1
|
Sun T, Wang K, Ma Y, Liu X, Ji D, Zhang Z, Xie X, Yuan Z, Wang L, Liu GQ, Ling Y. Novel one-/two-photon excited carbazole/quinolinium photosensitizers manifest nanomolar and hypoxia-resistant tumor photodynamic therapy by accelerating apoptosis, ferroptosis, and autophagy. Eur J Med Chem 2025; 290:117523. [PMID: 40121867 DOI: 10.1016/j.ejmech.2025.117523] [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: 01/04/2025] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Photodynamic therapy (PDT) holds potential in cancer treatment, but the development of photosensitizers with high-efficient PDT remains a challenge. Herein, we designed and synthesized a series of novel tricyclic carbazole/quinolinium hybrids-KNKQ, KAKQ, and KPKQ-as photosensitizers, and subsequently evaluated their photodynamic anticancer activities and the associated mechanisms. Among them, KPKQ exhibited the most prominent one-/two-photon activated photodynamic characteristics, generating •O2-, •OH, and 1O2. Particularly, the 1O2 quantum yield of KPKQ was 3∼9-fold stronger than KNKQ and KAKQ. Most interestingly, KPKQ demonstrated nanomolar-level and hypoxic-overcoming single-photon phototoxicities with IC50 values of 27∼43 nM (PIs = 46-54), significantly surpassing existing tricyclic carbazole photosensitizers, and also exerted potent photodynamic therapeutic effects (IC50s = 0.13-0.20 μM) via two-photon excitation at 808 nm. Furthermore, KPKQ significantly promoted mitochondrial damage, cell apoptosis, and DNA lesion via reducing Bcl-2 level and increasing the levels of Bax, cleaved-Caspase-3, and γ-H2AX. Concurrently, KPKQ lowered GSH/GPX4 levels and elevated malondialdehyde to trigger ferroptosis. Additionally, KPKQ powerfully promoted autophagy through boosting LC3-II and Beclin-1 expression, thereby demonstrating a multiple anti-tumor mechanism. Ultimately, KPKQ achieved a 90.7 % tumor-inhibitory rate through in vivo PDT. Our findings may provide a promising framework for the discovery of novel tricyclic carbazole photosensitizers with high PDT efficacy.
Collapse
Affiliation(s)
- Tiantian Sun
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, PR China; Nantong Key Laboratory of Small Molecular Drug Innovation, School of Pharmacy, Nantong University, Nantong, 226001, PR China
| | - Kai Wang
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, PR China; Nantong Key Laboratory of Small Molecular Drug Innovation, School of Pharmacy, Nantong University, Nantong, 226001, PR China
| | - Yifan Ma
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, PR China; Nantong Key Laboratory of Small Molecular Drug Innovation, School of Pharmacy, Nantong University, Nantong, 226001, PR China
| | - Xiao Liu
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, PR China; Nantong Key Laboratory of Small Molecular Drug Innovation, School of Pharmacy, Nantong University, Nantong, 226001, PR China
| | - Dongliang Ji
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, PR China; Nantong Key Laboratory of Small Molecular Drug Innovation, School of Pharmacy, Nantong University, Nantong, 226001, PR China
| | - Zirui Zhang
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, PR China; Nantong Key Laboratory of Small Molecular Drug Innovation, School of Pharmacy, Nantong University, Nantong, 226001, PR China
| | - Xudong Xie
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, PR China; Nantong Key Laboratory of Small Molecular Drug Innovation, School of Pharmacy, Nantong University, Nantong, 226001, PR China
| | - Zhifei Yuan
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, PR China; Nantong Key Laboratory of Small Molecular Drug Innovation, School of Pharmacy, Nantong University, Nantong, 226001, PR China
| | - Lei Wang
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, PR China; Nantong Key Laboratory of Small Molecular Drug Innovation, School of Pharmacy, Nantong University, Nantong, 226001, PR China.
| | - Gong-Qing Liu
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, PR China; Nantong Key Laboratory of Small Molecular Drug Innovation, School of Pharmacy, Nantong University, Nantong, 226001, PR China.
| | - Yong Ling
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, PR China; Nantong Key Laboratory of Small Molecular Drug Innovation, School of Pharmacy, Nantong University, Nantong, 226001, PR China.
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Mersal EA, Morsi AA, Alkahtani J, Alhalal R, Alessa S, Shehab A, Sakr EM, Sabir DK, Dawood AF, Abdelmoneim AM. Pirfenidone targeted mechanisms for alleviating methotrexate-induced testiculopathy in Wistar rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:2003-2014. [PMID: 39222241 DOI: 10.1007/s00210-024-03407-x] [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: 06/15/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Testicular injury and affected spermatogenesis are major complications of methotrexate (MTX) use. Oxidative stress is one contributing process leading to inflammation and apoptosis induction. Pirfenidone (PFD) is a well-known anti-fibrotic drug prescribed for interstitial lung fibrosis, in addition to anti-inflammatory, antioxidative, and antiapoptotic capabilities. The study aimed to explore the potential protection afforded by PFD in a rat model of MTX-induced testiculopathy. The experimental design included four groups, each containing seven adult Wistar rats: control, PFD (500 mg/kg/day, orally)-, MTX (0.5 mg/kg, intraperitoneal, twice weekly)-, and PFD/MTX-treated groups. Treatment continued for 4 weeks. Blood and testicular samples were harvested for biochemical, histological, immunohistochemical, and polymerase chain reaction (PCR) analyses. Also, the testicular damage and spermatogenic activity were graded by the testicular injury and Johnsen scoring system, respectively. PFD positively affected the serum testosterone (TST) level, reduced the testicular inflammatory mediators [tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β)], reduced the testicular oxidative burden, increased superoxide dismutase (SOD), and protected the testicular histological structure. In addition, antifibrotic effects, anti-caspase-3, and PCNA enhancement activity were recorded. PFD exhibited a protective potential and mitigated the MTX-induced testiculopathy via suppression of testicular oxidative stress, inflammation, fibrosis, and apoptosis and retaining the testicular proliferative efficacy as confirmed by histological, immunohistochemical, and biochemical methods.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Deema Kamal Sabir
- Department of Medical Surgical Nursing, College of Nursing, Princess Nourah bint Abdulrahman University, P.O. Box 84428, 11671, Riyadh, Saudi Arabia
| | - Amal F Dawood
- Department of Basic Medical Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, P.O. Box 84428, 11671, Riyadh, Saudi Arabia
| | - Ahmed M Abdelmoneim
- Department of Physiology, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| |
Collapse
|
5
|
Luo G, Yang W, Geng Z, Cheng Y, Xu Y, Xiao Y, Liu J. Molecular mechanism of GSH metabolism and autophagy in NAC-promoted recombinant human serum albumin and follicle stimulating hormone beta fusion protein secretion in Pichia pastoris. J Biotechnol 2025; 398:146-157. [PMID: 39710118 DOI: 10.1016/j.jbiotec.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 12/24/2024]
Abstract
The Pichia pastoris expression system is a favorable platform for production of pharmaceutical proteins. Treatment of strains with N-acetyl-L-cysteine (NAC) has been shown to enhance the yield of recombinant proteins, thereby contributing to a reduction in production costs. However, the specific mechanism of action of NAC remains unclear. Previous research has indicated that glutathione (GSH) and autophagy are involved in the increased production of human serum albumin and porcine follicle-stimulating hormone β (HSA-pFSHβ) by NAC. This study investigated the potential interaction between GSH and autophagy in the production of HSA-pFSHβ. The findings indicated that sulfhydryl-free antioxidants such as melatonin, vitamin C, or vitamin E did not exhibit similar effects to NAC in enhancing HSA-pFSHβ yield. Moreover, NAC was found to enhance HSA-pFSHβ production by modulating GSH metabolism to reduce GSH consumption, increase total GSH levels, as well as glutathione peroxidase (GSH-Px) and glutathione reductase (GR) activities. Additionally, inhibition of autophagy through disruption of autophagy scaffolding proteins Atg1 or Atg11 led to an increase in recombinant HSA-pFSHβ production. Furthermore, NAC significantly decreased the phosphorylation of Slt2, and the absence of the SLT2 gene influenced the effect of NAC on HSA-pFSHβ secretion by modulating mitophagy and GSH metabolism. In conclusion, these results suggest a complex interplay between GSH metabolism and autophagy in the regulation of NAC-induced HSA-pFSHβ secretion.
Collapse
Affiliation(s)
- Gang Luo
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Wen Yang
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Zijian Geng
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Yiyi Cheng
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Yingqing Xu
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Yimeng Xiao
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Jiying Liu
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China.
| |
Collapse
|
6
|
Mahmoud NM, Elshazly SM, El-shaarawy F, Zaitone SA, Aldahish AA, Ahmed GA, Fawzy MS, Aloyouni SY, Abed SY, Saeedi T, El-Sayed SS. Nitazoxanide mitigates methotrexate hepatotoxicity in rats: role in inhibiting apoptosis and regulating endoplasmic reticulum stress. Front Pharmacol 2024; 15:1491249. [PMID: 39687303 PMCID: PMC11647085 DOI: 10.3389/fphar.2024.1491249] [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: 09/04/2024] [Accepted: 11/14/2024] [Indexed: 12/18/2024] Open
Abstract
Objectives Hepatotoxicity is a severe outcome of methotrexate (MTX) therapy, limiting its clinical use and contributing to its related morbidity and mortality. This study investigated the hepatoprotective effects of nitazoxanide (NTZ), an antiprotozoal drug, against MTX-induced hepatotoxicity and whether endoplasmic reticulum (ER) stress-modulation underlies the expected beneficial effects of NTZ. Methods Thirty-six rats were allocated to six groups, one control group and five MTX groups, where induction of hepatotoxicity was achieved via injecting MTX (20 mg/kg). Groups were assigned as MTX-vehicle, NTZ-100, and NTZ-200 groups (at 100 and 200 mg/kg/day, gavage, respectively), N-acetyl cysteine (NAC) group (500 mg/kg), and 4-phenyl butyric acid (4-PBA) group (150 mg/kg, i.p). Liver function enzymes in serum, hepatic oxidative stress, proinflammatory cytokines, apoptosis, and ER-stress biomarkers were assessed. A histopathological examination was performed. Results Treatment with NTZ lessened the serum liver enzymes, reduced malondialdehyde (lipid peroxidation product), enhanced antioxidant capacity, attenuated proinflammatory cytokines, and suppressed apoptosis. The protective effect of NTZ was dose-dependent, and the findings observed with the high-dose NTZ were similar to those obtained with the ER-stress inhibitor (4-PBA). Conclusion NTZ exerted a hepatoprotective effect in MTX-challenged rats that is mediated via modulation of ER stress and inhibiting apoptosis.
Collapse
Affiliation(s)
| | - Shimaa M. Elshazly
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Fatma El-shaarawy
- Department of Biochemistry, Faculty of Pharmacy, Sinai University, Arish, Egypt
| | - Sawsan A. Zaitone
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Afaf A. Aldahish
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Gehan A. Ahmed
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Manal S. Fawzy
- Department of Biochemistry, Faculty of Medicine, Northern Border University, Arar, Saudi Arabia
- Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | - Sheka Yagub Aloyouni
- Research Department, Natural and Health Sciences Research Center, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Sally Y. Abed
- Department of Respiratory Care, College of Applied Medical Science in Jubail, Imam Abdulrahman Bin Faisal University, Jubail, Saudi Arabia
| | - Tahani Saeedi
- Department of Pharmacology and Toxicology, School of Pharmacy, Taibah University, Medina, Saudi Arabia
| | - Shaimaa S. El-Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| |
Collapse
|
7
|
Cheng X, Jiang T, Huang Q, Ji L, Li J, Kong X, Zhu X, He X, Deng X, Wu T, Yu H, Shi Y, Liu L, Zhao X, Wang X, Chen H, Yu J. Exposure to Titanium Dioxide Nanoparticles Leads to Specific Disorders of Spermatid Elongation via Multiple Metabolic Pathways in Drosophila Testes. ACS OMEGA 2024; 9:23613-23623. [PMID: 38854533 PMCID: PMC11154731 DOI: 10.1021/acsomega.4c01140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 05/10/2024] [Accepted: 05/14/2024] [Indexed: 06/11/2024]
Abstract
Titanium dioxide nanoparticles (TiO2 NPs) have been extensively utilized in various applications. However, the regulatory mechanism behind the reproductive toxicity induced by TiO2 NP exposure remains largely elusive. In this study, we employed a Drosophila model to assess potential testicular injuries during spermatogenesis and conducted bulk RNA-Seq analysis to elucidate the underlying mechanisms. Our results reveal that while prolonged exposure to lower concentrations of TiO2 NPs (0.45 mg/mL) for 30 days did not manifest reproductive toxicity, exposure at concentrations of 0.9 and 1.8 mg/mL significantly impaired spermatid elongation in Drosophila testes. Notably, bulk RNA-seq analysis revealed that TiO2 NP exposure affected multiple metabolic pathways including carbohydrate metabolism and cytochrome P450. Importantly, the intervention of glutathione (GSH) significantly protected against reproductive toxicity induced by TiO2 NP exposure, as it restored the number of Orb-positive spermatid clusters in Drosophila testes. Our study provides novel insights into the specific detrimental effects of TiO2 NP exposure on spermatid elongation through multiple metabolic alterations in Drosophila testes and highlights the protective role of GSH in countering this toxicity.
Collapse
Affiliation(s)
- Xinmeng Cheng
- Institute
of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Ting Jiang
- Institute
of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Qiuru Huang
- Institute
of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Li Ji
- Institute
of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Jiaxin Li
- Institute
of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Xiuwen Kong
- Institute
of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Xiaoqi Zhu
- Institute
of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Xuxin He
- Institute
of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Xiaonan Deng
- Institute
of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Tong Wu
- Institute
of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Hao Yu
- Institute
of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Yi Shi
- Institute
of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Lin Liu
- Institute
of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Xinyuan Zhao
- Department
of Occupational Medicine and Environmental Toxicology, Nantong Key
Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Xiaorong Wang
- Center
for Reproductive Medicine, Affiliated Maternity
and Child Health Care Hospital of Nantong University, Nantong 226018, China
- Nantong
Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong 226018, China
- Nantong
Key Laboratory of Genetics and Reproductive Medicine, Nantong 226018, China
| | - Hao Chen
- Institute
of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Jun Yu
- Institute
of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| |
Collapse
|
8
|
Yuan Z, Ye J, Liu B, Zhang L. Unraveling the role of autophagy regulation in Crohn's disease: from genetic mechanisms to potential therapeutics. ADVANCED BIOTECHNOLOGY 2024; 2:14. [PMID: 39883213 PMCID: PMC11740883 DOI: 10.1007/s44307-024-00021-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 02/29/2024] [Accepted: 03/10/2024] [Indexed: 01/31/2025]
Abstract
Autophagy serves as the primary intracellular degradation mechanism in which damaged organelles and self-cytoplasmic proteins are transported to the lysosome for degradation. Crohn's disease, an idiopathic chronic inflammatory disorder of the gastrointestinal tract, manifests in diverse regions of the digestive system. Recent research suggests that autophagy modulation may be a new avenue for treating Crohn's disease, and several promising small-molecule modulators of autophagy have been reported as therapeutic options. In this review, we discuss in detail how mutations in autophagy-related genes function in Crohn's disease and summarize the modulatory effects on autophagy of small-molecule drugs currently used for Crohn's disease treatment. Furthermore, we delve into the therapeutic potential of small-molecule autophagy inducers on Crohn's disease, emphasizing the prospects for development in this field. We aim to highlight the significance of autophagy modulation in Crohn's disease, with the aspiration of contributing to the development of more efficacious treatments that can alleviate their suffering, and improve their quality of life.
Collapse
Affiliation(s)
- Ziyue Yuan
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Jing Ye
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bo Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Lan Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| |
Collapse
|
9
|
Fikry E, Orfali R, El-Sayed SS, Perveen S, Ghafar S, El-Shafae AM, El-Domiaty MM, Tawfeek N. Potential Hepatoprotective Effects of Chamaecyparis lawsoniana against Methotrexate-Induced Liver Injury: Integrated Phytochemical Profiling, Target Network Analysis, and Experimental Validation. Antioxidants (Basel) 2023; 12:2118. [PMID: 38136237 PMCID: PMC10740566 DOI: 10.3390/antiox12122118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
Methotrexate (MTX) therapy encounters significant limitations due to the significant concern of drug-induced liver injury (DILI), which poses a significant challenge to its usage. To mitigate the deleterious effects of MTX on hepatic function, researchers have explored plant sources to discover potential hepatoprotective agents. This study investigated the hepatoprotective effects of the ethanolic extract derived from the aerial parts of Chamaecyparis lawsoniana (CLAE) against DILI, specifically focusing on MTX-induced hepatotoxicity. UPLC-ESI-MS/MS was used to identify 61 compounds in CLAE, with 31 potential bioactive compounds determined through pharmacokinetic analysis. Network pharmacology analysis revealed 195 potential DILI targets for the bioactive compounds, including TP53, IL6, TNF, HSP90AA1, EGFR, IL1B, BCL2, and CASP3 as top targets. In vivo experiments conducted on rats with acute MTX-hepatotoxicity revealed that administering CLAE orally at 200 and 400 mg/kg/day for ten days dose-dependently improved liver function, attenuated hepatic oxidative stress, inflammation, and apoptosis, and reversed the disarrayed hepatic histological features induced by MTX. In general, the findings of the present study provide evidence in favor of the hepatoprotective capabilities of CLAE in DILI, thereby justifying the need for additional preclinical and clinical investigations.
Collapse
Affiliation(s)
- Eman Fikry
- Department of Pharmacognosy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; (E.F.); (A.M.E.-S.); (N.T.)
| | - Raha Orfali
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia;
| | - Shaimaa S. El-Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt;
| | - Shagufta Perveen
- Department of Chemistry, School of Computer, Mathematical and Natural Sciences, Morgan State University, Baltimore, MD 21251, USA;
| | - Safina Ghafar
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia;
| | - Azza M. El-Shafae
- Department of Pharmacognosy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; (E.F.); (A.M.E.-S.); (N.T.)
| | - Maher M. El-Domiaty
- Department of Pharmacognosy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; (E.F.); (A.M.E.-S.); (N.T.)
| | - Nora Tawfeek
- Department of Pharmacognosy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; (E.F.); (A.M.E.-S.); (N.T.)
| |
Collapse
|
10
|
Wasfey EF, Shaaban M, Essam M, Ayman Y, Kamar S, Mohasseb T, Rozik R, Khaled H, Eladly M, Elissawi M, Bassem A, Elshora SZ, Radwan SM. Infliximab Ameliorates Methotrexate-Induced Nephrotoxicity in Experimental Rat Model: Impact on Oxidative Stress, Mitochondrial Biogenesis, Apoptotic and Autophagic Machineries. Cell Biochem Biophys 2023; 81:717-726. [PMID: 37656380 PMCID: PMC10611839 DOI: 10.1007/s12013-023-01168-7] [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] [Accepted: 08/18/2023] [Indexed: 09/02/2023]
Abstract
Accumulating data confirms that Methotrexate (MTX), a well-known immunosuppressive and anticancer drug, causes nephrotoxicity. Infliximab (INF), the inhibitor of tumor necrosis factor-alpha (TNF-α), was proven to have anti-inflammatory properties. Thus, it may have potential in preventing MTX-induced nephrotoxicity. Therefore, this study aimed to inspect the prospective nephroprotective effect of INF on MTX-induced rat nephrotoxicity through investigating the possible molecular mechanisms, including its interference with different death routes, oxidative stress as well as mitochondrial biogenesis. Rats received an INF intraperitoneal single dose of 7 mg/kg 72 h prior to a single 20 mg/kg MTX injection. MTX nephrotoxicity was demonstrated by significantly increased serum levels of the renal indicators urea and creatinine as well as renal inflammatory markers TNF-α and Interleukin-6 (IL-6) and the renal oxidative stress marker malondialdehyde (MDA), while renal antioxidant enzyme superoxide dismutase (SOD) was significantly decreased compared to control. INF injection prior to MTX markedly reversed these MTX-induced effects. Besides, MTX impaired mitochondrial biogenesis, while INF attenuated this impairment, as indicated by increased expression of peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α). Finally, MTX triggered apoptotic and autophagic cascades in renal tissues as evidenced by reduced anti-apoptotic Bcl-2 protein expression as well as elevated expression of the pro-apoptotic protein Bax and both key regulators of autophagy; beclin-1 and LC-3, whereas INF pretreatment counteracted these apoptotic and autophagic effects of MTX. Summarily, these results suggest that INF provides protection against MTX-induced nephrotoxicity which could be elucidated by its antioxidant, anti-inflammatory, anti-apoptotic and anti-autophagic effects as well as upregulating mitochondrial biogenesis.
Collapse
Affiliation(s)
- Eman F Wasfey
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Marah Shaaban
- Drug Design Program, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Manalia Essam
- Drug Design Program, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Youssef Ayman
- Drug Design Program, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Salma Kamar
- Drug Design Program, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Tasneem Mohasseb
- Drug Design Program, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Rana Rozik
- Drug Design Program, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Huda Khaled
- Drug Design Program, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mohamed Eladly
- Drug Design Program, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mohammed Elissawi
- Drug Design Program, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Ahmed Bassem
- Drug Design Program, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Shimaa Z Elshora
- Histology Department, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Sara M Radwan
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
11
|
Radwan SM, Alqulaly M, Elsaeed MY, Elshora SZ, Atwa AH, Wasfey EF. L-carnitine reverses methotrexate-induced nephrotoxicity in experimental rat model: Insight on SIRT1/PGC-1α/Nrf2/HO-1 axis. J Appl Toxicol 2023; 43:1667-1675. [PMID: 37312617 DOI: 10.1002/jat.4503] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/15/2023]
Abstract
Methotrexate (MTX) is a chemotherapeutic agent used for treating several types of cancer as well as psoriasis and rheumatoid arthritis, but its use is limited due to its nephrotoxicity. The purpose of this research work was to observe ameliorative effects of L-carnitine (LC) toward renal toxicity caused by MTX and mechanisms responsible for these effects. Thirty-two male Sprague-Dawley rats were divided into four groups (eight rats/group), control group (received saline), MTX group (20 mg/kg/i.p. once), LC group (500 mg/kg/i.p. for 5 days), and MTX + LC group (received a single MTX dose 20 mg/kg/i.p. followed by LC 500 mg/kg/i.p. for 5 days). Histopathological examinations, lipid oxidation marker, malondialdehyde (MDA), and the antioxidant superoxide dismutase (SOD) as well as inflammatory (tumor necrosis factor-α [TNF-α] and interleukin-6 [IL-6]) and apoptotic markers (Bax, Bcl2, and caspase-3) were used to assess renal toxicity. Moreover, the protein levels of silent information regulator 1 (SIRT1) and its downstream signaling targets, peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), and nuclear factor erythroid 2-related factor 2 (Nrf2) in addition to heme oxygenase-1 (HO-1) were measured. LC significantly protected against MTX-induced nephrotoxicity. It ameliorated MTX-induced renal histopathological changes and diminished MTX-induced renal oxidative stress, renal inflammation, and apoptosis. LC also upregulated the expression of SIRT1 and PGC-1 as well as Nrf2 and HO-1. By controlling the expression of renal SIRT1/PGC-1/Nrf2/HO-1, LC displayed antioxidant, anti-inflammatory, and anti-apoptotic activities. Hence, using LC supplements may help prevent negative MTX side effects.
Collapse
Affiliation(s)
- Sara M Radwan
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mustafa Alqulaly
- Physiology Department, Damietta Faculty of Medicine, Al-Azhar University, Damietta, Egypt
| | - Magdy Y Elsaeed
- Physiology Department, Damietta Faculty of Medicine, Al-Azhar University, Damietta, Egypt
| | - Shimaa Z Elshora
- Histology Department, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Asmaa H Atwa
- Forensic medicine and clinical toxicology Department, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Eman F Wasfey
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
12
|
Zhao Z, Hua Z, Luo X, Li Y, Yu L, Li M, Lu C, Zhao T, Liu Y. Application and pharmacological mechanism of methotrexate in rheumatoid arthritis. Biomed Pharmacother 2022; 150:113074. [PMID: 35658215 DOI: 10.1016/j.biopha.2022.113074] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 11/19/2022] Open
Abstract
Methotrexate (MTX) has been used for the treatment of rheumatoid arthritis (RA) for about forty years and to date MTX remains the part of global standard of treatment for RA. The efficacy of MTX in RA is the result of multiple mechanisms of action. In order to summarize the possible pharmacological mechanisms of MTX in the treatment of RA, this review will elaborate on folate antagonism, promotion of adenosine accumulation, regulation of inflammatory signaling pathways, bone protection and maintenance of immune system function.
Collapse
Affiliation(s)
- Zixuan Zhao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zhenglai Hua
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xinyi Luo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yang Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Liuchunyang Yu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Ming Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Ting Zhao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Yuanyan Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
13
|
Aslan R, Taken K, Erbin A, Alp HH, Eryilmaz R, Sarilar O, Huyut Z. The synergistic effects of testosterone and phophodiesterase-5 inhibitor combination on oxidative stress markers, matrix metalloproteinases and oxidative DNA damage: A randomized controlled experimental study. Rev Int Androl 2022; 20:73-79. [PMID: 35115255 DOI: 10.1016/j.androl.2020.10.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 10/17/2020] [Indexed: 10/19/2022]
Abstract
PURPOSE To investigate the effects of combined tadalafil and testosterone usage on oxidative stress, DNA damage and MMPs in testosterone deficiency. METHODS Fifty rats were randomly divided into 5 groups (group-1: sham group-placebo, group-2: bilateral orchiectomy (ORX), group-3: bilateral ORX+tadalafil, group-4: bilateral ORX+testosterone, group-5: bilateral ORX+tadalafil+testosterone). Group-3 received tadalafil (5mg/kg/day, oral). Group-4 was administered testosterone undecanoate (100mg/kg i.m., single dose). Group-5 was administered a combination of tadalafil and testosterone undecanoate. All groups were compared with regard to serum nicotinamide adenine dinucleotide phosphate oxidase-4 (NOX-4), total thiol, matrix metalloproteinase-2 (MMP-2), MMP-3 and MMP-9, tissue inhibitor of metalloproteinases-1 (TIMP-1) and TIMP-2 and 8-hydroxy-2-deoxy guanosine (8-OHdG) levels. RESULTS Total thiol levels of group-2 were significantly lower than the other groups and thiol levels were higher in group-1 and group-5 than in the other groups. NOX4, MMP2 and 9 levels in group-2 were higher than in the other groups. MMP-9 levels in group-5 were lower than in groups 3 and 4 (p=.001). The level of 8-OHdG in groups 2 and 3 was higher than in the other groups (p=.001). In correlation analysis, 8-OHdG, MMP2, and 9 levels were negatively correlated with total thiol, whereas NOX4 and 8-OHdG levels were positively correlated with MMPs values. CONCLUSIONS The combination of testosterone with PDE-5 inhibitor suppresses MMP-9 levels and increases total thiol levels better than testosterone alone and tadalafil alone. Therefore, testosterone can be considered for use with PDE-5 inhibitor from the initial stage in case of testosterone deficiency.
Collapse
Affiliation(s)
- Rahmi Aslan
- Department of Urology, Faculty of Medicine, Van Yuzuncu Yıl University, Van, Turkey
| | - Kerem Taken
- Department of Urology, Faculty of Medicine, Van Yuzuncu Yıl University, Van, Turkey
| | - Akif Erbin
- Department of Urology, Haseki Training and Research Hospital, Istanbul, Turkey.
| | - Hamit Hakan Alp
- Department of Biochemistry, Faculty of Medicine, Van Yuzuncu Yıl University, Van, Turkey
| | - Recep Eryilmaz
- Department of Urology, Faculty of Medicine, Van Yuzuncu Yıl University, Van, Turkey
| | - Omer Sarilar
- Department of Urology, Haseki Training and Research Hospital, Istanbul, Turkey
| | - Zubeyir Huyut
- Department of Biochemistry, Faculty of Medicine, Van Yuzuncu Yıl University, Van, Turkey
| |
Collapse
|
14
|
Lactoferrin Alleviated AFM1-Induced Apoptosis in Intestinal NCM 460 Cells through the Autophagy Pathway. Foods 2021; 11:foods11010023. [PMID: 35010149 PMCID: PMC8750231 DOI: 10.3390/foods11010023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 11/25/2022] Open
Abstract
Aflatoxin M1 (AFM1) is the only mycotoxin with maximum residue limit in milk, which may result in serious human diseases. On the contrary, lactoferrin (Lf) is an active protein with multiple functions. Studies have confirmed that Lf has a powerful potential to protect the intestines, but the influence of Lf on mycotoxins is not clear. This study aims to explore whether Lf can protect the cytotoxicity induced by AFM1, and determine the underlying mechanisms in human normal colonic epithelial NCM460 cells. The results indicated that AFM1 decreased the cell viability, and increased the levels of apoptosis and autophagy of NCM460 cells. Lf can alleviate the cytotoxicity induced by AFM1 through enhancing cell viability, significantly down-regulated the expression of apoptotic genes and proteins (BAX, caspase3, caspase9, caspase3, and caspase9), and regulated the gene and protein expression of autophagy factors (Atg5, Atg7, Atg12, Beclin1, ULK1, ULK2, LC3, and p62). Furthermore, interference of the key gene Atg5 of autophagy can reduce AFM1-induced apoptosis, which is consistent with the role of Lf, implying that Lf may protect AFM1-induced intestinal injury by inhibiting excessive autophagy-mediated apoptosis. Taken together, our data indicated that Lf has a mitigating effect on apoptosis induced by AFM1 through the autophagy pathway.
Collapse
|
15
|
Bitter melon (Momordica charantia) fruit extract ameliorates methotrexate‐induced reproductive toxicity in male rats. MARMARA MEDICAL JOURNAL 2021. [DOI: 10.5472/marumj.988941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
16
|
Mahoutforoush A, Solouk A, Hamishehkar H, Haghbin Nazarpak M, Abbaspour-Ravasjani S. Novel decorated nanostructured lipid carrier for simultaneous active targeting of three anti-cancer agents. Life Sci 2021; 279:119576. [PMID: 33965376 DOI: 10.1016/j.lfs.2021.119576] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/18/2021] [Accepted: 04/26/2021] [Indexed: 12/18/2022]
Abstract
Cancer-targeted co-delivery of therapeutic agents has been recognized as an effective strategy for increasing efficacy and reducing side effects of therapeutic agents. In this study, we used methotrexate (MTX) alone as a targeting moiety and chemotherapeutic agent and in combination with docetaxel (DTX) and doxorubicin (DOX) as chemotherapeutic agents to stop cancer cell proliferation with the aid of newly designed nanostructured lipid carriers (NLCs). The physicochemical properties of our designed nanocomplexes were evaluated by DLS, FT-IR spectroscopy, SEM, and TEM. Moreover, the targeting efficiency of the designed and synthesized nanoplatforms was evaluated on the folate receptor (FR) positive human breast cancer cell line (MCF-7) and FR negative human alveolar basal epithelial cells (A549). The NLCs/DTX/DOX/CS and NLCs/DTX/DOX/CS-MTX complexes significantly increased the cell cytotoxicity and the cell apoptosis rate. However, the complexes significantly reduced the capability of colony formation and cell migration. Our results revealed that NLCs/DTX/DOX/CS-MTX had synergistic cytotoxicity, reactive oxygen spaces, autophagy, and the apoptosis induction ability with an enhanced cellular internalization rate in FR-positive cancer cells, thorough MTX recognition capability. We conclude that the NLCs/DTX/DOX/CS-MTX complex is a new promising paradigm for breast cancer-targeted co-delivery.
Collapse
Affiliation(s)
- Amin Mahoutforoush
- Department of Biomedical Engineering, Central Tehran Branch, Islamic Azad University, Tehran 13185/768, Iran
| | - Atefeh Solouk
- Biomedical Engineering Department, Amirkabir University of Technology (Tehran Polytechnic), Tehran 1591634311, Iran
| | - Hamed Hamishehkar
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Masoumeh Haghbin Nazarpak
- New Technologies Research Center (NTRC), Amirkabir University of Technology, Tehran 1591634653, Iran
| | | |
Collapse
|
17
|
Zhao Y, Zheng W. Deciphering the antitumoral potential of the bioactive metabolites from medicinal mushroom Inonotus obliquus. JOURNAL OF ETHNOPHARMACOLOGY 2021; 265:113321. [PMID: 32877719 DOI: 10.1016/j.jep.2020.113321] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 08/09/2020] [Accepted: 08/22/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The crude extracts of the medicinal mushroom Inonotus obliquus have been used as an effective traditional medicine to treat malicious tumors, gastritis, gastric ulcers, and other inflammatory conditions in Russia and most Baltic countries. AIM OF THIS REVIEW Deciphering the antitumoral potential of the bioactive metabolites from I. obliquus and addressing its possibility to be used as effective agents for tumor treatment, restoration of compromised immunity and protection of gastrointestinal damage caused by chemotherapy. MATERIALS AND METHODS We analysed the current achievements and dilemma in tumor chemo- or immunotherapy. In this context, we searched the published literatures on I. obliquus covering from 1990 to 2020, and summarized the activities of antitumor, antioxidation, and immunomodulation by the polysaccharides, triterpenoids, small phenolic compounds, and hispidin polyphenols. By comparing the merits and shortcomings of current and traditional methodology for tumor treatment, we further addressed feasibility for the use of I. obliquus as an effective natural drug for tumor treatment and prevention. RESULTS The diverse bioactive metabolites confer I. obliquus great potential to inhibit tumor growth and metastasis. Its antitumor activities are achieved either through suppressing multiple oncogenic signals including but not limited to the activation of NF-κB and FAK, and the expression of RhoA/MMP-9 via ERK1/2 and PI3K/Akt signaling pathway. The antitumor activities can also be achieved by inhibiting tyrosinase activity via PAK1-dependent signaling pathway or altering lysosomal membrane permeabilization through blocking tubulin polymerization and/or disturbing energy metabolism through LKB1/AMPK pathway. In addition, the metabolites from I. obliquus also harbour the potentials to reverse MDR either through selective inhibition on P-gp/ABCB1 or MRP1/ABCC1 proteins or the induction of G2/M checkpoint arrest in tumor cells of chemoresistant phenotypes mediated by Nox/ROS/NF-kB/STAT3 signaling pathway. In addition to the eminent effects in tumor inhibition, the metabolites in I. obliquus also exhibit immunomodulatory potential to restore the compromised immunity and protect against ulcerative damage of GI tract caused by chemotherapy. CONCLUSIONS I. obliquus possesses the potential to reduce incidence of tumorigenesis in healthy people. For those whose complete remission has been achieved by chemotherapy, administration of the fungus will inhibit the activation of upstream oncogenic signals and thereby prevent metastasis; for those who are in the process of chemotherapy administration of the fungus will not only chemosensitize the tumor cells and thereby increasing the chemotherapeutic effects, but also help to restore the compromised immunity and protect against ulcerative GI tract damage and other side-effects induced by chemotherapy.
Collapse
Affiliation(s)
- Yanxia Zhao
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, Jiangsu Normal University, Xuzhou, 221116, China.
| | - Weifa Zheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, Jiangsu Normal University, Xuzhou, 221116, China.
| |
Collapse
|
18
|
Zhang X, Chen C, Zhong Y, Zeng X. lncRNA involved in triptonide-induced cytotoxicity in mouse germ cells. Reprod Toxicol 2020; 98:218-224. [PMID: 33045310 DOI: 10.1016/j.reprotox.2020.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 09/16/2020] [Accepted: 10/04/2020] [Indexed: 01/29/2023]
Abstract
Triptonide reportedly has strong antitumor and anti-inflammatory activities. However, its severe toxicity, including to the reproductive system, has greatly restricted its use in patients who wish to preserve fertility. lncRNAs play crucial roles in male fertility and reportedly regulate triptonide's antitumor activity. However, whether lncRNAs are involved in triptonide-induced reproductive toxicity is unknown. Here, we showed that triptonide induced significant cytotoxicity, as demonstrated by reduced cell viability and induction of apoptosis and autophagy in mouse germ cells (a spermatocyte cell line, GC2). The expression levels of numerous lncRNAs and mRNAs in GC2 cells were altered at the transcriptome level after treatment with triptonide for 24 h, as determined by RNA sequencing. Gene ontology and pathway analyses showed that the functions of the differentially expressed lncRNAs and mRNAs were closely linked with many processes, including gene expression regulation, cell death, cell cycle regulation, cell proliferation and development and others. After validating our RNA-seq data, we selected one lncRNA, Obox4-ps35, dramatically induced by triptonide for further investigation. Obox4-ps35 knock-out aggravated triptonide-induced cytotoxicity by decreasing cell survival and increasing apoptosis and autophagy rates. These data suggest that germ cells exposed to triptonide overexpress Obox4-ps35 to protect against triptonide-induced cytotoxicity. This study provides preliminary evidence and novel directions for exploring roles of lncRNAs in triptonide-induced cytotoxicity, especially in reproductive toxicity.
Collapse
Affiliation(s)
- Xiaoning Zhang
- Medical School, Institute of Reproductive Medicine, Nantong University, Nantong, China; Institute of Life Science, Nanchang University, Nanchang, China
| | - Chen Chen
- Medical School, Institute of Reproductive Medicine, Nantong University, Nantong, China
| | - Yuanyuan Zhong
- Institute of Life Science, Nanchang University, Nanchang, China
| | - Xuhui Zeng
- Medical School, Institute of Reproductive Medicine, Nantong University, Nantong, China.
| |
Collapse
|