1
|
Faheem H, Alawadhi R, Basha EH, Ismail R, Ibrahim HA, Elshamy AM, Motawea SM, Seleem MA, Elkordy A, Homouda AA, Khaled HE, Aboeida RA, Abdel Ghafar MT, Rizk FH, El-Harty YM. Ameliorating immune-dependent inflammation and apoptosis by targeting TLR4/MYD88/NF-κB pathway by celastrol mitigates the diabetic reproductive dysfunction. Physiol Genomics 2025; 57:103-114. [PMID: 39510137 DOI: 10.1152/physiolgenomics.00072.2024] [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: 06/10/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 11/15/2024] Open
Abstract
This study aimed to examine the protective effect of celastrol on testicular dysfunction in diabetic rats and the potential underlying mechanisms. All rats included in the study were divided into four groups: a control group treated with sodium citrate buffer and vehicle), a celastrol-treated control group, a streptozotocin (STZ)-induced diabetic group following insulin resistance, and a celastrol-treated diabetic group. Serum glucose, triglyceride, total cholesterol, high-density lipoprotein cholesterol, interleukin (IL)-1β, tumor necrosis factor-α, and testosterone levels were measured. In addition, the levels of testicular homogenate superoxide dismutase and malondialdehyde were assessed. Furthermore, testicular tissue relative toll-like receptor 4 (TLR4), nuclear factor kappa B (NF-κB), and myeloid differentiation factor 88 (MYD88) expressions were quantitatively measured using polymerase chain reaction. Histopathological and immunohistochemical studies were also conducted. The results revealed that treatment with celastrol significantly reduced TLR4, MyD88, and NF-κB expressions, and the levels of inflammatory mediators such as tumor necrosis factor-α and IL-1β in the testicular tissue of treated rats. These findings suggest that celastrol has the potential to be effective in the treatment of diabetes-induced testicular injury by inhibiting testicular inflammation, apoptosis, and oxidative stress.NEW & NOTEWORTHY Celastrol inhibits the production of proinflammatory cytokines in the testicular tissue by specifically targeting the TLR4/MyD88/NF-κB signaling cascade pathways. This indicates that celastrol may serve as a promising new therapeutic target for treating diabetic reproductive dysfunction.
Collapse
Affiliation(s)
- Heba Faheem
- Department of Physiology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Rana Alawadhi
- Science Department, College of Basic Education, PAAET, Ardhiya, Kuwait
| | - Eman H Basha
- Department of Physiology, Faculty of Medicine, Tanta University, Tanta, Egypt
- Department of Basic Medical Sciences-Physiology, Faculty of Medicine, Ibn Sina University for Medical Sciences, Amman, Jordan
| | - Radwa Ismail
- Department of Anatomy and Embryology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Hoda A Ibrahim
- Department of Medical Biochemistry, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Amira M Elshamy
- Department of Medical Biochemistry, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Shaimaa M Motawea
- Department of Anatomy and Embryology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Monira A Seleem
- Department of Medical Pharmacology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Alaa Elkordy
- Department of Neuropsychiatry, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Abdallah A Homouda
- Department of Urology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Howayda E Khaled
- Department of Zoology, Faculty of Science, Suez University, Suez, Egypt
| | - Reham A Aboeida
- Department of Internal Medicine, Faculty of Medicine, Tanta University, Tanta, Egypt
| | | | - Fatma H Rizk
- Department of Physiology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Yasmeen M El-Harty
- Department of Physiology, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
2
|
Zhu Y, Zhang F, Li Z, Zhou Y, Shu Y, Ruan J, Chen G. Chinese and western medicine treatment of myocardial fibrosis drugs. Front Cardiovasc Med 2025; 11:1477601. [PMID: 39882321 PMCID: PMC11774883 DOI: 10.3389/fcvm.2024.1477601] [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: 08/08/2024] [Accepted: 12/16/2024] [Indexed: 01/31/2025] Open
Abstract
Myocardial fibrosis (MF) is a common pathological manifestation of many cardiovascular diseases, such as myocardial infarction, myocardial ischemia, and sudden cardiac death. It is characterized by excessive proliferation and activation of fibroblasts, transformation into myofibroblasts, and, eventually, excessive deposition of the extracellular matrix, resulting in heart damage. Currently, modern drugs such as angiotensin-converting enzyme inhibitors, diuretics, and β-blockers can improve myocardial fibrosis in clinical treatment, but their therapeutic effect on this disease is limited, with obvious side effects and high cost. Traditional Chinese medicine (TCM) has the advantages of multiple targets, low cost, and few side effects. Traditional Chinese medicines, such as Salvia miltiorrhiza, Astragalus, and Angelica extracts, and patent Chinese medicines, such as Qiliqiangxin capsules, Shenqi Yiqi dropping pills, and Tongxinluo capsules, can improve myocardial fibrosis. In this review, current Chinese and Western medicine methods for treating myocardial fibrosis are discussed. The signaling pathways and targets of Chinese and Western medicine are involved in the treatment of myocardial fibrosis. This review aimed to provide valuable insights and ideas for both clinical treatment and basic research on myocardial fibrosis.
Collapse
Affiliation(s)
- Yuxi Zhu
- Department of Acupuncture, Bao’an Authentic TCM Therapy Hospital, Shenzheng, China
- Graduate School, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Fangmei Zhang
- Fever Clinic, The 334 Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhongcheng Li
- Department of Acupuncture, Bao’an Authentic TCM Therapy Hospital, Shenzheng, China
| | - Yu Zhou
- Department of Acupuncture, Bao’an Authentic TCM Therapy Hospital, Shenzheng, China
| | - Yi Shu
- Department of Acupuncture, Bao’an Authentic TCM Therapy Hospital, Shenzheng, China
| | - Jian Ruan
- Graduate School, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Guo Chen
- Department of Acupuncture, Bao’an Authentic TCM Therapy Hospital, Shenzheng, China
| |
Collapse
|
3
|
Badr AM, Alotaibi HN, El-Orabi N. Dibenzazepine, a γ-Secretase Enzyme Inhibitor, Protects Against Doxorubicin-Induced Cardiotoxicity by Suppressing NF-κB, iNOS, and Hes1/Hey1 Expression. Inflammation 2024:10.1007/s10753-024-02046-x. [PMID: 39078585 DOI: 10.1007/s10753-024-02046-x] [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: 03/29/2024] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 07/31/2024]
Abstract
Doxorubicin (DOX) is an effective chemotherapeutic drug; however, its cardiotoxicity and resistance compromise its therapeutic index. The Notch pathway was reported to contribute to DOX cancer resistance. The role of Notch pathway in DOX cardiotoxicity has not been identified yet. Notch receptors are characterized by their extracellular (NECD) and intracellular (NICD) domains (NICD). The γ-secretase enzyme helps in the release of NICD. Dibenzazepine (DBZ) is a γ-secretase inhibitor. The present study investigated the effect of Notch pathway inhibition on DOX cardiotoxicity. Twenty-four male Wistar rats were divided into four groups: control group, DOX group, acute cardiotoxicity was induced by a single dose of DOX (20 mg/kg) i.p., DOX (20 mg/kg) plus DBZ group, and DBZ group. The third and fourth groups received i.p. injection of DBZ daily for 14 days at 2 mg/kg dose. DOX cardiotoxicity increased the level of serum creatine kinase-MB and cardiac troponin I, and it was confirmed by the histopathological examination. Moreover, the antioxidants glutathione peroxidase and superoxide dismutase levels were markedly decreased, and the inflammatory markers, inducible nitric oxide synthase, nuclear factor-ķB, and tumor necrosis factor-α were markedly increased. Furthermore, DOX increased BAX protein and downregulated BCL-2. In addition, DOX upregulated Notch pathway-related parameters: Hes1 and Hey1 mRNA levels, and increased Hes1 protein levels. DBZ ameliorated DOX-induced cardiotoxicity, evidenced by reducing the cardiac injury biomarkers, improving cardiac histopathological changes, correcting antioxidant levels, and reducing inflammatory and apoptotic proteins. Our study indicates the protective effect of Notch inhibitor against DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Amira M Badr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Saud University, Riyadh, 11211, Saudi Arabia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Hind N Alotaibi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Saud University, Riyadh, 11211, Saudi Arabia
| | - Naglaa El-Orabi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt.
| |
Collapse
|
4
|
Sabra RT, Bekhit AA, Sabra NT, Abd El-Moeze NA, Fathy M. Nebivolol ameliorates sepsis-evoked kidney dysfunction by targeting oxidative stress and TGF-β/Smad/p53 pathway. Sci Rep 2024; 14:14735. [PMID: 38926458 PMCID: PMC11208533 DOI: 10.1038/s41598-024-64577-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Sepsis is a potential fetal organ destruction brought on through an overzealous immunologic reaction to infection, causing severe inflammation, septic shock, and damage to different organs. Although there has been progress in the identification and controlling of clinical sepsis, the fatality rates are still significant. This study, for the first time, intended to examine the possible ameliorative impact of Nebivolol, a β1-adrenergic antagonist antihypertensive drug, against nephrotoxicity resulted from cecal ligation and puncture (CLP)-induced sepsis in rats, on molecular basis. Sixty male Wistar albino rats were chosen. Oxidative stress indicators and biochemical markers of kidney activity were evaluated. Inflammatory mediators, fibrosis- and apoptosis-related proteins and gene expressions were investigated. Moreover, renal histopathological investigation was performed. CLP-induced nephrotoxicity characterized by markedly elevated serum levels of creatinine, blood urea nitrogen, uric acid, and renal malondialdhyde. On the other hand, it decreased serum total protein level, renal superoxide dismutase activity and reduced glutathione level. Additionally, it significantly elevated the renal inflammatory mediators (tumor necrosis factor-alpha, ilnerlukin (IL)-6, and IL-1β) and Caspase-3 protein, reduced IL-10 level, amplified the expression of transforming growth factor-beta 1 (TGF-β1), p-Smad2/3 and alpha-smooth-muscle actin proteins, downregulated the B cell lymphoma-2 (Bcl-2) gene and elevated the transcription of Bcl-2-associated X-protein (Bax), p53 and Nuclear factor-kappa B (NF-κB) genes. Furtheremor, kidney tissues exhibited significant histopathological changes with CLP. On the contrary, Nebivolol significantly improved all these biochemical changes and enhanced the histopathological alterations obtained by CLP. This research showed, for the first time, that Nebivolol effectively mitigated the CLP-induced kidney dysfunction via its antioxidant, antifibrotic and anti-apoptotic activity through modulation of oxidative stress, TGF-β/NF-κB and TGF-β/Smad/p53 signaling pathways.
Collapse
Affiliation(s)
- Rahma Tharwat Sabra
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | | | - Nourhan Tharwat Sabra
- Department of Anatomy and Embryology, Faculty of Medicine, Beni-Suef University, Beni-Suef, 62514, Egypt
| | | | - Moustafa Fathy
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt.
| |
Collapse
|
5
|
Otçu S, Deveci E, Özgökçe Ç, Gürsoy GT, Tuncer MC. Protective effect of nebivolol on rat ovary exposed to deltamethrin toxicity. Acta Cir Bras 2023; 38:e385423. [PMID: 37878988 PMCID: PMC10629476 DOI: 10.1590/acb385423] [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: 05/03/2023] [Accepted: 06/29/2023] [Indexed: 10/27/2023] Open
Abstract
PURPOSE We aimed to investigate the antioxidant activity of nebivolol against possible damage to the ovarian tissue due to the application of deltamethrin as a toxic agent, by evaluating histopathological proliferating cell nuclear antigen (PCNA) and tumor necrosis factor-alpha (TNF-α) signal molecules immunohistochemically. METHODS The animals were divided into three groups as control, deltamethrin and deltamethrin + nebivolol groups. Vaginal smears were taken after the animals were mated and detected on the first day of pregnancy. After the sixth day, deltamethrin (0.5 mL of 30 mg/kg BW undiluted ULV), and 2 mL of sterile nebivolol solution were administered intraperitoneally every day for 6-21 periods. After routine histopathological follow-up, the ovarian tissue was stained with hematoxylin and eosin stain. RESULTS Control group showed normal histology of ovarium. In deltamethrin group, hyperplasic cells, degenerative follicles, pyknotic nuclei, inflammation and hemorrhagic areas were observed. Nebivolol treatment restored these pathologies. Deltamethrin treatment increased TNF-α and PCNA reaction. However, nebivolol decreased the expression. CONCLUSIONS It was thought that deltamethrin toxicity adversely affected follicle development by inducing degeneration and apoptotic process in preantral and antra follicle cells, and nebivolol administration might reduce inflammation and slow down the apoptotic signal in the nuclear phase and regulate reorganization.
Collapse
Affiliation(s)
- Serap Otçu
- Health Sciences University – Diyarbakır Gazi Yaşargil, Training and Research Hospital – Department of Obstetrics and Gynecology – Diyarbakır – Turkey
| | - Engin Deveci
- Dicle University – Medical School – Department of Histology and Embryology – Diyarbakır – Turkey
| | - Çağdaş Özgökçe
- Zeynep Kamil Hospital – Department of Obstetrics and Gynecology – Perinatology Department – Istanbul – Turkey
| | - Görkem Tutal Gürsoy
- Healt Ministry of Türkiye Republic – Ankara Bilkent City Hospital – Department of Neurology – Ankara –Turkey
| | - Mehmet Cudi Tuncer
- Dicle University – Medical School – Department of Anatomy – Diyarbakır – Turkey
| |
Collapse
|
6
|
Al-Amir H, Janabi A, Hadi NR. Ameliorative effect of nebivolol in doxorubicin-induced cardiotoxicity. J Med Life 2023; 16:1357-1363. [PMID: 38107721 PMCID: PMC10719778 DOI: 10.25122/jml-2023-0090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 05/27/2023] [Indexed: 12/19/2023] Open
Abstract
This study aimed to investigate the potential of nebivolol in preventing doxorubicin-induced cardiotoxicity by targeting the inflammatory, oxidative, and apoptotic pathways. Twenty-eight male rats were randomly divided into four groups, each consisting of seven rats. The control group received standard diets and unrestricted access to water. The rats in the normal saline (N/S) group were administered a 0.9% normal saline solution for two weeks. The doxorubicin group (the "induced group") received doxorubicin at a dosage of 2.5 mg/kg three times per week for two weeks. The nebivolol group received an oral dose of 4 mg/kg of nebivolol for the same duration. The cardiac tissues of rats treated with doxorubicin exhibited increased levels of tumor necrosis factor, interleukin-1, malondialdehyde, and caspase-3 compared to the normal saline control group (p<0.05), along with decreased levels of total antioxidant capacity and Bcl-2. These results show that doxorubicin is harmful to the heart. The administration of nebivolol significantly reduced the cardiotoxic effects induced by doxorubicin, as indicated by a statistically significant decrease in the levels of inflammatory markers, specifically tumor necrosis factor-alpha (TNF-α) and interleukin-1 beta (IL-1β) (p<0.05). The nebivolol group exhibited a significant decrease in malondialdehyde levels, which serves as a signal of oxidation, in cardiac tissue compared to the doxorubicin-only group (p<0.05). Additionally, the nebivolol group showed a significant increase in overall antioxidant capacity. Nebivolol dramatically attenuated doxorubicin-induced cardiotoxicity in rats, likely by interfering with oxidative stress, the inflammatory response, and the apoptotic pathway.
Collapse
Affiliation(s)
| | - Ali Janabi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Kufa, Najaf, Iraq
| | - Najah Rayish Hadi
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, University of Kufa, Najaf, Iraq
| |
Collapse
|
7
|
Abdelrazik E, Hassan HM, Hamza E, Ezz Elregal FM, Elnagdy MH, Abdulhai EA. Beneficial role of rosemary extract on oxidative stress-mediated neuronal apoptosis in rotenone-induced attention deficit hyperactivity disease in juvenile rat model. ACTA BIO-MEDICA : ATENEI PARMENSIS 2023; 94:e2023104. [PMID: 37326266 PMCID: PMC10308472 DOI: 10.23750/abm.v94i3.14260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 02/23/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND AND AIM Attention deficit hyperactivity disorder (ADHD) is heterogeneous neurobehavioral disorders that co-exist with cognitive and learning deficits affecting 3-7% of children. We study the role of rosemary in the protection of the prefrontal cortical neurons against rotenone-induced ADHD in juvenile rats. METHODS Twenty-four juvenile rats were divided into four groups (n=6): control group, received olive oil 0.5 ml/kg/day/ I.P. for 4 weeks, rosemary group received rosemary 75 mg/kg/day/ I.P. for 4 weeks, rotenone group received rotenone 1 mg/kg/day/ I.P. dissolved in olive oil for 4 days and combined group received rotenone 1 mg/kg/day/ I.P. for 4 days and rosemary 75 mg/kg/day/ I.P. for 4 weeks. RESULTS Rotenone group showed higher impulsivity with reduction in the recognition index and total locomotor activity. However, combined group showed significant improvement in the recognition index and the total locomotor activity. Neurochemical analysis disclosed that rotenone decreased levels of GSH and significantly increased lipid peroxidation and oxidative stress. The administration of rosemary amended these neurochemical changes. Rotenone caused a significant increase in serum amyloid protein A and C-reactive protein levels indicating a marked state of inflammation. Rosemary ameliorated these biochemical changes. The immunohistochemical expression of tyrosine hydroxylase was decreased in the rotenone group. On the other hand, caspase-3 was increased in the rotenone group. PCR confirmed immunohistochemical results for gene expression. CONCLUSIONS The findings of the behavioral, neurochemical, biochemical, immunohistochemical and molecular outcomes suggested that rosemary could fight oxidative stress, inflammation and apoptosis in the prefrontal cortex of rotenone-induced ADHD in juvenile rats.
Collapse
Affiliation(s)
- Eman Abdelrazik
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt. .
| | - Hend M Hassan
- Department of Human Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt. .
| | - Eman Hamza
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt/ Department of Biochemistry and Molecular Biology, Horus University, Damietta, Egypt..
| | - Farah M Ezz Elregal
- Medical Student, Faculty of Medicine, Mansoura University, Mansoura, Egypt. .
| | - Marwa H Elnagdy
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt..
| | - Eman A Abdulhai
- Department of Pediatrics (pediatric neurology), Faculty of Medicine, Mansoura University, Mansoura, Egypt. .
| |
Collapse
|
8
|
Shi S, Chen Y, Luo Z, Nie G, Dai Y. Role of oxidative stress and inflammation-related signaling pathways in doxorubicin-induced cardiomyopathy. Cell Commun Signal 2023; 21:61. [PMID: 36918950 PMCID: PMC10012797 DOI: 10.1186/s12964-023-01077-5] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 02/12/2023] [Indexed: 03/16/2023] Open
Abstract
Doxorubicin (DOX) is a powerful and commonly used chemotherapeutic drug, used alone or in combination in a variety of cancers, while it has been found to cause serious cardiac side effects in clinical application. More and more researchers are trying to explore the molecular mechanisms of DOX-induced cardiomyopathy (DIC), in which oxidative stress and inflammation are considered to play a significant role. This review summarizes signaling pathways related to oxidative stress and inflammation in DIC and compounds that exert cardioprotective effects by acting on relevant signaling pathways, including the role of Nrf2/Keap1/ARE, Sirt1/p66Shc, Sirt1/PPAR/PGC-1α signaling pathways and NOS, NOX, Fe2+ signaling in oxidative stress, as well as the role of NLRP3/caspase-1/GSDMD, HMGB1/TLR4/MAPKs/NF-κB, mTOR/TFEB/NF-κB pathways in DOX-induced inflammation. Hence, we attempt to explain the mechanisms of DIC in terms of oxidative stress and inflammation, and to provide a theoretical basis or new idea for further drug research on reducing DIC. Video Abstract.
Collapse
Affiliation(s)
- Saixian Shi
- Department of Pharmacy, Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Luzhou, 646000, Sichuan Province, China.,School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Ye Chen
- Department of Pharmacy, Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Luzhou, 646000, Sichuan Province, China.,School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Zhijian Luo
- Department of Ultrasound, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Guojun Nie
- The First Outpatient Department of People's Liberation Army Western Theater General Hospital, Chengdu, 610000, Sichuan Province, China
| | - Yan Dai
- Department of Pharmacy, Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Luzhou, 646000, Sichuan Province, China.
| |
Collapse
|
9
|
Kuşçu GC, Gürel Ç, Buhur A, Karabay Yavaşoğlu NÜ, Köse T, Yavaşoğlu A, Oltulu F. Fluvastatin alleviates doxorubicin-induced cardiac and renal toxicity in rats via regulation of oxidative stress, inflammation, and apoptosis associated genes expressions. Drug Chem Toxicol 2023; 46:400-411. [PMID: 35209778 DOI: 10.1080/01480545.2022.2043351] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Doxorubicin (DOXO) is a cytostatic agent used in the chemotherapy protocol of several cancers for more than 40 years, but usage of this drug in cancer treatment has been limited due to severe renal and cardiac tissue toxicities that may result in death in patients. Fluvastatin (FV) is a fully synthetic hydroxymethyl glutaryl coenzyme A (HMG-CoA) reductase inhibitor used as a cholesterol-lowering agent in patients with hypercholesterolemia. Previous studies revealed that FV also exhibits antioxidant, anti-inflammatory, and antitumor activity. Additionally, our previous study indicated that FV exerts a prophylactic effect on DOXO-induced testicular toxicity by preventing lipid peroxidation, supporting the antioxidant system, and regulating the blood-testis barrier-associated genes expression. Herein, we purposed to evaluate the possible therapeutic and the protective effects of FV on the DOXO-induced cardiac and renal toxicitiy model by histochemical, immunohistochemical, biochemical, and real-time polymerase chain reaction (real-time PCR) analyses. Results point out protective use of FV exerts a beneficial effect by repressing lipid peroxidation and by regulating the inducible nitric oxide synthase (iNOS), nitric oxide synthase endothelial (eNOS), nuclear factor kappa-B (NF-κB), and Caspase-3 (Casp3) protein and mRNA expressions, which play an important role in mediating DOXO-induced renal and cardiac toxicity mechanisms. In conclusion, FV may be a candidate agent for the prevention of renal and cardiac toxicities in cancer patients receiving DOXO chemotherapy.
Collapse
Affiliation(s)
- Gökçe Ceren Kuşçu
- Department of Histology and Embryology, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Çevik Gürel
- Department of Histology and Embryology, Faculty of Medicine, Ege University, Izmir, Turkey.,Department of Histology and Embryology, Faculty of Medicine, Harran University, Şanlıurfa, Turkey
| | - Aylin Buhur
- Department of Histology and Embryology, Faculty of Medicine, Ege University, Izmir, Turkey
| | | | - Timur Köse
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Altuğ Yavaşoğlu
- Department of Histology and Embryology, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Fatih Oltulu
- Department of Histology and Embryology, Faculty of Medicine, Ege University, Izmir, Turkey
| |
Collapse
|
10
|
Morelli MB, Bongiovanni C, Da Pra S, Miano C, Sacchi F, Lauriola M, D’Uva G. Cardiotoxicity of Anticancer Drugs: Molecular Mechanisms and Strategies for Cardioprotection. Front Cardiovasc Med 2022; 9:847012. [PMID: 35497981 PMCID: PMC9051244 DOI: 10.3389/fcvm.2022.847012] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/03/2022] [Indexed: 12/13/2022] Open
Abstract
Chemotherapy and targeted therapies have significantly improved the prognosis of oncology patients. However, these antineoplastic treatments may also induce adverse cardiovascular effects, which may lead to acute or delayed onset of cardiac dysfunction. These common cardiovascular complications, commonly referred to as cardiotoxicity, not only may require the modification, suspension, or withdrawal of life-saving antineoplastic therapies, with the risk of reducing their efficacy, but can also strongly impact the quality of life and overall survival, regardless of the oncological prognosis. The onset of cardiotoxicity may depend on the class, dose, route, and duration of administration of anticancer drugs, as well as on individual risk factors. Importantly, the cardiotoxic side effects may be reversible, if cardiac function is restored upon discontinuation of the therapy, or irreversible, characterized by injury and loss of cardiac muscle cells. Subclinical myocardial dysfunction induced by anticancer therapies may also subsequently evolve in symptomatic congestive heart failure. Hence, there is an urgent need for cardioprotective therapies to reduce the clinical and subclinical cardiotoxicity onset and progression and to limit the acute or chronic manifestation of cardiac damages. In this review, we summarize the knowledge regarding the cellular and molecular mechanisms contributing to the onset of cardiotoxicity associated with common classes of chemotherapy and targeted therapy drugs. Furthermore, we describe and discuss current and potential strategies to cope with the cardiotoxic side effects as well as cardioprotective preventive approaches that may be useful to flank anticancer therapies.
Collapse
Affiliation(s)
| | - Chiara Bongiovanni
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Silvia Da Pra
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Carmen Miano
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
| | - Francesca Sacchi
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Mattia Lauriola
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Gabriele D’Uva
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
- *Correspondence: Gabriele D’Uva,
| |
Collapse
|
11
|
Osipova OA, Gosteva EV, Belousova ON, Gorelik SG, Klyushnikov NI, Lykov YA, Khachaturov AN, Bukatov VV. Dynamics of fibrotic and vascular endothelial dysfunction markers in elderly hypertensive patients after ischemic stroke receiving beta-blockers. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2022. [DOI: 10.15829/1728-8800-2021-3087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Aim. To compare the effect of beta-blocker therapy (bisoprolol and nebivolol) on the dynamics of fibrotic and vascular endothelial dysfunction markers in elderly hypertensive patients after ischemic stroke (IS).Material and methods. This prospective cohort study included 75 hypertensive patients who were admitted to the hospital due to IS. The mean age of patients was 67±6 years. The average National Institutes of Health Stroke Scale (NIHSS) score was 7±3. The followup period was 6 months. The control group consisted of 20 elderly people with hypertension without prior myocardial infarction. The patients were divided into groups based on received therapy: group 1 (n=38) — bisoprolol; group 2 (n=37) — nebivolol. The level of matrix metalloproteinase-9 (MMP-9) and tissue inhibitor of metalloproteinase-1 (TIMP-1) was determined by enzyme-linked immunosorbent assay (ELISAKit, USA). Vascular ultrasound was carried out using a LOGIQP9 (GE) system according to the Celermajer method.Results. After 6-month nebivolol, we revealed a decrease in the level of MMP-9 by 30,2% (p<0,01), TIMP-1 by 15,6% (p<0,05). After 6-month bisoprolol therapy, the level of MMP-9 decreased by 14,5% (p<0,05), while TIMP-1 did not change. Intergroup comparison found that when using nebivolol, there was a higher decrease in the level of MMP-9 by 15,7% (p<0,05), TIMP-1 by 9,7% (p<0,05), MMP-9/TIMP-1 by 7,8% (p<0,05) than with bisoprolol therapy. After 6-month bisoprolol therapy, there was a decrease in the proportion of patients with severe endothelial dysfunction (ED) by 7,9% (p<0,05). Two patients from the nebivolol group moved into mild ED category. The number of patients with moderate ED increased by 19% (p<0,01), while prevalence of severe ED decreased by 24,4% (p<0,01).Conclusion. The results obtained indicate that the beta-blocker nebivolol at an average dose of 8,55+1,75 mg/day significantly reduces the vascular fibrosis, normalizes the ratio of collagen synthesis and degradation markers, improves the vasodilation brachial artery properties in comparison with bisoprolol in elderly hypertensive patients after IS.
Collapse
Affiliation(s)
| | | | | | | | - N. I. Klyushnikov
- Belgorod National Research University; Voronezh City Clinical Emergency Hospital № 8
| | | | | | | |
Collapse
|
12
|
Wróbel AF, Stępniak A, Bańczerowska-Górska M, Stangel-Wójcikiewicz K, Czuczwar P. The efficacy of nebivolol on spontaneously hypertensive rats with overactive bladder - an experimental study. Arch Med Sci 2022; 18:1342-1350. [PMID: 36160351 PMCID: PMC9479596 DOI: 10.5114/aoms.2019.88279] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 05/03/2019] [Indexed: 11/21/2022] Open
Abstract
INTRODUCTION Overactive bladder (OAB) is a disease that significantly decreases quality of life. Adverse effects of the drugs currently used in OAB treatment limit their use in patients with hypertension. Nebivolol (a β1-adrenergic receptor blocker) is approved for hypertension treatment, but also shows agonistic activity on β3-adrenergic receptor, located in the urinary bladder. The aim of this study was to assess the impact of nebivolol on OAB symptoms and cardiovascular parameters in spontaneously hypertensive rats (SHR) - an animal model of OAB and hypertension. MATERIAL AND METHODS Female Wistar-Kyoto (WHY) and SHR rats were used in the experiments. Nebivolol was administered intra-arterially at a single daily dose of 0.05 mg/kg for 14 days. Subsequently, cystometry and bladder blood flow assessment were performed. Then, 24-hour measurement of heart rate, blood pressure, and urine production was carried out. In addition, the bladders of experimental rats were removed and processed to conduct biochemical analyses. RESULTS The main finding of this study is that the treatment with nebivolol resulted in an improvement of cystometric parameters characteristic for OAB in SHR. Nebivolol normalised blood pressure in SHR, while in WHY the cardiovascular parameters remained unchanged. Biomarkers characteristic for OAB were elevated in SHR compared to WHY, and nebivolol decreased their values in SHR while it had no influence on WHY. CONCLUSIONS Nebivolol alleviates OAB symptoms and normalises blood pressure in SHRs. These results suggest that nebivolol may be a useful treatment alternative for OAB patients with pre-existing hypertension.
Collapse
Affiliation(s)
- Andrzej F. Wróbel
- 2 Chair and Department of Gynecology, Medical University of Lublin, Lublin, Poland
| | - Anna Stępniak
- 3 Chair and Department of Gynecology, Medical University of Lublin, Lublin, Poland
| | | | | | - Piotr Czuczwar
- 3 Chair and Department of Gynecology, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
13
|
Osipova OA, Gosteva EV, Golivets TP, Belousova ON, Zemlyansky OA, Pokrovsky MV, Golovin AI, Grigorenko NV, Merezhko AA. Changes of myocardial fibrosis markers with the use of beta-blockers and mineralocorticoid receptor antagonists in patients with heart failure with mid-range ejection fraction of ischemic origin. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2021. [DOI: 10.15829/1728-8800-2021-3068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Aim. To compare the effect of 12-month pharmacotherapy with a betablocker (BB) (bisoprolol and nebivolol) and a combination of BB with a mineralocorticoid receptor antagonist (bisoprolol+eplerenone, nebivolol+eplerenone) on following fibrosis markers: matrix metalloproteinases 1 and 9 (MMP-1, MMP-9) and tissue inhibitor of MMP-1 (TIMP-1) in patients with heart failure with mid-range ejection fraction (HFmrEF) of ischemic origin.Material and methods. The study included 135 patients, including 40 (29,6%) women and 95 (70,4%) men aged 45-60 years (mean age, 53,1±5,7 years). Patients were randomized into subgroups based on pharmacotherapy with BB (bisoprolol or nebivolol) and their combination with eplerenone. The enzyme-linked immunosorbent assay was used to determine the level of MMP-1, MMP-9, TIMP-1 (ng/ml) using the commercial test system “MMP-1 ELISA”, “MMP-9 ELISA”, “Human TIMP-1 ELISA” (“Bender Medsystems “, Austria).Results. In patients with HFmrEF of ischemic origin, there were following downward changes in serum level of myocardial fibrosis markers, depending on the therapy: bisoprolol — MMP-1 decreased by 35% (p<0,01), MMP-9 — by 56,3% (p<0,001), TIMP-1 — by 17,9% (p<0,01); nebivolol — MMP-1 decreased by 45% (p<0,001), MMP-9 — by 57,1% (p<0,001), TIMP-1 — by 30,1% (p<0,01); combination of bisoprolol with eplerenone — MMP-1 decreased by 43% (p<0,001), MMP-9 — by 51,2% (p<0,001), TIMP-1 — by 25,1% (p<0,01); combination of nebivolol with eplerenone — MMP-1 decreased by 53% (p<0,001), MMP-9 — by 64,3% (p<0,001), TIMP-1 — by 39% (p<0,01). In patients with NYHA class I HFmrEF after 12-month therapy, the decrease in MMP-1 level was 39,9% (p<0,01), MMP-9 — 57,5% (p<0,001). In class II, the decrease in MMP-1 level was 47% (p<0,001), MMP-9 — 49,7% (p<0,001). A significant decrease in TIMP-1 level was revealed in patients with class I by 29% (p<0,01), in patients with class II by 27,1% (p<0,01) compared with the initial data.Conclusion. A significant decrease in the levels of myocardial fibrosis markers (MMP-1, MMP-9, TIMP-1) was demonstrated in patients with HFmrEF of ischemic origin receiving long-term pharmacotherapy. The most pronounced effect was determined in patients with NYHA class I HF.
Collapse
|
14
|
Li L, Zhang Q, Li J, Tian Y, Kang Y, Ren G, Liu W, Wang H, Wang B, Yan L, Guo L, Diao H. Targeted Delivery of Doxorubicin Using Transferrin-Conjugated Carbon Dots for Cancer Therapy. ACS APPLIED BIO MATERIALS 2021; 4:7280-7289. [PMID: 35006957 DOI: 10.1021/acsabm.1c00811] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A transferrin receptor (TfR)-targeted nanodrug [green fluorescence emission carbon dot (GCD)-polyethylene glycol (PEG)-transferrin (Tf)@doxorubicin (Dox)] for cancer therapy was developed by functionalizing GCDs with PEG, Tf, and Dox. GCDs were synthesized by the one-step hydrothermal method, followed by conjugating PEG and Tf by covalent bonds and loading Dox by electrostatic interactions. The nanodrug exhibits high stability under neutral conditions and effectively releases Dox at pH of 5.5. GCD-PEG-Tf@Dox can be selectively internalized by TfR-overexpressed tumor cells (MCF-7 and K150) via receptor-mediated endocytosis and further release Dox to the nuclei. As a result, GCD-PEG-Tf@Dox exhibits significant lethality to tumor cells (MCF-7 and K150) but greatly reduced toxicity to normal cells [Chinese hamster ovary cell line (CHO)] compared with free Dox. In vivo studies have confirmed that GCD-PEG-Tf@Dox can effectively inhibit tumor proliferation with negligible side effects.
Collapse
Affiliation(s)
- Lihong Li
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, PR China.,College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, PR China.,Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, Taiyuan 030001, PR China
| | - Qi Zhang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, PR China
| | - Jinyao Li
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, PR China
| | - Yafei Tian
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, PR China
| | - Yu Kang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, PR China
| | - Guodong Ren
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, PR China
| | - Wen Liu
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, PR China.,Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, Taiyuan 030001, PR China
| | - Haojiang Wang
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, PR China
| | - Bin Wang
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, PR China
| | - Lili Yan
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, PR China
| | - Lixia Guo
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, PR China
| | - Haipeng Diao
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, PR China.,College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, PR China.,Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, Taiyuan 030001, PR China
| |
Collapse
|
15
|
Modulation of vigabatrin induced cerebellar injury: the role of caspase-3 and RIPK1/RIPK3-regulated cell death pathways. J Mol Histol 2021; 52:781-798. [PMID: 34046766 DOI: 10.1007/s10735-021-09984-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 05/24/2021] [Indexed: 01/11/2023]
Abstract
Vigabatrin is the drug of choice in resistant epilepsy and infantile spasms. Ataxia, tremors, and abnormal gait have been frequently reported following its use indicating cerebellar involvement. This study aimed, for the first time, to investigate the involvement of necroptosis and apoptosis in the VG-induced cerebellar cell loss and the possible protective role of combined omega-3 and vitamin B12 supplementation. Fifty Sprague-Dawley adult male rats (160-200 g) were divided into equal five groups: the control group received normal saline, VG200 and VG400 groups received VG (200 mg or 400 mg/kg, respectively), VG200 + OB and VG400 + OB groups received combined VG (200 mg or 400 mg/kg, respectively), vitamin B12 (1 mg/kg), and omega-3 (1 g/kg). All medications were given daily by gavage for four weeks. Histopathological changes were examined in H&E and luxol fast blue (LFB) stained sections. Immunohistochemical staining for caspase-3 and receptor-interacting serine/threonine-protein kinase-1 (RIPK1) as well as quantitative real-time polymerase chain reaction (qRT-PCR) for myelin basic protein (MBP), caspase-3, and receptor-interacting serine/threonine-protein kinase-3 (RIPK3) genes were performed. VG caused a decrease in the granular layer thickness and Purkinje cell number, vacuolations, demyelination, suppression of MBP gene expression, and induction of caspases-3, RIPK1, and RIPK3 in a dose-related manner. Combined supplementation with B12 and omega-3 improved the cerebellar histology, increased MBP, and decreased apoptotic and necroptotic markers. In conclusion, VG-induced neuronal cell loss is dose-dependent and related to both apoptosis and necroptosis. This could either be ameliorated (in low-dose VG) or reduced (in high-dose VG) by combined supplementation with B12 and omega-3.
Collapse
|
16
|
Erfan OS, Sonpol HMA, Abd El-Kader M. Protective effect of rapamycin against acrylamide-induced hepatotoxicity: The associations between autophagy, apoptosis, and necroptosis. Anat Rec (Hoboken) 2021; 304:1984-1998. [PMID: 33480149 DOI: 10.1002/ar.24587] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 12/21/2020] [Accepted: 12/25/2020] [Indexed: 12/28/2022]
Abstract
Acrylamide (ACRL) was demonstrated to induce hepatotoxicity and programmed cell death (PCD). Rapamycin (RAPA)-induced autophagy had been reported to limit the progression of hepatocellular injury in experimental models. This research was designed to study two death pathways involved in ACRL-induced hepatotoxicity and the modulating effect of RAPA on the resulting hepatic injury. Thirty-six adult male rats were divided into three groups: control group, ACRL-treated group (20 mg kg/day), and the last group co-treated with ACRL plus RAPA (0.5 mg kg/day). Drugs were administered for 21 days via oral gavage. Blood samples were collected to assess alanine aminotransferase (ALT) and aspartate aminotransferase (AST). Livers were dissected; parts were used for detection of superoxide dismutase (SOD) and malondialdehyde (MDA) tissue levels. Other parts were processed for hematoxylin and eosin, Masson's trichrome staining, immunostaining for microtubule-associated proteins 1A/1B light chain 3B (LC3), ubiquitin-binding protein (p62), caspase-3, and receptor-interacting protein kinase 1 (RIPK1). ACRL induced a significant elevation in ALT, AST, MDA levels, and reduction in the SOD level. ACRL also induced hepatocellular injury, fibrosis, and defective autophagy indicated by elevation of LC3 and p62 and increased p62/LC3 ratio. Moreover, it increased the apoptotic (caspase-3) and necroptotic (RIPK1) markers expression. RAPA significantly reduced liver enzymes, oxidative stress, fibrosis, and improved liver histology. Moreover, RAPA decreased p62/LC3 ratio indicated enhanced autophagy, and significantly reduced caspase-3 and RIPK1 expression. In conclusion, RAPA maintained autophagic activity which may save the hepatocytes from PCD and enhance cell viability.
Collapse
Affiliation(s)
- Omnia S Erfan
- Anatomy and embryology department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Hany M A Sonpol
- Anatomy and embryology department, Faculty of Medicine, Mansoura University, Mansoura, Egypt.,Basic medical sciences department, College of Medicine, University of Bisha, Bisha, Saudi Arabia
| | - Marwa Abd El-Kader
- Anatomy and embryology department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
17
|
Owumi SE, Nwozo SO, Arunsi UO, Oyelere AK, Odunola OA. Co-administration of Luteolin mitigated toxicity in rats' lungs associated with doxorubicin treatment. Toxicol Appl Pharmacol 2021; 411:115380. [PMID: 33358696 DOI: 10.1016/j.taap.2020.115380] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/11/2020] [Accepted: 12/17/2020] [Indexed: 02/06/2023]
Abstract
Doxorubicin (DOX), is a drug against lung malignancies with undesirable side effect including oxidative, inflammatory and apoptotic effects. Luteolin (LUT), present in fruits and vegetables is pharmacologically active against oxido-inflammatory and apoptotic responses. The present study examined the effect of LUT on DOX-induced lungs and blood dysfunction in Wistars rat (sex: male; 10 weeks old, 160 ± 5 g). Randomly grouped (n = 10) rats were treated as follows: control, LUT alone (100 mg/kg; per os), DOX (2 mg/kg; i. p), and co-treated rats with LUT (50 or 100 mg/kg) and DOX for two consecutive weeks. DOX alone adversely altered the final body and relative organ weights, red and white blood cell and platelet counts. DOX significantly (p > 0.05) reduced lungs antioxidant capacity, and anti-inflammatory cytokines; increased biomarkers of oxidative stress, caspase-3 activity, and pro-inflammatory cytokine. Morphological damages accompanied these biochemical alterations in the lung of experimental rats. Co-treatment with LUT, dose-dependently reversed DOX-mediated changes in rats' survival, toxic responses, and diminished oxidative stress in rat's lungs. Furthermore, co-treatment with LUT resulted in the reduction of pro-inflammatory cytokines and apoptotic biomarkers, increased red and white blood cell, platelet counts and abated pathological injuries in rat lungs treated with DOX alone. In essence, our findings indicate that LUT dose-dependently mitigated DOX-induced toxicities in the lungs and haematopoietic systems. Supplementation of patients on DOX-chemotherapy with phytochemicals exhibiting antioxidant activities, specifically LUT, could circumvent the onset of unintended toxic responses in the lungs and haematopoietic system exposed to DOX.
Collapse
Affiliation(s)
- Solomon E Owumi
- Cancer Research and Molecular Biology Laboratories, University of Ibadan, Ibadan, Nigeria.
| | - Sarah O Nwozo
- Nutrition and Industrial Biochemistry Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, Nigeria
| | - Uche O Arunsi
- Cancer Immunology and Biotechnology Center, The University of Nottingham, Nottingham NG8 1AF, UK
| | - Adegboyega K Oyelere
- School of Chemistry & Biochemistry, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA
| | - Oyeronke A Odunola
- Cancer Research and Molecular Biology Laboratories, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
18
|
Colak S, Gurlek B, Topcu A, Tumkaya L, Mercantepe T, Yilmaz A. Protective effects of nebivolol on ovarian ischemia-reperfusion injury in rat. J Obstet Gynaecol Res 2020; 46:2407-2416. [PMID: 32961615 DOI: 10.1111/jog.14503] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 07/14/2020] [Accepted: 08/29/2020] [Indexed: 12/12/2022]
Abstract
AIM Ovarian torsion is a common gynecological emergency of reproductive ages, occurring at rates of 2.7-7.4%. This study aimed to evaluate the antioxidant effects of Nebivolol (NEB) and histopathological changes in experimental ischemic (I) and ischemic-reperfusion (I/R) injury in rat ovaries. METHODS Forty-eight adult female rats were randomly separated into six groups as group 1 (control) receiving an oral saline solution for 3 days; group 2 (I) that underwent ischemia for 3 h with the application of atraumatic vascular clips; group 3 (I/R); group 4 (I + NEB) receiving 10 mg/kg NEB by oral gavage 30 min prior to the ischemia induction; group 5 (I/R + NEB) receiving 10 mg/kg NEB, and group 6 (control + NEB) receiving oral 10 mg/kg NEB for 3 days before ischemia induction followed by consequent reperfusion. Ovarian tissue damage was scored by histopathological analysis. Ovarian tissue malondialdehyde (MDA) and glutathione (GSH) levels were measured biochemically. RESULTS The levels of MDA and tumor necrosis factor-alpha (TNF-α), and TUNEL assay positivity scores increased in the I and I/R groups. GSH levels decreased in all case groups (P < 0.05). The oral administration of NEB (10 mg/kg) to the I- and I/R-groups reduced the levels of MDA and TNF-α and TUNEL assay immunopositivity scores (P < 0.05). GSH levels increased in the treatment groups. CONCLUSION The current experimental ovarian torsion study suggests a protective role for NEB against I and I/R injury in rat ovaries. NEB may be a novel agent for decreasing ovarian I/R injury.
Collapse
Affiliation(s)
- Sabri Colak
- Department of Obstetrics and Gynecology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Beril Gurlek
- Department of Obstetrics and Gynecology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Atilla Topcu
- Department of Pharmacology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Levent Tumkaya
- Department of Histology and Embryology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Tolga Mercantepe
- Department of Histology and Embryology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Adnan Yilmaz
- Department of Medical Biochemistry, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| |
Collapse
|
19
|
Tedesco L, Rossi F, Ragni M, Ruocco C, Brunetti D, Carruba MO, Torrente Y, Valerio A, Nisoli E. A Special Amino-Acid Formula Tailored to Boosting Cell Respiration Prevents Mitochondrial Dysfunction and Oxidative Stress Caused by Doxorubicin in Mouse Cardiomyocytes. Nutrients 2020; 12:nu12020282. [PMID: 31973180 PMCID: PMC7071384 DOI: 10.3390/nu12020282] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/23/2019] [Accepted: 01/02/2020] [Indexed: 12/24/2022] Open
Abstract
Anthracycline anticancer drugs, such as doxorubicin (DOX), can induce cardiotoxicity supposed to be related to mitochondrial damage. We have recently demonstrated that a branched-chain amino acid (BCAA)-enriched mixture (BCAAem), supplemented with drinking water to middle-aged mice, was able to promote mitochondrial biogenesis in cardiac and skeletal muscle. To maximally favor and increase oxidative metabolism and mitochondrial function, here we tested a new original formula, composed of essential amino acids, tricarboxylic acid cycle precursors and co-factors (named α5), in HL-1 cardiomyocytes and mice treated with DOX. We measured mitochondrial biogenesis, oxidative stress, and BCAA catabolic pathway. Moreover, the molecular relevance of endothelial nitric oxide synthase (eNOS) and mechanistic/mammalian target of rapamycin complex 1 (mTORC1) was studied in both cardiac tissue and HL-1 cardiomyocytes. Finally, the role of Krüppel-like factor 15 (KLF15), a critical transcriptional regulator of BCAA oxidation and eNOS-mTORC1 signal, was investigated. Our results demonstrate that the α5 mixture prevents the DOX-dependent mitochondrial damage and oxidative stress better than the previous BCAAem, implying a KLF15/eNOS/mTORC1 signaling axis. These results could be relevant for the prevention of cardiotoxicity in the DOX-treated patients.
Collapse
Affiliation(s)
- Laura Tedesco
- Center for Study and Research on Obesity, Department of Medical Biotechnology and Translational Medicine, University of Milan, 20129 Milan, Italy
| | - Fabio Rossi
- Center for Study and Research on Obesity, Department of Medical Biotechnology and Translational Medicine, University of Milan, 20129 Milan, Italy
| | - Maurizio Ragni
- Center for Study and Research on Obesity, Department of Medical Biotechnology and Translational Medicine, University of Milan, 20129 Milan, Italy
| | - Chiara Ruocco
- Center for Study and Research on Obesity, Department of Medical Biotechnology and Translational Medicine, University of Milan, 20129 Milan, Italy
| | - Dario Brunetti
- Center for Study and Research on Obesity, Department of Medical Biotechnology and Translational Medicine, University of Milan, 20129 Milan, Italy
| | - Michele O. Carruba
- Center for Study and Research on Obesity, Department of Medical Biotechnology and Translational Medicine, University of Milan, 20129 Milan, Italy
| | - Yvan Torrente
- Department of Pathophysiology and Transplantation, University of Milan, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, 20122 Milan, Italy
| | - Alessandra Valerio
- Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy
- Correspondence: (A.V.); (E.N.); Tel.: +39-030-3717504 (A.V.); +39-02-50316956 (E.N.); Fax: +39-030-3717529 (A.V.); +39-02-50317118 (E.N.)
| | - Enzo Nisoli
- Center for Study and Research on Obesity, Department of Medical Biotechnology and Translational Medicine, University of Milan, 20129 Milan, Italy
- Correspondence: (A.V.); (E.N.); Tel.: +39-030-3717504 (A.V.); +39-02-50316956 (E.N.); Fax: +39-030-3717529 (A.V.); +39-02-50317118 (E.N.)
| |
Collapse
|
20
|
Gothandam K, Gomathinayagam S. Amelioration of doxorubicin-induced cardiotoxicity by oral lyophilized Dunaliella salina supplement in a female wistar rat model with active estrous cycle and surgical menopause. Pharmacogn Mag 2020. [DOI: 10.4103/pm.pm_52_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
|
21
|
Li J, Liu X, Wang W, Li C. miR-133a-3p promotes apoptosis and induces cell cycle arrest by targeting CREB1 in retinoblastoma. Arch Med Sci 2020; 16:941-956. [PMID: 32542098 PMCID: PMC7286343 DOI: 10.5114/aoms.2019.86901] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 12/25/2018] [Indexed: 12/29/2022] Open
Abstract
INTRODUCTION Retinoblastoma (RB) is a malignant tumor that is derived from photoreceptors. It is common in children under 3 years old with a family genetic predisposition. MicroRNA-133a-3p (miR-133a-3p) is one of the tumor-related miRNAs that interprets a critical function in the genesis and development of various tumors. This study investigated the effects and underlying mechanisms of miR-133a-3p in RB. MATERIAL AND METHODS Quantitative reverse-transcription polymerase chain reaction (qRT-PCR) analysis was used to assess the miR-133a-3p expression in RB tissues and a cell model. MTT assay, western blot, flow cytometry and luciferase reporter assay were performed to evaluate the effect of miR-133a-3p on cell viability, apoptosis and the cell cycle. An RB xenograft model was established to assess the in vivo influence of miR-133a-3p on RB growth. RESULTS MiR-133a-3p level was reduced in RB tissues and the cell model (p < 0.01 or p < 0.001). Addition of miR-133a-3p reduced cell viability, and increased apoptosis and cell cycle arrest (p < 0.001). Additionally, CREB1 was identified to be the target of miR-133a-3p in RB cell lines (p < 0.001). Cell viability reduction, apoptosis and cell cycle arrest increases mediated by miR-133a-3p were attenuated by CREB1 overexpression (p < 0.001). MiR-133a-3p inhibited tumor growth of RB in vivo (p < 0.001). CONCLUSIONS Our results reveal that miR-133a-3p exhibits anti-cancer effects by targeting CREB1 in RB. This study provides a new direction for effective targeted treatment of this disease.
Collapse
Affiliation(s)
| | - Xiuming Liu
- Corresponding author: Xiuming Liu, Department of Ophthalmology the Affiliated Huai’an, No. 1 People’s Hospital of Nanjing Medical University, 1 Huanghe Road West, Huaiyin District, Huai’an, Jiangsu, 223300, China, Phone: +86 0517 80872120, Fax: +86 0517 80872120, E-mail:
| | | | | |
Collapse
|
22
|
Zhao S, Shao L, Wang Y, Meng Q, Yu J. Ketamine exhibits anti-gastric cancer activity via induction of apoptosis and attenuation of PI3K/Akt/mTOR. Arch Med Sci 2020; 16:1140-1149. [PMID: 32864003 PMCID: PMC7444715 DOI: 10.5114/aoms.2019.85146] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 01/27/2019] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION Gastric cancer (GC) is the most widespread type of cancer after lung and liver cancer in men and after breast cancer in women. Thus, the present study was intended to evaluate the effect of ketamine (KET) on gastric cancer cells. MATERIAL AND METHODS The effect of KET was analyzed in vitro by the MTT assay against human gastric cancer cell lines BGC-823, MKN-45 and MKN-28. The effect KET on apoptosis, cell migration and cell cycle arrest was also quantified. Western blot analysis was performed to estimate the effect of KET on apoptosis mediators and PI3K/AKT/mTOR pathway mediators. A mouse xenograft assay was also conducted to further confirm the anticancer activity. RESULTS KET causes reduction of cellular viability of BGC-823, MKN-45 and MKN-28, with a more significant effect against BGC-823 cells. The KET treatment showed a dose-dependent increase in apoptotic cells among BGC-823 cells. KET causes a significant dose-dependent decline in migration of treated cells. It causes induction of apoptosis mediated via the mitochondrial pathway, where it causes a decline in Bcl2 and mitochondrial cytochrome c level together with increase in expression of Bax, cytosolic cytochrome c and cytosolic apoptotic protease activating factor-1 (Apaf-1). The level of p-PI3K, p-mTOR, p-GSK3β and p-AKT was found to be downregulated in a dose-dependent manner in KET-treated cells. In a mouse xenograft model, KET causes a reduction in relative tumour volume and tumour weight. CONCLUSIONS Our results suggest that ketamine has the ability to inhibit progression of gastric cancer via induction of apoptosis and attenuation of PI3K/Akt/mTOR.
Collapse
Affiliation(s)
- Shiling Zhao
- Department of Anesthesiology, The Third People's Hospital of Dalian, Dalian, China
| | - Lin Shao
- Department of Anesthesiology, The Third People's Hospital of Dalian, Dalian, China
| | - Yingwei Wang
- Department of Anesthesiology, The Third People's Hospital of Dalian, Dalian, China
| | - Qingtao Meng
- Department of Spinal, Department of Anesthesiology, The Third People's Hospital of Dalian, Dalian, China
| | - Jinning Yu
- Department of General Surgery, The Third People's Hospital of Dalian, Dalian, China
| |
Collapse
|
23
|
Yoon CS, Kim HK, Mishchenko NP, Vasileva EA, Fedoreyev SA, Stonik VA, Han J. Spinochrome D Attenuates Doxorubicin-Induced Cardiomyocyte Death via Improving Glutathione Metabolism and Attenuating Oxidative Stress. Mar Drugs 2018; 17:md17010002. [PMID: 30577438 PMCID: PMC6356724 DOI: 10.3390/md17010002] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 12/13/2018] [Accepted: 12/15/2018] [Indexed: 12/15/2022] Open
Abstract
Doxorubicin, an anthracycline from Streptomyces peucetius, exhibits antitumor activity against various cancers. However, doxorubicin is cardiotoxic at cumulative doses, causing increases in intracellular reactive oxygen species in the heart. Spinochrome D (SpD) has a structure of 2,3,5,6,8-pentahydroxy-1,4-naphthoquinone and is a structural analogue of well-known sea urchin pigment echinochrome A. We previously reported that echinochrome A is cardioprotective against doxorubicin toxicity. In the present study, we assessed the cardioprotective effects of SpD against doxorubicin and determined the underlying mechanism. ¹H-NMR-based metabolomics and mass spectrometry-based proteomics were utilized to characterize the metabolites and proteins induced by SpD in a human cardiomyocyte cell line (AC16) and human breast cancer cell line (MCF-7). Multivariate analyses identified 12 discriminating metabolites (variable importance in projection > 1.0) and 1814 proteins from SpD-treated AC16 cells. Proteomics and metabolomics analyses showed that glutathione metabolism was significantly influenced by SpD treatment in AC16 cells. SpD treatment increased ATP production and the oxygen consumption rate in D-galactose-treated AC16 cells. SpD protected AC16 cells from doxorubicin cytotoxicity, but it did not affect the anticancer properties. With SpD treatment, the mitochondrial membrane potential and mitochondrial calcium localization were significantly different between cardiomyocytes and cancer cell lines. Our findings suggest that SpD could be cardioprotective against the cytotoxicity of doxorubicin.
Collapse
Affiliation(s)
- Chang Shin Yoon
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center (CMDC), Inje University, Busan 614-735, Korea.
| | - Hyoung Kyu Kim
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center (CMDC), Inje University, Busan 614-735, Korea.
| | - Natalia P Mishchenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, Vladivostok 690022, Russia.
| | - Elena A Vasileva
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, Vladivostok 690022, Russia.
| | - Sergey A Fedoreyev
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, Vladivostok 690022, Russia.
| | - Valentin A Stonik
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, Vladivostok 690022, Russia.
| | - Jin Han
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center (CMDC), Inje University, Busan 614-735, Korea.
| |
Collapse
|