1
|
Al-Maamari A, Sultan M, Wu S, Zhang T, Wang C, Han B, Duan Y, Ding SS, Chen N, Zhang H, Sun F, Chen X, Yu D, Su S. Activation of sigma 1 receptor attenuates doxorubicin-induced cardiotoxicity by alleviating oxidative stress, mitochondria dysfunction, ER stress-related apoptosis, and autophagy impairment. Int J Biol Macromol 2025; 310:143549. [PMID: 40294674 DOI: 10.1016/j.ijbiomac.2025.143549] [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: 01/03/2025] [Revised: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 04/30/2025]
Abstract
The cardiotoxic effects of doxorubicin (DOX) are a significant clinical challenge. This study explored the mechanisms underlying the cardioprotective effects of fluvoxamine (FLU) in DOX-induced cardiotoxicity. In vitro and in vivo models of DOX-induced cardiotoxicity were established using H9c2 cardiomyocytes and BALB/c mice, respectively. The cardioprotective effects of FLU were evaluated using echocardiography, electrocardiography, HE staining, myocardial injury indicators, immunohistochemical staining, immunofluorescence staining, and western blotting. Small interfering RNA (siRNA) targeting the sigma-1 receptor (Sig1R), the Sig1R inhibitor haloperidol (HALO), and the nuclear factor erythroid 2-related factor 2 inhibitor ML385 were used to verify the mechanism of FLU in H9c2 cells, and molecular docking was performed to compare the binding affinities of DOX and FLU to Sig1R. DOX treatment significantly decreased Sig1R expression, leading to cardiac dysfunction, endoplasmic reticulum stress, apoptosis, impaired autophagy, oxidative stress, and mitochondrial dysfunction. Treatment with FLU mitigated these effects. Molecular docking simulations revealed that FLU showed better binding affinity for Sig1R than DOX. Moreover, the beneficial effects of FLU in DOX-induced cardiotoxicity were reduced in the presence of HALO, ML385, or Sig1R siRNA. Our study indicates that FLU effectively reduces DOX-induced cardiotoxicity by activating Sig1R, highlighting its potential as a therapeutic agent against chemotherapy-induced cardiotoxicity.
Collapse
Affiliation(s)
- Ahmed Al-Maamari
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Marwa Sultan
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Shang Wu
- Breast Cancer Center, The Fourth Hospital, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Tao Zhang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Chuchu Wang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Boye Han
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Yuxin Duan
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Shan-Shan Ding
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Nannan Chen
- School of Pharmacy, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Huaxing Zhang
- Core Facilities and Centers, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Fangyi Sun
- Department of Cardiovascular Medicine, The Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Xueyan Chen
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Ding Yu
- Cardiovascular Intensive Care Unit, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050023, PR China
| | - Suwen Su
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China.
| |
Collapse
|
2
|
Fligor SC, Tsikis ST, Hirsch TI, Jain A, Sun L, Rockowitz S, Gura KM, Puder M. Inflammation drives pathogenesis of early intestinal failure-associated liver disease. Sci Rep 2024; 14:4240. [PMID: 38378873 PMCID: PMC10879484 DOI: 10.1038/s41598-024-54675-9] [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: 07/26/2023] [Accepted: 02/15/2024] [Indexed: 02/22/2024] Open
Abstract
Patients with intestinal failure who receive long-term parenteral nutrition (PN) often develop intestinal failure-associated liver disease (IFALD). Although there are identified risk factors, the early pathogenesis is poorly understood and treatment options are limited. Here, we perform a transcriptomic analysis of liver tissue in a large animal IFALD model to generate mechanistic insights and identify therapeutic targets. Preterm Yorkshire piglets were provided PN or bottle-fed with sow-milk replacer for 14 days. Compared to bottle-fed controls, piglets receiving PN developed biochemical cholestasis by day of life 15 (total bilirubin 0.2 vs. 2.9 mg/dL, P = 0.01). RNA-Seq of liver tissue was performed. Ingenuity Pathway Analysis identified 747 differentially expressed genes (343 upregulated and 404 downregulated) with an adjusted P < 0.05 and a fold-change of > |1|. Enriched canonical pathways were identified, demonstrating broad activation of inflammatory pathways and inhibition of cell cycle progression. Potential therapeutics including infliximab, glucocorticoids, statins, and obeticholic acid were identified as predicted upstream master regulators that may reverse the PN-induced gene dysregulation. The early driver of IFALD in neonates may be inflammation with an immature liver; identified therapeutics that target the inflammatory response in the liver should be investigated as potential treatments.
Collapse
Affiliation(s)
- Scott C Fligor
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, USA
| | - Savas T Tsikis
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, USA
| | - Thomas I Hirsch
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, USA
| | - Ashish Jain
- Research Computing, Information Technology, Boston Children's Hospital, Boston, MA, USA
| | - Liang Sun
- Research Computing, Information Technology, Boston Children's Hospital, Boston, MA, USA
| | - Shira Rockowitz
- Harvard Medical School, Boston, MA, USA
- Research Computing, Information Technology, Boston Children's Hospital, Boston, MA, USA
- Division of Genetics and Genomics, and the Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, USA
| | - Kathleen M Gura
- Harvard Medical School, Boston, MA, USA
- Department of Pharmacy and the Division of Gastroenterology and Nutrition, Boston Children's Hospital, Boston, MA, USA
| | - Mark Puder
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital, Boston, MA, 02115, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
3
|
Patel D, Yadav P, Singh SK, Tanwar SS, Sehrawat A, Khurana A, Bhatti JS, Navik U. Betaine alleviates doxorubicin-induced nephrotoxicity by preventing oxidative insults, inflammation, and fibrosis through the modulation of Nrf2/HO-1/NLRP3 and TGF-β expression. J Biochem Mol Toxicol 2024; 38:e23559. [PMID: 37840533 DOI: 10.1002/jbt.23559] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 09/11/2023] [Accepted: 10/05/2023] [Indexed: 10/17/2023]
Abstract
Doxorubicin (Dox) is an anthracycline antibiotic used to treat various cancers and shows severe toxicity in multiple organ systems, including kidneys. Evidence shows that betaine's antioxidant and anti-inflammatory properties could prevent the onset of several disorders. Hence, the present study aims to investigate the therapeutic potential of betaine on Dox-induced nephrotoxicity (DIN). Nephrotoxicity was induced in male Sprague Dawley rats using Dox at a dose of 4 mg/kg (cumulative dose: 20 mg/kg) by the intraperitoneal route and cotreated with betaine through oral gavage (200 and 400 mg/kg) for 28 days. At the end of the experiment, biochemical, oxidative stress parameters, histopathology, and qRT-PCR were performed. DIN was indicated by elevated serum creatinine, urea, and decreased albumin levels representing kidney damage; the histopathological lesions (increased capsular space, renal tubule damage, and fibrosis) in renal tissues supported these biochemical findings. Interestingly, betaine treatment improves these alterations in Dox-treated rats. Further, betaine treatment decreases the lipid peroxidation and nitrite concentration and increases the superoxide dismutases and catalase enzyme concentration in Dox-treated rats. Fascinatingly, at the molecular level, DIN in rats shows upregulation of the Nrf2/HO-1 gene, while betaine treatment attenuated its expression along with the downregulation of inflammatory genes (NLRP3, TLR-4, TNF-α, and IL-6) and fibrosis-related genes (TGF-β and Acta2) expression in Dox-treated rats. These results showed that betaine has reno-protective properties by reducing inflammatory and fibrotic mediators and enhancing antioxidant capacity in the renal tissue of rats treated with Dox. We believe betaine can be exploited as a dietary supplement to attenuate DIN.
Collapse
Affiliation(s)
- Dhaneshvaree Patel
- Department of Pharmacology, Central University of Punjab, Bathinda, Punjab, India
| | - Poonam Yadav
- Department of Pharmacology, Central University of Punjab, Bathinda, Punjab, India
| | - Sumeet K Singh
- Department of Pharmacology, Central University of Punjab, Bathinda, Punjab, India
| | - Sampat S Tanwar
- Department of Pharmacology, Central University of Punjab, Bathinda, Punjab, India
| | - Abhishek Sehrawat
- Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Amit Khurana
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH Aachen University Hospital, Aachen, Germany
| | - Jasvinder S Bhatti
- Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Bathinda, Punjab, India
| |
Collapse
|
4
|
Li X. Doxorubicin-mediated cardiac dysfunction: Revisiting molecular interactions, pharmacological compounds and (nano)theranostic platforms. ENVIRONMENTAL RESEARCH 2023; 234:116504. [PMID: 37356521 DOI: 10.1016/j.envres.2023.116504] [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: 04/22/2023] [Revised: 06/17/2023] [Accepted: 06/23/2023] [Indexed: 06/27/2023]
Abstract
Although chemotherapy drugs are extensively utilized in cancer therapy, their administration for treatment of patients has faced problems that regardless of chemoresistance, increasing evidence has shown concentration-related toxicity of drugs. Doxorubicin (DOX) is a drug used in treatment of solid and hematological tumors, and its function is based on topoisomerase suppression to impair cancer progression. However, DOX can also affect the other organs of body and after chemotherapy, life quality of cancer patients decreases due to the side effects. Heart is one of the vital organs of body that is significantly affected by DOX during cancer chemotherapy, and this can lead to cardiac dysfunction and predispose to development of cardiovascular diseases and atherosclerosis, among others. The exposure to DOX can stimulate apoptosis and sometimes, pro-survival autophagy stimulation can ameliorate this condition. Moreover, DOX-mediated ferroptosis impairs proper function of heart and by increasing oxidative stress and inflammation, DOX causes cardiac dysfunction. The function of DOX in mediating cardiac toxicity is mediated by several pathways that some of them demonstrate protective function including Nrf2. Therefore, if expression level of such protective mechanisms increases, they can alleviate DOX-mediated cardiac toxicity. For this purpose, pharmacological compounds and therapeutic drugs in preventing DOX-mediated cardiotoxicity have been utilized and they can reduce side effects of DOX to prevent development of cardiovascular diseases in patients underwent chemotherapy. Furthermore, (nano)platforms are used comprehensively in treatment of cardiovascular diseases and using them for DOX delivery can reduce side effects by decreasing concentration of drug. Moreover, when DOX is loaded on nanoparticles, it is delivered into cells in a targeted way and its accumulation in healthy organs is prevented to diminish its adverse impacts. Hence, current paper provides a comprehensive discussion of DOX-mediated toxicity and subsequent alleviation by drugs and nanotherapeutics in treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Xiaofeng Li
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine Tongji University, Shanghai, 200072, China.
| |
Collapse
|
5
|
Demir M, Altinoz E, Koca O, Elbe H, Onal MO, Bicer Y, Karayakali M. Antioxidant and anti-inflammatory potential of crocin on the doxorubicin mediated hepatotoxicity in Wistar rats. Tissue Cell 2023; 84:102182. [PMID: 37523948 DOI: 10.1016/j.tice.2023.102182] [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: 04/14/2023] [Revised: 07/18/2023] [Accepted: 07/25/2023] [Indexed: 08/02/2023]
Abstract
Doxorubicin (DXR) is widely used in cancer treatment. However, it has not yet been possible to prevent the side effects of DXR. The aim of this study was to investigate the hepatoprotective effect of crocin against DXR used in cancer treatment. For this reason; forty Wistar rats (male-250-300 g) were allocated into four groups (n = 10/group): Control, Crocin, DXR and DXR+Crocin. Control and Crocin groups were administered saline and crocin (40 mg/kg, i.p) for 15 days, respectively. DXR group, cumulative dose 12 mg/kg DXR, was administered for 12 days via 48 h intervals in six injections (2 mg/kg each, i.p). DXR+Crocin group, crocin (40 mg/kg-i.p) was administered for 15 days, and DXR was given as in the DXR group. The results revealed that serum liver markers (alanine transaminase (ALT), aspartate transaminase (AST), and alkaline phosphatase (ALP) increased significantly after DXR administration but recovered after crocin therapy. In addition, lipid peroxidation (MDA), and inflammatory cytokine (TNF-α) increased after DXR application and the antioxidative defense system (GSH, SOD, CAT) significantly decreased and re-achieved by crocin treatment. Our results conclude that crocin treatment was related to ameliorated hepatocellular architecture and reduced hepatic oxidative stress and inflammation in rats with DXR-induced hepatotoxicity.
Collapse
Affiliation(s)
- M Demir
- Department of Physiology, Faculty of Medicine, Karabuk University, Karabuk, Turkey.
| | - E Altinoz
- Department of Medical Biochemistry, Faculty of Medicine, Karabuk University, Karabuk, Turkey
| | - O Koca
- Department of Biochemistry, Karabuk University Education and Research Hospital, Karabuk, Turkey
| | - H Elbe
- Department of Histology and Embryology, Faculty of Medicine, Mugla Sıtkı Kocman University, Mugla, Turkey
| | - M O Onal
- Department of Histology and Embryology, Faculty of Medicine, Mugla Sıtkı Kocman University, Mugla, Turkey
| | - Y Bicer
- Department of Medical Biochemistry, Faculty of Medicine, Karabuk University, Karabuk, Turkey
| | - M Karayakali
- Department of Medical Biochemistry, Faculty of Medicine, Karabuk University, Karabuk, Turkey
| |
Collapse
|