1
|
Chen Y, Cheng R, Lu W, Fan Y, Yu Y, Huang L, Wan Z, Zheng S. Metformin promotes the survival of random skin flaps via the activation of Nrf2/HO-1 signaling. Chem Biol Interact 2024; 401:111188. [PMID: 39121897 DOI: 10.1016/j.cbi.2024.111188] [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: 06/04/2024] [Revised: 07/25/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
The random flap is one of the commonly used techniques for tissue defect repair in surgery and orthopaedics, however the risk of ischaemic necrosis at the distal end of the flap limits its size and clinical application. Metformin (Met) is a first-line medication in the treatment of type 2 diabetes, with additional effects such as anti-tumor, anti-aging, and neuroprotective properties. In this study, we aimed to investigate the biological effects and potential mechanisms of Met in improving the survival of random skin flaps. Twenty-four male Sprague-Dawley rats and 12 male C57BL/6J mice underwent McFarlane flap surgery and divided into control (Ctrl) and Met groups (100 mg/kg). The survival rate of the flap were evaluated on day 7. Angiography, Laser doppler blood flow imaging, and H&E staining were used to assess blood flow supply and the levels of microvascular density. Then, reactive oxygen species (ROS) and malondialdehyde (MDA) levels, and the activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) were measured by test kits. Immunohistochemistry analysis was conducted to evaluate the expression of Vascular Endothelial Growth Factor A (VEGFA), Vascular endothelial cadherin (VE-cadherin) and CD31. Rats and mice in the Met group exhibited higher flap survival rate, microcirculatory flow, and higher expression levels of VEGFA and VE-cadherin compared with the Ctrl group. In addition, the level of oxidative stress was significantly lower in the met group. And then we demonstrated that the human umbilical vein endothelial cells (HUVECs) treated with Met can alleviate tert-butyl hydroperoxide (TBHP)-stimulated cellular dysfunction and oxidative stress injury. Mechanistically, Met markedly stimulated the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1), and promoted Nrf2 nuclear translocation. Silencing of Nrf2 partially abolished the antioxidant and therapeutic effects of Met. In summary, our data have confirmed that Met has a positive effect on flap survival and reduces necrosis. The mechanism of action involves the regulation of the Nrf2/HO-1 signaling pathway to combat oxidative stress and reduce damage.
Collapse
Affiliation(s)
- Yan Chen
- Key Laboratory of Emergency and Trauma of Ministry of Education, Engineering Research Center for Hainan Biological Sample Resources of Major Diseases, The Hainan Branch of National Clinical Research Center for Cancer & the First Affiliated Hospital, Hainan Medical University, Haikou, 570102, China; Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Hainan Medical University, Haikou, 571199, China
| | - Ruxin Cheng
- Emergency and Trauma College, Hainan Medical University, Haikou, 571199, China
| | - Wenyan Lu
- Key Laboratory of Emergency and Trauma of Ministry of Education, Engineering Research Center for Hainan Biological Sample Resources of Major Diseases, The Hainan Branch of National Clinical Research Center for Cancer & the First Affiliated Hospital, Hainan Medical University, Haikou, 570102, China; Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Hainan Medical University, Haikou, 571199, China
| | - Yonghao Fan
- Key Laboratory of Emergency and Trauma of Ministry of Education, Engineering Research Center for Hainan Biological Sample Resources of Major Diseases, The Hainan Branch of National Clinical Research Center for Cancer & the First Affiliated Hospital, Hainan Medical University, Haikou, 570102, China; Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Hainan Medical University, Haikou, 571199, China
| | - Ye Yu
- Hainan Medical University, Haikou, 571199, China
| | - Ling Huang
- Emergency and Trauma College, Hainan Medical University, Haikou, 571199, China; School of Hainan Provincial Drug Safety Evaluation Research Center, Hainan Medical University, Haikou, 571199, China.
| | - Zhenling Wan
- Department of Pathology, Hainan Women and Children Medical Center, Hainan Medical University, Haikou, 571199, China.
| | - Shaojiang Zheng
- Key Laboratory of Emergency and Trauma of Ministry of Education, Engineering Research Center for Hainan Biological Sample Resources of Major Diseases, The Hainan Branch of National Clinical Research Center for Cancer & the First Affiliated Hospital, Hainan Medical University, Haikou, 570102, China; Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Hainan Medical University, Haikou, 571199, China.
| |
Collapse
|
2
|
Si L, Lai Y. Pharmacological mechanisms by which baicalin ameliorates cardiovascular disease. Front Pharmacol 2024; 15:1415971. [PMID: 39185317 PMCID: PMC11341428 DOI: 10.3389/fphar.2024.1415971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 07/29/2024] [Indexed: 08/27/2024] Open
Abstract
Baicalin is a flavonoid glycoside obtained from the dried root of Scutellaria baicalensis Georgi, which belongs to the Labiatae family. Accumulating evidence indicates that baicalin has favorable therapeutic effects on cardiovascular diseases. Previous studies have revealed the therapeutic effects of baicalin on atherosclerosis, myocardial ischemia/reperfusion injury, hypertension, and heart failure through anti-inflammatory, antioxidant, and lipid metabolism mechanisms. In recent years, some new ideas related to baicalin in ferroptosis, coagulation and fibrinolytic systems have been proposed, and new progress has been made in understanding the mechanism by which baicalin protects cardiomyocytes. However, many relevant underlying mechanisms remain unexplained, and much experimental data is lacking. Therefore, further research is needed to determine these mechanisms. In this review, we summarize the mechanisms of baicalin, which include its anti-inflammatory and antioxidant effects; inhibition of endothelial cell apoptosis; modulation of innate immunity; suppression of vascular smooth muscle cells proliferation, migration, and contraction; regulation of coagulation and fibrinolytic systems; inhibition of myocardial hypertrophy; prevention of myocardial fibrosis; and anti-apoptotic effects on cardiomyocytes.
Collapse
Affiliation(s)
- Lujia Si
- Acupunture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu Lai
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
3
|
Yin X, Feng L, Hua Q, Ye J, Cai L. Progress in the study of mechanisms and pathways related to the survival of random skin flaps. Updates Surg 2024; 76:1195-1202. [PMID: 38308185 DOI: 10.1007/s13304-023-01746-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 12/29/2023] [Indexed: 02/04/2024]
Abstract
The clinical application of random flaps in wound repair has been a topic of discussion. Random flaps are prone to necrosis due to the lack of well-defined vascular blood supply during transfer surgery. Their clinical utility is restricted, financial and psychological burdens is imposed on patients due to this limitation. The survival of random skin flaps depends on factors such as ischemia-reperfusion injury, oxidative stress, local inflammatory response, and neovascularization. This review aims to provide an overview of the evidence supporting the use of random flaps in clinical practice. In addition, this review explores the impact of different medications on signaling pathways within the flap's local microcirculation and investigates the interconnections between these pathways.
Collapse
Affiliation(s)
- Xinghao Yin
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Liang Feng
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Qianqian Hua
- The First School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Jiangtian Ye
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Leyi Cai
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China.
- Department of Orthopaedics Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, No. 109, XueYuan West Road, Luheng District, Wenzhou, Zhejiang, 325000, People's Republic of China.
| |
Collapse
|
4
|
Lai Y, Yang N, Shi D, Ma X, Huang Y, Lu J, Zhang X, Zhou H, Gao W, Mao C, Wang L. Puerarin enhances TFEB-mediated autophagy and attenuates ROS-induced pyroptosis after ischemic injury of random-pattern skin flaps. Eur J Pharmacol 2024; 974:176621. [PMID: 38679118 DOI: 10.1016/j.ejphar.2024.176621] [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: 02/13/2024] [Revised: 04/25/2024] [Accepted: 04/25/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND AND AIM Necrosis of random-pattern flaps restricts their application in clinical practice. Puerarin has come into focus due to its promising therapeutic effects in ischemic diseases. Here, we employed Puerarin and investigated its role and potential mechanisms in flap survival. EXPERIMENTAL PROCEDURE The effect of Puerarin on the viability of human umbilical vein endothelial cells (HUVECs) was assessed by CCK-8, EdU staining, migration, and scratch assays. Survival area measurement and laser Doppler blood flow (LDBF) were utilized to assess the viability of ischemic injury flaps. Levels of molecules related to oxidative stress, pyroptosis, autophagy, transcription factor EB (TFEB), and the AMPK-TRPML1-Calcineurin signaling pathway were detected using western blotting, immunofluorescence, dihydroethidium (DHE) staining, RT-qPCR and Elisa. KEY RESULTS The findings demonstrated that Puerarin enhanced the survivability of ischemic flaps. Autophagy, oxidative stress, and pyroptosis were implicated in the ability of Puerarin in improving flap survival. Increased autophagic flux and augmented tolerance to oxidative stress contribute to Puerarin's suppression of pyroptosis. Additionally, Puerarin modulated the activity of TFEB through the AMPK-TRPML1-Calcineurin signaling pathway, thereby enhancing autophagic flux. CONCLUSIONS AND IMPLICATIONS Puerarin promoted flap survival from ischemic injury through upregulation of TFEB-mediated autophagy and inhibition of oxidative stress. Our findings offered valuable support for the clinical application of Puerarin in the treatment of ischemic diseases, including random-pattern flaps.
Collapse
Affiliation(s)
- Yingying Lai
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, 325027, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325027, China
| | - Ningning Yang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, 325027, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325027, China
| | - Donghao Shi
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xianhui Ma
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, 325027, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325027, China
| | - Yingying Huang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, 325027, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jingzhou Lu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, 325027, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xuzi Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, 325027, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325027, China
| | - Hui Zhou
- Graduate School, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330002, China
| | - Weiyang Gao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, 325027, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325027, China.
| | - Cong Mao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, 325027, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325027, China.
| | - Long Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, 325027, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
5
|
Hu F, Huang K, Zhang H, Hu W, Tong S, Xu H. IGF-PLGA microspheres promote angiogenesis and accelerate skin flap repair and healing by inhibiting oxidative stress and regulating the Ang 1/Tie 2 signaling pathway. Eur J Pharm Sci 2024; 193:106687. [PMID: 38176662 DOI: 10.1016/j.ejps.2023.106687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/27/2023] [Accepted: 12/29/2023] [Indexed: 01/06/2024]
Abstract
Random flaps are widely used in the treatment of injuries, tumors, congenital malformations, and other diseases. However, postoperative skin flaps are prone to ischemic necrosis, leading to surgical failure. Insulin-like growth factor- 1(IGF-1) belongs to the IGF family and exerts its growth-promoting effects in various tissues through autocrine or paracrine mechanisms. Its application in skin flaps and other traumatic diseases is relatively limited. Poly (lactic-co-glycolic acid) (PLGA) is a degradable high-molecular-weight organic compound commonly used in biomaterials. This study prepared IGF-PLGA sustained-release microspheres to explore their impact on the survival rate of flaps both in vitro and in vivo, as well as the mechanisms involved. The research results demonstrate that IGF-PLGA has a good sustained-release effect. At the cellular level, it can promote 3T3 cell proliferation by inhibiting oxidative stress, inhibit apoptosis, and enhance the tube formation ability of human umbilical vein endothelial cells (HUVEC) . At the animal level, it accelerates flap healing by promoting vascularization through the inhibition of oxidative stress. Furthermore, this study reveals the role of IGF-PLGA in activating the Angiopoietin-1(Ang1)/Tie2 signaling pathway in promoting flap vascularization, providing a strong theoretical basis and therapeutic target for the application of IGF-1 in flaps and other traumatic diseases.
Collapse
Affiliation(s)
- Fei Hu
- Cixi Biomedical Research Institute, Wenzhou Medical University, Cixi, Ningbo, China
| | - Kai Huang
- Department of Orthopaedic Surgery, Affiliated Cixi Hospital, Wenzhou Medical University, No. 999, South Second Ring Road, Hushan Street, Cixi, Ningbo 315300, China
| | - Hanbo Zhang
- Cixi Biomedical Research Institute, Wenzhou Medical University, Cixi, Ningbo, China
| | - Wenjie Hu
- Department of Orthopaedic Surgery, Affiliated Cixi Hospital, Wenzhou Medical University, No. 999, South Second Ring Road, Hushan Street, Cixi, Ningbo 315300, China
| | - Songlin Tong
- Department of Orthopaedic Surgery, Affiliated Cixi Hospital, Wenzhou Medical University, No. 999, South Second Ring Road, Hushan Street, Cixi, Ningbo 315300, China
| | - Hongming Xu
- Department of Orthopaedic Surgery, Affiliated Cixi Hospital, Wenzhou Medical University, No. 999, South Second Ring Road, Hushan Street, Cixi, Ningbo 315300, China.
| |
Collapse
|
6
|
Chen J, Lin C, Huang X, Bian W. Baicalin enhances proliferation and reduces inflammatory-oxidative stress effect in H 2O 2-induced granulosa cells apoptosis via USP48 protein regulation. BMC Complement Med Ther 2024; 24:42. [PMID: 38245760 PMCID: PMC10799411 DOI: 10.1186/s12906-024-04346-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 01/11/2024] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND Oxidative stress and inflammation can lead to apoptosis of ovarian granulosa cells (GCs), resulting in ovulation disorders and infertility. Baicalin (BAI) promotes cell proliferation and reduces inflammation and oxidative stress. However, the mechanisms by which BAI treatment affects oxidative stress and inflammation in GCs remain incompletely understood. METHODS KGN cells were treated with hydrogen peroxide (H2O2) to analyze the effect of oxidative stress on GCs in vitro. Subsequently, H2O2-stimulated KGN cells were treated with BAI. The levels of GSH-Px, CAT, and SOD were measured using an activity assay kit. The levels of MDA, IL-1β, IL-6, IL-8, and TNF-α were measured by ELISA. Proliferation, apoptosis, and mRNA and protein levels were measured using the CCK8, flow cytometry, qRT-PCR, and western blotting. RESULTS H2O2 treatment inhibited KGN cell proliferation and promoted apoptosis, accompanied by increased oxidative stress and inflammation. BAI promoted proliferation, inhibited apoptosis, and reduced oxidative stress and inflammation in H2O2-stimulated KGN cells. BAI treatment promoted USP48 protein expression, and USP48 knockdown abrogated the protective effects of BAI, indicating that USP48 is a downstream mediator of BAI. CONCLUSION BAI treatment enhanced cell proliferation and ameliorated oxidative stress and inflammation by enhancing USP48 protein expression. BAI, which is used clinically and as a dietary supplement, may alleviate oxidative stress-induced GC injury and ovarian disorders.
Collapse
Affiliation(s)
- Jun Chen
- Department of Traditional Chinese Medicine, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University; The First Affiliated Hospital of Southern University of Science and Technology), No. 1017, Dongmen North Road, Luohu District, Shenzhen, 518020, China
| | - Chuhua Lin
- Department of Traditional Chinese Medicine, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University; The First Affiliated Hospital of Southern University of Science and Technology), No. 1017, Dongmen North Road, Luohu District, Shenzhen, 518020, China
| | - Xiurong Huang
- Department of Rehabilitation Medicine, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University; The First Affiliated Hospital of Southern University of Science and Technology), Shenzhen, 518020, China
| | - Wei Bian
- Department of Traditional Chinese Medicine, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University; The First Affiliated Hospital of Southern University of Science and Technology), No. 1017, Dongmen North Road, Luohu District, Shenzhen, 518020, China.
| |
Collapse
|
7
|
Berry CE, Le T, An N, Griffin M, Januszyk M, Kendig CB, Fazilat AZ, Churukian AA, Pan PM, Wan DC. Pharmacological and cell-based treatments to increase local skin flap viability in animal models. J Transl Med 2024; 22:68. [PMID: 38233920 PMCID: PMC10792878 DOI: 10.1186/s12967-024-04882-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/10/2024] [Indexed: 01/19/2024] Open
Abstract
Local skin flaps are frequently employed for wound closure to address surgical, traumatic, congenital, or oncologic defects. (1) Despite their clinical utility, skin flaps may fail due to inadequate perfusion, ischemia/reperfusion injury (IRI), excessive cell death, and associated inflammatory response. (2) All of these factors contribute to skin flap necrosis in 10-15% of cases and represent a significant surgical challenge. (3, 4) Once flap necrosis occurs, it may require additional surgeries to remove the entire flap or repair the damage and secondary treatments for infection and disfiguration, which can be costly and painful. (5) In addition to employing appropriate surgical techniques and identifying healthy, well-vascularized tissue to mitigate the occurrence of these complications, there is growing interest in exploring cell-based and pharmacologic augmentation options. (6) These agents typically focus on preventing thrombosis and increasing vasodilation and angiogenesis while reducing inflammation and oxidative stress. Agents that modulate cell death pathways such as apoptosis and autophagy have also been investigated. (7) Implementation of drugs and cell lines with potentially beneficial properties have been proposed through various delivery techniques including systemic treatment, direct wound bed or flap injection, and topical application. This review summarizes pharmacologic- and cell-based interventions to augment skin flap viability in animal models, and discusses both translatability challenges facing these therapies and future directions in the field of skin flap augmentation.
Collapse
Affiliation(s)
- Charlotte E Berry
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, 257 Campus Drive West, Stanford, CA, 94305, USA
| | - Thalia Le
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, 257 Campus Drive West, Stanford, CA, 94305, USA
| | - Nicholas An
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, 257 Campus Drive West, Stanford, CA, 94305, USA
| | - Michelle Griffin
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, 257 Campus Drive West, Stanford, CA, 94305, USA
| | - Micheal Januszyk
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, 257 Campus Drive West, Stanford, CA, 94305, USA
| | - Carter B Kendig
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, 257 Campus Drive West, Stanford, CA, 94305, USA
| | - Alexander Z Fazilat
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, 257 Campus Drive West, Stanford, CA, 94305, USA
| | - Andrew A Churukian
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, 257 Campus Drive West, Stanford, CA, 94305, USA
| | - Phoebe M Pan
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, 257 Campus Drive West, Stanford, CA, 94305, USA
| | - Derrick C Wan
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, 257 Campus Drive West, Stanford, CA, 94305, USA.
| |
Collapse
|
8
|
Wang Y, Li X, Yan C, Xie L, Yang Y. Baicalin Exhibits a Protective Effect against Cisplatin-Induced Cytotoxic Damage in Canine Renal Tubular Epithelial Cells. Metabolites 2023; 13:1173. [PMID: 38132855 PMCID: PMC10745033 DOI: 10.3390/metabo13121173] [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: 10/19/2023] [Revised: 11/14/2023] [Accepted: 11/20/2023] [Indexed: 12/23/2023] Open
Abstract
Renal failure is a common chronic disease in dogs that substantially affects both their quality of life and longevity. The objective of this study was to assess the protective mechanisms of baicalin in cisplatin-induced Madin-Darby canine kidney (MDCK) epithelial cells' apoptosis model and explore the impacts of baicalin at varying doses on various indexes, such as cisplatin-induced MDCK cell apoptosis, oxidation and antioxidation, and inflammatory factors. (Methods) MDCK cells in the logarithmic growth phase were randomly divided into a control group, a model group (20 μmol/L cisplatin), and a baicalin-protection group (20 μmol/L cisplatin + 50, 25 μmol/L baicalin) and received the corresponding treatments for 24 h. The effects of cisplatin on MDCK cell apoptosis, oxidation and antioxidation, inflammatory factors, and other indicators were studied, and the relieving effect of baicalin on cisplatin-induced MDCK cell damage was explored. Calcein/PI staining and Annexin V-FITC/PI staining showed that cisplatin induced the apoptosis of MDCK cells, while baicalin effectively reduced the damage caused by cisplatin. The ELISA results demonstrated a significant elevation in the nitric oxide (NO) and malondialdehyde (MDA) levels within the MDCK cells following treatment with cisplatin (p < 0.01). In addition, superoxide dismutase (SOD), glutathione peroxidase (GSH), and catalase (CAT) activities remarkably declined (p < 0.01), while tumor necrosis factor α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6) expression within the MDCK cells were apparently elevated (p < 0.01). However, baicalin treatment resulted in opposite changes in these factors. The findings suggested that baicalin exhibits potential in mitigating cisplatin-induced oxidative stress and inflammation in MDCK cells. As revealed with the Western blot results, cisplatin promoted P62, P53, and BAX protein levels, increased mTOR phosphorylation, inhibited AMPK phosphorylation, and reduced Beclin1 and BCL-2 protein levels. However, a contrasting trend was observed following baicalin treatment. Cisplatin can inhibit the activity of MDCK cells, lead to abnormalities in oxidation and antioxidation functions and cell inflammatory factors, and accelerate cell apoptosis. Moreover, baicalin can significantly alleviate the damage of cisplatin to MDCK cells.
Collapse
Affiliation(s)
- Yao Wang
- College of Police Dog Technology, Criminal Investigation Police University of China, Shenyang 110854, China; (Y.W.); (C.Y.)
| | - Xiao Li
- Institute of Special Wild Economic Animals and Plants, Chinese Academy of Agricultural Sciences, Changchun 130112, China;
| | - Chuanguo Yan
- College of Police Dog Technology, Criminal Investigation Police University of China, Shenyang 110854, China; (Y.W.); (C.Y.)
| | - Liuwei Xie
- The Second Affiliated Hospital of Shenyang Medical College, Shenyang 110031, China
| | - Yang Yang
- The Second Affiliated Hospital of Shenyang Medical College, Shenyang 110031, China
| |
Collapse
|
9
|
Li H, Zhang Q. Research Progress of Flavonoids Regulating Endothelial Function. Pharmaceuticals (Basel) 2023; 16:1201. [PMID: 37765009 PMCID: PMC10534649 DOI: 10.3390/ph16091201] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/09/2023] [Accepted: 08/20/2023] [Indexed: 09/29/2023] Open
Abstract
The endothelium, as the guardian of vascular homeostasis, is closely related to the occurrence and development of cardiovascular diseases (CVDs). As an early marker of the development of a series of vascular diseases, endothelial dysfunction is often accompanied by oxidative stress and inflammatory response. Natural flavonoids in fruits, vegetables, and Chinese herbal medicines have been shown to induce and regulate endothelial cells and exert anti-inflammatory, anti-oxidative stress, and anti-aging effects in a large number of in vitro models and in vivo experiments so as to achieve the prevention and improvement of cardiovascular disease. Focusing on endothelial mediation, this paper introduces the signaling pathways involved in the improvement of endothelial dysfunction by common dietary and flavonoids in traditional Chinese medicine and describes them based on their metabolism in the human body and their relationship with the intestinal flora. The aim of this paper is to demonstrate the broad pharmacological activity and target development potential of flavonoids as food supplements and drug components in regulating endothelial function and thus in the prevention and treatment of cardiovascular diseases. This paper also introduces the application of some new nanoparticle carriers in order to improve their bioavailability in the human body and play a broader role in vascular protection.
Collapse
Affiliation(s)
| | - Qi Zhang
- The Basic Medical College, Shaanxi University of Chinese Medicine, Xianyang 712046, China;
| |
Collapse
|