1
|
Rahaman KA, Mukim MSI, Hasan ML, Kim H, Pan CH, Kwon OS, Song DG, Han HS. Protein to biomaterials: Unraveling the antiviral and proangiogenic activities of Ac-Tβ 1-17 peptide, a thymosin β4 metabolite, and its implications in peptide-scaffold preparation. Bioact Mater 2025; 49:437-455. [PMID: 40177110 PMCID: PMC11964602 DOI: 10.1016/j.bioactmat.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 01/20/2025] [Accepted: 02/04/2025] [Indexed: 04/05/2025] Open
Abstract
Peptide metabolites are emerging biomolecules with numerous possibilities in biomaterial-based regenerative medicine due to their inherent bioactivities. These small, naturally occurring compounds are intermediates or byproducts of larger proteins and peptides, and they can have profound effects, such as antiviral therapeutics, proangiogenic agents, and regenerative medicinal applications. This study is among the first to focus on using thymosin β4 protein-derived metabolites to pioneer novel applications for peptide metabolites in biomaterials. This study found that the novel peptide metabolite acetyl-thymosin β4 (amino acid 1-17) (Ac-Tβ1-17) exhibited significant protease inhibition activity against SARS-CoV-2, surpassing its precursor protein. Additionally, Ac-Tβ1-17 demonstrated beneficial effects, such as cell proliferation, wound healing, and scavenging of reactive oxygen species (ROS) in human umbilical vein endothelial cells (HUVEC). Integrating Ac-Tβ1-17 into a peptide-based scaffold facilitated cell growth and angiogenesis inside the scaffold and through gradual release into the surrounding environment. The Ac-Tβ1-17 peptide treatment induced significant biochemical responses in HUVEC, increasing Akt, ERK, PI3K, MEK, and Bcl-2 gene expression and proangiogenic proteins. Ac-Tβ1-17 peptide treatment showed similar results in ex vivo by enhancing mouse fetal metatarsal growth and angiogenesis. These findings highlight the potential of natural protein metabolites to generate biologically active peptides, offering a novel strategy for enhancing biomaterial compatibility. This approach holds promise for developing therapeutic biomaterials using peptide metabolites, presenting exciting prospects for future research and applications.
Collapse
Affiliation(s)
- Khandoker Asiqur Rahaman
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Md Sofequl Islam Mukim
- Center for Natural Product Systems Biology, KIST Gangneung Institute of Natural Products, Gangneung, 25451, Republic of Korea
- Natural Product Applied Science, KIST School, University of Science and Technology (UST), Gangneung, 25451, Republic of Korea
| | - Md Lemon Hasan
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, University of Science and Technology (UST), 113 Gwahangno, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Hyeok Kim
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Cheol-Ho Pan
- Center for Natural Product Systems Biology, KIST Gangneung Institute of Natural Products, Gangneung, 25451, Republic of Korea
- Natural Product Applied Science, KIST School, University of Science and Technology (UST), Gangneung, 25451, Republic of Korea
| | - Oh-Seung Kwon
- Doping Control Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, University of Science and Technology (UST), 113 Gwahangno, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Dae-Geun Song
- Center for Natural Product Systems Biology, KIST Gangneung Institute of Natural Products, Gangneung, 25451, Republic of Korea
- Natural Product Applied Science, KIST School, University of Science and Technology (UST), Gangneung, 25451, Republic of Korea
| | - Hyung-Seop Han
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, University of Science and Technology (UST), 113 Gwahangno, Yuseong-gu, Daejeon, 34113, Republic of Korea
- Research and Development Center, Elecell Corporation, Seoul, 02455, Republic of Korea
| |
Collapse
|
2
|
Li R, Zhang C, Xu W, Tao L, Cheng J, Li Z, Zhang Y. Natural pyrethrins induced developmental toxicity of zebrafish swim bladder in vivo and genotoxicity of lung cells in vitro. Toxicol In Vitro 2024; 100:105896. [PMID: 39025157 DOI: 10.1016/j.tiv.2024.105896] [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: 06/13/2024] [Revised: 07/08/2024] [Accepted: 07/13/2024] [Indexed: 07/20/2024]
Abstract
Natural pyrethrins (NPs) are insecticidal compounds isolated and extracted from pyrethrum flowers and are primarily use to control sanitary pests. The lungs become the main target after exposure, and its use may pose potential hazards to respiratory health. Therefore, in this paper, the toxic effects of NPs on human lung cells A549 were investigated and the risk of respiratory toxicity of NPs was studied using zebrafish swim bladder as a model. The results showed that NPs induced cytotoxicity, caused oxidative DNA damage and triggered mitochondria-mediated apoptosis. In addition, exposure to NPs decreased zebrafish embryo survival, hatchability, and heartbeat, and may inhibit normal swim bladder development by disrupting Wnt and Hedgehog signaling pathways. In conclusion, our results suggest that NPs can induce cytotoxicity in A549 in vitro and developmental toxicity in zebrafish in vivo. This study provides a conceptual basis for understanding the mechanisms of toxicity of NPs and assessing respiratory health risks in humans.
Collapse
Affiliation(s)
- Ruirui Li
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Cheng Zhang
- Department of Pathology, UT southwestern Medical Center, Dallas, TX 75390, United States
| | - Wenping Xu
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Liming Tao
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Jiagao Cheng
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Zhong Li
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yang Zhang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
3
|
Tang X, Xiong K, Zeng Y, Fang R. The Mechanism of Zinc Oxide in Alleviating Diarrhea in Piglets after Weaning: A Review from the Perspective of Intestinal Barrier Function. Int J Mol Sci 2024; 25:10040. [PMID: 39337525 PMCID: PMC11432186 DOI: 10.3390/ijms251810040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/12/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Weaning is one of the most challenging phases for piglets, and it is also the time when piglets are the most susceptible to diarrhea, which may result in significant economic losses for pig production. One of the dietary strategies for reducing post-weaning diarrhea (PWD) in piglets is to provide them with a pharmacological dose of zinc oxide (ZnO). However, excessive or long-term usage of high-dose ZnO has significant impacts on pig health and the ecological environment. Therefore, caution should be exercised when considering the use of high-dose ZnO for the prevention or treatment of PWD in piglets. In this paper, the significant role of zinc in animal health, the potential mode of action of ZnO in alleviating diarrhea, and the impact of innovative, highly efficient ZnO alternatives on the regulation of piglet diarrhea were reviewed to offer insights into the application of novel ZnO in pig production.
Collapse
Affiliation(s)
- Xiaopeng Tang
- State Engineering Technology Institute for Karst Desertfication Control, School of Karst Science, Guizhou Normal University, Guiyang 550025, China;
| | - Kangning Xiong
- State Engineering Technology Institute for Karst Desertfication Control, School of Karst Science, Guizhou Normal University, Guiyang 550025, China;
| | - Yan Zeng
- Key Laboratory for Information System of Mountainous Areas and Protection of Ecological Environment, Guizhou Normal University, Guiyang 550025, China;
| | - Rejun Fang
- College of Animal Science, Hunan Agricultural University, Changsha 410128, China
| |
Collapse
|
4
|
Wang B, Ma Y, Zhang Y, Yin X. Therapeutic potential of ASK1 activators in cancer treatment: Current insights and future directions. Biomed Pharmacother 2024; 178:117214. [PMID: 39079264 DOI: 10.1016/j.biopha.2024.117214] [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: 05/16/2024] [Revised: 07/18/2024] [Accepted: 07/26/2024] [Indexed: 08/25/2024] Open
Abstract
Apoptosis signal-regulated kinase 1 (ASK1) is a member of the mitogen-activated protein kinase kinase (MAP3K) family, whose activation and regulation are intricately associated with apoptosis. ASK1 is activated in response to oxidative stress, among other stimuli, subsequently triggering downstream JNK, p38 MAPK, and mitochondria-dependent apoptotic signaling, which participate in the initiation of tumor cell apoptosis induced by various stimuli. Research has shown that ASK1 plays a crucial role in the apoptosis of lung cancer, breast cancer, and liver cancer cells. Currently, the investigation of effective ASK1 activators is a hot topic in research on tumor cell apoptosis. Synthetic compounds such as human β-defensin, triazolothiazide derivatives and heat shock protein 27 inhibitors; natural compounds such as quercetin, Laminarina japonica polysaccharide-1 peptide and theabrownin; and nanomedicines such as cerium oxide nanoparticles, magnetite FeO nanoparticles and silver nanoparticles can activate ASK1 and induce apoptosis in various tumor cells. This review extensively investigates the roles and activation mechanisms of ASK1, explores its impact on a variety of apoptotic signaling pathways, and discusses the potential therapeutic applications of various ASK1 activators in cancer treatment. In addition, this paper provides an in-depth discussion of the future development of this field and proposes a promising method for further research and clinical progress.
Collapse
Affiliation(s)
- Bo Wang
- Department of Integrated Chinese and Western Medicine, Jilin Cancer Hospital, Changchun 130103, China
| | - Ying Ma
- Department of Integrated Chinese and Western Medicine, Jilin Cancer Hospital, Changchun 130103, China
| | - Yue Zhang
- Department of Integrated Chinese and Western Medicine, Jilin Cancer Hospital, Changchun 130103, China.
| | - Xunzhe Yin
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.
| |
Collapse
|
5
|
Jiang D, Guo J, Liu Y, Li W, Lu D. Glycolysis: an emerging regulator of osteoarthritis. Front Immunol 2024; 14:1327852. [PMID: 38264652 PMCID: PMC10803532 DOI: 10.3389/fimmu.2023.1327852] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 12/20/2023] [Indexed: 01/25/2024] Open
Abstract
Osteoarthritis (OA) has been a leading cause of disability in the elderly and there remains a lack of effective therapeutic approaches as the mechanisms of pathogenesis and progression have yet to be elucidated. As OA progresses, cellular metabolic profiles and energy production are altered, and emerging metabolic reprogramming highlights the importance of specific metabolic pathways in disease progression. As a crucial part of glucose metabolism, glycolysis bridges metabolic and inflammatory dysfunctions. Moreover, the glycolytic pathway is involved in different areas of metabolism and inflammation, and is associated with a variety of transcription factors. To date, it has not been fully elucidated whether the changes in the glycolytic pathway and its associated key enzymes are associated with the onset or progression of OA. This review summarizes the important role of glycolysis in mediating cellular metabolic reprogramming in OA and its role in inducing tissue inflammation and injury, with the aim of providing further insights into its pathological functions and proposing new targets for the treatment of OA.
Collapse
Affiliation(s)
- Dingming Jiang
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jianan Guo
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yingquan Liu
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Wenxin Li
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Hangzhou Linping District Nanyuan Street Community Health Center, Hangzhou, China
| | - Dezhao Lu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
6
|
Abdel Fattah S, Ibrahim MEED, El-Din SS, Emam HS, Algaleel WAA. Possible therapeutic role of zinc oxide nanoparticles versus vanillic acid in testosterone-induced benign prostatic hyperplasia in adult albino rat: A histological, immunohistochemical and biochemical study. Life Sci 2023; 334:122190. [PMID: 37866805 DOI: 10.1016/j.lfs.2023.122190] [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: 07/25/2023] [Revised: 10/12/2023] [Accepted: 10/16/2023] [Indexed: 10/24/2023]
Abstract
BACKGROUND The search for alternative therapies for treatment of Benign prostatic hyperplasia (BPH) has been increasingly studied to avoid the common adverse effects of the usual regimens. Therefore, this study aimed at delineating possible mechanisms of benign prostatic hyperplasia (BPH) and possible therapeutic role of zinc oxide nanoparticles (ZnO-NPs) versus vanillic acid. METHODS Forty rats were divided into five groups: control, sham control, Testosterone-induced BPH, BPH and Zn-NPs, and BPH and vanillic acid. Light microscopic, immune-histochemical; PCNA, Bcl-2, Bax, caspase-3, p-Akt and p-mTOR, histomorphometric analysis, MDA/SOD and GPx and were done. Gene expression of p-Akt, p-mTOR and survivin were evaluated. RESULTS Application of zinc oxide nanoparticles as well as vanillic acid significantly reduced prostatic index, epithelial thickness, stromal collagen fibers, expression of PCNA, Bcl2, p-Akt, p-mTOR and MDA tissue level (p < 0.05). Whereas expression of Bax and caspase 3, and tissue levels of SOD and GPx were significantly increased in groups treated with Zno-Nps and vanillic acid compared to that of BPH group. Zinc oxide nanoparticles showed a better effect than vanillic acid in alleviating BPH. CONCLUSION These findings suggested that ZnO-NPs as well as VA ameliorated the histolo-pathological and biochemical effects of induced BPH, moreover they improved the proapoptotic and antioxidant parameters which ere induced in BPH. It is recommended to search for new agents to prevent the development and progression of BPH.
Collapse
Affiliation(s)
- Shereen Abdel Fattah
- Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Egypt
| | | | - Shimaa Saad El-Din
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Egypt.
| | - Hadeel Sayed Emam
- Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Egypt
| | | |
Collapse
|
7
|
Wang X, Geng L, Wu M, Xu W, Cheng J, Li Z, Tao L, Zhang Y. Molecular mechanisms of cardiotoxicity induced by acetamide and its chiral isomers. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 900:166349. [PMID: 37598958 DOI: 10.1016/j.scitotenv.2023.166349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 08/22/2023]
Abstract
Acetamide (ACT) is used in a racemic form, and the considerable residues of this compound in the environment raise potential safety concerns for human health. We investigated the toxicity of ACT and its chiral isomers on human cardiomyocyte (AC16) cell line and zebrafish embryonic heart, and found that (+)-S-ACT was the main component causing cardiac toxicity. Our findings indicate that the IC50 of (±)-Rac-ACT on AC16 cells was 20.19 μg/mL. (-)-R-ACT, (±)-Rac-ACT, and (+)-S-ACT caused DNA damage and apoptosis in AC16 cells at this concentration. The underlying molecular mechanism may involve the induction of reactive oxygen species (ROS). The accumulation of ROS results in a decline in mitochondrial membrane potential (MMP) and prompts the release of cytochrome c (cyt c) from the mitochondria. This cascade of events ultimately activates the caspase-3 and caspase-9 signaling pathways, resulting in apoptosis. Furthermore, in vivo observations in zebrafish hearts demonstrated caspase-3 activation and the presence of the DNA damage marker (γH2AX), indicating that (+)-S-ACT is more toxic to cardiomyocytes than (-)-R-ACT and (±)-Rac-ACT. These findings suggest that (+)-S-ACT may be the primary component responsible for the toxicity of (±)-Rac-ACT in AC16 cells. Overall, these findings raise public awareness regarding the risks associated with chiral isomeric pesticides and provide a scientific foundation for their appropriate use.
Collapse
Affiliation(s)
- Xin Wang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Li Geng
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Mengqi Wu
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Wenping Xu
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Jiagao Cheng
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Zhong Li
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Liming Tao
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yang Zhang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
8
|
Jiang H, Zhang Y, Wang X, Wang G, Zhu J, Sun J, Zhang M, Li Y, Xu S, Hu J, Wang Y. Establishment and characterization of a liver cell line from silver pomfret (Pampus argenteus) for studying fish health. JOURNAL OF FISH DISEASES 2023; 46:1193-1205. [PMID: 37496293 DOI: 10.1111/jfd.13839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 07/28/2023]
Abstract
Fish cell lines have become a useful tool to study in resource conservation, genetic breeding, diseases control, and environmental pollutants detection. The silver pomfret (Pampus argenteus) is a high-valued marine fish species in aquaculture, which is seriously threatened by various fish diseases. In this study, a new cell line derived from P. argenteus liver (PaL) was established and characterized. PaL cells mainly consisted of fibroblast-like morphology and multiplied well in Leibovitz's L-15 medium supplemented with 15% foetal bovine serum and 3 ng/mL basic fibroblast growth factor at 22°C. Amplification of the Cyt b gene confirmed that the origin of PaL cells as P. argenteus. Chromosome analysis revealed that PaL cells had a diploid Karyotyp. The PaL cells were efficiently transfected with pEGFP-N3 plasmids, indicating its potential application in foreign gene manipulation studies. The PaL cells were found to be susceptible to red sea bream iridovirus (RSIV) and the expression of immune-related gene (TLR5) and apoptosis-related genes (Bax, Cyt c3, CASP9) were upregulated. Furthermore, lipopolysaccharide and palmitic acid (PA) treatments decreased cell viability and up-regulated the expression of inflammation related genes (IL-8, IL-1β). Meanwhile, PA incubation induced cell apoptosis by Bcl-2-regulated caspase activation. In conclusion, the newly established PaL cell line will be an appropriate in vitro tool for viral propagation and immune response.
Collapse
Affiliation(s)
- Huan Jiang
- Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China
- Key Laboratory of Aquacultural Biotechnology, Ningbo University, Ningbo, China
- Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Ningbo, China
| | - Youyi Zhang
- Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China
- Key Laboratory of Aquacultural Biotechnology, Ningbo University, Ningbo, China
- Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Ningbo, China
| | - Xiangbing Wang
- Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China
- Key Laboratory of Aquacultural Biotechnology, Ningbo University, Ningbo, China
- Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Ningbo, China
| | - Guanlin Wang
- Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China
- Key Laboratory of Aquacultural Biotechnology, Ningbo University, Ningbo, China
- Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Ningbo, China
| | - Jiajie Zhu
- Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China
- Key Laboratory of Aquacultural Biotechnology, Ningbo University, Ningbo, China
- Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Ningbo, China
| | - Jiachu Sun
- Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China
- Key Laboratory of Aquacultural Biotechnology, Ningbo University, Ningbo, China
- Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Ningbo, China
| | - Man Zhang
- Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China
- Key Laboratory of Aquacultural Biotechnology, Ningbo University, Ningbo, China
- Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Ningbo, China
| | - Yaya Li
- Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China
- Key Laboratory of Aquacultural Biotechnology, Ningbo University, Ningbo, China
- Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Ningbo, China
| | - Shanliang Xu
- Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China
- Key Laboratory of Aquacultural Biotechnology, Ningbo University, Ningbo, China
- Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Ningbo, China
| | - Jiabao Hu
- Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China
- Key Laboratory of Aquacultural Biotechnology, Ningbo University, Ningbo, China
- Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Ningbo, China
| | - Yajun Wang
- Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China
- Key Laboratory of Aquacultural Biotechnology, Ningbo University, Ningbo, China
- Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Ningbo, China
| |
Collapse
|
9
|
Han D, Chen R, Kan H, Xu Y. The bio-distribution, clearance pathways, and toxicity mechanisms of ambient ultrafine particles. ECO-ENVIRONMENT & HEALTH (ONLINE) 2023; 2:95-106. [PMID: 38074989 PMCID: PMC10702920 DOI: 10.1016/j.eehl.2023.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 06/02/2023] [Accepted: 06/09/2023] [Indexed: 02/17/2024]
Abstract
Ambient particles severely threaten human health worldwide. Compared to larger particles, ultrafine particles (UFPs) are highly concentrated in ambient environments, have a larger specific surface area, and are retained for a longer time in the lung. Recent studies have found that they can be transported into various extra-pulmonary organs by crossing the air-blood barrier (ABB). Therefore, to understand the adverse effects of UFPs, it is crucial to thoroughly investigate their bio-distribution and clearance pathways in vivo after inhalation, as well as their toxicological mechanisms. This review highlights emerging evidence on the bio-distribution of UFPs in pulmonary and extra-pulmonary organs. It explores how UFPs penetrate the ABB, the blood-brain barrier (BBB), and the placental barrier (PB) and subsequently undergo clearance by the liver, kidney, or intestine. In addition, the potential underlying toxicological mechanisms of UFPs are summarized, providing fundamental insights into how UFPs induce adverse health effects.
Collapse
Affiliation(s)
- Dongyang Han
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Renjie Chen
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Haidong Kan
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| | - Yanyi Xu
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai 200032, China
| |
Collapse
|
10
|
Kitala K, Tanski D, Godlewski J, Krajewska-Włodarczyk M, Gromadziński L, Majewski M. Copper and Zinc Particles as Regulators of Cardiovascular System Function-A Review. Nutrients 2023; 15:3040. [PMID: 37447366 DOI: 10.3390/nu15133040] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 06/30/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Copper and zinc are micronutrients that play a crucial role in many cellular pathways, act as cofactors in enzymatic systems, and hence, modulate enzyme activity. The regulation of these elements in homeostasis is precisely controlled by various mechanisms. Superoxide dismutase (SOD) is an enzyme requiring both copper and zinc for proper functioning. Additionally, there is an interaction between the concentrations of copper and zinc. Dietary ingestion of large amounts of zinc augments intestinal absorption of this trace element, resulting in copper deficiency secondary to zinc excess. The presence of an overabundance of copper and zinc has a detrimental impact on the cardiovascular system; however, the impact on vascular contractility varies. Copper plays a role in the modulation of vascular remodeling in the cardiac tissue, and the phenomenon of cuproptosis has been linked to the pathogenesis of coronary artery disease. The presence of copper has an observable effect on the vasorelaxation mediated by nitric oxide. The maintenance of proper levels of zinc within an organism influences SOD and is essential in the pathogenesis of myocardial ischemia/reperfusion injury. Recently, the effects of metal nanoparticles have been investigated due to their unique characteristics. On the other hand, dietary introduction of metal nanoparticles may result in vascular dysfunction, oxidative stress, and cellular DNA damage. Copper and zinc intake affect cardiovascular function, but more research is needed.
Collapse
Affiliation(s)
- Klaudia Kitala
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Warmia and Mazury in Olsztyn, 10-082 Olsztyn, Poland
| | - Damian Tanski
- Department of Human Histology and Embryology, Faculty of Medicine, University of Warmia and Mazury in Olsztyn, 10-082 Olsztyn, Poland
| | - Janusz Godlewski
- Department of Human Histology and Embryology, Faculty of Medicine, University of Warmia and Mazury in Olsztyn, 10-082 Olsztyn, Poland
| | - Magdalena Krajewska-Włodarczyk
- Department of Mental and Psychosomatic Diseases, Faculty of Medicine, University of Warmia and Mazury in Olsztyn, 10-082 Olsztyn, Poland
| | - Leszek Gromadziński
- Department of Cardiology and Internal Medicine, Faculty of Medicine, University of Warmia and Mazury in Olsztyn, 10-082 Olsztyn, Poland
| | - Michał Majewski
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Warmia and Mazury in Olsztyn, 10-082 Olsztyn, Poland
| |
Collapse
|
11
|
Ma JY, Li WY, Yang ZY, Su JZ, Li L, Deng YR, Tuo YF, Niu YY, Xiang P. The spatial distribution, health risk, and cytotoxicity of metal(loid)s in contaminated field soils: The role of Cd in human gastric cells damage. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 878:162942. [PMID: 36940749 DOI: 10.1016/j.scitotenv.2023.162942] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/10/2023] [Accepted: 03/14/2023] [Indexed: 05/13/2023]
Abstract
The spatial distribution and pollution level of heavy metal(loid)s in soil (0-6 m) from a typical industrial region in Jiangmen City, Southeast China was investigated. Their bioaccessibility, health risk, and human gastric cytotoxicity in topsoil were also evaluated using an in vitro digestion/human cell model. The average concentrations of Cd (87.52 mg/kg), Co (106.9 mg/kg), and Ni (1007 mg/kg) exceeded the risk screening values. The distribution profiles of metal(loid)s showed a downward migration trend to reach a depth of 2 m. The highest contamination was found in topsoil (0-0.5 m), with the concentrations of As, Cd, Co, and Ni being 46.98, 348.28, 317.44, and 2395.60 mg/kg, respectively, while Cd showed the highest bioaccessibility in the gastric phase (72.80 %), followed by Co (21.08 %), Ni (18.27 %), and As (5.26 %) and unacceptable carcinogenic risk. Moreover, the gastric digesta of topsoil suppressed the cell viability and triggered cell apoptosis, evidenced by disruption of mitochondrial transmembrane potential and increase of Cytochrome c (Cyt c) and Caspases 3/9 mRNA expression. Bioaccessible Cd in topsoil was responsible for those adverse effects. Our data suggest the importance to reduce Cd in the soil to decrease its adverse impacts on the human stomach.
Collapse
Affiliation(s)
- Jiao-Yang Ma
- Yunnan Province Innovative Research Team of Environmental pollution, Food Safety, and Human Health, Institute of Environmental Remediation and Human Health, School of Ecology and Environment, Southwest Forestry University, Kunming 650224, China
| | - Wei-Yu Li
- Yunnan Province Innovative Research Team of Environmental pollution, Food Safety, and Human Health, Institute of Environmental Remediation and Human Health, School of Ecology and Environment, Southwest Forestry University, Kunming 650224, China; Guangdong Key Laboratory of Contaminated Environmental Management and Remediation, Guangdong Provincial Academy of Environmental Science, Guangzhou 510000, China
| | - Zi-Yue Yang
- Yunnan Province Innovative Research Team of Environmental pollution, Food Safety, and Human Health, Institute of Environmental Remediation and Human Health, School of Ecology and Environment, Southwest Forestry University, Kunming 650224, China
| | - Jin-Zhou Su
- Yunnan Province Innovative Research Team of Environmental pollution, Food Safety, and Human Health, Institute of Environmental Remediation and Human Health, School of Ecology and Environment, Southwest Forestry University, Kunming 650224, China
| | - Li Li
- Precious Metal Testing Co. LTD of Yunnan Gold Mining Group, Kunming 650215, China
| | - Yi-Rong Deng
- Guangdong Key Laboratory of Contaminated Environmental Management and Remediation, Guangdong Provincial Academy of Environmental Science, Guangzhou 510000, China
| | - Yun-Fei Tuo
- Yunnan Province Innovative Research Team of Environmental pollution, Food Safety, and Human Health, Institute of Environmental Remediation and Human Health, School of Ecology and Environment, Southwest Forestry University, Kunming 650224, China
| | - You-Ya Niu
- School of Basic Medical Sciences, Hunan University of Medicine, Huaihua 418000, China.
| | - Ping Xiang
- Yunnan Province Innovative Research Team of Environmental pollution, Food Safety, and Human Health, Institute of Environmental Remediation and Human Health, School of Ecology and Environment, Southwest Forestry University, Kunming 650224, China.
| |
Collapse
|
12
|
Misra SK, Rosenholm JM, Pathak K. Functionalized and Nonfunctionalized Nanosystems for Mitochondrial Drug Delivery with Metallic Nanoparticles. Molecules 2023; 28:4701. [PMID: 37375256 DOI: 10.3390/molecules28124701] [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: 05/03/2023] [Revised: 06/04/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Background: The application of metallic nanoparticles as a novel therapeutic tool has significant potential to facilitate the treatment and diagnosis of mitochondria-based disorders. Recently, subcellular mitochondria have been trialed to cure pathologies that depend on their dysfunction. Nanoparticles made from metals and their oxides (including gold, iron, silver, platinum, zinc oxide, and titanium dioxide) have unique modi operandi that can competently rectify mitochondrial disorders. Materials: This review presents insight into the recent research reports on exposure to a myriad of metallic nanoparticles that can alter the dynamic ultrastructure of mitochondria (via altering metabolic homeostasis), as well as pause ATP production, and trigger oxidative stress. The facts and figures have been compiled from more than a hundred PubMed, Web of Science, and Scopus indexed articles that describe the essential functions of mitochondria for the management of human diseases. Result: Nanoengineered metals and their oxide nanoparticles are targeted at the mitochondrial architecture that partakes in the management of a myriad of health issues, including different cancers. These nanosystems not only act as antioxidants but are also fabricated for the delivery of chemotherapeutic agents. However, the biocompatibility, safety, and efficacy of using metal nanoparticles is contested among researchers, which will be discussed further in this review.
Collapse
Affiliation(s)
- Shashi Kiran Misra
- School of Pharmaceutical Sciences, CSJM University Kanpur, Kanpur 208024, India
| | - Jessica M Rosenholm
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, BioCity (3rd Floor), Tykistökatu, 6A, 20520 Turku, Finland
| | - Kamla Pathak
- Faculty of Pharmacy, Uttar Pradesh University of Medical Sciences, Saifai, Etawah 206130, India
| |
Collapse
|
13
|
Liu S, Pan Y, Li T, Zou M, Liu W, Li Q, Wan H, Peng J, Hao L. The Role of Regulated Programmed Cell Death in Osteoarthritis: From Pathogenesis to Therapy. Int J Mol Sci 2023; 24:ijms24065364. [PMID: 36982438 PMCID: PMC10049357 DOI: 10.3390/ijms24065364] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
Osteoarthritis (OA) is a worldwide chronic disease that can cause severe inflammation to damage the surrounding tissue and cartilage. There are many different factors that can lead to osteoarthritis, but abnormally progressed programmed cell death is one of the most important risk factors that can induce osteoarthritis. Prior studies have demonstrated that programmed cell death, including apoptosis, pyroptosis, necroptosis, ferroptosis, autophagy, and cuproptosis, has a great connection with osteoarthritis. In this paper, we review the role of different types of programmed cell death in the generation and development of OA and how the different signal pathways modulate the different cell death to regulate the development of OA. Additionally, this review provides new insights into the radical treatment of osteoarthritis rather than conservative treatment, such as anti-inflammation drugs or surgical operation.
Collapse
Affiliation(s)
- Suqing Liu
- Department of Orthopedics, Second Affifiliated Hospital of Nanchang University, Nanchang 330006, China
- Queen Marry College, Nanchang University, Nanchang 330006, China
| | - Yurong Pan
- Department of Orthopedics, Second Affifiliated Hospital of Nanchang University, Nanchang 330006, China
- Queen Marry College, Nanchang University, Nanchang 330006, China
| | - Ting Li
- Department of Orthopedics, Second Affifiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Mi Zou
- Department of Orthopedics, Second Affifiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Wenji Liu
- Department of Orthopedics, Second Affifiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Qingqing Li
- Department of Orthopedics, Second Affifiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Huan Wan
- Department of Orthopedics, Second Affifiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Jie Peng
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
- Correspondence: (J.P.); (L.H.); Tel.: +86-15983280459 (J.P.); +86-13607008562 (L.H.)
| | - Liang Hao
- Department of Orthopedics, Second Affifiliated Hospital of Nanchang University, Nanchang 330006, China
- Correspondence: (J.P.); (L.H.); Tel.: +86-15983280459 (J.P.); +86-13607008562 (L.H.)
| |
Collapse
|
14
|
Sun X, Zhang B, Luo L, Yang Y, He B, Zhang Q, Wang L, Xu S, Zheng P, Zhu W. Design, synthesis and pharmacological evaluation of 2-arylurea-1,3,5-triazine derivative (XIN-9): A novel potent dual PI3K/mTOR inhibitor for cancer therapy. Bioorg Chem 2022; 129:106157. [PMID: 36209563 DOI: 10.1016/j.bioorg.2022.106157] [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: 07/10/2022] [Revised: 08/25/2022] [Accepted: 09/12/2022] [Indexed: 01/03/2023]
Abstract
Blocking the PI3K/AKT/mTOR pathway has been widely recognized as an attractive cancer therapeutic strategy because of its crucial role in cell growth and survival. In this study, a novel series of 2-arylurea-1,3,5-triazine derivatives had been synthesized and evaluated as highly potent PI3K and mTOR inhibitors. The new compounds exhibited cytotoxic activities against MCF-7, Hela and A549 cancer cell lines (IC50 = 0.03-36.54 μM). The most promising compound XIN-9 exhibited potent inhibition against PI3K and mTOR kinase (IC50 = 23.8 and 10.9 nM). Mechanistic study using real-time PCR revealed the ability of XIN-9 to inhibit PI3K and mTOR. In addition, compound XIN-9 arrested the cell cycle of MCF-7 cells at the G0/G1 phase. XIN-9 also caused a significant dose-dependent increase of early and late apoptotic events. Molecular docking analysis revealed a high binding affinity for XIN-9 toward PI3K and mTOR. Following in vitro studies, XIN-9 was further evaluated in MCF-7 xenograft models to show significant in vivo anticancer efficacies with tumor growth inhibitions of 41.67% (po, 75 mg/kg). Overall, this work indicated that compound XIN-9 represents a potential anticancer targeting PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Xin Sun
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi 330013, China
| | - Binliang Zhang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi 330013, China; School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510000, China
| | - Leixuan Luo
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi 330013, China
| | - Yang Yang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi 330013, China
| | - Bin He
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi 330013, China
| | - Qian Zhang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi 330013, China; School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510000, China
| | - Linxiao Wang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi 330013, China
| | - Shan Xu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi 330013, China.
| | - Pengwu Zheng
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi 330013, China.
| | - Wufu Zhu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi 330013, China.
| |
Collapse
|
15
|
Liu R, Huang SS, Shi H, Chang S, Ge J. Alpha-lipoic acid protects against aortic aneurysm and dissection by improving vascular smooth muscle cell function. Life Sci 2022; 311:121159. [DOI: 10.1016/j.lfs.2022.121159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/26/2022] [Accepted: 11/05/2022] [Indexed: 11/10/2022]
|
16
|
Zhao Y, Zhang H, Hao D, Wang J, Zhang D, Sun Z, Liu C. Selenium Alleviates Chromium(VI)-Induced Ileum Damage and Cecal Microbial Disturbances in Mice. Biol Trace Elem Res 2022; 200:4750-4761. [PMID: 35031963 DOI: 10.1007/s12011-021-03061-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 12/08/2021] [Indexed: 01/14/2023]
Abstract
Hexavalent chromium [Cr(VI)] is one of the most common environmental contaminants caused by its broad industrial applications. Importantly, exposure to Cr(VI) induces oxidative damage and apoptosis in animal cells. Studies have shown that selenium (Se) can alleviate the toxic effects of Cr(VI) by functioning as an antioxidant and/or by chelating Cr(VI) into biologically inert complexes, but the underlying mechanism remains unknown. Here, we evaluated whether Se can ameliorate ileum damage and cecal microbial disturbances induced by Cr(VI) in vivo. Mice administered Cr(VI) for 30 days presented histopathological damage, reduced responses to oxidative stress, and increased expression of apoptosis-related genes in the ileum compared with those in the control (non-exposed) group. Se alleviated the histopathological damage and decreased the oxidative stress and apoptosis induced by Cr(VI) in the ileum. In addition, Cr(VI) disturbed cecal microflora, and it was partially reversed by Se treatment. These findings demonstrate that the damaging and potentially pathological effects of Cr(VI) on the ileum and cecal microflora can be effectively alleviated with Se treatment.
Collapse
Affiliation(s)
- Yanbing Zhao
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, 030801, People's Republic of China
| | - Huan Zhang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, 030801, People's Republic of China
| | - Dezheng Hao
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, 030801, People's Republic of China
| | - Jinglu Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, 030801, People's Republic of China
| | - Ding Zhang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, 030801, People's Republic of China
| | - Zilong Sun
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, 030801, People's Republic of China
| | - Ci Liu
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, 030801, People's Republic of China.
| |
Collapse
|
17
|
Zhang XH, Li Y, Zhou L, Tian GP. Interleukin-38 in atherosclerosis. Clin Chim Acta 2022; 536:86-93. [PMID: 36150521 DOI: 10.1016/j.cca.2022.09.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 09/15/2022] [Accepted: 09/15/2022] [Indexed: 11/25/2022]
Abstract
Chronic inflammation caused by immune cells and their mediators is a characteristic of atherosclerosis. Interleukin-38 (IL-38), a member of the IL-1 family, exerts multiple anti-inflammatory effects via specific ligand-receptor interactions. Upon recognizing a specific receptor, IL-38 restrains mitogen-activated protein kinase (MAPK), nuclear factor kappa B (NK-κB), or other inflammation-related signaling pathways in inflammatory disease. Further research has shown that IL-38 also displays anti-atherosclerotic effects and reduces the occurrence and risk of cardiovascular events. On the one hand, IL-38 can regulate innate and adaptive immunity to inhibit inflammation, reduce pathological neovascularization, and inhibit apoptosis. On the other hand, it can curb obesity, reduce hyperlipidemia, and restrain insulin resistance to reduce cardiovascular disease risk. Therefore, this article expounds on the vital function of IL-38 in the development of atherosclerosis to provide a theoretical basis for further in-depth studies of IL-38 and insights on the prophylaxis and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Xiao-Hong Zhang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yu Li
- Department of Orthopaedics, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430016, China
| | - Li Zhou
- Department of Pathology, Chongqing Public Health Medical Center, Southwest University Public Health Hospital, Chongqing 400036, China.
| | - Guo-Ping Tian
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
18
|
Luo X, Xie D, Wu T, Xu W, Meng Q, Cao K, Hu J. Evaluation of the protective roles of alpha-lipoic acid supplementation on nanomaterial-induced toxicity: A meta-analysis of in vitro and in vivo studies. Front Nutr 2022; 9:991524. [PMID: 36147302 PMCID: PMC9486203 DOI: 10.3389/fnut.2022.991524] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/17/2022] [Indexed: 01/02/2023] Open
Abstract
Extensive exposure to nanomaterials causes oxidative stress and inflammation in various organs and leads to an increased risk of adverse health outcomes; therefore, how to prevent the toxic effects are of great concern to human. Alpha-lipoic acid (ALA) has anti-oxidant and anti-inflammatory activities, suggesting it may be effective to prevent nanomaterial-induced toxicity. However, the results obtained in individual studies remained controversial. We aimed to comprehensively evaluate the effects of ALA supplementation on nanomaterial-induced toxicity by performing a meta-analysis. Databases of PubMed, EMBASE, and Cochrane Library were searched up to May 2022. STATA 15.0 software was used for statistical analysis. Twelve studies were included. Meta-analysis of eight in vivo studies showed ALA supplementation could exert significant effects on nanomaterial-induced oxidative stress (by reducing MDA, ROS and increasing GSH, CAT, GPx, and SOD), inflammation (by downregulating NO, IgG, TNF-α, IL-6, and CRP), apoptosis (by activation of pro-apoptotic caspase-3), DNA damage (by a reduction in the tail length) and organ damage (by a decrease in the liver biomarker ALT and increases in brain neuron biomarker AChE and heart biomarker CPK). Pooled analysis of four in vitro studies indicated ALA intervention increased cell viability, decreased ROS levels, inhibited cell apoptosis and chelated metal ions. Subgroup analyses revealed changing the levels of GSH, IL-6, and metal ions were the main protective mechanisms of ALA supplementation because they were not changed by any subgroup factors. In conclusion, ALA supplementation may represent a potential strategy for the prevention of the toxicity induced by nanomaterials.
Collapse
Affiliation(s)
- Xiaogang Luo
- College of Textile and Clothing Engineering, Soochow University, Suzhou, China
- *Correspondence: Xiaogang Luo,
| | - Dongli Xie
- College of Textile and Clothing Engineering, Soochow University, Suzhou, China
| | - Tong Wu
- Shanghai Jing Rui Yang Industrial Co., Ltd, Shanghai, China
| | - Wei Xu
- Shanghai Nutri-woods Bio-Technology Co., Ltd, Shanghai, China
| | - Qingyang Meng
- Shanghai Pechoin Daily Chemical Co., Ltd, Shanghai, China
| | - Kangli Cao
- Shanghai Institute of Spacecraft Equipment, Shanghai, China
| | - Jianchen Hu
- College of Textile and Clothing Engineering, Soochow University, Suzhou, China
- Jianchen Hu,
| |
Collapse
|
19
|
Abstract
The constant evolution and applications of metallic nanoparticles (NPs) make living organisms more susceptible to being exposed to them. Among the most used are zinc oxide nanoparticles (ZnO-NPs). Therefore, understanding the molecular effects of ZnO-NPs in biological systems is extremely important. This review compiles the main mechanisms that induce cell toxicity by exposure to ZnO-NPs and reported in vitro research models, with special attention to mitochondrial damage. Scientific evidence indicates that in vitro ZnO-NPs have a cytotoxic effect that depends on the size, shape and method of synthesis of ZnO-NPs, as well as the function of the cells to which they are exposed. ZnO-NPs come into contact with the extracellular region, leading to an increase in intracellular [Zn2+] levels. The mechanism by which intracellular ZnO-NPs come into contact with organelles such as mitochondria is still unclear. The mitochondrion is a unique organelle considered the “power station” in the cells, participates in numerous cellular processes, such as cell survival/death, multiple biochemical and metabolic processes, and holds genetic material. ZnO-NPs increase intracellular levels of reactive oxygen species (ROS) and, in particular, superoxide levels; they also decrease mitochondrial membrane potential (MMP), which affects membrane permeability and leads to cell death. ZnO-NPs also induced cell death through caspases, which involve the intrinsic apoptotic pathway. The expression of pro-apoptotic genes after exposure to ZnO-NPs can be affected by multiple factors, including the size and morphology of the NPs, the type of cell exposed (healthy or tumor), stage of development (embryonic or differentiated), energy demand, exposure time and, no less relevant, the dose. To prevent the release of pro-apoptotic proteins, the damaged mitochondrion is eliminated by mitophagy. To replace those mitochondria that underwent mitophagy, the processes of mitochondrial biogenesis ensure the maintenance of adequate levels of ATP and cellular homeostasis.
Collapse
|
20
|
Cameron SJ, Sheng J, Hosseinian F, Willmore WG. Nanoparticle Effects on Stress Response Pathways and Nanoparticle-Protein Interactions. Int J Mol Sci 2022; 23:7962. [PMID: 35887304 PMCID: PMC9323783 DOI: 10.3390/ijms23147962] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/01/2022] [Accepted: 07/11/2022] [Indexed: 12/12/2022] Open
Abstract
Nanoparticles (NPs) are increasingly used in a wide variety of applications and products; however, NPs may affect stress response pathways and interact with proteins in biological systems. This review article will provide an overview of the beneficial and detrimental effects of NPs on stress response pathways with a focus on NP-protein interactions. Depending upon the particular NP, experimental model system, and dose and exposure conditions, the introduction of NPs may have either positive or negative effects. Cellular processes such as the development of oxidative stress, the initiation of the inflammatory response, mitochondrial function, detoxification, and alterations to signaling pathways are all affected by the introduction of NPs. In terms of tissue-specific effects, the local microenvironment can have a profound effect on whether an NP is beneficial or harmful to cells. Interactions of NPs with metal-binding proteins (zinc, copper, iron and calcium) affect both their structure and function. This review will provide insights into the current knowledge of protein-based nanotoxicology and closely examines the targets of specific NPs.
Collapse
Affiliation(s)
- Shana J. Cameron
- Department of Chemistry, Carleton University, Ottawa, ON K1S 5B6, Canada; (S.J.C.); (F.H.)
| | - Jessica Sheng
- Department of Biology, Carleton University, Ottawa, ON K1S 5B6, Canada;
| | - Farah Hosseinian
- Department of Chemistry, Carleton University, Ottawa, ON K1S 5B6, Canada; (S.J.C.); (F.H.)
| | - William G. Willmore
- Department of Chemistry, Carleton University, Ottawa, ON K1S 5B6, Canada; (S.J.C.); (F.H.)
- Department of Biology, Carleton University, Ottawa, ON K1S 5B6, Canada;
- Institute of Biochemistry, Carleton University, Ottawa, ON K1S 5B6, Canada
| |
Collapse
|
21
|
Metal nanoparticles: biomedical applications and their molecular mechanisms of toxicity. CHEMICAL PAPERS 2022. [DOI: 10.1007/s11696-022-02351-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
22
|
Novel Insight into the Potential Role of Acylglycerophosphate Acyltransferases Family Members on Triacylglycerols Synthesis in Buffalo. Int J Mol Sci 2022; 23:ijms23126561. [PMID: 35743005 PMCID: PMC9224252 DOI: 10.3390/ijms23126561] [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] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/04/2022] [Accepted: 06/10/2022] [Indexed: 02/01/2023] Open
Abstract
Acylglycerophosphate acyltransferases (AGPATs) are the rate-limiting enzymes for the de novo pathway of triacylglycerols (TAG) synthesis. Although AGPATs have been extensively explored by evolution, expression and functional studies, little is known on functional characterization of how many members of the AGPAT family are involved in TAG synthesis and their impact on the cell proliferation and apoptosis. Here, 13 AGPAT genes in buffalo were identified, of which 12 AGPAT gene pairs were orthologous between buffalo and cattle. Comparative transcriptomic analysis and real-time quantitative reverse transcription PCR (qRT-PCR) further showed that both AGPAT1 and AGPAT6 were highly expressed in milk samples of buffalo and cattle during lactation. Knockdown of AGPAT1 or AGPAT6 significantly decreased the TAG content of buffalo mammary epithelial cells (BuMECs) and bovine mammary epithelial cells (BoMECs) by regulating lipogenic gene expression (p < 0.05). Knockdown of AGPAT1 or AGPAT6 inhibited proliferation and apoptosis of BuMECs through the expression of marker genes associated with the proliferation and apoptosis (p < 0.05). Our data confirmed that both AGPAT1 and AGPAT6 could regulate TAG synthesis and growth of mammary epithelial cells in buffalo. These findings will have important implications for understanding the role of the AGPAT gene in buffalo milk performance.
Collapse
|
23
|
Aqueous Extract and Polysaccharide of Aconiti Lateralis Radix Induce Apoptosis and G0/G1 Phase Cell Cycle Arrest by PI3K/AKT/mTOR Signaling Pathway in Mesangial Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:3664696. [PMID: 35497917 PMCID: PMC9054446 DOI: 10.1155/2022/3664696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/03/2022] [Accepted: 03/16/2022] [Indexed: 12/03/2022]
Abstract
Mesangial proliferative glomerulonephritis (MesPGN) is a common renal disease that lacks effective drug intervention. Aconiti Lateralis Radix (Fuzi), a natural Chinese medical herb, is found with significant therapeutic effects on various diseases in the clinic. However, its effects on MesPGN have not been reported. This study is aimed to discuss the therapeutic effects of the aqueous extract of Aconiti Lateralis Radix (ALR) and the polysaccharides of Aconiti Lateralis Radix (PALR) on MesPGN as well as the underlying mechanism. In this study, we, firstly, studied the anti-MesPGN mechanism of ALR and PALR. ALR and PALR inhibit the proliferation of the mesangial cells through the PI3K/AKT/mTOR pathway, induce the G0/G1 phase of block and apoptosis, inhibit the activity of Cyclin E and CDK2, increase the expression of Bax, cleaved caspase-8/caspase-8, and cleaved caspase-3/caspase-3 proteins, and effectively inhibit the growth of the mesangial cells. Overall, our data suggest that ALR and PALR may be potential candidates for MesPGN and that PALR is more effective than ALR.
Collapse
|
24
|
Vascular Graft Infections: An Overview of Novel Treatments Using Nanoparticles and Nanofibers. FIBERS 2022. [DOI: 10.3390/fib10020012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Vascular disease in elderly patients is a growing health concern, with an estimated prevalence of 15–20% in patients above 70 years old. Current treatment for vascular diseases requires the use of a vascular graft (VG) to revascularize lower or upper extremities, create dialysis access, treat aortic aneurysms, and repair dissection. However, postoperative infection is a major complication associated with the use of these VG, often necessitating several operations to achieve complete or partial graft excision, vascular coverage, and extra-anatomical revascularization. There is also a high risk of morbidity, mortality, and limb loss. Therefore, it is important to develop a method to prevent or reduce the incidence of these infections. Numerous studies have investigated the efficacy of antibiotic- and antiseptic-impregnated grafts. In comparison to these traditional methods of creating antimicrobial grafts, nanotechnology enables researchers to design more efficient VG. Nanofibers and nanoparticles have a greater surface area compared to bulk materials, allowing for more efficient encapsulation of antibiotics and better control over their temporo-spatial release. The disruptive potential of nanofibers and nanoparticles is exceptional, and they could pave the way for a new generation of prosthetic VG. This review aims to discuss how nanotechnology is shaping the future of cardiovascular-related infection management.
Collapse
|
25
|
Astragaloside IV protects diabetic cardiomyopathy against inflammation and apoptosis via regulating TLR4/MyD88/NF-κB signaling pathway. J Funct Foods 2022. [DOI: 10.1016/j.jff.2021.104905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
26
|
Liu D, Cheng Y, Tang Z, Chen J, Xia Y, Xu C, Cao X. Potential mechanisms of methylglyoxal-induced human embryonic kidney cells damage: Regulation of oxidative stress, DNA damage, and apoptosis. Chem Biodivers 2021; 19:e202100829. [PMID: 34962083 DOI: 10.1002/cbdv.202100829] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/27/2021] [Indexed: 11/09/2022]
Abstract
Methylglyoxal (MGO) is a reactive carbonyl species that can cause cellular damage and is closely related to kidney disease, especially diabetic nephropathy. The toxic effect of MGO (0.5, 1, and 2 mM) on human embryonic kidney (HEK293) cells and its underlying mechanism were explored in this study. Cell viability, apoptosis and the signaling pathways were measured with MTT, fluorescent staining and western blot experiments, the results showed that MGO could induce oxidative stress and cell inflammation, the level of reactive oxygen species (ROS) increased, and p38MAPK, JNK and NF-κB signaling pathways were activated. Meanwhile, MGO also induced DNA damage. The expression of DNA oxidative damage marker 8-hydroxy-2'-deoxyguanosine (8-OHdG) increased, the expression of double-strand break marker γH2AX increased significantly, and ATM/Chk2/p53 DNA damage response signaling pathway was activated. Furthermore, the expression of the receptor for advanced glycation end products (RAGE) also increased. Finally, mitochondrial membrane potential (MMP) decreased, fluorescence intensity of Hoechst33258 increased, and the protein expression ratio of Bax/Bcl-2 increased significantly after the treatment of MGO. These results demonstrated that MGO might induce HEK293 cells damage by regulating oxidative stress, inflammation, DNA damage, and cell apoptosis, which revealed the specific mechanism of MGO-induced damage to HEK293 cells.
Collapse
Affiliation(s)
- Dan Liu
- Liaoning University, School of life science, 66 Chongshan Road, Huanggu District, Shenyang, CHINA
| | - Ye Cheng
- Liaoning University, School of life science, 66 Chongshan Road, Huanggu District, Shenyang, CHINA
| | - Zhipeng Tang
- Liaoning University, School of life science, 66 Chongshan Road, Huanggu District, Shenyang, CHINA
| | - Junliang Chen
- Liaoning University, School of life science, 66 Chongshan Road, Huanggu District, Shenyang, CHINA
| | - Ying Xia
- Liaoning University, School of life science, 66 Chongshan Road, Huanggu District, Shenyang, CHINA
| | - Chengbin Xu
- Liaoning University, School of environment science, 66 Chongshan Road, Huanggu District, Shenyang, CHINA
| | - Xiangyu Cao
- Liaoning University, School of Life Science, 66 Chongshan Road, Huanggu District, 110036, Shenyang, CHINA
| |
Collapse
|
27
|
Li M, Kong Y, Wu X, Yin Z, Niu X, Wang G. Dietary α-lipoic acid can alleviate the bioaccumulation, oxidative stress, cell apoptosis, and inflammation induced by lead (Pb) in Channa argus. FISH & SHELLFISH IMMUNOLOGY 2021; 119:249-261. [PMID: 34653663 DOI: 10.1016/j.fsi.2021.10.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/04/2021] [Accepted: 10/11/2021] [Indexed: 06/13/2023]
Abstract
This study aims to evaluate the effects of dietary α-lipoic acid (α-LA) on bioaccumulation, oxidative stress, apoptosis, and inflammation in Channa argus after 28 d of lead (Pb) exposure. A total of 300 fish were divided into five groups: the first group was the control group and the other four groups were exposed to waterborne Pb (800 ppb) and fed α-LA diets supplemented with 0, 300, 600, and 900 mg/kg. The results demonstrated that dietary α-LA effectively reduced the Pb accumulation in the liver, kidney, gill, intestine, and muscle of C. argus after exposure to Pb. Meanwhile, dietary α-LA reversed alterations in the biochemical parameters (Alanine aminotransferase (ALT), lactate dehydrogenase (LDH), aspartate aminotransferase (AST), blood urea nitrogen (BUN), cortisol (COR), and creatinine (CRE)) and immunity parameters (myeloperoxidase (MPO), complement 3 (C3), lysozyme (LYS), complement 4 (C4), C-reactive protein (CRP), and immunoglobulin M (IgM)) in the serum of fish caused by Pb. Pb-induced reduction of antioxidant enzyme activities (Catalase (CAT), glutathione reductase (GR), superoxide dismutase (SOD), glutathione (GSH), glutathione peroxidase (GSH-Px)) was inhibited by dietary α-LA. And malondialdehyde (MDA) and protein carbonyl (PC) content exhibited an opposite trend. Meanwhile, dietary supplemented with α-LA was found to relieve Pb-induced oxidative stress by downregulating Keap1 mRNA expression levels and upregulating the expression levels of CAT, nuclear factor erythroid 2-related factor 2 (Nrf2), GSH-Px, and Cu/Zn SOD. Furthermore, α-LA supplementation reversed Pb-induced upregulation of pro-inflammatory genes (interleukin (IL)-6, IL-1β, tumor necrosis factor α (TNF-α), and nuclear factor kappa B (NF-κB)), Pro-apoptotic genes (Bcl-2-associated X (Bax), caspase (Cas)-3, and tumor protein p53 (p53)) and Hsp70, and downregulation of anti-inflammatory genes (IL-10, inhibitor of κBα (IκBα), and transforming growth factor β (TGF-β)) and anti-apoptosis gene (B-cell lymphoma-2 (Bcl-2)). Overall, dietary α-LA supplementation could enhance the innate immunity and antioxidant capacity of fish, attenuating the Pb accumulation, and cell apoptosis after being exposed to Pb. Furthermore, dietary α-LA could relieve Pb-induced inflammatory response and oxidative stress of fish via regulating NF-κB and Nrf2 signaling, respectively.
Collapse
Affiliation(s)
- Min Li
- College of Animal Science and Technology, Jilin Agriculture University, Changchun, 130118, China; Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Changchun, Jilin, 130118, China; Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agriculture University, Changchun, 130118, China; Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, 130118, China
| | - Yidi Kong
- College of Animal Science and Technology, Jilin Agriculture University, Changchun, 130118, China; Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Changchun, Jilin, 130118, China; Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agriculture University, Changchun, 130118, China; Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, 130118, China
| | - Xueqin Wu
- College of Animal Science and Technology, Jilin Agriculture University, Changchun, 130118, China; Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Changchun, Jilin, 130118, China; Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agriculture University, Changchun, 130118, China; Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, 130118, China
| | - Zhuang Yin
- College of Animal Science and Technology, Jilin Agriculture University, Changchun, 130118, China; Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Changchun, Jilin, 130118, China; Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agriculture University, Changchun, 130118, China; Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, 130118, China
| | - Xiaotian Niu
- College of Animal Science and Technology, Jilin Agriculture University, Changchun, 130118, China; Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Changchun, Jilin, 130118, China; Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agriculture University, Changchun, 130118, China; Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, 130118, China.
| | - Guiqin Wang
- College of Animal Science and Technology, Jilin Agriculture University, Changchun, 130118, China; Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Changchun, Jilin, 130118, China; Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agriculture University, Changchun, 130118, China; Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
28
|
Zhu W, Zhang R, Liu S, Tian J, Lv X, Yu F, Xin H. The effect of nanoparticles of cobalt-chromium on human aortic endothelial cells in vitro. J Appl Toxicol 2021; 41:1966-1979. [PMID: 33959985 DOI: 10.1002/jat.4177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/19/2021] [Indexed: 11/08/2022]
Abstract
Despite advances in stent technology for vascular interventions, in-stent restenosis (ISR) remains a main complication. The corrosion of cobalt-chromium (CoCr) alloy coronary stents has been identified to be associated with ISR, whereas its role in ISR has not been elucidated. In the current work, CoCr nanoparticles, simulated corrosion products of CoCr alloy, were used to investigate their effect on the endothelial cells. It has been demonstrated that the cell viability declines and the cell membrane is damaged, indicating the cytotoxicity of CoCr nanoparticles. The expression of GRP78, CHOP, and cleaved-caspase12 proteins has increased when exposed to CoCr nanoparticles, suggesting that CoCr nanoparticles induced cell apoptosis through endoplasmic reticulum (ER) stress-mediated apoptotic pathway. An increased release of adhesion and inflammatory mediators was also induced by CoCr nanoparticles, including ICAM-1, VCAM-1, IL-1β, IL-6, and TNF-α. Our results demonstrated that CoCr nanoparticles could trigger apoptosis, adhesion, and inflammation. These findings indicated potential damaging effects of CoCr nanoparticles on the vascular endothelium, which suggested corrosion of CoCr alloy may promote the progression and development of ISR.
Collapse
Affiliation(s)
- Wenxiu Zhu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China.,Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Rui Zhang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Song Liu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Jiawei Tian
- Department of Emergency Internal Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaobing Lv
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Fei Yu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Hui Xin
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| |
Collapse
|
29
|
Wang S, Zhang Y, Gao J, Zhang J, Tao L, Xu W. The enantioselective study of the toxicity effects of chiral acetochlor in HepG2 cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 218:112261. [PMID: 33964548 DOI: 10.1016/j.ecoenv.2021.112261] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 04/07/2021] [Accepted: 04/12/2021] [Indexed: 06/12/2023]
Abstract
Acetochlor is one of the most widely used chiral herbicides in the world, and it is usually produced and used as racemic form (Rac). The potential effects of acetochlor in human body are mainly induced by its residue in agriculture food. The direct target exposed is the liver in human body. However, the potential toxic and mechanism threat to human liver cells caused by chiral acetochlor has been rarely reported. The purpose of this study is to explore the potential mechanism of the toxicity caused by chiral acetochlor in HepG2 cells. The results revealed that acetochlor and its enantiomers could inhibit cell activity and cause DNA damage in HepG2 cells. The toxicity of Rac was higher than that of the two enantiomers, mainly derived from S configuration. The mechanism is through inducing decreased membrane potential (△Ψ), up-regulated Bax/BcL-2 expression, caused a cascade reaction, activated casepase-3 and casepase-9 and cleaved PARP, which maybe lead to cell death through apoptotic-signaling pathway in the end. These results illuminate that the genotoxic and cytotoxic risks of chiral acetochlor are major coming from S configuration. It provides a theoretical basis for the production of single pesticide to reduce the effects of human health.
Collapse
Affiliation(s)
- Susu Wang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yang Zhang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Jufang Gao
- College of Life, Shanghai Normal University, Shanghai 200234, China
| | | | - Liming Tao
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Wenping Xu
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
30
|
Estronca L, Francisco V, Pitrez P, Honório I, Carvalho L, Vazão H, Blersch J, Rai A, Nissan X, Simon U, Grãos M, Saúde L, Ferreira L. Induced pluripotent stem cell-derived vascular networks to screen nano-bio interactions. NANOSCALE HORIZONS 2021; 6:245-259. [PMID: 33576750 DOI: 10.1039/d0nh00550a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The vascular bioactivity/safety of nanomaterials is typically evaluated by animal testing, which is of low throughput and does not account for biological differences between animals and humans such as ageing, metabolism and disease profiles. The development of personalized human in vitro platforms to evaluate the interaction of nanomaterials with the vascular system would be important for both therapeutic and regenerative medicine. A library of 30 nanoparticle (NP) formulations, in use in imaging, antimicrobial and pharmaceutical applications, was evaluated in a reporter zebrafish model of vasculogenesis and then tested in personalized humanized models composed of human-induced pluripotent stem cell (hiPSC)-derived endothelial cells (ECs) with "young" and "aged" phenotypes in 3 vascular network formats: 2D (in polystyrene dish), 3D (in Matrigel) and in a blood vessel on a chip. As a proof of concept, vascular toxicity was used as the main readout. The results show that the toxicity profile of NPs to hiPSC-ECs was dependent on the "age" of the endothelial cells and vascular network format. hiPSC-ECs were less susceptible to the cytotoxicity effect of NPs when cultured in flow than in static conditions, the protective effect being mediated, at least in part, by glycocalyx. Overall, the results presented here highlight the relevance of in vitro hiPSC-derived vascular systems to screen vascular nanomaterial interactions.
Collapse
Affiliation(s)
- Luís Estronca
- Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Zhang J, Wang C, Kang K, Liu H, Liu X, Jia X, Yu K. Loganin Attenuates Septic Acute Renal Injury with the Participation of AKT and Nrf2/HO-1 Signaling Pathways. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:501-513. [PMID: 33603340 PMCID: PMC7886113 DOI: 10.2147/dddt.s294266] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/12/2021] [Indexed: 01/23/2023]
Abstract
Purpose Sepsis, a destructive inflammatory response syndrome, is the principal reason to induce death in the intensive care unit. Loganin has been proved to possess the property of anti-inflammation, antioxidant, neuroprotection, and sedation. The primary aim of this study was to evaluate whether Loganin could alleviate acute kidney injury (AKI) during sepsis and investigate the latent mechanisms. Methods Septic AKI models were established by cecal ligation and puncture (CLP) surgery in mice and given Loganin (20, 40, 80 mg/kg) by gavage. Lipopolysaccharides (LPS)-stimulated human kidney proximal tubular (HK2) cells incubated in Loganin (5, 10, 20 μ M) were used to explore the accurate mechanisms. Survival rate, renal function (creatinine and blood urea nitrogen), and renal pathological changes were detected in septic mice. Oxidative stress markers (SOD, GSH-Px, MDA, and SOD), mitochondrial membrane potential, mitochondrial calcium overload, and nuclear factor E2-related factor 2 (Nrf2)/heme-oxygenase 1 (HO-1) pathway activation in vivo and in vitro were determined by commercial kits and Western blot. Cell apoptosis, apoptotic-related protein (cleaved caspase-3, Bcl-2, and Bax) expression and protein kinase B (AKT) phosphorylation in vivo and in vitro were measured by TUNEL staining and Western blot. Finally, AKT blockage by 10 μM LY294002 or Nrf2 inhibition by10 μ M ML385 were utilized to prove the involvement of AKT and Nrf2/HO-1 pathway in AKI during sepsis. Results We found Loganin treatment (20, 40, 80 mg/kg) mitigated septic AKI reflected by elevated renal function and palliative pathological changes. Oxidative stress and apoptosis in the kidney and LPS-treated HK2 cells were also inhibited by Loganin administration, which was accompanied by AKT and Nrf2/HO-1 pathway activation. Besides, the protective effects of Loganin could be diminished by AKT or Nrf2 blockage, indicating the involvement of AKT and Nrf2/HO-1 pathway. Conclusion The results suggested that the protective effects of Loganin on AKI during sepsis might be mediated by AKT and Nrf2/HO-1 pathway signaling activation in kidney proximal tubular cells.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, Heilongjiang 150001, People's Republic of China
| | - Changsong Wang
- Department of Critical Care Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, People's Republic of China
| | - Kai Kang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, Heilongjiang 150001, People's Republic of China
| | - Haitao Liu
- Department of Critical Care Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, People's Republic of China
| | - Xiaowei Liu
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, Heilongjiang 150001, People's Republic of China
| | - Xiaonan Jia
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, Heilongjiang 150001, People's Republic of China
| | - Kaijiang Yu
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, Heilongjiang 150001, People's Republic of China
| |
Collapse
|
32
|
Yan BF, Chen X, Liu J, Liu SJ, Zhang JZ, Zeng QQ, Duan JA. Asiatic Acid Induces Mitochondrial Apoptosis via Inhibition of JAK2/STAT3 Signalling Pathway in Human Osteosarcoma. Folia Biol (Praha) 2021; 67:108-117. [PMID: 35151244 DOI: 10.14712/fb2021067030108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Osteosarcoma (OS), a severe malignant bone tumour, usually occurs in adolescents and children and has a poor prognosis. Asiatic acid (AA), an active component isolated from Centella asiatica (L.) Urb., exhibits appreciable anti-oxidant and anti-tumour activities. So far, the effects and underlying mechanisms of AA against OS have not been clarified. Here, we explored the anti-tumour effects of AA against human OS and the involved mechanism mediating its actions. To evaluate effects of AA on the cell proliferation of human OS cells, cell viability and colony formation assays were performed. Flow cytometry was used to evaluate apoptosis in OS cells exposed to AA and mitochondrial membrane potential. Western blotting and RT-PCR were applied to determine expression of the relevant proteins and their mRNA levels. Our explorations showed that AA inhibits proliferation of human OS cells in a concentration- and time-dependent manner, and induces apoptosis of OS cells by the intrinsic (mitochondrial) pathway. Importantly, we found that inhibition of the AA-induced phosphorylation of JAK2/STAT3 signalling molecules and the decrease in MCL-1 contributed to the anti-tumour efficacy of AA. Collectively, our results suggest that AA could evoke mitochondrial- induced apoptosis in human OS cells by suppression of the JAK2/STAT3 pathway and MCL-1 expression. These results strongly demonstrate that AA could be a potential anti-tumour agent for OS treatment.
Collapse
Affiliation(s)
- B F Yan
- Jiangsu Health Vocational College; Nanjing, China
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Key Laboratory of Chinese Medicinal Resources Recycling Utilization, State Administration of Traditional Chinese Medicine, Nanjing University of Chinese Medicine; Nanjing, China
| | - X Chen
- Jiangsu College of Nursing; Huaian, China
| | - J Liu
- Jiangsu Health Vocational College; Nanjing, China
| | - S J Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Key Laboratory of Chinese Medicinal Resources Recycling Utilization, State Administration of Traditional Chinese Medicine, Nanjing University of Chinese Medicine; Nanjing, China
| | - J Z Zhang
- Jiangsu Health Vocational College; Nanjing, China
| | - Q Q Zeng
- Jiangsu Health Vocational College; Nanjing, China
| | - J A Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Key Laboratory of Chinese Medicinal Resources Recycling Utilization, State Administration of Traditional Chinese Medicine, Nanjing University of Chinese Medicine; Nanjing, China
| |
Collapse
|
33
|
Czyżowska A, Barbasz A. Cytotoxicity of zinc oxide nanoparticles to innate and adaptive human immune cells. J Appl Toxicol 2020; 41:1425-1437. [PMID: 33368402 DOI: 10.1002/jat.4133] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/18/2020] [Accepted: 12/11/2020] [Indexed: 11/06/2022]
Abstract
Zinc oxide nanoparticles (ZnO-NPs) are widely used in almost every area of life. Therefore, exposure to them is unavoidable, which makes it necessary to assess their safety for humans. This paper aims to determine toxicity of ZnO-NPs of two diameters toward human immune cells responsible for: innate response (U-937 and HL-60) and acquired response (COLO-720L and HUT-78). Mitochondrial activity, membrane integrity, degree of cellular lipid oxidation, induction of inflammation, and activation of the apoptosis pathway were evaluated to determine differences in cellular response to the tested nanoparticles. ZnO-NPs with a diameter of 100 and 130 nm cause significant cell mortality at 25 and 12 mg/L, respectively. Mitochondrial damage leads to the activation of the caspase cascade and, consequently, to cell apoptosis. ZnO-NPs also cause peroxidation of membrane lipids. Due to the photocatalytic properties of ZnO-NPs, the effect of ultraviolet (UV) irradiation on the degree of their toxicity was also investigated. However, UV irradiation enhances the toxic effect of nanoparticles mainly by weakening the cell's defense capabilities. ZnO-NPs are cytotoxic to human immune system, and they may cause both long-term and short-term negative effects.
Collapse
Affiliation(s)
- Agnieszka Czyżowska
- Department of Biochemistry and Biophysics, Institute of Biology, Pedagogical University of Cracow, Kraków, Poland
| | - Anna Barbasz
- Department of Biochemistry and Biophysics, Institute of Biology, Pedagogical University of Cracow, Kraków, Poland
| |
Collapse
|
34
|
Mao X, Fu P, Wang L, Xiang C. Mitochondria: Potential Targets for Osteoarthritis. Front Med (Lausanne) 2020; 7:581402. [PMID: 33324661 PMCID: PMC7726420 DOI: 10.3389/fmed.2020.581402] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/19/2020] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is a common and disabling joint disorder that is mainly characterized by cartilage degeneration and narrow joint spaces. The role of mitochondrial dysfunction in promoting the development of OA has gained much attention. Targeting endogenous molecules to improve mitochondrial function is a potential treatment for OA. Moreover, research on exogenous drugs to improve mitochondrial function in OA based on endogenous molecular targets has been accomplished. In addition, stem cells and exosomes have been deeply researched in the context of cartilage regeneration, and these factors both reverse mitochondrial dysfunctions. Thus, we hypothesize that biomedical approaches will be applied to the treatment of OA. Furthermore, we have summarized the global status of mitochondria and osteoarthritis research in the past two decades, which will contribute to the research field and the development of novel treatment strategies for OA.
Collapse
Affiliation(s)
- Xingjia Mao
- Department of Orthopedic, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Panfeng Fu
- Department of Respiratory and Critical Care, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, China
| | - Linlin Wang
- Department of Basic Medicine Sciences, The School of Medicine of Zhejiang University, Hangzhou, China
| | - Chuan Xiang
- Department of Orthopedic, The Second Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
35
|
Wang Z, Tang M. Research progress on toxicity, function, and mechanism of metal oxide nanoparticles on vascular endothelial cells. J Appl Toxicol 2020; 41:683-700. [DOI: 10.1002/jat.4121] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/05/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Zhihui Wang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health Southeast University Nanjing China
| | - Meng Tang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health Southeast University Nanjing China
| |
Collapse
|
36
|
Czyżowska A, Dyba B, Rudolphi-Szydło E, Barbasz A. Structural and biochemical modifications of model and native membranes of human immune cells in response to the action of zinc oxide nanoparticles. J Appl Toxicol 2020; 41:458-469. [PMID: 33103261 DOI: 10.1002/jat.4057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/03/2020] [Accepted: 08/13/2020] [Indexed: 12/22/2022]
Abstract
The development of nanotechnology has led to the increased production of zinc oxide nanoparticles (ZnO-NPs) and their application in a wide variety of everyday products. It creates the need for a full assessment of their safety for humans. The aim of the study was to assess the toxic effects of ZnO-NPs on model human cells of the immune system: U-937, HL-60, HUT-78, and COLO-720L. Particular attention was paid to the direct interaction of the nanoparticles with membrane lipids and the role of zinc ions in the mechanism of their toxicity. Cell viability, lipid peroxidation, cell membrane integrity, and the amount of zinc ions released from nanoparticles were tested. Disruption in cell metabolism was noted for ZnO-NPs concentrations from 6.25 mg/L. Contact with ZnO-NPs caused lipid peroxidation of all cells and correlated with membrane disruption of HL-60, HUT-78, and COLO-720L cells. Model monolayers (Langmuir technique) were used to assess the interaction of the nanoparticles with the studied lipids. Physicochemical parameters, such as the area per molecule at maximal layer compression, the pressure at which the monolayer collapses, and the static compression modulus, were calculated. The models of the HL-60 and U-937 cell membranes under ZnO-NPs treatment reacted in a different way. It has also been shown that Zn2+ are not the main causative factor of ZnO-NPs toxicity. Investigating the early stages of mechanism of nanoparticles toxicity will allow for a more complete risk assessment and development of methods for a safer synthesis of engineering nanomaterials.
Collapse
Affiliation(s)
- Agnieszka Czyżowska
- Department of Biochemistry and Biophysics, Institute of Biology, Pedagogical University of Cracow, Kraków, Poland
| | - Barbara Dyba
- Department of Biochemistry and Biophysics, Institute of Biology, Pedagogical University of Cracow, Kraków, Poland
| | - Elżbieta Rudolphi-Szydło
- Department of Biochemistry and Biophysics, Institute of Biology, Pedagogical University of Cracow, Kraków, Poland
| | - Anna Barbasz
- Department of Biochemistry and Biophysics, Institute of Biology, Pedagogical University of Cracow, Kraków, Poland
| |
Collapse
|
37
|
Mostafa A, Korayem HE, Fekry E, Hosny S. The Effect of Intra-articular versus Intravenous Injection of Mesenchymal Stem Cells on Experimentally-Induced Knee Joint Osteoarthritis. J Microsc Ultrastruct 2020; 9:31-38. [PMID: 33850710 PMCID: PMC8030543 DOI: 10.4103/jmau.jmau_2_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 02/22/2020] [Indexed: 11/04/2022] Open
Abstract
Background Osteoarthritis (OA) is a chronic degenerative debilitating disease, primarily affects joints, particularly weight-bearing areas. The surface layer of the articular cartilage breaks down and wears away leading to rubbing of bones, pain, swelling, and joint stiffness. Aim and Objectives This study investigates the possible therapeutic effects of intra-articular versus intravenous injection of umbilical cord blood mesenchymal stem cells (UCB-MSCs) against mono-iodoacetate-induced OA of the knee joints in male albino rats, using histological and immunohistochemical techniques. Materials and Methods Thirty male adult albino rats were randomized into five groups as follows: Group (I) and (II): Served as control. Group (III): Osteoarthritic group. Group IV: Osteoarthritic and intraarticularly-injected MSCs. Group V: Osteoarthritic and intravenously-injected MSCs. Animals were sacrificed 1 month after stem cell injection, the right knee was prepared for histological techniques (Hematoxylin and Eosin and Toluidine blue stains) and immunohistochemical technique (Bax stain). Prussian blue stain was used to assess homing of MSCs in Groups IV and V. Results Knee joint surface was irregular, fissured, and fragmented in Group III. In Groups IV and V, affected area was filled with newly formed tissue. Toluidine blue showed a decrease in matrix staining in Group III compared to both control and MSCs-treated groups. Chondrocytes in Group III showed strong Bax immunoreactivity and this reaction decreased in Group IV and V; however, Group V immunoreactivity was more than Group IV. Prussian blue stain showed labeled UCB-MSCs in many chondrocytes in Group IV and few chondrocytes in Group V. Conclusion Intraarticularly-injected UCB-MSCs showed better healing of knee OA than intravenously-injected UCB-MSCs.
Collapse
Affiliation(s)
- Aya Mostafa
- Department of Histology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Horeya E Korayem
- Department of Histology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Ereny Fekry
- Department of Histology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Somaya Hosny
- Department of Histology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
38
|
Samet JM, Chen H, Pennington ER, Bromberg PA. Non-redox cycling mechanisms of oxidative stress induced by PM metals. Free Radic Biol Med 2020; 151:26-37. [PMID: 31877355 PMCID: PMC7803379 DOI: 10.1016/j.freeradbiomed.2019.12.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 12/13/2019] [Accepted: 12/20/2019] [Indexed: 12/27/2022]
Abstract
Metallic compounds contribute to the oxidative stress of ambient particulate matter (PM) exposure. The toxicity of redox inert ions of cadmium, mercury, lead and zinc, as well as redox-active ions of vanadium and chromium is underlain by dysregulation of mitochondrial function and loss of signaling quiescence. Central to the initiation of these effects is the interaction of metal ions with cysteinyl thiols on glutathione and key regulatory proteins, which leads to impaired mitochondrial electron transport and persistent pan-activation of signal transduction pathways. The mitochondrial and signaling effects are linked by the production of H2O2, generated from mitochondrial superoxide anion or through the activation of NADPH oxidase, which extends the range and amplifies the magnitude of the oxidative effects of the metals. This oxidative burden can be further potentiated by inhibitory effects of the metals on the enzymes of the glutathione and thioredoxin systems. Along with the better-known Fenton-based mechanisms, the non-redox cycling mechanisms of oxidative stress induced by metals constitute significant pathways for cellular injury induced by PM inhalation.
Collapse
Affiliation(s)
- James M Samet
- Environmental Public Health Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Chapel Hill, NC, USA.
| | - Hao Chen
- Oak Ridge Institute for Science and Education, Oak Ridge, TN, USA
| | | | - Philip A Bromberg
- Center for Environmental Medicine, Asthma and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
39
|
Li Y, Li F, Zhang L, Zhang C, Peng H, Lan F, Peng S, Liu C, Guo J. Zinc Oxide Nanoparticles Induce Mitochondrial Biogenesis Impairment and Cardiac Dysfunction in Human iPSC-Derived Cardiomyocytes. Int J Nanomedicine 2020; 15:2669-2683. [PMID: 32368048 PMCID: PMC7183345 DOI: 10.2147/ijn.s249912] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 04/01/2020] [Indexed: 12/18/2022] Open
Abstract
Background Zinc oxide nanoparticles (ZnO NPs) are one of the most widely used nanomaterials in a variety of fields such as industrial, pharmaceutical, and household applications. Increasing evidence suggests that ZnO NPs could elicit unignorable harmful effect to the cardiovascular system, but the potential deleterious effects to human cardiomyocytes remain to be elucidated. Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have been increasingly used as a promising in vitro model of cardiomyocyte in various fields such as drug cardiac safety evaluation. Herein, the present study was designed to elucidate the cardiac adverse effects of ZnO NPs and explore the possible underlying mechanism using hiPSC-CMs. Methods ZnO NPs were characterized by transmission electron microscopy and dynamic light scattering. The cytotoxicity induced by ZnO NPs in hiPSC-CMs was evaluated by determination of cell viability and lactate dehydrogenase release. Cellular reactive oxygen species (ROS) and mitochondrial membrane potential were measured by high-content analysis (HCA). Mitochondrial biogenesis was assayed by detection of mtDNA copy number and PGC-1α pathway. Moreover, microelectrode array techniques were used to investigate cardiac electrophysiological alterations. Results We demonstrated that ZnO NPs concentration- and time-dependently elicited cytotoxicity in hiPSC-CMs. The results from HCA revealed that ZnO NPs exposure at low-cytotoxic concentrations significantly promoted ROS generation and induced mitochondrial dysfunction. We further demonstrated that ZnO NPs could impair mitochondrial biogenesis and inhibit PGC-1α pathway. In addition, ZnO NPs at insignificantly cytotoxic concentrations were found to trigger cardiac electrophysiological alterations as evidenced by decreases of beat rate and spike amplitude. Conclusion Our findings unveiled the potential harmful effects of ZnO NPs to human cardiomyocytes that involve mitochondrial biogenesis and the PGC-1α pathway that could affect cardiac electrophysiological function.
Collapse
Affiliation(s)
- Yujie Li
- Graduate School, Academy of Military Medical Sciences, Beijing, People's Republic of China.,Department of Operational Medical Protection, PLA Center for Disease Control and Prevention, Beijing, People's Republic of China
| | - Fengxiang Li
- Department of Operational Medical Protection, PLA Center for Disease Control and Prevention, Beijing, People's Republic of China
| | - Lincong Zhang
- Department of Operational Medical Protection, PLA Center for Disease Control and Prevention, Beijing, People's Republic of China
| | - Chi Zhang
- Department of Operational Medical Protection, PLA Center for Disease Control and Prevention, Beijing, People's Republic of China
| | - Hui Peng
- Department of Operational Medical Protection, PLA Center for Disease Control and Prevention, Beijing, People's Republic of China
| | - Feng Lan
- Beijing Key Laboratory for Cardiovascular Precision Medicines, Anzhen Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Shuangqing Peng
- Department of Operational Medical Protection, PLA Center for Disease Control and Prevention, Beijing, People's Republic of China
| | - Chao Liu
- Graduate School, Academy of Military Medical Sciences, Beijing, People's Republic of China.,Department of Operational Medical Protection, PLA Center for Disease Control and Prevention, Beijing, People's Republic of China
| | - Jiabin Guo
- Graduate School, Academy of Military Medical Sciences, Beijing, People's Republic of China.,Department of Operational Medical Protection, PLA Center for Disease Control and Prevention, Beijing, People's Republic of China
| |
Collapse
|
40
|
Sheng W, Yang H, Niu Z, Yin H. Anti-apoptosis effect of heme oxygenase-1 on lung injury after cardiopulmonary bypass. J Thorac Dis 2020; 12:1393-1403. [PMID: 32395277 PMCID: PMC7212168 DOI: 10.21037/jtd.2020.03.48] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Background This study aimed to investigate the anti-apoptosis effects of heme oxygenase-1 (HO-1) on lung injury (LI) after cardiopulmonary bypass (CPB) and its probable mechanisms. Methods One hundred and forty-four male Wistar rats were divided into 3 groups randomly: group A (control group), group B (cobalt protoporphyrin, CoPP), and group C [CoPP plus zinc protoporphyrin (ZnPP)]. Lung tissues were harvested at different time: before CPB (T0), 0 min after CPB (T1), 2 h after CPB (T2), 6 h (T3), 12 h (T4), and 24 h (T5). Results The HO-1 protein expressions in lung tissue in group B were higher than those in group A and group C in any given time, and the same as HO-1 activity (P<0.05). The expressions of Bcl-2 protein in group B at all time point after bypass were higher than those in group A and group C, and the difference was statistically significant (P<0.05). Apoptosis index (AI) in group B at any time point after bypass were lower than those in group A and group C (P<0.05). Conclusions CoPP can significantly increase the expression of HO-1 protein in lung tissue. HO-1 is still highly expressed after CPB, so as to play an important part in anti-apoptosis, and reduce LI.
Collapse
Affiliation(s)
- Wei Sheng
- Department of Cardiovascular Surgery, Qingdao Municipal Hospital, Medical College of Qingdao University, Qingdao 266071, China
| | - Haiqin Yang
- Department of Mental Intervention, Qingdao Preferential Hospital, Qingdao 266071, China
| | - Zhaozhuo Niu
- Department of Cardiovascular Surgery, Qingdao Municipal Hospital, Medical College of Qingdao University, Qingdao 266071, China
| | - Hong Yin
- Department of Cardiovascular Surgery, Qingdao Municipal Hospital, Medical College of Qingdao University, Qingdao 266071, China
| |
Collapse
|
41
|
Wang L, Guo D, Wang Z, Yin X, Wei H, Hu W, Chen R, Chen C. Zinc oxide nanoparticles induce human tenon fibroblast apoptosis through reactive oxygen species and caspase signaling pathway. Arch Biochem Biophys 2020; 683:108324. [PMID: 32112740 DOI: 10.1016/j.abb.2020.108324] [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: 09/26/2019] [Revised: 01/23/2020] [Accepted: 02/25/2020] [Indexed: 12/27/2022]
Abstract
Glaucoma is the leading cause of irreversible blindness in the world and trabeculectomy remains still the most commonly performed filtration surgery. Failure of trabeculectomy is due to the formation of scarring, which is associated with the increased fibroblast proliferation, activation, and collagen deposition at the site of the drainage channel with subconjunctival fibrosis. Our previous study has revealed that zinc oxide (ZnO) nanoparticles could efficiently decrease the expressions of TGF-β1 and inhibit fibroblast-mediated collagen lattice contraction. However, the mechanism underlying ZnO nanoparticle-induced fibroblast apoptosis is still unclear. In the present study, we investigated the effect of ZnO nanoparticles on the reactive oxygen species (ROS) and mitochondrial membrane potential (Δψm) in human Tenon fibroblasts (HTFs). Moreover, we also explored the influence of ZnO nanoparticles on the expression of Caspase-3, Caspase-9, apoptotic protease-activating factor-1 (Apaf-1), fibroblast-specific protein-1 (FSP-1), collagen III, and E-cadherin. The results indicated that ZnO nanoparticles markedly inhibit HTFs viability and decrease the Δψm in a concentration-dependent pattern. Exposure of HTFs to ZnO nanoparticles could also induce the elevated Caspase-3, Caspase-9, and Apaf-1 expression, decrease the levels of FSP-1, collagen III, and E-cadherin expression, leading to HTFs apoptosis. Our results suggested that elevated ROS and activated Caspase signaling play a fundamental role in ZnO nanoparticle-induced HTFs apoptosis.
Collapse
Affiliation(s)
- Ling Wang
- Jining Medical University, Jining, 272000, Shandong Province, China
| | - Dadong Guo
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Eye Institute of Shandong University of Traditional Chinese Medicine, Jinan, 250002, Shandong Province, China
| | - Zhe Wang
- Department of Ophthalmology, Zaozhuang Hospital of Traditional Chinese Medicine, Zaozhuang, 277000, China
| | - Xuewei Yin
- The Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| | - Huixia Wei
- The Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| | - Wanli Hu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, No. 4655#, Daxue Road, Jinan, 250355, China
| | - Ruihong Chen
- Jining Medical University, Jining, 272000, Shandong Province, China
| | - Chao Chen
- Department of Ophthalmology, the First People's Hospital of Jining, Jining, 272002, Shandong Province, China.
| |
Collapse
|
42
|
Shi L, Duan Y, Yao X, Song R, Ren Y. Effects of selenium on the proliferation and apoptosis of sheep spermatogonial stem cells in vitro. Anim Reprod Sci 2020; 215:106330. [PMID: 32216931 DOI: 10.1016/j.anireprosci.2020.106330] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 02/08/2020] [Accepted: 02/21/2020] [Indexed: 01/06/2023]
Abstract
The objective of this study was to investigate effects of selenium (Se) on proliferation and apoptosis of sheep spermatogonial stem cells (SSC) in vitro. The SSC were assigned to five treatment groups (0, 2.0, 4.0, 8.0 and 16.0 μmol/L Se). After treatment with Se for 96 h, cell proliferation and apoptosis were evaluated. The relative abundance of P53 mRNA transcript and protein, cell cycle and apoptosis-related genes were detected using real-time PCR and Western blot quantifications, respectively. The results indicate there were the least cell proliferation rates in the Se16.0 group. Treatments with relatively greater Se concentrations (8.0 and 16.0 μmol/L) resulted in a greater percentage of apoptotic cells, which was consistent with the relative abundances of P53, P21, P27 and pro-apoptosis mRNA transcripts. There were relatively greater ROS concentrations in the control, Se8.0 and Se16.0 groups. Compared with the control group, treatment with the Se concentration of 16.0 μmol/L resulted in an increased abundance of P53, P21, P27 and BAX proteins. Treatment with Pifithrin-α suppressed the increase in abundance of P53 and P21 proteins induced by the relatively greater concentration of Se (16.0 μmol/L), however, did not result in a change in abundances of P27 and BAX proteins. These results indicate the regulatory functions of Se on proliferation and apoptosis of sheep SSC is associated with the P21-mediated P53 signalling pathway. The P27 and BAX proteins have limited functions during the apoptotic process of SSC induced by the relatively greater concentrations of Se.
Collapse
Affiliation(s)
- Lei Shi
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, PR China; Laboratory of Animal Reproductive Biotechnology, Shanxi Agricultural University, Taigu, 030801, PR China
| | - Yunli Duan
- Laboratory of Animal Reproductive Biotechnology, Shanxi Agricultural University, Taigu, 030801, PR China
| | - Xiaolei Yao
- Jiangsu Engineering Technology Research Center of Mutton Sheep and Goat Industry, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Ruigao Song
- Laboratory of Animal Reproductive Biotechnology, Shanxi Agricultural University, Taigu, 030801, PR China
| | - Youshe Ren
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, PR China; Laboratory of Animal Reproductive Biotechnology, Shanxi Agricultural University, Taigu, 030801, PR China.
| |
Collapse
|
43
|
Wu D, Ma Y, Cao Y, Zhang T. Mitochondrial toxicity of nanomaterials. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 702:134994. [PMID: 31715400 DOI: 10.1016/j.scitotenv.2019.134994] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/11/2019] [Accepted: 10/14/2019] [Indexed: 05/11/2023]
Abstract
In recent years, nanomaterials have been widely applied in electronics, food, biomedicine and other fields, resulting in increased human exposure and consequent research focus on their biological and toxic effects. Mitochondria, the main target organelle for nanomaterials (NM), play a critical role in their toxic activities. Several studies to date have shown that nanomaterials cause alterations in mitochondrial morphology, mitochondrial membrane potential, opening of the mitochondrial permeability transition pore (MPTP) and mitochondrial respiratory function, and promote cytochrome C release. An earlier mitochondrial toxicity study of NMs additionally reported induction of mitochondrial dynamic changes. Here, we have reviewed the mitochondrial toxicity of NMs and provided a scientific basis for the contribution of mitochondria to the toxicological effects of different NMs along with approaches to reduce mitochondrial and, consequently, overall toxicity of NMs.
Collapse
Affiliation(s)
- Daming Wu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Ying Ma
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Yuna Cao
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Ting Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China.
| |
Collapse
|
44
|
Bai C, Sun Y, Pan X, Yang J, Li X, Wu A, Qin D, Cao S, Zou W, Wu J. Antitumor Effects of Trimethylellagic Acid Isolated From Sanguisorba officinalis L. on Colorectal Cancer via Angiogenesis Inhibition and Apoptosis Induction. Front Pharmacol 2020; 10:1646. [PMID: 32047442 PMCID: PMC6997556 DOI: 10.3389/fphar.2019.01646] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 12/16/2019] [Indexed: 12/24/2022] Open
Abstract
Previous studies have demonstrated that tannin could inhibit the proliferation and angiogenesis of cancer cells. However, the mechanism(s) associated with its antitumor effect remains unclear. Here, we investigated the effects of 3,3',4'-trimethylellagic acid (TMEA), a tannin compound isolated from Sanguisorba officinalis L., on the proliferation, angiogenesis, and apoptosis in cancer cells, as well as the underlying mechanism(s) related to its antitumor activity. TMEA was isolated from Sanguisorba officinalis L. by silica gel column chromatography. Molecular docking was carried out to assess active pocket binding between TMEA and vascular endothelial growth factor receptor 2 (VEGFR2). The antiangiogenic effect of TMEA on the migration and tube formation was detected in HUVECs by wound healing and tube formation assays, respectively. The antitumor effects of TMEA on the cell proliferation were determined in HepG2, A549, and SW620 cells by MTS assay in vitro and on the tumor growth of SW620 xenografts bearing in nude mice in vivo. The mRNA expression of Bcl-2, Bax, caspase-3, VEGF, PI3K, and mTOR were measured by qRT-PCR and protein expression of Bcl-2, Bax, caspase-3, VEGF, PI3K, and mTOR by Western blotting, and the protein expression of Bcl-2, Bax, caspase-3 and CD31 were detected by immunohistochemical analysis in vivo, respectively. The results showed that TMEA combined with VEGFR2 in the functional pockets of Asn223A, Gly922A, and Leu840A and inhibited the proliferation, migration, tube formation, and expression of VEGF and its downstream signaling mediators in HUVECs. TMEA also significantly inhibited the proliferation of HepG2, A549, and SW620 cancer cells in vitro, and suppressed the growth of SW620 tumors in vivo. Moreover, TMEA upregulated the expression of proapoptotic factors Bax and caspase-3 and downregulated the expression of antiapoptotic factors CD31 and Bcl-2 in cancer cells and/or tumor tissues. The data indicate that TMEA executes its anticancer activity by inducing apoptosis and inhibiting angiogenesis in cancer cells in vitro and tumor growth in vivo. The underlying anticancer mechanism is associated with the apoptotic and VEGF/PI3K/AKT/mTOR pathways.
Collapse
Affiliation(s)
- Chongfei Bai
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Department of Chinese Materia Medica, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yueshan Sun
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xianchao Pan
- Department of Medicine, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jing Yang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Institute of Cardiovascular Research, Key Laboratory of Medical Electrophysiology, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Luzhou, China
| | - Xiaoxuan Li
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Anguo Wu
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Institute of Cardiovascular Research, Key Laboratory of Medical Electrophysiology, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Luzhou, China
| | - Dalian Qin
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Institute of Cardiovascular Research, Key Laboratory of Medical Electrophysiology, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Luzhou, China
| | - Shousong Cao
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Wenjun Zou
- Department of Chinese Materia Medica, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianming Wu
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Institute of Cardiovascular Research, Key Laboratory of Medical Electrophysiology, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Luzhou, China.,Department of Pharmacy, Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
45
|
Xiang W, Wang L, Cheng S, Zhou Y, Ma L. Protective Effects of α-Lipoic Acid on Vascular Oxidative Stress in Rats with Hyperuricemia. Curr Med Sci 2019; 39:920-928. [PMID: 31845223 DOI: 10.1007/s11596-019-2124-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 10/06/2019] [Indexed: 02/07/2023]
Abstract
The aim of the present study was to observe the protective effects of α-lipoic acid (ALA) on vascular injury in rats with hyperuricemia (HUA). The ALA treatment groups (10, 30 and 90 mg/kg, respectively) were administered with ALA via gavage for 2 weeks. Subsequently, the levels of blood urea nitrogen (BUN), creatinine (CREA), uric acid (UA), total cholesterol (TC), high density lipoprotein-C (HDL-C) and low density lipoprotein-C (LDL-C) were measured; the activities of glutathione peroxidase (GSH-Px), catalase (CAT), malonaldehyde (MDA), superoxide dismutase (SOD) and xanthine oxidase (XOD) were also determined. The thoracic aorta of rats in each experimental group was observed under a light microscope; ultrastructural analysis was performed. SOD and CAT protein contents were investigated by Western blotting. The results revealed that: i) Compared with the model group, the levels of UA were decreased in the ALA groups and the levels of BUN, CREA, TC, and LDL-C decreased in the 30 and 90 mg/kg ALA groups (P<0.05); ii) compared with the model group, the activities of GSH-Px, SOD and XOD were increased and the levels of MDA were reduced in the 90 mg/kg ALA group (P<0.05); and iii) in the model and 10 mg/kg ALA groups, edema and shedding were observed in endothelial cells. Compared with the model and 10 mg/kg ALA groups, the 30 and 90 mg/kg ALA groups exhibited fewer swollen endothelial cells. In summary, the results of the present study indicated that HUA resulted in vascular oxidative stress injury and decreased the activity of antioxidative enzymes, which leads to endothelial cell damage and vascular lesions. ALA may serve as a therapeutic agent for the treatment of HUA-induced endothelial dysfunction.
Collapse
Affiliation(s)
- Wei Xiang
- Department of Nutrition and Food Hygiene, School of Public Health, Xinjiang Medical University, Urumqi, 830011, China
| | - Li Wang
- Community Health Service Management Center, Dong cheng District, Beijing, 100000, China
| | - Shi Cheng
- Department of Nutrition and Food Hygiene, School of Public Health, Xinjiang Medical University, Urumqi, 830011, China
| | - Yong Zhou
- Department of Medical Cell Biology and Genetics, College of Preclinical Medicine, Southwest Medical University, Luzhou, 646000, China
| | - Ling Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
46
|
Cao M, Zhu B, Sun Y, Zhao X, Qiu G, Fu W, Jiang H. TBX3 deficiency accelerates apoptosis in cardiomyoblasts through regulation of P21 expression. Life Sci 2019; 239:117040. [PMID: 31704448 DOI: 10.1016/j.lfs.2019.117040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/23/2019] [Accepted: 10/31/2019] [Indexed: 01/22/2023]
Abstract
Congenital heart disease (CHD) is the most common birth defect in newborns. There is increasing evidence that apoptosis and remodeling of the cardiomyoblasts are the major pathology of CHD. Previous research found that T-box transcription factor 3 (TBX3) was compulsory for the regulation of proliferation, cell cycle arrest and apoptosis in various cells. Hence, TBX3 might be involved in the treatment of CHD. The primary aim of this study was to study the effects of TBX3 on apoptosis in aged cardiomyoblasts and investigate the latent mechanism. In the present study, we found TBX3 knockdown induced proliferation inhibition, cell cycle arrest and apoptosis accompanied by mitochondrial dysfunction in cardiomyoblasts at passage 10 to 15. Apoptosis-inducing effects of TBX3 silence could be neutralized by silencing P21 using specific siRNA. In addition, the mRNA and protein expression levels of TBX3 in the heart tissues of sporadic type CHD donors were obviously down-regulated. In conclusion, we demonstrated that TBX3 deficiency accelerated apoptosis via directly regulating P21 expression in senescent cardiomyoblasts.
Collapse
Affiliation(s)
- Meiling Cao
- Department of Neonatology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Binlu Zhu
- Department of Pediatrics, The First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Yuanyuan Sun
- Department of Medical Genetics, College of Basic Medical Sciences, China Medical University, Shenyang, 110122, People's Republic of China
| | - Xueqi Zhao
- Department of Pediatrics, The First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Guangrong Qiu
- Department of Medical Genetics, College of Basic Medical Sciences, China Medical University, Shenyang, 110122, People's Republic of China
| | - Weineng Fu
- Department of Medical Genetics, College of Basic Medical Sciences, China Medical University, Shenyang, 110122, People's Republic of China
| | - Hongkun Jiang
- Department of Pediatrics, The First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China.
| |
Collapse
|
47
|
Liu J, Wang J, Ning Y, Chen F. The inhibition of miR‑101a‑3p alleviates H/R injury in H9C2 cells by regulating the JAK2/STAT3 pathway. Mol Med Rep 2019; 21:89-96. [PMID: 31746349 PMCID: PMC6896302 DOI: 10.3892/mmr.2019.10793] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 08/30/2019] [Indexed: 12/18/2022] Open
Abstract
Hypoxia/reoxygenation (H/R) is used as an in vivo model of ischemia/reperfusion injury, and myocardial ischemia can lead to heart disease. Therefore, it is necessary to prevent myocardial H/R injury to avoid the risk of heart disease. The aim of the present study was to investigate whether inhibiting microRNA (miR)-101a-3p attenuated H9C2 cell H/R injury, apoptosis mechanisms and key target proteins. Cell viability and apoptosis were determined by Cell Counting Kit-8 assays and flow cytometry using a cell apoptosis kit, respectively. The contents of creatine kinase (CK) and lactate dehydrogenase (LDH) were detected using colorimetric assays. Dual luciferase assays were carried out to determine if miR-101a-3p inhibited Janus kinase (JAK)2. Western blot analysis and reverse transcription-quantitative PCR were used to determine proteins levels and mRNAs expression. It was found that the inhibition of miR-101a-3p increased the growth of H9C2 cells and decreased H9C2 cell apoptosis during H/R injury. The inhibition of miR-101a-3p reduced the amounts of CK and LDH in H/R model H9C2 cells. The inhibition of miR-101a-3p lowered the levels of Bax, interleukin-6 and tumor necrosis factor-α, but raised the levels of phosphorylated (p)-STAT3 and p-JAK2 in H9C2 cells subjected to H/R injury treatment. miR-101a-3p mimic was found to inhibit H9C2 cell viability, raise p-JAK2 level and slightly increase p-STAT3 during H/R injury. AG490 induced H9C2 cell apoptosis, and decreased the levels of p-JAK2 and p-STAT3 during H/R injury. The data indicated that inhibiting miR-101a-3p reduced H/R damage in H9C2 cells and decreased apoptosis via Bax/Bcl-2 signaling during H/R injury. In addition, it was suggested that the inhibition of miR-101a-3p decreased H/R injury in H9C2 cell by regulating the JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Jingying Liu
- Emergency Department, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, P.R. China
| | - Juanjuan Wang
- Emergency Department, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, P.R. China
| | - Yuzhen Ning
- Emergency Department, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, P.R. China
| | - Fengying Chen
- Emergency Department, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, P.R. China
| |
Collapse
|
48
|
Bi Y, Fu Y, Wang S, Chen X, Cai X. Schizandrin A exerts anti-tumor effects on A375 cells by down-regulating H19. ACTA ACUST UNITED AC 2019; 52:e8385. [PMID: 31618367 PMCID: PMC6787960 DOI: 10.1590/1414-431x20198385] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 08/14/2019] [Indexed: 02/07/2023]
Abstract
Malignant melanoma (MM) is one of the malignant tumors with highly metastatic and aggressive biological actions. Schizandrin A (SchA) is a bioactive lignin compound with strong anti-oxidant and anti-aging properties, which is stable at room temperature and is often stored in a cool dry place. Hence, we investigated the effects of SchA on MM cell line A375 and its underlying mechanism. A375 cells were used to construct an in vitro MM cell model. Cell viability, proliferation, apoptosis, and migration were detected by Cell Counting Kit-8, BrdU assay, flow cytometry, and transwell two-chamber assay, respectively. The cell cycle-related protein cyclin D1 and cell apoptotic proteins (Bcl-2, Bax, cleaved-caspase-3, and cleaved-caspase-9) were analyzed by western blot. Alteration of H19 expression was achieved by transfecting with pEX-H19. PI3K/AKT pathway was measured by detecting phosphorylation of PI3K and AKT. SchA significantly decreased cell viability in a dose-dependent manner. Furthermore, SchA inhibited cell proliferation and cyclin D1 expression. SchA increased cell apoptosis along with the up-regulation of pro-apoptotic proteins (cleaved-caspase-3, cleaved-caspase-9, and Bax) and the down-regulation of anti-apoptotic protein (Bcl-2). Besides, SchA decreased migration and down-regulated matrix metalloproteinases (MMP)-2 and MMP-9. SchA down-regulated lncRNA H19. Overexpression of H19 blockaded the inhibitory effects of SchA on A375 cells. SchA decreased the phosphorylation of PI3K and AKT while H19 overexpression promoted the phosphorylation of PI3K and AKT. SchA inhibited A375 cell growth, migration, and the PI3K/AKT pathway through down-regulating H19.
Collapse
Affiliation(s)
- Yiming Bi
- Department of Oncology, Binzhou People's Hospital, Binzhou, China
| | - Yan Fu
- Department of Dermatology, Binzhou People's Hospital, Binzhou, China
| | - Shuyan Wang
- Department of Oncology, Binzhou People's Hospital, Binzhou, China
| | - Xingxiu Chen
- Department of Oncology, Binzhou People's Hospital, Binzhou, China
| | - Xiaoping Cai
- Department of Oncology, Binzhou People's Hospital, Binzhou, China
| |
Collapse
|
49
|
Gamasaee NA, Muhammad HA, Tadayon E, Ale-Ebrahim M, Mirpour M, Sharifi M, Salihi A, Shekha MS, Alasady AAB, Aziz FM, Akhtari K, Hasan A, Falahati M. The effects of nickel oxide nanoparticles on structural changes, heme degradation, aggregation of hemoglobin and expression of apoptotic genes in lymphocytes. J Biomol Struct Dyn 2019; 38:3676-3686. [PMID: 31476976 DOI: 10.1080/07391102.2019.1662850] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Nickel oxide nanoparticles (NiO NPs) have received great interests in medical and biotechnological applications. However, their adverse impacts against biological systems have not been well-explored. Herein, the influence of NiO NPs on structural changes, heme degradation and aggregation of hemoglobin (Hb) was evaluated by UV-visible (Vis) spectroscopy, circular dichroism (CD) spectroscopy, fluorescence spectroscopy, transmission electron microscopy (TEM), and molecular modeling investigations. Also, the morphological changes and expression of Bax/Bcl-2 mRNA in human lymphocyte cell exposed to NiO NPs were assayed by DAPI staining and quantitative real-time PCR (qPCR), respectively. The UV-Vis study depicted that NiO NPs resulted in the displacement of aromatic residues and heme groups and production of the pro-aggregatory species. Intrinsic and Thioflavin T (ThT) fluorescence studies revealed that NiO NPs resulted in heme degradation and amorphous aggregation of Hb, respectively, which the latter result was also confirmed by TEM study. Moreover, far UV-CD study depicted that NiO NPs lead to substantial secondary structural changes of Hb. Furthermore, near UV-CD displayed that NiO NPs cause quaternary conformational changes of Hb as well as heme displacement. Molecular modelling study also approved that NiO NPs resulted in structural alterations of Hb and heme deformation. Moreover, morphological and genotoxicity assays revealed that the DNA fragmentation and expression ratio of Bax/Bcl-2 mRNA increased in lymphocyte cells treated with NiO NPs for 24 hr. In conclusion, this study indicates that NiO NPs may affect the biological media and their applications should be limited.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Niusha Abbasi Gamasaee
- Department of Genetics, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hawzheen A Muhammad
- Department of Microbiology, College of Medicine, University of Sulaimani, Sulaimani, Kurdistan Region, Iraq
| | - Elahe Tadayon
- Faculty of Specialized Veterinary Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mahsa Ale-Ebrahim
- Department of Physiology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mirsasan Mirpour
- Department of Microbiology, Faculty of Basic Sciences, Lahijan Branch, Islamic Azad University (IAU), Lahijan, Guilan, Iran
| | - Majid Sharifi
- Department of Nanotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Abbas Salihi
- Department of Biology, College of Science, Salahaddin University-Erbil, Kurdistan Region, Iraq.,Department of Medical Analysis, Faculty of Science, Tishk International University, Erbil, Iraq
| | - Mudhir Sabir Shekha
- Department of Biology, College of Science, Salahaddin University-Erbil, Kurdistan Region, Iraq.,Department of Pathological Analysis, College of Science, Knowledge University, Erbil, Kurdistan Region, Iraq
| | - Asaad A B Alasady
- Anatomy, Histology, and Embryology Unit, College of Medicine, University of Duhok, Kurdistan Region, Iraq
| | - Falah Mohammad Aziz
- Department of Biology, College of Science, Salahaddin University-Erbil, Kurdistan Region, Iraq
| | - Keivan Akhtari
- Department of Physics, University of Kurdistan, Sanandaj, Iran
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha, Qatar.,Biomedical Research Centre (BRC), Qatar University, Doha, Qatar
| | - Mojtaba Falahati
- Department of Nanotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
50
|
Li HY, Li P, Yang HG, Wang YZ, Huang GX, Wang JQ, Zheng N. Investigation and comparison of the anti-tumor activities of lactoferrin, α-lactalbumin, and β-lactoglobulin in A549, HT29, HepG2, and MDA231-LM2 tumor models. J Dairy Sci 2019; 102:9586-9597. [PMID: 31447140 DOI: 10.3168/jds.2019-16429] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 03/23/2019] [Indexed: 12/27/2022]
Abstract
To investigate the anti-tumor activities of lactoferrin, α-lactalbumin, and β-lactoglobulin, 4 types of human tumor cells (lung tumor cell A549, intestinal epithelial tumor cell HT29, hepatocellular cell HepG2, and breast cancer cell MDA231-LM2) were exposed to 3 proteins, respectively. The effects on cell proliferation, migration, and apoptosis were detected in vitro, and nude mice bearing tumors were administered the 3 proteins in vivo. Results showed that the 3 proteins (20 g/L) inhibited viability and migration, as well as induced apoptosis, in 4 tumor cells to different degrees (compared with the control). In vivo, tumor weights in the HT29 group (0.84 ± 0.22 g vs. control 2.05 ± 0.49 g) and MDA231-LM2 group (1.11 ± 0.25 g vs. control 2.49 ± 0.57 g) were significantly reduced by lactoferrin; tumor weights in the A549 group (1.07 ± 0.19 g vs. control 3.11 ± 0.73 g) and HepG2 group (2.32 ± 0.46 g vs. control 3.50 ± 0.74 g) were significantly reduced by α-lactalbumin. Moreover, the roles of lactoferrin, α-lactalbumin, and β-lactoglobulin in regulating apoptotic proteins were validated. In summary, lactoferrin, α-lactalbumin, and β-lactoglobulin were proven to inhibit growth and development of A549, HT29, HepG2, and MDA231-LM2 tumors to different degrees via induction of cell apoptosis.
Collapse
Affiliation(s)
- H Y Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China; Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China; Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| | - P Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China; Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China; Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| | - H G Yang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China; Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China; Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| | - Y Z Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China; Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China; Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| | - G X Huang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China; Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China; Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| | - J Q Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China; Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China; Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| | - N Zheng
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China; Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China; Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China.
| |
Collapse
|