histopathological examination were performed. To assess oxidative stress, ovarian MDA, GSH, and SOD were evaluated. The ovarian contents of caspase-3 and inflammatory markers were assessed using the ELISA method. The expression of ovarian NF-κB p65 was examined using an immunohistochemical technique. RT-qPCR was used to measure Bax and Bcl-2 mRNA expression. Utilizing a Western blot, the TLR4, p38 MAPK, α-Klotho, and cleaved caspase-3 levels were estimated. The estrous cycle was also monitored. FINDINGS LVM attenuated CPA-induced ovarian toxicity by regulating hormones and alleviating histopathological aberrations. It also raised SOD and GSH levels and lowered MDA's ovarian content. Moreover, Bcl-2 levels were raised, Bax and caspase-3 expression levels were reduced, and IL-18, IL-1β, and TNF-α levels were all reduced. LVM-induced ovarian protection by diminishing TLR4/p38 MAPK/NF-κB p65 expression and boosting the protein levels of α-Klotho. SIGNIFICANCE LVM mitigated POI caused by CPA by downregulating TLR4/p38 MAPK/NF-κB p65, enhancing the α-Klotho level and attenuating caspase-3 derived apoptosis.
Collapse
Affiliation(s)
- Remon Roshdy Rofaeil
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Deraya University, Minia, 61111, Egypt; Department of Medical Pharmacology, Faculty of Medicine, Minia University, Minia, 61519, Egypt.
| | - Ehab E Sharata
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Deraya University, Minia, 61111, Egypt.
| | - Mina Ezzat Attya
- Department of Pathology, Faculty of Medicine, Minia University, Minia, 61519, Egypt.
| | - Amira M Abo-Youssef
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt.
| | - Ramadan Am Hemeida
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Deraya University, Minia, 61111, Egypt.
| | - Marwa M Khalaf
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt.
| |
Collapse
|
2
|
Khallaf WAI, Sharata EE, Attya ME, Rofaeil RR, Khalaf MM, Hemeida RAM, Abo-Youssef AM. Buspirone ameliorates premature ovarian insufficiency evoked by cyclophosphamide in female rats; attention to AMPK/Nrf2/HO-1, α-Klotho/NLRP3/Caspase-1, and Caspase-3-mediated apoptosis interplay. Toxicol Appl Pharmacol 2025; 500:117373. [PMID: 40345558 DOI: 10.1016/j.taap.2025.117373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 04/11/2025] [Accepted: 05/02/2025] [Indexed: 05/11/2025]
Abstract
This study aims to investigate the protective impact of buspirone (BUS) against cyclophosphamide (CPA)-induced premature ovarian insufficiency (POI) by focusing on pyroptosis, apoptosis, and the AMPK/Nrf2/HO-1 signaling pathway. POI was achieved by i.p. injection of CPA in female Wistar albino rats. CPA toxicity was evaluated using biochemical analysis of the serum hormones (AMH, FSH, inhibin B, and estrogen) and histopathological examination. Oxidative stress markers (MDA, SOD) were also evaluated. Levels of inflammatory indicators (TNF-α, IL-1β, and IL-18), apoptotic marker (caspase-3), ovarian p-AMPK, ovarian NF-κB, Nrf2, and HO-1 were evaluated. RT-qPCR was used to measure Bax and Bcl-2 mRNA expression. A western blot assay was used to determine the expression of α-Klotho, NLRP3, and caspase 1. The estrous cycle and the weights of the body and ovaries were also observed. BUS, in a dose-dependent manner, attenuated CPA-induced ovarian toxicity by regulating hormonal and estrous cycle irregularities and alleviating the histopathological aberrations. It also lowered MDA levels and increased SOD activity. Furthermore, it reduced NF-κB, TNF-α, IL-1β, and IL-18 levels, as well as BAX and caspase-3 expression, while raising Bcl-2 levels. Additionally, BUS enhanced Nrf2 and HO-1 expression and boosted the protein levels of p-AMPK and α-Klotho. As well, it diminished pyroptosis by decreasing NLRP3 and caspase-1 expression. BUS attenuated POI induced by CPA, showing potential for effective protection via increasing the activity of Nrf2/HO-1 and reducing the activity of NLRP3/Caspase-1 through the participation of α-Klotho and p-AMPK, as well as inhibiting caspase-3-driven apoptosis.
Collapse
Affiliation(s)
- Waleed A I Khallaf
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt.
| | - Ehab E Sharata
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Deraya University, Minia 61111, Egypt.
| | - Mina Ezzat Attya
- Department of Pathology, Faculty of Medicine, Minia University, Minia 61519, Egypt.
| | - Remon Roshdy Rofaeil
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Deraya University, Minia 61111, Egypt; Department of Medical Pharmacology, Faculty of Medicine, Minia University, Minia 61519, Egypt.
| | - Marwa M Khalaf
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt.
| | - Ramadan A M Hemeida
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Deraya University, Minia 61111, Egypt.
| | - Amira M Abo-Youssef
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt.
| |
Collapse
|
3
|
Saleh DO, Abo El Nasr NME, Hussien YA, El-Baset MA, Ahmed KA. Cyclophosphamide-induced testicular injury: the role of chrysin in mitigating iron overload and ferroptosis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:5475-5489. [PMID: 39565397 PMCID: PMC11985597 DOI: 10.1007/s00210-024-03519-4] [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: 07/28/2024] [Accepted: 10/06/2024] [Indexed: 11/21/2024]
Abstract
This study evaluated the beneficial effects of chrysin against cyclophosphamide (CP)-induced testicular toxicity in rats across several parameters, including hormones, oxidative stress, inflammation, apoptosis, and protein expression. Rats were pretreated with oral doses of chrysin at 25, 50, or 100 mg/kg daily for 7 days. On the 8th day, all groups except controls received CP (200 mg/kg) injection. Chrysin doses continued for 7 more days. Hormones, oxidative stress markers, inflammatory cytokines, apoptosis regulators, and iron regulatory proteins were assessed. CP decreased testosterone, inhibin B, GSH, and GPx4 and increased FSH, cholesterol, MDA, IL-6, and BAX. It also drastically reduced TfR1, liprin, and IREB2. Chrysin dose-dependently counteracted these effects. The highest 100 mg/kg chrysin dose increased testosterone, inhibin B, GSH, GPx4, BCL2, TfR1, liprin, and IREB2 while decreasing FSH, cholesterol, MDA, IL-6, and BAX close to control levels. There were also significant incremental benefits for testosterone, inhibin B, and other parameters with higher chrysin doses. Chrysin dose-dependently attenuated CP-induced hormonal dysfunction, oxidative stress, inflammation, apoptosis, and iron-regulatory protein suppression. The maximum dose showed the most optimal protective effects in restoring the testicular toxicity markers. These results validate the promising spermatoprotective properties of chrysin against chemotherapeutic germ cell damage.
Collapse
Affiliation(s)
- Dalia O Saleh
- Pharmacology Depatrment, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Nesma M E Abo El Nasr
- Pharmacology Depatrment, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt.
| | - Yosra A Hussien
- Pharmacology Depatrment, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Marawan Abd El-Baset
- Pharmacology Depatrment, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Kawkab A Ahmed
- Pathology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
4
|
Khattab MA, Ahmed SM, Salama HG, Mekawy NH. Effect of olanzapine on testes of adult albino rats and the possible role of granulocyte colony stimulating factor versus umbelliferone light and electron microscopic study. Ultrastruct Pathol 2025; 49:265-287. [PMID: 40275515 DOI: 10.1080/01913123.2025.2495159] [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: 03/05/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/26/2025]
Abstract
Olanzapine (OLZ) is one of atypical antipsychotic drugs (second generation) used for treating schizophrenia, manic, and mixed episodes of bipolar disorder. Continuous evaluation of its effects is necessary, and there is a need to explore alternative natural products as G-CSF and UMB to manage potential side effects. This research designed to mitigate the atypical antipsychotic drugs' adverse effects through biochemical analyses, light and electron microscopic studies. Fifty-six rats were divided into five groups: Control, OLZ, G-CSF, UMB, and Recovery groups. End body and testicular weights, serum testosterone, testicular MDA levels, and seminal analysis were recorded. Testicular specimens were processed to evaluate histological structure, PCNA, and CD34 immune expression. Morphometric and statistical analyses were also performed. OLZ group exhibited a distorted testicular structure, a significant increase in end body and a decline in testicular weight, a significant decline in the serum level of testosterone level, testicular MDA, and seminal analysis parameters. Furthermore, disturbed histoarchitecture, reduction in PCNA, and elevation in CD34 immunoreaction were observed. These alterations were partially attenuated by G-CSF therapy, whereas UMB significantly improved all parameters. In conclusion, UMB, and to a lesser degree G-CSF, appeared to be superior therapeutic options by attenuating oxidative stress and restoring intact histological structure and biochemical parameters.
Collapse
Affiliation(s)
- Maha A Khattab
- Medical Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig City, Egypt
| | - Samah M Ahmed
- Medical Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig City, Egypt
| | - Haidy G Salama
- Medical Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig City, Egypt
| | - Noura H Mekawy
- Medical Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig City, Egypt
| |
Collapse
|
5
|
Sharata EE, Attya ME, Khalaf MM, Rofaeil RR, Abo-Youssef AM, Hemeida RAM. Levomilnacipran alleviates cyclophosphamide-induced hepatic dysfunction in male Wistar albino rats; emerging role of α-Klotho/TLR4/p38-MAPK/NF-κB p65 and caspase-3-driven apoptosis trajectories. Int Immunopharmacol 2025; 152:114384. [PMID: 40056515 DOI: 10.1016/j.intimp.2025.114384] [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: 12/10/2024] [Revised: 02/23/2025] [Accepted: 02/26/2025] [Indexed: 03/10/2025]
Abstract
AIM This study aims to investigate the potential protective effect of levomilnacipran (LVM) against cyclophosphamide (CPA)-induced hepatotoxicity by targeting α-Klotho/TLR4/p38-MAPK/NF-κB p65 and Caspase-3-dependent apoptosis signaling pathways. MAIN METHODS The toxicity of CPA was assessed using biochemical analysis of the serum hepatotoxicity parameters (AST, ALT, and direct bilirubin) and histopathological examination. Hepatic MDA and SOD were evaluated. The ELISA procedure was employed to evaluate the levels of hepatic TNF-α, IL-1β, and IL-18, hepatic caspase-3, and serum α-Klotho. The expression of hepatic TLR4 and NF-κB p65 was examined using an immunohistochemical technique. A western blot assay was used to determine the expression of MYD88, and p38-MAPK. KEY FINDINGS LVM abrogated CPA-induced hepatotoxicity by reducing the elevated hepatoxicity markers and mitigating the histopathological aberrations. It also lowered MDA content and increased SOD activity. Furthermore, it reduced TNF-α, IL-1β, and IL-18 contents, as well as caspase-3 activity. Additionally, LVM diminished TLR4, MYD88, NF-κB p65, and p38 MAPK expression and boosted the levels of α-Klotho. SIGNIFICANCE LVM alleviated hepatic injury generated by CPA via downregulating TLR4/p38 MAPK/NF-κB p65 signaling cascade through the participation of α-Klotho, as well as inhibiting caspase-3-driven apoptosis.
Collapse
Affiliation(s)
- Ehab E Sharata
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Deraya University, Minia 61111, Egypt.
| | - Mina Ezzat Attya
- Department of Pathology, Faculty of Medicine, Minia University, Minia 61519, Egypt.
| | - Marwa M Khalaf
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt.
| | - Remon Roshdy Rofaeil
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Deraya University, Minia 61111, Egypt; Department of Medical Pharmacology, Faculty of Medicine, Minia University, Minia 61519, Egypt.
| | - Amira M Abo-Youssef
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt.
| | - Ramadan A M Hemeida
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Deraya University, Minia 61111, Egypt.
| |
Collapse
|
6
|
Almuqati AF. Cyclophosphamide-induced testicular injury is associated with inflammation, oxidative stress, and apoptosis in mice: Protective role of taxifolin. Reprod Biol 2025; 25:100990. [PMID: 39764890 DOI: 10.1016/j.repbio.2024.100990] [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: 10/19/2024] [Revised: 11/27/2024] [Accepted: 12/16/2024] [Indexed: 02/26/2025]
Abstract
Testicular damage is a major complication of chemotherapeutic cyclophosphamide (CP) compound. Taxifolin (TX), a natural flavonoid with well-established anti-inflammatory and antioxidant properties, is commonly found in various medicinal plants and foods. This study investigated the protective effect of TX against testicular damage in CP-administered mice. Mice were administered with TX at the doses of 10, 25, and 50 mg/kg for 15 days followed by a single CP injection on the 16th day. CP-administered mice demonstrated significantly decreased testosterone levels and low sperm parameters (count, viability, motility). TX administration significantly improved sperm parameters and testosterone levels and effectively mitigated histopathological testicular changes in CP-administered animals. Moreover, TX administration decreased oxidative stress markers and boosted antioxidants (superoxide dismutase, catalase, and reduced glutathione), suppressed and NF-κB p65 and pro-inflammatory cytokines [TNF-α (tumor necrosis factor-alpha) and IL-6 (interleukin-6)], and reduced apoptosis as depicted by testicular levels of caspase-3, Bcl-2, and Bax. Thus, TX could be a highly potent compound to counter CP-linked testicular damage through modulation of oxidative stress, inflammation, and apoptosis, warranting further studies to evaluate the role of TX in human CP-induced testicular injury.
Collapse
Affiliation(s)
- Afaf F Almuqati
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hafr Al-Batin, Hafr Al-Batin 31991, Saudi Arabia.
| |
Collapse
|
7
|
El-Marasy SA, Mostafa RE, Mabrok HB, Khattab MS, Awdan SAE. Protective effect of irbesartan against hepatic ischemia-reperfusion injury in rats: role of ERK, STAT3, and PPAR-γ inflammatory pathways in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:1681-1693. [PMID: 39167169 PMCID: PMC11825560 DOI: 10.1007/s00210-024-03301-6] [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: 03/25/2024] [Accepted: 07/15/2024] [Indexed: 08/23/2024]
Abstract
This study aimed to elucidate the possible hepatocellular protective role of irbesartan during hepatic ischemia-reperfusion injury (HIRI) and the probable underlying mechanisms. Wistar rats were allocated into four groups: sham; HIRI (control); irbesartan (50 mg/kg) + HIRI; irbesartan (100 mg/kg) + HIRI; irbesartan + GW9662 (1 mg/kg, i.p.) + HIRI. Rats pretreated orally with irbesartan or vehicle for 14 days underwent 45-min hepatic ischemia followed by 60-min reperfusion. Irbesartan preconditioning diminished alanine transaminase (ALT) and aspartate transaminase (AST) serum levels, and reduced extracellular signal-regulated kinase (ERK) and signal transducer and activator of transcription 3 (STAT3). Irbesartan decreased proapoptotic BAX (bcl-2-like protein 4), increased anti-apoptotic B-cell lymphoma 2 (BCL2) hepatic content, and thereby reduced BAX/BCL2 ratio. Moreover, irbesartan preconditioning reduced autophagy-related proteins Beclin1 and LC3 II, and elevated p62 (protein responsible for autophagosome degradation). It elevated proliferator-activated receptor γ (PPAR-γ), and reduced tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) hepatic gene expression. Also, hepatic protein expressions of nuclear factor kappa-B p65 (NF-κB p65) and caspase-3 were lessoned by irbesartan pretreatment in HIRI rats. However, GW9662 abrogated irbesartan's effect on HIRI. The protective effect of irbesartan on HIRI may be mediated by alleviation of ERK, STAT3, and PPAR-γ inflammatory pathways, exerting anti-apoptotic and anti-autophagic effects in HIRI in rats.
Collapse
Affiliation(s)
- Salma A El-Marasy
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt.
| | - Rasha E Mostafa
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Hoda B Mabrok
- Nutrition and Food Science Department, Food Industries and Nutrition Research Institute, National Research Centre, Giza, Egypt
| | - Marwa S Khattab
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Sally A El Awdan
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| |
Collapse
|
8
|
Wang H, Zhang H, Miao L, Wang C, Teng H, Li X, Zhang X, Yang G, Wang S, Zeng X. α-amanitin induces hepatotoxicity via PPAR-γ inhibition and NLRP3 inflammasome activation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117749. [PMID: 39862693 DOI: 10.1016/j.ecoenv.2025.117749] [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: 11/07/2024] [Revised: 01/07/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
Mushroom poisoning, predominantly caused by α-amanitin, is a critical food safety concern in worldwide, with severe cases leading to hepatotoxicity and fatalities. This study delves into the hepatotoxic effects of α-amanitin, focusing on the NLRP3 inflammasome and PPAR-γ's regulatory role in inflammation. In vitro studies with L-02 cells showed that α-amanitin reduces cell viability and triggers NLRP3 inflammasome activation, increasing NF-κB phosphorylation and pro-inflammatory cytokines IL-18 and IL-1β. The NLRP3 inhibitor MCC950 mitigated these effects without impacting NF-κB. Conversely, PPAR-γ knockdown intensified the inflammatory response. In vivo, α-amanitin induced dose-dependent liver injury in mice, evident by elevated serum ALT and AST, and histological liver damage. MCC950 pretreatment offered protection against hepatotoxicity, while PPAR-γ inhibition with GW9662 worsened the condition. The study highlights the interplay between α-amanitin, NLRP3, and PPAR-γ in hepatotoxicity, proposing potential therapeutic targets for mushroom poisoning-induced liver diseases.
Collapse
Affiliation(s)
- Haowei Wang
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Huijie Zhang
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Lin Miao
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Chan Wang
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Hanxin Teng
- Department of Pathogen Biology and Immunology, School of Basic Medical Science, Kunming Medical University, Kunming, China
| | - Xiaodong Li
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Xiaoxing Zhang
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Genmeng Yang
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China.
| | - Shangwen Wang
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China.
| | - Xiaofeng Zeng
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China.
| |
Collapse
|
9
|
Ahmed HA, Gatea FK, Hussein ZA. Azilsartan as a preventive agent against cyclophosphamide-induced testicular injury in male rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:979-990. [PMID: 39093466 DOI: 10.1007/s00210-024-03339-6] [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: 06/05/2024] [Accepted: 07/26/2024] [Indexed: 08/04/2024]
Abstract
Cyclophosphamide (CP) is a popular cancer treatment; however, despite its efficacy, it is known to cause harm to the testicles. To mitigate the reproductive damage caused by CP in male rats, we examined the protective effect of azilsartan (AZ) on CP-induced testicular damage. Thirty Sprague-Dawley male rats were equally divided into three groups: normal control group: received 0.5% CMC suspension for 13 days; induction group: received a single dose of 200 mg/kg of CP on day 6 by intraperitoneal (IP) injection, azilsartan group: received azilsartan (4 mg/kg) orally for 5 days followed by a single dose of 200 mg/kg of (CP) on day 6 by IP injection, then azilsartan administered again for 7 days. Animals were sacrificed on day 14, and sperm characteristics, testosterone levels, and testicular histopathology were evaluated. Induction with CP caused a significant reduction in median value compared to normal control in sperm count (12.0 vs. 22.0 × 106/mm3), sperm motility (30 vs. 90%), abnormal sperm (30.32 vs. 14.43%), dead sperm count (32.43 vs. 10.49 × 106/mm3), DNA fragmentation (21.57 vs. 5.49%); meanwhile, azilsartan prevent these effects on median sperm count (17.0 × 106/mm3), sperm motility (70.0%), abnormal sperm (23.19%), dead sperm count (26.17 × 106/mm3), DNA fragmentation (13.81%), and improved plasmatic testosterone levels compared to the CP group and prevented histopathological alterations of the testes. Azilsartan's mitigation of CP's effects suggests it can prevent male rats' reproductive damage caused by CP. One possible explanation for AZ's protective effects is that it inhibits lipid peroxidation and has antioxidant properties.
Collapse
Affiliation(s)
- Haneen Alaa Ahmed
- Department of Pharmacology, College of Medicine, Al-Nahrain University, Baghdad, Iraq.
| | - Fouad Kadhim Gatea
- Department of Pharmacology, College of Medicine, Al-Nahrain University, Baghdad, Iraq
| | - Zeena Ayad Hussein
- Department of Pharmacology, College of Medicine, Al-Nahrain University, Baghdad, Iraq
| |
Collapse
|
10
|
Abd Elmaaboud MA, Kabel AM, Borg HM, Magdy AA, Kabel SM, Arafa ESA, Alsufyani SE, Arab HH. Omarigliptin/rosinidin combination ameliorates cyclophosphamide-induced lung toxicity in rats: The interaction between glucagon-like peptide-1, TXNIP/NLRP3 inflammasome signaling, and PI3K/Akt/FoxO1 axis. Biomed Pharmacother 2024; 177:117026. [PMID: 38936197 DOI: 10.1016/j.biopha.2024.117026] [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: 04/23/2024] [Revised: 06/12/2024] [Accepted: 06/21/2024] [Indexed: 06/29/2024] Open
Abstract
Cyclophosphamide is an anti-neoplastic drug that has shown competence in the management of a broad range of malignant tumors. In addition, it represents a keystone agent for management of immunological conditions. Despite these unique properties, induction of lung toxicity may limit its clinical use. Omarigliptin is one of the dipeptidyl peptidase-4 inhibitors that has proven efficacy in management of diabetes mellitus. Rosinidin is an anthocyanidin flavonoid that exhibited promising results in management of diseases characterized by oxidative stress, inflammation, and apoptosis. The present work investigated the possible effects of omarigliptin with or without rosinidin on cyclophosphamide-induced lung toxicity with an exploration of the molecular mechanisms that contribute to these effects. In a rodent model of cyclophosphamide elicited lung toxicity, the potential efficacy of omarigliptin with or without rosinidin was investigated at both the biochemical and the histopathological levels. Both omarigliptin and rosinidin exhibited a synergistic ability to augment the tissue antioxidant defenses, mitigate the inflammatory pathways, restore glucagon-like peptide-1 levels, modulate high mobility group box 1 (HMGB1)/receptors of advanced glycation end products (RAGE)/nuclear factor kappa B (NF-κB) axis, downregulate the fibrogenic mediators, and create a balance between the pathways involved in apoptosis and the autophagy signals in the pulmonary tissues. In conclusion, omarigliptin/rosinidin combination may be introduced as a novel therapeutic modality that attenuates the different forms of lung toxicities induced by cyclophosphamide.
Collapse
Affiliation(s)
- Maaly A Abd Elmaaboud
- Department of Pharmacology, Faculty of Medicine, Tanta University, Tanta 31527, Egypt
| | - Ahmed M Kabel
- Department of Pharmacology, Faculty of Medicine, Tanta University, Tanta 31527, Egypt.
| | - Hany M Borg
- Physiology Department, Faculty of Medicine, Kafrelsheikh University, Kafr El-Shaikh 33516, Egypt
| | - Amr A Magdy
- Anesthesia and ICU Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt
| | - Shaimaa M Kabel
- Zoology Department, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - El-Shaimaa A Arafa
- College of Pharmacy and Health Sciences, Ajman University, Ajman 346, United Arab Emirates; Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman 346, United Arab Emirates
| | - Shuruq E Alsufyani
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Hany H Arab
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
11
|
Abu-Baih RH, Ibrahim MFG, Elhamadany EY, Abu-Baih DH. Irbesartan mitigates the impact of cyclophosphamide-induced acute neurotoxicity in rats: Shedding highlights on NLRP3 inflammasome/CASP-1 pathway-driven immunomodulation. Int Immunopharmacol 2024; 135:112336. [PMID: 38801809 DOI: 10.1016/j.intimp.2024.112336] [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: 03/14/2024] [Revised: 05/09/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
IIrbesartan (IRB), an angiotensin II type 1 receptor (AT1R) antagonist, has been widely employed in the medical field for its effectiveness in managing hypertension. However, there have been no documented investigations regarding the immunostimulatory properties of IRB. To address this gap, this study has been performed to assess the neuroprotective impact of IRB as an immunostimulatory agent in mitigating acute neurotoxicity induced by cyclophosphamide (CYP) in rats. mRNA levels of nuclear factor erythroid 2 (Nrf-2), interleukin (IL)-18, IL-1β, and MMP-1 have been assessed using quantitative real-time polymerase chain reaction (qRT-PCR). Additionally, the levels of malondialdehyde (MDA), reduced glutathione (GSH), and superoxide dismutase (SOD) has been evaluated to assess the oxidative stress. Additionally, macrophage inflammatory protein 2 (MIP2) has been evaluated using enzyme-linked immunosorbent assay (ELISA). Western blotting has been used to investigate the protein expression of nucleotide binding oligomerization domain-like receptor protein 3 (NLRP3) and caspase-1 (CASP-1), along with an assessment of histopathological changes. Administration of IRB protected against oxidative stress by augmenting the levels of GSH and SOD as well as reducing MDA level. Also, administration of IRB led to a diminishment in the brain levels of MIP2 and MMP1. Furthermore, it led to a suppression of IL-1β and IL-18 levels, which are correlated with a reduction in the abundance of NLRP3 and subsequently CASP-1. This study provides new insights into the immunomodulatory effects of IRB in the context of CYP-induced acute neurotoxicity. Specifically, IRB exerts its effects by reducing oxidative stress, neuroinflammation, inhibiting chemokine recruitment, and mitigating neuronal degeneration through the modulation of immune markers. Therefore, it can be inferred that the use of IRB as an immunomodulator has the potential to effectively mitigate immune disorders associated with inflammation.
Collapse
Affiliation(s)
- Rania H Abu-Baih
- Drug Information Center, Faculty of Pharmacy, Minia University, Minia 61519, Egypt.
| | | | - Eyad Y Elhamadany
- Deraya Center for Scientific Research, Deraya University, Minia 61111, Egypt.
| | - Dalia H Abu-Baih
- Deraya Center for Scientific Research, Deraya University, Minia 61111, Egypt; Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Deraya University, Minia 61111, Egypt.
| |
Collapse
|
12
|
Yu W, Zhang Y, Sun L, Huang W, Li X, Xia N, Chen X, Wikana LP, Xiao Y, Chen M, Han S, Wang Z, Pu L. Myeloid Trem2 ameliorates the progression of metabolic dysfunction-associated steatotic liver disease by regulating macrophage pyroptosis and inflammation resolution. Metabolism 2024; 155:155911. [PMID: 38609037 DOI: 10.1016/j.metabol.2024.155911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/06/2024] [Accepted: 04/07/2024] [Indexed: 04/14/2024]
Abstract
BACKGROUND The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) is increasing year by year and has become one of the leading causes of end-stage liver disease worldwide. Triggering Receptor Expressed on Myeloid Cells 2 (Trem2) has been confirmed to play an essential role in the progression of MASLD, but its specific mechanism still needs to be clarified. This study aims to explore the role and mechanism of Trem2 in MASLD. METHODS Human liver tissues were obtained from patients with MASLD and controls. Myeloid-specific knockout mice (Trem2mKO) and myeloid-specific overexpression mice (Trem2TdT) were fed a high-fat diet, either AMLN or CDAHFD, to establish the MASLD model. Relevant signaling molecules were assessed through lipidomics and RNA-seq analyses after that. RESULTS Trem2 is upregulated in human MASLD/MASH-associated macrophages and is associated with hepatic steatosis and inflammation progression. Hepatic steatosis and inflammatory responses are exacerbated with the knockout of myeloid Trem2 in MASLD mice, while mice overexpressing Trem2 exhibit the opposite phenomenon. Mechanistically, Trem2mKO can aggravate macrophage pyroptosis through the PI3K/AKT signaling pathway and amplify the resulting inflammatory response. At the same time, Trem2 promotes the inflammation resolution phenotype transformation of macrophages through TGFβ1, thereby promoting tissue repair. CONCLUSIONS Myeloid Trem2 ameliorates the progression of Metabolic dysfunction-associated steatotic liver disease by regulating macrophage pyroptosis and inflammation resolution. We believe targeting myeloid Trem2 could represent a potential avenue for treating MASLD.
Collapse
Affiliation(s)
- Wenjie Yu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary cancers, Nanjing 210029, Jiangsu Province, China
| | - Yu Zhang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary cancers, Nanjing 210029, Jiangsu Province, China
| | - Linfeng Sun
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary cancers, Nanjing 210029, Jiangsu Province, China
| | - Wei Huang
- Department of General Surgery, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Ili & Jiangsu Joint Institute of Health, Ili, China
| | - Xiangdong Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary cancers, Nanjing 210029, Jiangsu Province, China
| | - Nan Xia
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary cancers, Nanjing 210029, Jiangsu Province, China
| | - Xuejiao Chen
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary cancers, Nanjing 210029, Jiangsu Province, China
| | - Likalamu Pascalia Wikana
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary cancers, Nanjing 210029, Jiangsu Province, China
| | - Yuhao Xiao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary cancers, Nanjing 210029, Jiangsu Province, China
| | - Minhao Chen
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary cancers, Nanjing 210029, Jiangsu Province, China
| | - Sheng Han
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary cancers, Nanjing 210029, Jiangsu Province, China
| | - Ziyi Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary cancers, Nanjing 210029, Jiangsu Province, China
| | - Liyong Pu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary cancers, Nanjing 210029, Jiangsu Province, China.
| |
Collapse
|
13
|
Osama HM, Khadrawy SM, El-Nahass ES, Othman SI, Mohamed HM. Eltroxin and Hesperidin mitigate testicular and renal damage in hypothyroid rats: amelioration of oxidative stress through PPARγ and Nrf2/HO-1 signaling pathway. Lab Anim Res 2024; 40:19. [PMID: 38745206 PMCID: PMC11092223 DOI: 10.1186/s42826-024-00204-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 04/22/2024] [Accepted: 04/28/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Thyroid hormones (THs) regulate growth, development and function of different tissues. Hypothyroidism is a common clinical disorder characterized by deficiency in THs and adversely affects the development and functions of several organs. This work aimed to investigate the ameliorative effect of eltroxin (ELT), a hypothyroidism medication, and hesperidin (HSP), a flavonoid, against testicular and renal toxicity in hypothyroid rats. Twenty-four rats were divided into four groups and treated orally for 12 weeks. Group I (control), group II (hypothyroidism) received 20 mg/kg carbimazole (CBZ), group III received CBZ and 0.045 mg/kg ELT, and group IV received CBZ and 200 mg/kg HSP. RESULTS CBZ administration induced biochemical and histopathological changes in testis and kidney. Co-administration of ELT or HSP significantly (P < 0.05) ameliorated THs, reduced urea and creatinine while raised follicle stimulating hormone (FSH), Luteinizing hormone (LH), and testosterone in serum. Testicular and renal malondialdehyde level as a lipid peroxidation indicator, tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) were significantly (P < 0.05) decreased while glutathione content, glutathione peroxidase, and glutathione-s-transferase activities were significantly (P < 0.05) increased. The histopathological changes were also diminished. Decreased mRNA and protein expressions of nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), and peroxisome proliferator-activated receptor gamma(PPARγ) in hypothyroid rats were up-regulated after ELT or HSP treatment. CONCLUSIONS ELT and HSP showed antioxidant and anti-inflammatory effects against CBZ-induced testicular and renal toxicity, and these effects may be promoted via activating Nrf2/HO-1 and PPARγ signaling pathways.
Collapse
Affiliation(s)
- Hadeel M Osama
- Genetics and Molecular Biology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Sally M Khadrawy
- Genetics and Molecular Biology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt.
| | - El-Shaymaa El-Nahass
- Pathology Department, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Sarah I Othman
- Biology Department, College of Science, Princess Nourah bint Abdulrahman University, P.O. BOX 84428, Riyadh, 11671, Saudi Arabia
| | - Hanaa M Mohamed
- Genetics and Molecular Biology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
14
|
Fani F, Hosseinimehr SJ, Zargari M, Mirzaei M, Karimpour Malekshah A, Talebpour Amiri F. Piperine mitigates oxidative stress, inflammation, and apoptosis in the testicular damage induced by cyclophosphamide in mice. J Biochem Mol Toxicol 2024; 38:e23696. [PMID: 38528700 DOI: 10.1002/jbt.23696] [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: 11/25/2023] [Revised: 01/19/2024] [Accepted: 03/12/2024] [Indexed: 03/27/2024]
Abstract
Although cyclophosphamide (CP) has been approved as an anticancer drug, its toxic effect on most organs, especially the testis, has been established. Piperine (PIP) is an alkaloid that has antioxidant, antiapoptotic, and anti-inflammatory activities. This study was investigated the protective effects of PIP on CP-induced testicular toxicity in the mice. In this experimental study, 48 adult male BALB/c mice (30-35 g) were divided into six groups (n = 8), receiving normal saline (C), 5 mg/kg of PIP (PIP5), 10 mg/kg of PIP (PIP10), 200 mg/kg of CP, 200 mg/kg of CP + PIP5, and 200 mg/kg of CP + PIP10. On the eighth day of the study, blood and testis samples were prepared for serum testosterone hormone quantification, sperm analysis, histological, and immunohistochemical assays. The results of this study showed that CP induced testicular toxicity with the decrease of sperm count, motility, and viability. Also, CP treatment caused histological structure alterations in the testis, including exfoliation, degeneration, vacuolation of spermatogenic cells, and reducing the thickness of the epithelium and the diameter of the seminiferous tubule. In addition, CP decreased glutathione (GSH) levels, increased malondialdehyde (MDA) levels, Caspase-3, and NF-κB. At the same time, PIP treatment reduced testicular histopathological abnormalities, oxidative stress, and apoptosis that were induced by CP. These results showed that PIP improved CP-induced testicular toxicity in mice, which can be related to its antioxidant, antiapoptotic, and anti-inflammatory activities.
Collapse
Affiliation(s)
- Fatemeh Fani
- Department of Anatomy, Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Student Research Committee, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mehryar Zargari
- Department of Biochemistry, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mansoureh Mirzaei
- Department of Anatomy, Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abbasali Karimpour Malekshah
- Department of Anatomy, Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Fereshteh Talebpour Amiri
- Department of Anatomy, Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
15
|
AbdElrazek DA, Hassan NH, Ibrahim MA, Hassanen EI, Farroh KY, Abass HI. Ameliorative effects of rutin and rutin-loaded chitosan nanoparticles on testicular oxidative stress and histological damage induced by cyclophosphamide in male rats. Food Chem Toxicol 2024; 184:114436. [PMID: 38211767 DOI: 10.1016/j.fct.2024.114436] [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: 10/13/2023] [Revised: 01/01/2024] [Accepted: 01/02/2024] [Indexed: 01/13/2024]
Abstract
Cyclophosphamide (CP) is broadly used to kill various tumor cells; however, its repeated uses have been reported to cause reproductive dysfunction and infertility. Natural flavonoid, rutin (RUT), possesses strong antioxidant and antiapoptotic activity that is attributed to ameliorate the reproductive dysfunction induced by CP. Many previous studies proved that the formulation of flavonoids in nanoemulsion has a promising perspective in mitigating the side effects of chemotherapy. Therefore, the main objective of this study was to investigate the ameliorative effects of RUT and RUT-loaded chitosan nanoparticles (RUT-CH NPs) against CP-induced reproductive dysfunction in male rats. For this aim, thirty-six male albino rats were randomly allocated into six groups as follows: control, RUT, RUT-CH NPs, CP, CP + RUT, and CP + RUT-CH NPs. In the CP groups, a single intraperitoneal injection of CP (150 mg/kg bwt) was administered on the first day of the experiment. RUT and RUT-CH NPs were orally administered either alone or with CP injection at a dose of 10 mg/kg bwt per day for 60 days. The results revealed that CP administration caused significant testicular oxidative stress damage through increasing the nitric oxide and malondialdehyde levels as well as decreasing the total antioxidant capacity and reduced glutathione contents. It also impaired spermatogenesis and steroidogenesis via altering the transcription levels of CYP11A1, HSD-3b, StAR, Bax, bcl-2, and Nrf-2 genes. Otherwise, the oral intake of either RUT or RUT-CH NPs with CP injection effectively attenuated these alterations and significantly improved the microscopic appearance of testicular tissue. In conclusion, this study highlights the potential of RUT either free or NPs in mitigating CP-induced testicular dysfunction via its antioxidant and anti-apoptotic properties.
Collapse
Affiliation(s)
- Dina A AbdElrazek
- Physiology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Neven H Hassan
- Physiology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Marwa A Ibrahim
- Biochemistry Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt.
| | - Eman I Hassanen
- Pathology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Khaled Y Farroh
- Nanotechnology and Advanced Materials Central Lab, Agricultural Research Center, Giza, Egypt
| | - H I Abass
- Physiology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
16
|
Mo P, Zhao Z, Ke X, Fan Y, Li C. Effects of clinical medications on male fertility and prospects for stem cell therapy. Front Cell Dev Biol 2023; 11:1258574. [PMID: 37791073 PMCID: PMC10543686 DOI: 10.3389/fcell.2023.1258574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/07/2023] [Indexed: 10/05/2023] Open
Abstract
An increasing number of men require long-term drug therapy for various diseases. However, the effects of long-term drug therapy on male fertility are often not well evaluated in clinical practice. Meanwhile, the development of stem cell therapy and exosomes treatment methods may provide a new sight on treating male infertility. This article reviews the influence and mechanism of small molecule medications on male fertility, as well as progress of stem cell and exosomes therapy for male infertility with the purpose on providing suggestions (recommendations) for evaluating the effect of drugs on male fertility (both positive and negative effect on male fertility) in clinical application and providing strategies for diagnosis and treatment of male infertility.
Collapse
Affiliation(s)
| | | | | | - Yong Fan
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Department of Obstetrics and Gynecology, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Chaohui Li
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Department of Obstetrics and Gynecology, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
17
|
Samadi M, Kamrani A, Nasiri H, Shomali N, Heris JA, Shahabi P, Ghahremanzadeh K, Mohammadinasab R, Sadeghi M, Sadeghvand S, Shotorbani SS, Akbari M. Cancer immunotherapy focusing on the role of interleukins: A comprehensive and updated study. Pathol Res Pract 2023; 249:154732. [PMID: 37567033 DOI: 10.1016/j.prp.2023.154732] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/29/2023] [Accepted: 07/30/2023] [Indexed: 08/13/2023]
Abstract
Cytokines bind to specific receptors on target cells to activate intracellular signaling pathways that control diverse cellular functions, such as proliferation, differentiation, migration, and death. They are essential for the growth, activation, and operation of immune cells and the control of immunological reactions to pathogens, cancer cells, and other dangers. Based on their structural and functional properties, cytokines can be roughly categorized into different families, such as the tumor necrosis factor (TNF) family, interleukins, interferons, and chemokines. Leukocytes produce interleukins, a class of cytokines that have essential functions in coordinating and communicating with immune cells. Cancer, inflammation, and autoimmunity are immune-related disorders brought on by dysregulation of cytokine production or signaling. Understanding cytokines' biology to create novel diagnostic, prognostic, and therapeutic methods for various immune-related illnesses is crucial. Different immune cells, including T cells, B cells, macrophages, and dendritic cells, and other cells in the body, including epithelial cells and fibroblasts, generate and secrete interleukins. The present study's main aim is to fully understand interleukins' roles in cancer development and identify new therapeutic targets and strategies for cancer treatment.
Collapse
Affiliation(s)
- Mahmoud Samadi
- Pediatrics Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amin Kamrani
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Nasiri
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran
| | - Navid Shomali
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Ahmadian Heris
- Department of Allergy and Clinical Immunology, Pediatric Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parviz Shahabi
- Stem Cell and Regenerative Medicine Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Reza Mohammadinasab
- Department of History of Medicine, School of Traditional Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammadreza Sadeghi
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahram Sadeghvand
- Pediatrics Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Morteza Akbari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
18
|
Alqahtani MJ, Negm WA, Saad HM, Salem EA, Hussein IA, Ibrahim HA. Fenofibrate and Diosmetin in a rat model of testicular toxicity: New insight on their protective mechanism through PPAR-α/NRF-2/HO-1 signaling pathway. Biomed Pharmacother 2023; 165:115095. [PMID: 37413905 DOI: 10.1016/j.biopha.2023.115095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/27/2023] [Accepted: 06/27/2023] [Indexed: 07/08/2023] Open
Abstract
One of the most significant chemotherapeutic side effects of cisplatin (Cis) that limits its use and efficacy is testicular toxicity. Thus, the objective of the present study was to investigate the possible ameliorative effect of Fenofibrate (Fen), Diosmetin (D), and their combination against cis-mediated testicular damage. Fifty-four adult male albino rats were randomly allocated into nine groups (6 rats each): Control group, Fen (100 mg/kg), D20 (20 mg/kg), D40 (40 mg/kg), Cis group (7 mg/kg), Cis +Fen group (7 mg/kg+100 mg/kg), Cis+D20 group (7 mg/kg+20 mg/kg), Cis+D40 group (7 mg/kg+40 mg/kg), Cis+Fen+D40 treated group (7 mg/kg+100 mg/kg+40 mg/kg). Relative testicular weight, epididymal sperm count and viability, serum testosterone level, testicular oxidative stress indices, mRNA expression of peroxisome proliferator-activated receptor alpha (PPAR-α), nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase 1 (HO-1), histopathological, and immunohistochemical alterations were assessed. Our results revealed that cis administration induced testicular oxidative and inflammatory damage as indicated by a substantial reduction in relative testicular weight, sperm parameters, serum testosterone levels, the antioxidant enzyme activity of catalase, and Johnson's histopathological score, PPAR-α/NRF-2/HO-1 and proliferating cell nuclear antigen (PCNA) immunoexpression with marked increment in malondialdehyde (MDA), Cosentino's score, nuclear factor kappa B (NF-κβ p65), interleukin (IL)- 1β and caspase 3 in testicular tissue. Interestingly, Fen and D diminished the harmful effects of cis on testes via upregulation of the antioxidant activities and downregulation of lipid peroxidation, apoptosis, and inflammation. Moreover, the combination therapy Fen/D40 also exhibited a more pronounced enhancement of previous markers than either treatment alone. In conclusion, because of their antioxidant, anti-inflammatory, and anti-apoptotic properties, cotreatment with Fen or D or their combination could be beneficial in reducing the harmful impacts of cis on testicular tissue, particularly in patients that receive cis chemotherapy.
Collapse
Affiliation(s)
- Moneerah J Alqahtani
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia.
| | - Walaa A Negm
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | - Hebatallah M Saad
- Department of Pathology, Faculty of veterinary medicine, Matrouh University, Marsa Matrouh, Egypt.
| | - Esraa A Salem
- Department of Medical Physiology, Faculty of Medicine, Menoufia University, Shebeen ElKom 32511, Egypt.
| | - Ismail A Hussein
- Department of Pharmacognosy and Medicinal Plants, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt.
| | - Hanaa A Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tanta, Egypt.
| |
Collapse
|
19
|
Ye Y, Fang C, Li L, Liu D, Wang Y, Huang F, Gong X, Xu Y, Yao Y, Ye S, Feng D, Luo F. Protective Effect of l-Theanine on Cyclophosphamide-Induced Testicular Toxicity in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:8050-8060. [PMID: 37198140 DOI: 10.1021/acs.jafc.3c01010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
l-Theanine is the most abundant free amino acid present in tea. Several tea components have been studied for their impact on male fertility, but little is known about the effects of l-theanine. Cyclophosphamide (CP) is an antineoplastic and immunosuppressive agent that reduces fertility in males. In the present study, we evaluated the effect of l-theanine on CP-induced testicular toxicity in male mice. A single dosage of 50 mg/kg saline or CP was administered intraperitoneally over the course of 5 days. Mice were administered l-theanine (80 mg/kg) or saline by gavage for 30 days. Animals were euthanized 24 h after the last l-theanine administration, and the testes were removed for histopathological and transmission electron microscopy analysis. Histological evaluation and transmission electron microscopy showed that administration of l-theanine alleviated CP-induced damage to the testicles, including spermatogonial cells, epithelial cells, seminiferous tubules, and basement membrane. An integrated proteomics and metabolomics investigation of testes revealed that l-theanine therapy substantially affected the quantity of 719 proteins (395 upregulated and 324 downregulated) and 196 metabolites (75 upregulated and 111 downregulated). The top three enriched Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways for these proteins and metabolites were purine metabolism, choline metabolism in cancer, and arachidonic acid metabolism. This is the first study to reveal the protective effect of l-theanine on CP-induced testicular toxicity. l-Theanine could be a potential natural active substance for resistance to the testis toxicity induced by CP.
Collapse
Affiliation(s)
- Yulong Ye
- Tea Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu 610066, P. R. China
| | - Chunyan Fang
- Tea Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu 610066, P. R. China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, P. R. China
| | - Lanying Li
- Tea Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu 610066, P. R. China
| | - Dongna Liu
- Tea Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu 610066, P. R. China
| | - Yingchun Wang
- Tea Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu 610066, P. R. China
| | - Fan Huang
- Tea Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu 610066, P. R. China
| | - Xuejiao Gong
- Tea Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu 610066, P. R. China
| | - Yaqiong Xu
- Tea Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu 610066, P. R. China
| | - Yu Yao
- Tea Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu 610066, P. R. China
| | - Shanrong Ye
- National Institute of Measurement and Testing Technology, Chengdu 610021, P. R. China
| | - Dejian Feng
- National Institute of Measurement and Testing Technology, Chengdu 610021, P. R. China
| | - Fan Luo
- Tea Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu 610066, P. R. China
| |
Collapse
|
20
|
El-Beheiry KM, El-Sayed El-Sayad M, El-Masry TA, Elsisi AE. Combination of metformin and hesperidin mitigates cyclophosphamide-induced hepatotoxicity. Emerging role of PPAR-γ/Nrf-2/NF-κB signaling pathway. Int Immunopharmacol 2023; 117:109891. [PMID: 36812672 DOI: 10.1016/j.intimp.2023.109891] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/02/2023] [Accepted: 02/11/2023] [Indexed: 02/22/2023]
Abstract
Cyclophosphamide (CP) is widely used as an immunosuppressive and chemotherapeutic drug. However, its therapeutic application is restricted by its adverse effects, particularly hepatotoxicity. Both metformin (MET) and hesperidin (HES) have promising antioxidant, anti-inflammatory, and anti-apoptotic effects. Therefore, the principal aim of the current study is to investigate the hepatoprotective effects of MET, HES, and their combinations on the CP-induced hepatotoxicity model. Hepatotoxicity was evoked by a single (I.P) injection of CP (200 mg/kg) on day 7. For this study, 64 albino rats were randomly categorized into eight equal groups; naïve, control vehicle, untreated CP (200 mg/kg, IP), and CP 200 groups treated with MET 200, HES 50, HES 100 or a combination of MET 200 with HES 50 and HES 100 respectively orally daily for 12 days. At the end of the study, the liver function biomarkers, oxidative stress, inflammatory parameters, histopathological and immunohistochemical analysis of PPAR-γ, Nrf-2, NF-κB, Bcl-2, and caspase3 were assessed. CP significantly increased serum ALT, AST, total bilirubin, hepatic MDA, NO content, NF-κB, and TNF-α. Otherwise, albumin, hepatic GSH content, Nrf-2, and PPAR-γ expression decreased considerably compared to the control vehicle group. The combinations of MET200 with HES50 or HES100 induced pronounced hepatoprotective, anti-oxidative, anti-inflammatory, and anti-apoptotic effects on CP-treated rats. The possible explanation of such hepatoprotective effects may be mediated via upregulation of Nrf-2, PPAR-γ, Bcl-2 expression, hepatic GSH content, and marked suppression of TNF-α and NF-κB expression. In conclusion, the current study showed that combining MET and HES revealed a remarkable hepatoprotective effect against CP-induced hepatotoxicity.
Collapse
Affiliation(s)
- Kareman M El-Beheiry
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt.
| | - Magda El-Sayed El-Sayad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Thanaa A El-Masry
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Alaa E Elsisi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
21
|
Abdul-Hamid M, Abdel-Reheim ES, Hegazy W, Allam AA, Othman SI, ALqhtani H, Abdel-Kawi SH. Impact of gervital against histopathological, ultrastructural, and biochemical alterations caused by methotrexate or azathioprine in albino rat testis. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:21914-21926. [PMID: 36279059 PMCID: PMC9938037 DOI: 10.1007/s11356-022-23588-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 10/08/2022] [Indexed: 06/16/2023]
Abstract
Methotrexate (MTX) and azathioprine (AZA) are chemotherapeutic, immunosuppressive, cytotoxic drugs with reported adverse effects, including oxidative damage to testis. This study aims to evaluate the potential effect of grape seed extract (GSE; gervital) to prevent testicular damage caused by MTX and AZA. Male albino rats were separated into six groups: group I, normal control group; group II, GSE (150 mg/kg/day); group III, MTX (8 mg/kg/week); group IV, AZA (15 mg/kg/day); group V, GSE (150 mg/kg/day) + MTX (8 mg/kg/week); group VI, GSE (150 mg/kg/day) + AZA (15 mg/kg/day). All rats were sacrificed, blood samples were obtained for testosterone analysis, and testis was removed for histological and ultrastructural studies and oxidation measurements. A reduction in relative body and testis weight, along with a significant decrease in testosterone levels, was observed. Histopathological and ultrastructural alterations induced by MTX or AZA included reduced spermatozoa, sloughing, marked reduction of spermatogenic cells, and pyknosis of some nuclei. Significant oxidative stress manifested as reduced glutathione (GSH) levels and catalase (CAT) and superoxide dismutase (SOD) activities, as well as increased malondialdehyde (MDA) levels. GSE administration showed an ameliorative effect on testosterone levels and histopathological and ultrastructural changes. GSE treatment also suppressed the increases in MDA levels and the decreases in GSH levels and CAT and SOD activities. In conclusion, these findings confirm that GSE is an effective antioxidant that protects testis from histopathological and ultrastructural damage induced by MTX and AZA. Therefore, GSE is a promising candidate for future use to minimize and alleviate MTX and AZA risks.
Collapse
Affiliation(s)
- Manal Abdul-Hamid
- Zoology Department, Faculty of Science, Histology and Cell Biology Division, Beni-Suef University, Beni-Suef, Egypt
| | - Eman S Abdel-Reheim
- Zoology Department, Faculty of Science, Molecular Physiology Division, Beni-Suef University, Beni-Suef, Egypt
| | - Walaa Hegazy
- Basic Science Department, Faculty of Physical Therapy, Histology Division, Nahda University, Beni-Suef, Egypt
| | - Ahmed A Allam
- Department of Zoology, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt.
| | - Sarah I Othman
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh, 11671, Saudi Arabia
| | - Haifa ALqhtani
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh, 11671, Saudi Arabia
| | - Samraa H Abdel-Kawi
- Medical Histology & Cell Biology Department, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
22
|
Yang X, Liu P, Cui Y, Song M, Zhang X, Zhang C, Jiang Y, Li Y. T-2 Toxin Caused Mice Testicular Inflammation Injury via ROS-Mediated NLRP3 Inflammasome Activation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:14043-14051. [PMID: 36260425 DOI: 10.1021/acs.jafc.2c05317] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
T-2 toxin treatment causes male reproduction system dysfunction, although the exact mechanism remains unclear. In this research, male Kunming mice and TM4 cells were treated with varying concentrations of the T-2 toxin for evaluating the adverse effect of T-2 toxin on male reproductive function. MCC950 or NAC was used to block NLRP3 inflammasome activation and eliminate reactive oxygen species (ROS) accumulation in the TM4 cell, respectively. The results showed that: (1) T-2 toxin caused testicular atrophy, destroyed the microstructure and ultrastructure of the testis, and caused sperm deformities; (2) T-2 toxin increased the content and gene expressions of TNF-α and IL-6 and decreased the IL-10 content and gene expression, causing testis and TM4 cell inflammatory injury; (3) T-2 toxin activated NLRP3 inflammasome in the testis and TM4 cells and caused ROS accumulation in the testis; (4) suppressing NLRP3 inflammasome activation using 20 nM MCC950 alleviated the TM4 cell inflammatory damage caused via the T-2 toxin; nevertheless, 20 nM MCC950 did not reduce ROS accumulation in TM4 cells; and (5) NAC relieved the inflammatory damage in TM4 cells by inhibiting NLRP3 inflammasome activation. Taken together, T-2 toxin caused testicular inflammation injury through ROS-mediated NLRP3 inflammasome activation, resulting in male reproductive dysfunction.
Collapse
Affiliation(s)
- Xu Yang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, Henan, China
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Pengli Liu
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Yilong Cui
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- College of Animal Science and Technology, Inner Mongolia University for Nationalities, Tongliao 028000, China
| | - Miao Song
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Xuliang Zhang
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Cong Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, Henan, China
| | - Yibao Jiang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, Henan, China
| | - Yanfei Li
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
23
|
Arab HH, Eid AH, El-Sheikh AAK, Arafa ESA, Ashour AM. Irbesartan reprofiling for the amelioration of ethanol-induced gastric mucosal injury in rats: Role of inflammation, apoptosis, and autophagy. Life Sci 2022; 308:120939. [PMID: 36115582 DOI: 10.1016/j.lfs.2022.120939] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/28/2022] [Accepted: 09/05/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND Pronounced anti-inflammatory and anti-apoptotic features have been characterized for the angiotensin receptor blocker irbesartan. Yet, its effect on ethanol-induced gastropathy has not been studied. The present work explored the potential modulation of inflammatory, apoptotic, and autophagic events by irbesartan for the attenuation of ethanol-evoked gastric mucosal injury. METHODOLOGY Wistar rats were divided into control, control + irbesartan, ethanol, ethanol + irbesartan, and ethanol + omeprazole groups. Macroscopic examination, histopathology, immunohistochemistry, and biochemical assays were applied to examine the gastric tissues. KEY FINDINGS Irbesartan administration (50 mg/kg; by gavage) in ethanol-evoked gastropathy improved the gastric pathological manifestations (area of gastric lesion and ulcer index scores), histopathological changes, and microscopic damage scores. These beneficial effects were interceded by suppression of the HMGB1-associated inflammatory events and the linked downregulation of the nuclear NF-κBp65 protein expression. In the meantime, curtailing of the NLRP3 inflammasome by irbesartan was observed with consequent decline of the pro-inflammatory cytokine IL-1β. In tandem, upregulation of the antioxidant Nrf2 and the cytoprotective PPAR-γ were seen. Together, suppression of the pro-inflammatory cues and pro-oxidant signals attenuated the pro-apoptotic events as evidenced by Bcl-2 upregulation, Bax downregulation, and caspase 3 dampened activity. Regarding gastric autophagy signals, irbesartan diminished SQSTM-1/p62 accumulation and upregulated Beclin 1. This was associated with gastric AMPK/mTOR pathway activation evidenced by increased AMPK (Ser487) phosphorylation and lowered mTOR (Ser2448) phosphorylation. CONCLUSION Suppression of the inflammatory and apoptotic signals and upregulation of the pro-autophagy events may advocate the promising gastroprotective actions of irbesartan against ethanol-induced gastric injury.
Collapse
Affiliation(s)
- Hany H Arab
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.
| | - Ahmed H Eid
- Department of Pharmacology, Egyptian Drug Authority (EDA), formerly NODCAR, Giza, Egypt
| | - Azza A K El-Sheikh
- Basic Health Sciences Department, College of Medicine, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - El-Shaimaa A Arafa
- College of Pharmacy and Health Sciences, 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 Ashour
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al Qura University, P.O. Box 13578, Makkah 21955, Saudi Arabia
| |
Collapse
|