1
|
Tonon CR, Pereira AG, Ferreira NF, Monte MG, Vieira NM, Fujimori ASS, Ballin PDS, de Paiva SAR, Zornoff LAM, Minicucci MF, Polegato BF. The Gut-Heart Axis and Its Role in Doxorubicin-Induced Cardiotoxicity: A Narrative Review. Microorganisms 2025; 13:855. [PMID: 40284691 PMCID: PMC12029146 DOI: 10.3390/microorganisms13040855] [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: 03/13/2025] [Revised: 04/04/2025] [Accepted: 04/06/2025] [Indexed: 04/29/2025] Open
Abstract
Doxorubicin is a widely used chemotherapy for the treatment of several types of cancer. However, its application is restricted due to adverse effects, particularly cardiotoxicity, which can progress to heart failure-a chronic and debilitating condition. Several mechanisms have been identified in the pathophysiology of doxorubicin-induced cardiotoxicity, including oxidative stress, mitochondrial dysfunction, inflammation, and disruption of collagen homeostasis. More recently, dysbiosis of the gut microbiota has been implicated in the development and perpetuation of cardiac injury. Studies have reported alterations in the composition and abundance of the microbiota during doxorubicin treatment. Therefore, as of recent, there is a new field of research in order to develop strategies involving the gut microbiota to prevent or attenuate cardiotoxicity since there is no effective therapy at the moment. This narrative review aims to provide an update on the role of gut microbiota and intestinal permeability in the pathophysiology of cardiovascular diseases, and more specifically doxorubicin-induced cardiotoxicity. Additionally, it seeks to establish a foundation for future research targeting gut microbiota to alleviate cardiotoxicity.
Collapse
Affiliation(s)
- Carolina Rodrigues Tonon
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, Brazil (B.F.P.)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2
|
Yang Y, Wang L, Yu L, Chang C, Zhang H, Hu L, Liu J, Zhang Y, Han H, Zhang H, Zhou Y, Wang J. Monocytes Expressing IL-36G Play a Crucial Role in Atopic Dermatitis. J Cell Mol Med 2025; 29:e70503. [PMID: 40159643 PMCID: PMC11955416 DOI: 10.1111/jcmm.70503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/06/2025] [Accepted: 03/11/2025] [Indexed: 04/02/2025] Open
Abstract
Atopic dermatitis (ad) is a chronic inflammatory skin disease, with recent studies indicating that immune cells, such as monocytes and inflammatory cytokines, play a crucial role. By retrieving datasets from public databases and analysing immune cell infiltration in lesional skin using CIBERSORT, we found that monocytes and M2 macrophages were significantly upregulated in atopic dermatitis. Differentially expressed gene (DEG) functional enrichment analysis revealed that cytokine-cytokine receptor interaction was the most significantly enriched pathway. Further analysis of cytokines and their receptors, along with their correlation with infiltrating immune cells, identified IL36G-expressing monocytes as a key target in atopic dermatitis. We compared immune cell infiltration and cytokine-related targets in similar inflammatory skin diseases, such as psoriasis and urticaria, to evaluate similarities and differences among these three skin conditions. The analysis revealed that IL36G-expressing monocytes were also highly expressed in psoriasis but did not play a pivotal role in urticaria. Finally, we used molecular docking to predict and validate drugs targeting IL36G. Our study highlights IL36G-expressing monocytes as a common key target in atopic dermatitis and psoriasis, offering novel insights and therapeutic strategies for these related diseases.
Collapse
Affiliation(s)
- Yitao Yang
- School of MedicineShanghai UniversityShanghaiChina
| | - Lei Wang
- Hubei Shizhen LaboratoryHubei University of Chinese MedicineWuhanChina
| | - Longmei Yu
- National Institute of TCM Constitution and Preventive MedicineBeijing University of Chinese MedicineBeijingChina
| | - Chenxi Chang
- National Institute of TCM Constitution and Preventive MedicineBeijing University of Chinese MedicineBeijingChina
| | - Honglei Zhang
- National Institute of TCM Constitution and Preventive MedicineBeijing University of Chinese MedicineBeijingChina
| | - Linhan Hu
- National Institute of TCM Constitution and Preventive MedicineBeijing University of Chinese MedicineBeijingChina
| | - Juntong Liu
- National Institute of TCM Constitution and Preventive MedicineBeijing University of Chinese MedicineBeijingChina
| | - Yihang Zhang
- National Institute of TCM Constitution and Preventive MedicineBeijing University of Chinese MedicineBeijingChina
| | - Hui Han
- National Institute of TCM Constitution and Preventive MedicineBeijing University of Chinese MedicineBeijingChina
| | - Haiyun Zhang
- School of MedicineShanghai UniversityShanghaiChina
| | - Yumei Zhou
- National Institute of TCM Constitution and Preventive MedicineBeijing University of Chinese MedicineBeijingChina
| | - Ji Wang
- National Institute of TCM Constitution and Preventive MedicineBeijing University of Chinese MedicineBeijingChina
| |
Collapse
|
3
|
Li Y, Liu X, Lin R, Peng X, Wang X, Meng F, Jin S, Lv W, Liu X, Du Z, Wen S, Bai R, Ruan Y, Zhou H, Zou R, Tang R, Liu N. Ibrutinib Promotes Atrial Fibrillation by Disrupting A-Kinase Anchoring Protein 1-Mediated Mitochondrial Quality Surveillance in Cardiomyocytes. RESEARCH (WASHINGTON, D.C.) 2024; 7:0509. [PMID: 39469220 PMCID: PMC11518619 DOI: 10.34133/research.0509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/20/2024] [Accepted: 09/29/2024] [Indexed: 10/30/2024]
Abstract
Background: Ibrutinib, a potent Bruton's tyrosine kinase inhibitor with marked efficacy against hematological malignancies, is associated with the heightened risk of atrial fibrillation (AF). Although ibrutinib-induced AF is linked to enhanced oxidative stress, the underlying mechanisms remain unclear. Objective: This research aimed to explore the molecular mechanism and regulatory target in ibrutinib-induced AF. Methods: We performed in vivo electrophysiology studies using ibrutinib-treated mice, and then employed proteomic and single-cell transcriptomic analyses to identify the underlying targets and mechanisms. The effects of A-kinase anchoring protein 1 (AKAP1) depletion on mitochondrial quality surveillance (MQS) were evaluated using both in vivo and ex vivo AKAP1 overexpression models. Results: Atrial AKAP1 expression was significantly reduced in ibrutinib-treated mice, leading to inducible AF, atrial fibrosis, and mitochondrial fragmentation. These pathological changes were effectively mitigated in an overexpression model of ibrutinib-treated mice injected with an adeno-associated virus carrying Akap1. In ibrutinib-treated atrial myocytes, AKAP1 down-regulation promoted dynamin-related protein 1 (DRP1) translocation into mitochondria by facilitating DRP1 dephosphorylation at Ser637, thereby mediating excessive mitochondrial fission. Impaired MQS was also suggested by defective mitochondrial respiration, mitochondrial metabolic reprogramming, and suppressed mitochondrial biogenesis, accompanied by excessive oxidative stress and inflammatory activation. The ibrutinib-mediated MQS disturbance can be markedly improved with the inducible expression of the AKAP1 lentiviral system. Conclusions: Our findings emphasize the key role of AKAP1-mediated MQS disruption in ibrutinib-induced AF, which explains the previously observed reactive oxygen species overproduction. Hence, AKAP1 activation can be employed to prevent and treat ibrutinib-induced AF.
Collapse
Affiliation(s)
- Yukun Li
- Department of Cardiology, Beijing Anzhen Hospital,
Capital Medical University, Beijing 100012, China
- National Clinical Research Center for Cardiovascular Diseases, Beijing 100012, China
| | - Xinmeng Liu
- Department of Cardiology, Beijing Anzhen Hospital,
Capital Medical University, Beijing 100012, China
- National Clinical Research Center for Cardiovascular Diseases, Beijing 100012, China
| | - Rong Lin
- Department of Cardiology, Beijing Anzhen Hospital,
Capital Medical University, Beijing 100012, China
- National Clinical Research Center for Cardiovascular Diseases, Beijing 100012, China
| | - Xiaodong Peng
- Department of Cardiology, Beijing Anzhen Hospital,
Capital Medical University, Beijing 100012, China
- National Clinical Research Center for Cardiovascular Diseases, Beijing 100012, China
| | - Xuesi Wang
- Department of Cardiology, Beijing Anzhen Hospital,
Capital Medical University, Beijing 100012, China
- National Clinical Research Center for Cardiovascular Diseases, Beijing 100012, China
| | - Fanchao Meng
- Department of Cardiology, Beijing Anzhen Hospital,
Capital Medical University, Beijing 100012, China
- National Clinical Research Center for Cardiovascular Diseases, Beijing 100012, China
| | - Shuqi Jin
- Department of Cardiology, Beijing Anzhen Hospital,
Capital Medical University, Beijing 100012, China
- National Clinical Research Center for Cardiovascular Diseases, Beijing 100012, China
| | - Wenhe Lv
- Department of Cardiology, Beijing Anzhen Hospital,
Capital Medical University, Beijing 100012, China
- National Clinical Research Center for Cardiovascular Diseases, Beijing 100012, China
| | - Xiaoying Liu
- Department of Cardiology, Beijing Anzhen Hospital,
Capital Medical University, Beijing 100012, China
- National Clinical Research Center for Cardiovascular Diseases, Beijing 100012, China
| | - Zhuohang Du
- Department of Cardiology, Beijing Anzhen Hospital,
Capital Medical University, Beijing 100012, China
- National Clinical Research Center for Cardiovascular Diseases, Beijing 100012, China
| | - Songnan Wen
- Department of Cardiovascular Medicine,
Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Rong Bai
- Banner University Medical Center Phoenix,
College of Medicine University of Arizona, Phoenix, AZ 85123, USA
| | - Yanfei Ruan
- Department of Cardiology, Beijing Anzhen Hospital,
Capital Medical University, Beijing 100012, China
- National Clinical Research Center for Cardiovascular Diseases, Beijing 100012, China
| | - Hao Zhou
- Department of Cardiology,
Chinese PLA General Hospital, Beijing 100853, China
- Xianning Medical College,
Hubei University of Science and Technology, Xianning 437000, China
| | - Rongjun Zou
- Department of Cardiovascular Surgery,
the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong, China
| | - Ribo Tang
- Department of Cardiology, Beijing Anzhen Hospital,
Capital Medical University, Beijing 100012, China
- National Clinical Research Center for Cardiovascular Diseases, Beijing 100012, China
| | - Nian Liu
- Department of Cardiology, Beijing Anzhen Hospital,
Capital Medical University, Beijing 100012, China
- National Clinical Research Center for Cardiovascular Diseases, Beijing 100012, China
| |
Collapse
|
4
|
Nevins S, McLoughlin CD, Oliveros A, Stein JB, Rashid MA, Hou Y, Jang MH, Lee KB. Nanotechnology Approaches for Prevention and Treatment of Chemotherapy-Induced Neurotoxicity, Neuropathy, and Cardiomyopathy in Breast and Ovarian Cancer Survivors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2300744. [PMID: 37058079 PMCID: PMC10576016 DOI: 10.1002/smll.202300744] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/05/2023] [Indexed: 06/19/2023]
Abstract
Nanotechnology has emerged as a promising approach for the targeted delivery of therapeutic agents while improving their efficacy and safety. As a result, nanomaterial development for the selective targeting of cancers, with the possibility of treating off-target, detrimental sequelae caused by chemotherapy, is an important area of research. Breast and ovarian cancer are among the most common cancer types in women, and chemotherapy is an essential treatment modality for these diseases. However, chemotherapy-induced neurotoxicity, neuropathy, and cardiomyopathy are common side effects that can affect breast and ovarian cancer survivors quality of life. Therefore, there is an urgent need to develop effective prevention and treatment strategies for these adverse effects. Nanoparticles (NPs) have extreme potential for enhancing therapeutic efficacy but require continued research to elucidate beneficial interventions for women cancer survivors. In short, nanotechnology-based approaches have emerged as promising strategies for preventing and treating chemotherapy-induced neurotoxicity, neuropathy, and cardiomyopathy. NP-based drug delivery systems and therapeutics have shown potential for reducing the side effects of chemotherapeutics while improving drug efficacy. In this article, the latest nanotechnology approaches and their potential for the prevention and treatment of chemotherapy-induced neurotoxicity, neuropathy, and cardiomyopathy in breast and ovarian cancer survivors are discussed.
Collapse
Affiliation(s)
- Sarah Nevins
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| | - Callan D. McLoughlin
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| | - Alfredo Oliveros
- Department of Neurosurgery, Robert Wood Johnson Medical
School, Rutgers University, the State University of New Jersey, 661 Hoes Ln W,
Piscataway, NJ, 08854, U.S.A
| | - Joshua B. Stein
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| | - Mohammad Abdur Rashid
- Department of Neurosurgery, Robert Wood Johnson Medical
School, Rutgers University, the State University of New Jersey, 661 Hoes Ln W,
Piscataway, NJ, 08854, U.S.A
| | - Yannan Hou
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| | - Mi-Hyeon Jang
- Department of Neurosurgery, Robert Wood Johnson Medical
School, Rutgers University, the State University of New Jersey, 661 Hoes Ln W,
Piscataway, NJ, 08854, U.S.A
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| |
Collapse
|
5
|
Chen L, Chen X, Ruan B, Yang H, Yu Y. Tirzepatide protects against doxorubicin-induced cardiotoxicity by inhibiting oxidative stress and inflammation via PI3K/Akt signaling. Peptides 2024; 178:171245. [PMID: 38801993 DOI: 10.1016/j.peptides.2024.171245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Doxorubicin (DOX) is a highly effective and widely used cytotoxic agent with application for various malignancies, but it's clinically limited due to its cardiotoxicity Oxidative stress and inflammation were reported to take part in DOX-induced cardiotoxicity. Tirzepatide, a dual glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) receptor agonist has been approved to treat type 2 diabetes. However, its role in DOX-induced cardiotoxicity and the underlying mechanisms has not been explored. METHODS The cardioprotective properties of Tirzepatide against DOX-induced cardiotoxicity are examined in this work both in vivo and in vitro. For four weeks, an intraperitoneal injection of 4 mg/kg DOX was used to cause cardiotoxicity in C57BL/6 mice. To ascertain the cardioprotective function and underlying mechanisms of Tirzepatide against DOX-induced cardiotoxicity, mice and H9c2 cells were treated with and without Tirzepatide. RESULTS Tirzepatide treatment significantly inhibited DOX-induced oxidative stress, inflammation and cardiac injury. Mechanistically, PI3K/Akt signaling pathway contributes to the protective effect of Tirzepatide against DOX-induced cardiotoxicity and inhibited PI3K/Akt signaling pathway with LY294002 almost blocked its therapeutic effect. CONCLUSIONS Collectively, Tirzepatide could alleviate DOX-induced oxidative stress, inflammation and cardiac injury via activating PI3K/Akt signaling pathway and Tirzepatide may be a novel therapeutic target for DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Ling Chen
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China
| | - Xi Chen
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China
| | - Bing Ruan
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China
| | - Hongjie Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Yang Yu
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China.
| |
Collapse
|
6
|
Savran M, Asci S, Gulle K, Aslankoc R, Asci H, Karakuyu NF, Erzurumlu Y, Kaynak M. Agomelatine ameliorates doxorubicin-induced cortical and hippocampal brain injury via inhibition of TNF-alpha/NF-kB pathway. Toxicol Mech Methods 2024; 34:359-368. [PMID: 38093452 DOI: 10.1080/15376516.2023.2291123] [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: 08/11/2023] [Accepted: 11/29/2023] [Indexed: 04/20/2024]
Abstract
Side effects of doxorubicin (DOX) are mainly due to oxidative stress, with the involvement of inflammatory and apoptotic mechanisms. Agomelatine (AGO) is a melatonin receptor agonist with antioxidant, anti-inflammatory, and anti-apoptotic features. This study aimed to evaluate the effects of AGO with different doses on DOX-induced neurotoxicity. Rats were divided into four groups as control, DOX (40 mg/kg, intraperitoneal single dose), DOX + AGO20 (20 mg/kg AGO oral gavage for 14 days), and DOX + AGO40 (40 mg/kg AGO oral gavage for 14 days). On day 14, brain tissues were collected for biochemical, histopathological, and genetic examinations. DOX significantly increased malondialdehyde and decreased superoxide dismutase and catalase (CAT) levels. CAT levels were significantly increased only in the DOX + AGO40 group compared to the DOX group (p = 0.040) while other changes in oxidant and antioxidant indicators were insignificant. DOX-induced significant increases in TNF-alpha and NF-κB were reversed following both low and high-dose AGO administration in a dose-dependent manner (p < 0.001 for both doses). Cellular shrinkage, pycnotic change, and vacuolization in apoptotic bodies were apparent in the cortical and hippocampal areas of DOX-treated samples. Both doses of AGO alleviated these histopathological changes (p = 0.01 for AGO20 and p = 0.05 for AGO40). Significantly increased apoptosis shown with caspase-3 immunostaining in the DOX group was alleviated following AGO administration, with additional improvement after high-dose treatment (p < 0.01 for DOX compared to both AGO groups and p < 0.05 for AGO40 compared to AGO20). AGO can be protective against DOX-induced neurotoxicity by antioxidant, anti-inflammatory, and anti-apoptotic mechanisms in a dose-dependent manner.
Collapse
Affiliation(s)
- Mehtap Savran
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Sanem Asci
- Department of Neurology, Private MEDDEM Hospital, Isparta, Turkey
| | - Kanat Gulle
- Department of Histology and Embryology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Rahime Aslankoc
- Department of Physiology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Halil Asci
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Nasif Fatih Karakuyu
- Department of Pharmacology, Faculty of Pharmacy, Suleyman Demirel University, Isparta, Turkey
| | - Yalçın Erzurumlu
- Department of Biochemistry, Faculty of Pharmacy, Suleyman Demirel University, Isparta, Turkey
| | - Mine Kaynak
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| |
Collapse
|
7
|
Zhang GL, Liu Y, Liu YF, Huang XT, Tao Y, Chen ZH, Lai HL. Teneligliptin mitigates diabetic cardiomyopathy by inhibiting activation of the NLRP3 inflammasome. World J Diabetes 2024; 15:724-734. [PMID: 38680706 PMCID: PMC11045420 DOI: 10.4239/wjd.v15.i4.724] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 12/20/2023] [Accepted: 02/27/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND Diabetic cardiomyopathy (DCM), which is a complication of diabetes, poses a great threat to public health. Recent studies have confirmed the role of NLRP3 (NOD-like receptor protein 3) activation in DCM development through the inflammatory response. Teneligliptin is an oral hypoglycemic dipeptidyl peptidase-IV inhibitor used to treat diabetes. Teneligliptin has recently been reported to have anti-inflammatory and protective effects on myocardial cells. AIM To examine the therapeutic effects of teneligliptin on DCM in diabetic mice. METHODS Streptozotocin was administered to induce diabetes in mice, followed by treatment with 30 mg/kg teneligliptin. RESULTS Marked increases in cardiomyocyte area and cardiac hypertrophy indicator heart weight/tibia length reductions in fractional shortening, ejection fraction, and heart rate; increases in creatine kinase-MB (CK-MB), aspartate transaminase (AST), and lactate dehydrogenase (LDH) levels; and upregulated NADPH oxidase 4 were observed in diabetic mice, all of which were significantly reversed by teneligliptin. Moreover, NLRP3 inflammasome activation and increased release of interleukin-1β in diabetic mice were inhibited by teneligliptin. Primary mouse cardiomyocytes were treated with high glucose (30 mmol/L) with or without teneligliptin (2.5 or 5 µM) for 24 h. NLRP3 inflammasome activation. Increases in CK-MB, AST, and LDH levels in glucose-stimulated cardiomyocytes were markedly inhibited by teneligliptin, and AMP (p-adenosine 5'-monophosphate)-p-AMPK (activated protein kinase) levels were increased. Furthermore, the beneficial effects of teneligliptin on hyperglycaemia-induced cardiomyocytes were abolished by the AMPK signaling inhibitor compound C. CONCLUSION Overall, teneligliptin mitigated DCM by mitigating activation of the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Gu-Lao Zhang
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330000, Jiangxi Province, China
| | - Yuan Liu
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330000, Jiangxi Province, China
| | - Yan-Feng Liu
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330000, Jiangxi Province, China
| | - Xian-Tao Huang
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330000, Jiangxi Province, China
| | - Yu Tao
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330000, Jiangxi Province, China
| | - Zhen-Huan Chen
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330000, Jiangxi Province, China
| | - Heng-Li Lai
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330000, Jiangxi Province, China
| |
Collapse
|
8
|
Wang W, Zhang J. Teneligliptin alleviates diabetes-related cognitive impairment by inhibiting the endoplasmic reticulum (ER) stress and NLRP3 inflammasome in mice. Aging (Albany NY) 2023; 16:8336-8347. [PMID: 38127000 PMCID: PMC11131981 DOI: 10.18632/aging.205333] [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: 07/14/2023] [Accepted: 10/23/2023] [Indexed: 12/23/2023]
Abstract
Diabetes mellitus (DM) significantly influences the normal health of patients with its severe complications, including diabetes-related cognitive impairment (CI). Recently, neuroinflammation and oxidative stress (OS) have been reported to participate in the pathogenesis of diabetes-related CI. Teneligliptin, an inhibitor of DDP-IV, was developed for treating DM and is claimed with promising effects against inflammation. Herein, in the current study, we examined the potential therapeutic function of Teneligliptin against diabetes-related CI. Db/m or diabetic mice were orally administered with teneligliptin (60 mg/kg/day) for 10 weeks. Elevated levels of total cholesterol (TC), triglyceride (TG), and low-density lipoprotein cholesterol (LDL-C), increased escape latency, declined time in the platform quadrant and decreased number of platform crossings in the Morris water maze test, reduced freezing index in the fear conditioning test, and lessened time spent in the novel arm and percentage of alterations in the Y-maze test were observed in diabetic mice, all of which were sharply improved by teneligliptin. Furthermore, increased levels of inflammatory cytokines and activated OS state were observed in the hippocampus of diabetic mice, which were markedly repressed by Teneligliptin. Lastly, the activation of the NOD-like receptor family pyrin domain containing 3 (NLRP3) signaling and the endoplasmic reticulum (ER) stress pathway in the hippocampus of diabetic mice were notably inhibited by teneligliptin. Collectively, teneligliptin mitigated diabetes-related CI by repressing the ER stress and NLRP3 inflammasome in diabetic mice.
Collapse
Affiliation(s)
- Weifeng Wang
- Department of Endocrinology, Laizhou City People’s Hospital, Yantai, Shandong 261400, China
| | - Juanjuan Zhang
- Department of Endocrinology, Laizhou City People’s Hospital, Yantai, Shandong 261400, China
| |
Collapse
|
9
|
Kuthati Y, Rao VN, Huang WH, Busa P, Wong CS. Teneligliptin Co-Infusion Alleviates Morphine Tolerance by Inhibition of Spinal Microglial Cell Activation in Streptozotocin-Induced Diabetic Rats. Antioxidants (Basel) 2023; 12:1478. [PMID: 37508016 PMCID: PMC10376493 DOI: 10.3390/antiox12071478] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/17/2023] [Accepted: 07/22/2023] [Indexed: 07/30/2023] Open
Abstract
Morphine (MOR) is a commonly prescribed drug for the treatment of moderate to severe diabetic neuropathic pain (DNP). However, long-term MOR treatment is limited by morphine analgesic tolerance (MAT). The activation of microglial cells and the release of glia-derived proinflammatory cytokines are known to play an important role in the development of MAT. In this study, we aimed to investigate the effects of the dipeptidyl peptidase-4 inhibitor (DPP-4i) teneligliptin (TEN) on MOR-induced microglial cell activation and MAT in DNP rats. DNP was induced in four groups of male Wistar rats through a single intraperitoneal injection of streptozotocin (STZ) (50 mg/kg, freshly dissolved in 5 mmol/L citrate buffer, pH 4.5). Sham rats were administered with the vehicle. Seven days after STZ injection, all rats were implanted with an intrathecal (i.t) catheter connected to a mini-osmotic pump, divided into five groups, and infused with the following combinations: sham + saline (1 µL/h, i.t), DNP + saline (1 µL/h, i.t), DNP + MOR (15 µg/h, i.t), DNP + TEN (2 µg/h, i.t), and DNP + MOR (15 µg/h, i.t) + TEN (2 µg/h, i.t) for 7 days at a rate of 1 μL/h. The MAT was confirmed through the measurement of mechanical paw withdrawal threshold and tail-flick tests. The mRNA expression of neuroprotective proteins nuclear factor erythroid 2-related factor (Nrf2) and heme oxygenase-1 (HO-1) in the dorsal horn was evaluated by quantitative PCR (qPCR). Microglial cell activation and mononucleate cell infiltration in the spinal cord dorsal horn were assessed by immunofluorescence assay (IFA) and Western blotting (WB). The results showed that co-infusion of TEN with MOR significantly attenuated MAT in DNP rats through the restoration of neuroprotective proteins Nrf2 and HO-1 and suppression of microglial cell activation in the dorsal horn. Though TEN at a dose of 2 μg has mild antinociceptive effects, it is highly effective in limiting MAT.
Collapse
Affiliation(s)
- Yaswanth Kuthati
- Department of Anesthesiology, Cathy General Hospital, Taipei 106, Taiwan
| | - Vaikar Navakanth Rao
- PhD Program in Pharmacology and Toxicology, School of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Wei-Hsiu Huang
- Department of Anesthesiology, Cathy General Hospital, Taipei 106, Taiwan
| | - Prabhakar Busa
- Department of Anesthesiology, Cathy General Hospital, Taipei 106, Taiwan
| | - Chih-Shung Wong
- Department of Anesthesiology, Cathy General Hospital, Taipei 106, Taiwan
- National Defense Medical Center, Institute of Medical Sciences, Taipei 114, Taiwan
| |
Collapse
|
10
|
Qi P, Li P, Qiao L, Xue H, Ma Y, Wei S, Yang X, Zhang H, Zhang Y, Wang Y, He S, Quan H, Zhang W. Simultaneous quantification of pirarubicin, doxorubicin, cyclophosphamide, and vincristine in human plasma of patients with non-Hodgkin's lymphoma by LC-MS/MS method. J Chromatogr B Analyt Technol Biomed Life Sci 2023; 1224:123754. [PMID: 37229818 DOI: 10.1016/j.jchromb.2023.123754] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 05/05/2023] [Accepted: 05/11/2023] [Indexed: 05/27/2023]
Abstract
Pirarubicin (THP), doxorubicin (DOX), cyclophosphamide (CTX), and vincristine (VCR) are widely used in the treatment of patients with non-Hodgkin's Lymphoma. Herein, a precise and sensitive method was developed for the determination of THP, DOX, CTX and VCR in human plasma by high-performance liquid-chromatography-tandem mass spectrometry (LC-MS/MS). Liquid-liquid extraction was applied to extract THP, DOX, CTX, VCR, and the internal standard (IS, Pioglitazone) in plasma. Agilent Eclipse XDB-C18 (3.0 mm × 100 mm) was utilized and chromatographic separation was obtained in eight minutes. Mobile phases were composed of methanol and buffer (10 mM ammonium formate containing 0.1% formic acid). The method was linear within the concentration range of 1-500 ng/mL for THP, 2-1000 ng/mL for DOX, 2.5-1250 ng/mL for CTX, and 3-1500 ng/mL for VCR. The intra- and inter-day precisions of QC samples were found to be below 9.31 and 13.66%, and accuracy ranged from -0.2 to 9.07%, respectively. THP, DOX, CTX, VCR and the internal standard were stable in several conditions. Finally, this method was successfully utilized to simultaneously determine THP, DOX, CTX and VCR in human plasma of 15 patients with non-Hodgkin's Lymphoma after intravenous administration. Finally, the method was successfully employed in the clinical determination of THP, DOX, CTX, and VCR in patients with non-Hodgkin lymphoma after administration of RCHOP (rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone) regimens.
Collapse
Affiliation(s)
- Peng Qi
- Institute of Clinical Pharmacology, Department of Pharmacy, General Hospital of Ningxia Medical University, Ningxia, China; Ningxia Medical University School of Pharmacy, Ningxia, China.
| | - Ping Li
- Cancer Hospital, General Hospital of Ningxia Medical University, Ningxia, China.
| | - Lijiao Qiao
- Cancer Hospital, General Hospital of Ningxia Medical University, Ningxia, China
| | - Huaqian Xue
- Institute of Clinical Pharmacology, Department of Pharmacy, General Hospital of Ningxia Medical University, Ningxia, China; Ningxia Medical University School of Pharmacy, Ningxia, China
| | - Yanni Ma
- Institute of Clinical Pharmacology, Department of Pharmacy, General Hospital of Ningxia Medical University, Ningxia, China
| | - Shijie Wei
- Institute of Clinical Pharmacology, Department of Pharmacy, General Hospital of Ningxia Medical University, Ningxia, China
| | - Xiaoying Yang
- Institute of Clinical Pharmacology, Department of Pharmacy, General Hospital of Ningxia Medical University, Ningxia, China
| | - Hao Zhang
- Institute of Clinical Pharmacology, Department of Pharmacy, General Hospital of Ningxia Medical University, Ningxia, China
| | - Yuxin Zhang
- Institute of Clinical Pharmacology, Department of Pharmacy, General Hospital of Ningxia Medical University, Ningxia, China
| | - Yifan Wang
- Institute of Clinical Pharmacology, Department of Pharmacy, General Hospital of Ningxia Medical University, Ningxia, China; Ningxia Medical University School of Pharmacy, Ningxia, China
| | - Shaolong He
- Institute of Clinical Pharmacology, Department of Pharmacy, General Hospital of Ningxia Medical University, Ningxia, China
| | - Hongfeng Quan
- Ningxia Medical University School of Pharmacy, Ningxia, China
| | - Wenping Zhang
- Institute of Clinical Pharmacology, Department of Pharmacy, General Hospital of Ningxia Medical University, Ningxia, China.
| |
Collapse
|
11
|
Becker MMC, Arruda GFA, Berenguer DRF, Buril RO, Cardinale D, Brandão SCS. Anthracycline cardiotoxicity: current methods of diagnosis and possible role of 18F-FDG PET/CT as a new biomarker. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2023; 9:17. [PMID: 36973762 PMCID: PMC10041777 DOI: 10.1186/s40959-023-00161-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 02/01/2023] [Indexed: 03/29/2023]
Abstract
Despite advances in chemotherapy, the drugs used in cancer treatment remain rather harmful to the cardiovascular system, causing structural and functional cardiotoxic changes. Positron-emission tomography associated with computed tomography (PET/CT) has emerged like a promising technique in the early diagnosis of these adverse drug effects as the myocardial tissue uptake of fluorodeoxyglucose labeled with fluorine-18 (18F-FDG), a glucose analog, is increased after their use. Among these drugs, anthracyclines are the most frequently associated with cardiotoxicity because they promote heart damage through DNA breaks, and induction of an oxidative, proinflammatory, and toxic environment. This review aimed to present the scientific evidence available so far regarding the use of 18F-FDG PET/CT as an early biomarker of anthracycline-related cardiotoxicity. Thus, it discusses the physiological basis for its uptake, hypotheses to justify its increase in the myocardium affected by anthracyclines, importance of 18F-FDG PET/CT findings for cardio-oncology, and primary challenges of incorporating this technique in standard clinical oncology practice.
Collapse
Affiliation(s)
- Mônica M C Becker
- Postgraduate Program in Surgery, Federal University of Pernambuco, Recife, State of Pernambuco, Brazil
| | - Gustavo F A Arruda
- Recife Medical School, Federal University of Pernambuco, Recife, State of Pernambuco, Brazil
| | - Diego R F Berenguer
- Postgraduate Program in Translational Health, Federal University of Pernambuco, Recife, State of Pernambuco, Brazil
| | - Roberto O Buril
- Postgraduate Program in Surgery, Federal University of Pernambuco, Recife, State of Pernambuco, Brazil
| | - Daniela Cardinale
- Cardioncology Unit, European Institute of Oncology, I.R.C.C.S., Milan, Italy
| | - Simone C S Brandão
- Postgraduate Program in Surgery, Federal University of Pernambuco, Recife, State of Pernambuco, Brazil.
- Recife Medical School, Federal University of Pernambuco, Recife, State of Pernambuco, Brazil.
- Nuclear Medicine Department, Hospital das Clínicas, Federal University of Pernambuco, 1st floor, 1235 Avenida Professor Moraes Rego, Recife, State of Pernambuco, 50670-901, Brazil.
| |
Collapse
|
12
|
Xie Y, Zhou Q, He Q, Wang X, Wang J. Opportunities and challenges of incretin-based hypoglycemic agents treating type 2 diabetes mellitus from the perspective of physiological disposition. Acta Pharm Sin B 2022. [DOI: 10.1016/j.apsb.2022.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
13
|
Sun X, Zhu Y, Li F, Li M, Wan G. Cardioprotective Mechanism and Active Compounds of Folium Ginkgo on Adriamycin-Induced Cardiotoxicity: A Network Pharmacology Study. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:4338260. [PMID: 36213575 PMCID: PMC9534669 DOI: 10.1155/2022/4338260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 09/19/2022] [Indexed: 12/06/2022]
Abstract
Objective To investigate the mechanism of Folium Ginkgo (FG) against adriamycin-induced cardiotoxicity (AIC) through a network pharmacology approach. Methods Active ingredients of FG were screened by TCMSP, and the targets of active ingredient were collected by Genclip3 and HERB databases. AIC-related target genes were predicted by Genecards, OMIM, and CTD databases. Protein-protein interaction (PPI) network was constructed by STRING platform and imported into Cytoscape software to construct the FG-active ingredients-targets-AIC network, and CytoNCA plug-in was used to analyze and identify the core target genes. The Metascape platform was used for transcription factor, GO and signaling pathway enrichment analysis. Results 27 active ingredients of FG and 1846 potential targets were obtained and 358 AIC target genes were retrieved. The intersection of FG and AIC targets resulted in 218 target genes involved in FG action. The top 5 active ingredients with most targets were quercetin, luteolin, kaempferol, isorhamnetin, and sesamin. After constructing the FG-active ingredients-targets-AIC network, CytoNCA analysis yielded 51 core targets, of which the top ranked target was STAT3. Ninety important transcription factors were enriched by transcription factor enrichment analysis, including RELA, TP53, NFKB1, SP1, JUN, STAT3, etc. The results of GO enrichment analysis showed that the effective active ingredient targets of FG were involved in apoptotic signaling, response to growth factor, cellular response to chemical stress, reactive oxygen species metabolic process, etc. The signaling pathway enrichment analysis showed that there were many signaling pathways involved in AIC, mainly including pathways in cancer, FOXO signaling pathway, AGE-RAGE signaling pathway in diabetic complications, signaling by interleukins, and PI3K-AKT signaling pathway,. Conclusions The study based on a network pharmacology approach demonstrates that the possible mechanisms of FG against AIC are the involvement of multicomponents, multitargets, and multipathways, and STAT3 may be a key target. Further experiments are needed to verify the results.
Collapse
Affiliation(s)
- Xue Sun
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, 39# Chaoyang Road, Shiyan, Hubei 442000, China
| | - Yiming Zhu
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, 39# Chaoyang Road, Shiyan, Hubei 442000, China
| | - Fang Li
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, 39# Chaoyang Road, Shiyan, Hubei 442000, China
| | - Min Li
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, 39# Chaoyang Road, Shiyan, Hubei 442000, China
| | - Guoxing Wan
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, 39# Chaoyang Road, Shiyan, Hubei 442000, China
- Institute of Cancer, Renmin Hospital of Hubei University of Medicine, Shiyan 442000, China
| |
Collapse
|
14
|
Endogenous Hydrogen Sulfide Persulfidates Caspase-3 at Cysteine 163 to Inhibit Doxorubicin-Induced Cardiomyocyte Apoptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6153772. [PMID: 35571249 PMCID: PMC9095366 DOI: 10.1155/2022/6153772] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/31/2022] [Accepted: 04/13/2022] [Indexed: 02/07/2023]
Abstract
Doxorubicin (DOX) is an efficient antitumor anthracycline drug, but its cardiotoxicity adversely affects the prognosis of the patients. In this study, we explored whether endogenous gasotransmitter hydrogen sulfide (H2S) could protect against DOX-induced cardiomyocyte apoptosis and its mechanisms. The results indicated that DOX significantly downregulated endogenous H2S production and endogenous synthetase cystathionine γ-lyase (CSE) expression and obviously stimulated the apoptosis in H9C2 cells. The supplement of H2S donor sodium hydrosulfide (NaHS) or overexpression of CSE inhibited DOX-induced H9C2 cell apoptosis. DOX enhanced the activities of caspase family members in cardiomyocytes, while NaHS attenuated DOX-enhanced caspase-3, caspase-2, and caspase-9 activities by 223.1%, 73.94%, and 52.29%, respectively. Therefore, taking caspase-3 as a main target, we demonstrated that NaHS or CSE overexpression alleviated the cleavage of caspase-3, suppressed caspase-3 activity, and inhibited the cleavage of poly ADP-ribose polymerase (PARP). Mechanistically, we found that H2S persulfidated caspase-3 in H9C2 cells and human recombinant caspase-3 protein, while the thiol-reducing agent dithiothreitol (DTT) abolished H2S-induced persulfidation of caspase-3 and thereby prevented the antiapoptotic effect of H2S on caspase-3 in H9C2 cells. The mutation of caspase-3 C148S and C170S failed to block caspase-3 persulfidation by H2S in H9C2 cells. However, caspase-3 C163S mutation successfully abolished the effect of H2S on caspase-3 persulfidation and the corresponding protection of H9C2 cells. Collectively, these findings indicate that endogenous H2S persulfidates caspase-3 at cysteine 163, inhibiting its activity and cardiomyocyte apoptosis. Sufficient endogenous H2S might be necessary for the protection against myocardial cell apoptosis induced by DOX. The results of the study might open new avenues with respect to the therapy of DOX-stimulated cardiomyopathy.
Collapse
|
15
|
Fabiani I, Aimo A, Grigoratos C, Castiglione V, Gentile F, Saccaro LF, Arzilli C, Cardinale D, Passino C, Emdin M. Oxidative stress and inflammation: determinants of anthracycline cardiotoxicity and possible therapeutic targets. Heart Fail Rev 2021; 26:881-890. [PMID: 33319255 PMCID: PMC8149360 DOI: 10.1007/s10741-020-10063-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/01/2020] [Indexed: 12/04/2022]
Abstract
Chemotherapy with anthracycline-based regimens remains a cornerstone of treatment of many solid and blood tumors but is associated with a significant risk of cardiotoxicity, which can manifest as asymptomatic left ventricular dysfunction or overt heart failure. These effects are typically dose-dependent and cumulative and may require appropriate screening strategies and cardioprotective therapies in order to minimize changes to anticancer regimens or even their discontinuation. Our current understanding of cardiac damage by anthracyclines includes a central role of oxidative stress and inflammation. The identification of these processes through circulating biomarkers or imaging techniques might then be helpful for early diagnosis and risk stratification. Furthermore, therapeutic strategies relieving oxidative stress and inflammation hold promise to prevent heart failure development or at least to mitigate cardiac damage, although further evidence is needed on their efficacy, either alone or as part of combination therapies with neurohormonal antagonists, which are the current adopted standard.
Collapse
Affiliation(s)
- Iacopo Fabiani
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Alberto Aimo
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy.
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy.
| | | | | | | | - Luigi F Saccaro
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | | | - Daniela Cardinale
- Cardioncology Unit, European Institute of Oncology, IRCCS, Milan, Italy
| | - Claudio Passino
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Michele Emdin
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| |
Collapse
|
16
|
Ozone Attenuated H9c2 Cell Injury Induced by Doxorubicin. J Cardiovasc Pharmacol 2021; 78:e86-e93. [PMID: 33929391 DOI: 10.1097/fjc.0000000000001043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/23/2021] [Indexed: 12/26/2022]
Abstract
ABSTRACT Doxorubicin (DOX) is a commonly used drug in the treatment of cancers, whereas its application in the clinical stage is restricted because of side effects such as cardiomyocyte injury. Increasing studies indicated that ozone may protect cardiomyocytes from injuries. This study aimed to explore the effects of ozone on cardiotoxicity induced by DOX treatment. Rat heart myoblasts (H9c2) were treated with increasing concentrations of DOX (0.5, 1, 1.5, and 2 μM) to induce cell injury. 3-(4,5)-dimethylthiahiazo(-2)-3,5-diphenytetrazoliumromide assay and flow cytometry analysis were used to measure the viability and apoptosis of H9c2 cells. The mRNA and protein levels of proinflammatory cytokines [tumor necrosis factor-α (TNF-α), interleukin-(IL)1β, and IL-6, matrix metalloproteinases (MMP-2 and MMP-9), and the key factors on the TLR4/NF-kB signaling (TLR4, p-p65, and p65) were measured by reverse transcription quantitative polymerase chain reaction, enzyme-linked immunosorbent assay, and western blot. The result showed that DOX promoted apoptosis and increased the expression of TNF-α (by 3.65-fold changes), IL-1β (by 4.98-fold changes), IL-6 (by 3.44-fold changes), MMP-2 (by 1.98-fold changes), and MMP-9 (by 1.98-fold changes) levels in H9c2 cells. Moreover, the introduction of ozone reversed these changes in gene expression and suppressed the activation of the TLR4/NF-kB signaling, which indicated that ozone may exert protective effects on H9c2 heart myoblasts by relieving the cardiotoxicity induced by DOX. Our study provides theoretical basis for the significance of ozone in managing doxorubicin-induced H9c2 heart myoblast injury.
Collapse
|
17
|
Du J, Zhang A, Li J, Liu X, Wu S, Wang B, Wang Y, Jia H. Doxorubicin-Induced Cognitive Impairment: The Mechanistic Insights. Front Oncol 2021; 11:673340. [PMID: 34055643 PMCID: PMC8158153 DOI: 10.3389/fonc.2021.673340] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/08/2021] [Indexed: 12/21/2022] Open
Abstract
Chemotherapy can significantly prolong the survival of patients with breast cancer; Nevertheless, the majority of patients receiving chemotherapy such as doxorubicin may have cognitive deficits that manifest as impairments in learning, reasoning, attention, and memory. The phenomenon of chemotherapy-induced cognitive decline is termed as chemotherapy-related cognitive impairment (CRCI) or chemo-brain. Doxorubicin (DOX), a commonly used drug in adjuvant chemotherapy for patients with breast cancer, has been reported to induce chemo-brain through a variety of mechanisms including DNA damage, oxidative stress, inflammation, dysregulation of apoptosis and autophagy, changes in neurotransmitter levels, mitochondrial dysfunction, glial cell interactions, neurogenesis inhibition, and epigenetic factors. These mechanisms do not operate independently but are inter-related, coordinately contributing to the development of chemo-brain. Here we review the relationships of these mechanisms and pathways in attempt to provide mechanistic insights into the doxorubicin-induced cognitive impairment.
Collapse
Affiliation(s)
- Jiajia Du
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Aoxue Zhang
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Jing Li
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Xin Liu
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Shuai Wu
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Bin Wang
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yanhong Wang
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, China
| | - Hongyan Jia
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
18
|
Cheng F, Jiang W, Xiong X, Chen J, Xiong Y, Li Y. Ethanol Extract of Chinese Hawthorn (Crataegus pinnatifida) Fruit Reduces Inflammation and Oxidative Stress in Rats with Doxorubicin-Induced Chronic Heart Failure. Med Sci Monit 2020; 26:e926654. [PMID: 33232307 PMCID: PMC7697658 DOI: 10.12659/msm.926654] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background Chinese hawthorn (Crataegus pinnatifida) fruit is a traditional Chinese medicine for treatment of digestive system and cardiovascular diseases. The fruit contains polyphenol compounds, such as epicatechin, that have anti-inflammatory activity. This study aimed to investigate the effects of an alcohol extract of hawthorn fruit (HAE) on inflammation and oxidative stress in rats with doxorubicin-induced chronic heart failure (CHF). Material/Methods Rats were intraperitoneally injected with doxorubicin to induce CHF and subsequently treated with HAE intragastrically once daily for 6 weeks. At the end of the experiment, echocardiographic and hemodynamic parameters were assessed, and enzyme-linked immunoassays were used to detect the levels of cardiac injury markers (brain natriuretic peptide, creatine kinase-MB, aspartate aminotransferase, lactate dehydrogenase, copeptin, and adrenomedullin), oxidative stress markers (glutathione peroxidase and malondialdehyde), and inflammatory cytokines (interleukin [IL]-6, IL-8, IL-1β, and tumor necrosis factor-α). The IL-1β, IL-6, glutathione peroxidase-1, and catalase mRNA levels were also measured by quantitative real-time polymerase chain reaction. Results Our findings indicated that HAE exerts a cardioprotective effect, as shown by improved echocardiographic and hemodynamic parameters, decreased activity of serum myocardial enzymes, reduced serum levels of CHF markers, and inhibited inflammatory response in cardiac tissue. In addition, HAE treatment downregulated the mRNA expression of IL-1β and tumor necrosis factor-α and upregulated the mRNA expression of glutathione peroxidase-1 and catalase compared with untreated doxorubicin-induced CHF rats. Conclusions HAE shows promise for the prevention and treatment of CHF. The cardioprotective effect of HAE appears to be related to inhibition of both the inflammatory response and oxidative stress in vivo.
Collapse
Affiliation(s)
- Fangzhou Cheng
- Department of Cardiology, Shenzhen Yantian People's Hospital, ShenzhenShenzhen, Guangdong, China (mainland)
| | - Wenlong Jiang
- Department of Cardiology, Shenzhen Yantian People's Hospital, Shenzhen, Guangdong, China (mainland)
| | - Xiaoshuan Xiong
- Department of Cardiology, Shenzhen Yantian People's Hospital, Shenzhen, Guangdong, China (mainland)
| | - Juan Chen
- Department of Cardiology, Shenzhen Yantian People's Hospital, Shenzhen, Guangdong, China (mainland)
| | - Yunzhi Xiong
- Department of Cardiology, Shenzhen Yantian People's Hospital, Shenzhen, Guangdong, China (mainland)
| | - Yinghong Li
- The Central Laboratory, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China (mainland)
| |
Collapse
|
19
|
Zhang L, Yuan W, Kong X, Zhang B. Teneligliptin protects against ischemia/reperfusion-induced endothelial permeability in vivo and in vitro. RSC Adv 2020; 10:3765-3774. [PMID: 35492650 PMCID: PMC9048428 DOI: 10.1039/c9ra08810e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 01/09/2020] [Indexed: 12/13/2022] Open
Abstract
Ischemic stroke is a leading cause of disability and mortality worldwide, especially among the elderly population. Ischemia and reperfusion cause damage to cells and initiate an acute inflammatory response, which leads to cerebral endothelial dysfunction, increased endothelial permeability, and potentially permanent disability. Teneligliptin is a dipeptidyl peptidase-4 (DPP-4) inhibitor that has been used almost exclusively in the treatment of type 2 diabetes mellitus. However, it is still unknown whether teneligliptin possesses a protective effect in brain endothelial dysfunction in the context of ischemic stroke. In the present work, we demonstrate the potential of teneligliptin treatment to protect against ischemia/reperfusion-induced damage using a series of both in vivo and in vitro experiments. Our key findings are that administration of teneligliptin could reduce brain infarct volume, ameliorate neurological damage, and improve brain permeability by increasing the expression of the tight junction protein occludin in middle cerebral artery occlusion (MCAO) mice models. Importantly, teneligliptin displayed a robust protective effect against oxygen–glucose deprivation/reperfusion (OGD/R)-induced cell death of primary human brain microvascular endothelial cells (HBMVECs) in vitro. Notably, teneligliptin prevented OGD/R-induced increased endothelial monolayer permeability in HBMVECs by increasing the expression of occludin, which was mediated by the ERK5/KLF2 signaling pathway. These findings suggest that teneligliptin might serve as a potential therapeutic agent for the treatment of stroke Ischemic stroke is a leading cause of disability and mortality worldwide, especially among the elderly population.![]()
Collapse
Affiliation(s)
- Lei Zhang
- Department of Neurology, The First Affiliated Hospital of Xi'an Medical University No. 48 Fenghao West Road, Lianhu District Xi'an Shaanxi province China +86-29-84277356 +86-29-84277356.,Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University Xi'an China
| | - Weiqiong Yuan
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University Xi'an Shaanxi China
| | - Xiangli Kong
- Department of Neurology, The First Affiliated Hospital of Xi'an Medical University No. 48 Fenghao West Road, Lianhu District Xi'an Shaanxi province China +86-29-84277356 +86-29-84277356.,Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University Xi'an China
| | - Bei Zhang
- Department of Neurology, The First Affiliated Hospital of Xi'an Medical University No. 48 Fenghao West Road, Lianhu District Xi'an Shaanxi province China +86-29-84277356 +86-29-84277356.,Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University Xi'an China
| |
Collapse
|