51
|
Pennisi R, Musarra-Pizzo M, Velletri T, Mazzaglia A, Neri G, Scala A, Piperno A, Sciortino MT. Cancer-Related Intracellular Signalling Pathways Activated by DOXorubicin/Cyclodextrin-Graphene-Based Nanomaterials. Biomolecules 2022; 12:63. [PMID: 35053211 PMCID: PMC8773469 DOI: 10.3390/biom12010063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/29/2021] [Accepted: 12/30/2021] [Indexed: 02/07/2023] Open
Abstract
In the last decade, nanotechnological progress has generated new opportunities to improve the safety and efficacy of conventional anticancer therapies. Compared with other carriers, graphene nanoplatforms possess numerous tunable functionalities for the loading of multiple bioactive compounds, although their biocompatibility is still a debated concern. Recently, we have investigated the modulation of genes involved in cancer-associated canonical pathways induced by graphene engineered with cyclodextrins (GCD). Here, we investigated the GCD impact on cells safety, the HEp-2 responsiveness to Doxorubicin (DOX) and the cancer-related intracellular signalling pathways modulated by over time exposure to DOX loaded on GCD (GCD@DOX). Our studies evidenced that both DOX and GCD@DOX induced p53 and p21 signalling resulting in G0/G1 cell cycle arrest. A genotoxic behaviour of DOX was reported via detection of CDK (T14/Y15) activation and reduction of Wee-1 expression. Similarly, we found a cleavage of PARP by DOX within 72 h of exposure. Conversely, GCD@DOX induced a late cleavage of PARP, which could be indicative of less toxic effect due to controlled release of the drug from the GCD nanocarrier. Finally, the induction of the autophagy process supports the potential recycling of DOX with the consequent limitation of its toxic effects. Together, these findings demonstrate that GCD@DOX is a biocompatible drug delivery system able to evade chemoresistance and doxorubicin toxicity.
Collapse
Affiliation(s)
- Rosamaria Pennisi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (M.M.-P.); (G.N.); (A.S.); (A.P.)
| | - Maria Musarra-Pizzo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (M.M.-P.); (G.N.); (A.S.); (A.P.)
| | - Tania Velletri
- IFOM-Cogentech Società Benefit srl; via Adamello 16, 20139 Milan, Italy;
| | - Antonino Mazzaglia
- Istituto per lo Studio dei Materiali Nanostrutturati, Consiglio Nazionale delle Ricerche (ISMN-CNR), V.le F. Stagno d’Alcontres 31, 98166 Messina, Italy;
| | - Giulia Neri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (M.M.-P.); (G.N.); (A.S.); (A.P.)
| | - Angela Scala
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (M.M.-P.); (G.N.); (A.S.); (A.P.)
| | - Anna Piperno
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (M.M.-P.); (G.N.); (A.S.); (A.P.)
| | - Maria Teresa Sciortino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (M.M.-P.); (G.N.); (A.S.); (A.P.)
| |
Collapse
|
52
|
Xu J, Wang SJ, Bu SS, Guo XQ, Ge H. Theaflavin promoted apoptosis in nasopharyngeal carcinoma unexpectedly via inducing autophagy in vitro. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:68-74. [PMID: 35656444 PMCID: PMC9118283 DOI: 10.22038/ijbms.2021.59190.13143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 12/15/2021] [Indexed: 11/17/2022]
Abstract
Objectives This study aimed to investigate the mechanism of the anticancer effect of theaflavin (TF) in nasopharyngeal carcinoma. Materials and Methods CNE2 cells were used to study the anticancer effect of TF. This study used Cell Counting Kit-8 (CCK8) assay on proliferation and used flow cytometry to detect apoptosis. The protein expression of Bcl-2, Bax, caspase 3, and caspase 9 was detected by Western blot, and autophagy-related proteins were also detected. Results TF inhibited proliferation of CNE2 cells, promoted apoptosis, and up-regulated the expression of caspase 3, caspase 9, and Bax, and decreased the level of Bcl-2. Unexpectedly, TF induced autophagy rather than inhibiting autophagy through up-regulating the levels of the autophagy marker light chain 3 (LC3) and Lysosomal-associated membrane protein 1 (LAMP1) and reducing levels of the autophagosome cargo protein p62, and the effect was via the mTOR pathway. Besides, autophagy inhibitor Chloroquine (CQ) suppressed the effect of TF on Bax, Bcl-2 and activation of caspase 3 and caspase 9. Conclusion TF promoted apoptosis of nasopharyngeal carcinoma cells, the mechanism was unexpectedly involved in inducing autophagy.
Collapse
Affiliation(s)
- Jing Xu
- Department of Radiotherapy, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan province, China
| | - Shu-Juan Wang
- Department of Radiotherapy, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan province, China
| | - Shan-Shan Bu
- Department of Radiotherapy, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan province, China
| | - Xiao-Qi Guo
- Department of Radiotherapy, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan province, China
| | - Hong Ge
- Department of Radiotherapy, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan province, China,Corresponding author: Hong Ge. Department of Radiotherapy, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan province, China.
| |
Collapse
|
53
|
Epigenetic Mechanisms in Understanding Nanomaterial-Induced Toxicity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1357:195-223. [DOI: 10.1007/978-3-030-88071-2_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
54
|
Wei L, Zuo Z, Yang Z, Yin H, Yang Y, Fang J, Cui H, Du Z, Ouyang P, Chen X, Chen J, Geng Y, Zhu Y, Chen Z, Huang C, Wang F, Guo H. Mitochondria damage and ferroptosis involved in Ni-induced hepatotoxicity in mice. Toxicology 2021; 466:153068. [PMID: 34921910 DOI: 10.1016/j.tox.2021.153068] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/13/2021] [Accepted: 12/14/2021] [Indexed: 02/07/2023]
Abstract
Nickel (Ni) is an environmental toxicant that can cause toxic damage to humans and animals. Although the hepatotoxicity of Ni has been confirmed, its precise mechanism is still unclear. In this study, the results showed that nickel chloride (NiCl2)-treatment could induce mice hepatotoxicity including hepatic histopathological alterations and up-regulation of serum AST and ALT. According to the results, NiCl2 increased malondialdehyde (MDA) production while reducing total antioxidant capacity (T-AOC) activity and glutathione (GSH) content. Additionally, NiCl2 induced mitochondrial damage which was featured by increase in mitochondrial ROS (mt-ROS) and mitochondrial membrane potential (MMP) depolarization. The mitochondrial respiratory chain complexes I-IV and ATP content were decreased in the liver of NiCl2-treated mice. Meanwhile, NiCl2 caused hepatic ferroptosis accompanied by increased iron content in the liver and up-regulation of cyclooxygenase 2 (COX-2) protein and mRNA expression levels, down-regulation of glutathione eroxidase 4 (GPX4), ferritin heavy chain 1 (FTH1) and nuclear receptor coactivator 4 (NCOA4) protein and mRNA expression levels. Altogether, the above mentioned results indicate that NiCl2 treatment may induce hepatic damage through mitochondrial damage and ferroptosis.
Collapse
Affiliation(s)
- Ling Wei
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
| | - Zhicai Zuo
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
| | - Zhuangzhi Yang
- Chengdu Academy of Agriculture and Forestry Sciences, Chengdu, Sichuan, 611130, China
| | - Heng Yin
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
| | - Yue Yang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
| | - Jing Fang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China.
| | - Hengmin Cui
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
| | - Zongjun Du
- College of Animal Science and Technology, Sichuan Agriculture University, Wenjiang, Chengdu, 611130, China
| | - Ping Ouyang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
| | - Xia Chen
- Chengdu Academy of Agriculture and Forestry Sciences, Chengdu, Sichuan, 611130, China
| | - Jian Chen
- Chengdu Academy of Agriculture and Forestry Sciences, Chengdu, Sichuan, 611130, China
| | - Yi Geng
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
| | - Yanqiu Zhu
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
| | - Zhengli Chen
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
| | - Chao Huang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
| | - Fengyuan Wang
- College of Animal Science and Veterinary Medicine, Southwest Minzu University, Chengdu, Sichuan, 610041, China
| | - Hongrui Guo
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China.
| |
Collapse
|
55
|
Ginsenoside Rg1 Prevents Cognitive Impairment and Hippocampal Neuronal Apoptosis in Experimental Vascular Dementia Mice by Promoting GPR30 Expression. Neural Plast 2021; 2021:2412220. [PMID: 34899899 PMCID: PMC8664545 DOI: 10.1155/2021/2412220] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 11/19/2021] [Indexed: 01/02/2023] Open
Abstract
This study is aimed at investigating the potential roles of G protein-coupled estrogen receptor 1 (GPER, also known as GPR30) in the preventive effect of ginsenoside Rg1 against cognitive impairment and hippocampal cell apoptosis in experimental vascular dementia (VD) in mice. The effects of bilateral common carotid artery stenosis (BCAS) on GPR30 expression at mRNA level were evaluated. Thereafter, the BCAS mouse model was utilized to evaluate the protection of Rg1 (0.1, 1, 10 mg/kg, 14 days, ip). Spatial memory was evaluated by water Morris Maze 7 days post BCAS. After behavioral tests, neuronal apoptosis was detected by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay, and potential mechanisms were determined using western blotting and quantitative real-time PCR. Our results showed that GPR30 expression in the hippocampal region at mRNA level was promoted 30 min, 3 h, 6 h, and 24 h following BCAS. Ginsenoside Rg1 (1 or 10 mg/kg, 14 days, ip) promoted GPR30 expression in the hippocampus of model mice (after behavioral tests) but did not alter GPR30 expression in the hippocampus of control mice. Moreover, treatment of ginsenoside Rg1 (10 mg/kg) or G1 (5 μg/kg), a GPR30 agonist, prevented BCAS-induced memory impairment and hippocampal neuronal loss and apoptosis and promoted the ratio of Bcl-2 to Bax expression in the hippocampus (after behavioral tests). On the contrary, G15 (185 μg/kg), an antagonist of GPR30, aggravated BCAS-induced hippocampal neuronal loss and apoptosis. Finally, drug-target molecular docking pointed that Rg1 had a lower binding energy with GPR30 compared with Bax and Bcl-2. Together, our data implicate that ginsenoside Rg1 prevents cognitive impairment and hippocampal neuronal apoptosis in VD mice, likely through promoting GPR30 expression. These results would provide important implications for the application of Rg1 in the treatment of VD.
Collapse
|
56
|
Hu W, Gao W, Miao J, Xu Z, Sun L. Alamandine, a derivative of angiotensin-(1-7), alleviates sepsis-associated renal inflammation and apoptosis by inhibiting the PI3K/Ak and MAPK pathways. Peptides 2021; 146:170627. [PMID: 34400214 DOI: 10.1016/j.peptides.2021.170627] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 08/06/2021] [Accepted: 08/10/2021] [Indexed: 12/15/2022]
Abstract
Sepsis is a frequent cause of kidney injury. The present study investigated whether Alamandine (Ala) could alleviate sepsis-associated renal injury by reducing inflammation and apoptosis. In addition, we investigated downstream signaling pathways modulated by Ala. Studies were performed in mice treated with lipopolysaccharide (LPS) and in the human proximal tubular epithelial cell line HK-2. The increase in serum creatinine, blood urea nitrogen, cystatin C and Fg, and neutrophil gelatinase-associated lipocalin and kidney injury molecule-1 in the kidneys of mice treated with LPS were reduced after administration of Ala. Exposure to LPS increased interleukin-1 beta (IL-1β), IL-6, and tumor necrosis factor alpha (TNF-α) in mice and HK-2 cells, but were reduced after Ala treatment. Furthermore, increased levels of cleaved caspase 3, cleaved caspase 7, cleaved caspase 9, cleaved poly (ADP-ribose) polymerase (PARP) and Bax and reduced levels of Bcl2 in LPS-treated mice and HK-2 cells were reversed after Ala administration. In addition, LPS increased the levels of p-PI3K/PI3K, p-Akt/Akt, p-ERK/ERK, p-JNK/JNK, p-p38/p38 and p-FoxO1 in HK-2 cells, and all were reversed after Ala administration. These results indicate that Ala could improve renal function and inhibit inflammation and apoptosis in LPS induced sepsis mouse models. We demonstrated that Ala attenuated LPS induced sepsis by inhibiting the PI3K/Akt and MAPK signaling pathways.
Collapse
Affiliation(s)
- Wei Hu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, China
| | - Wenchuang Gao
- Department of Thoracic Surgery, Lian Shui People's Hospital, Huaian, China
| | - Jiayi Miao
- Department of Nephrology, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, China
| | - Ziheng Xu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, China
| | - Lei Sun
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, China.
| |
Collapse
|
57
|
Ye C, Zheng S, Jiang D, Lu J, Huang Z, Liu Z, Zhou H, Zhuang C, Li J. Initiation and Execution of Programmed Cell Death and Regulation of Reactive Oxygen Species in Plants. Int J Mol Sci 2021; 22:ijms222312942. [PMID: 34884747 PMCID: PMC8657872 DOI: 10.3390/ijms222312942] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/19/2021] [Accepted: 11/24/2021] [Indexed: 12/21/2022] Open
Abstract
Programmed cell death (PCD) plays crucial roles in plant development and defence response. Reactive oxygen species (ROS) are produced during normal plant growth, and high ROS concentrations can change the antioxidant status of cells, leading to spontaneous cell death. In addition, ROS function as signalling molecules to improve plant stress tolerance, and they induce PCD under different conditions. This review describes the mechanisms underlying plant PCD, the key functions of mitochondria and chloroplasts in PCD, and the relationship between mitochondria and chloroplasts during PCD. Additionally, the review discusses the factors that regulate PCD. Most importantly, in this review, we summarise the sites of production of ROS and discuss the roles of ROS that not only trigger multiple signalling pathways leading to PCD but also participate in the execution of PCD, highlighting the importance of ROS in PCD.
Collapse
Affiliation(s)
- Chanjuan Ye
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China; (C.Y.); (S.Z.); (D.J.); (J.L.); (Z.H.); (Z.L.); (H.Z.); (C.Z.)
- Key Laboratory of Plant Functional Genomics and Biotechnology of Guangdong Provincial Higher Education Institutions, South China Agricultural University, Guangzhou 510642, China
- College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Shaoyan Zheng
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China; (C.Y.); (S.Z.); (D.J.); (J.L.); (Z.H.); (Z.L.); (H.Z.); (C.Z.)
- Key Laboratory of Plant Functional Genomics and Biotechnology of Guangdong Provincial Higher Education Institutions, South China Agricultural University, Guangzhou 510642, China
- College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Dagang Jiang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China; (C.Y.); (S.Z.); (D.J.); (J.L.); (Z.H.); (Z.L.); (H.Z.); (C.Z.)
- Key Laboratory of Plant Functional Genomics and Biotechnology of Guangdong Provincial Higher Education Institutions, South China Agricultural University, Guangzhou 510642, China
- College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Jingqin Lu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China; (C.Y.); (S.Z.); (D.J.); (J.L.); (Z.H.); (Z.L.); (H.Z.); (C.Z.)
- Key Laboratory of Plant Functional Genomics and Biotechnology of Guangdong Provincial Higher Education Institutions, South China Agricultural University, Guangzhou 510642, China
- College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Zongna Huang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China; (C.Y.); (S.Z.); (D.J.); (J.L.); (Z.H.); (Z.L.); (H.Z.); (C.Z.)
- Key Laboratory of Plant Functional Genomics and Biotechnology of Guangdong Provincial Higher Education Institutions, South China Agricultural University, Guangzhou 510642, China
- College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Zhenlan Liu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China; (C.Y.); (S.Z.); (D.J.); (J.L.); (Z.H.); (Z.L.); (H.Z.); (C.Z.)
- Key Laboratory of Plant Functional Genomics and Biotechnology of Guangdong Provincial Higher Education Institutions, South China Agricultural University, Guangzhou 510642, China
- College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Hai Zhou
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China; (C.Y.); (S.Z.); (D.J.); (J.L.); (Z.H.); (Z.L.); (H.Z.); (C.Z.)
- Key Laboratory of Plant Functional Genomics and Biotechnology of Guangdong Provincial Higher Education Institutions, South China Agricultural University, Guangzhou 510642, China
- College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Chuxiong Zhuang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China; (C.Y.); (S.Z.); (D.J.); (J.L.); (Z.H.); (Z.L.); (H.Z.); (C.Z.)
- Key Laboratory of Plant Functional Genomics and Biotechnology of Guangdong Provincial Higher Education Institutions, South China Agricultural University, Guangzhou 510642, China
- College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Jing Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China; (C.Y.); (S.Z.); (D.J.); (J.L.); (Z.H.); (Z.L.); (H.Z.); (C.Z.)
- Key Laboratory of Plant Functional Genomics and Biotechnology of Guangdong Provincial Higher Education Institutions, South China Agricultural University, Guangzhou 510642, China
- College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
- Correspondence:
| |
Collapse
|
58
|
Nephrotoxicity induced by cisplatin is primarily due to the activation of the 5-hydroxytryptamine degradation system in proximal renal tubules. Chem Biol Interact 2021; 349:109662. [PMID: 34560070 DOI: 10.1016/j.cbi.2021.109662] [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: 05/26/2021] [Revised: 08/16/2021] [Accepted: 09/16/2021] [Indexed: 01/07/2023]
Abstract
As a widely used anticancer drug in the clinic, cisplatin has obvious side effects, especially nephrotoxicity. Previous studies have suggested that the accumulation of intracellular reactive oxygen species (ROS) is a hallmark of cisplatin-induced acute kidney injury. This study aimed to investigate the relationship between ROS accumulation induced by cisplatin and 5-HT degradation. In vivo, by HE and TUNEL staining, we found that cisplatin-induced renal lesions and apoptotic regions, which were located in proximal tubular epithelial cells, were also the regions in which tryptophan hydroxylase 1 (Tph1), aromatic l-amino acid decarboxylase (AADC), 5-HT2A receptor (5-HT2AR) and monoamine oxidase A (MAO-A) were overexpressed, as determined by immunohistochemistry. Notably, the 5-HT2AR antagonist sarpogrelate hydrochloride (SH) and the AADC inhibitor carbidopa (CDP) significantly attenuated cisplatin-induced increases in serum creatinine and blood urea nitrogen levels, renal ROS levels, oxidative stress (SOD activity and MDA), proinflammatory cytokine levels (NF-κB, TNF-α and IL-1β), proapoptotic factor levels (Bax, Bcl-2, C-caspase 3 and C-caspase 9) and the phosphorylation of p38 and STAT3, as well as renal lesions and apoptosis. The combination of SH and CDP could almost abolish the effects of cisplatin challenge. In vitro, the effects of cisplatin challenge and the inhibitory effects of SH and CDP were also observed in HK-2 cells. Additionally, similar to the combination of SH and CDP, the MAO-A inhibitor clorgyline could also abolish the effects of cisplatin challenge. More importantly, by western blotting, we detected that the upregulation of Tph1, AADC and MAO-A expression induced by cisplatin both in vivo and in vitro could be obviously suppressed by SH to decrease 5-HT synthesis and mitochondrial 5-HT degradation. Altogether, these findings suggested that cisplatin-induced nephrotoxicity is due to the activation of the 5-HT degradation system in proximal tubular epithelial cells, including 5-HT2AR and 5-HT synthesis and degradation. 5-HT2AR plays a role by mediating the expression of MAO-A and the 5-HT synthases Tph1 and AADC.
Collapse
|
59
|
Povo-Retana A, Mojena M, Boscá A, Pedrós J, Peraza DA, Valenzuela C, Laparra JM, Calle F, Boscá L. Graphene Particles Interfere with Pro-Inflammatory Polarization of Human Macrophages: Functional and Electrophysiological Evidence. Adv Biol (Weinh) 2021; 5:e2100882. [PMID: 34590442 DOI: 10.1002/adbi.202100882] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 09/09/2021] [Indexed: 02/05/2023]
Abstract
The interaction of two types of fragmented graphene particles (30-160 nm) with human macrophages is studied. Since macrophages have significant phagocytic activity, the incorporation of graphene particles into cells has an effect on the response to functional polarization stimuli, favoring an anti-inflammatory profile. Incubation of macrophages with graphene foam particles, prepared by chemical vapor deposition, and commercially available graphene nanoplatelet particles does not affect cell viability when added at concentrations up to 100 µg mL-1 ; macrophages exhibit differential quantitative responses to each type of graphene particles. Although both materials elicit similar increases in the release of reactive oxygen species, the impact on the transcriptional regulation associated with the polarization profile is different; graphene nanoplatelets significantly modify this transcriptomic profile. Moreover, these graphene particles differentially affect the motility and phagocytosis of macrophages. After the incorporation of both graphene types into the macrophages, they exhibit specific responses in terms of the mitochondrial oxygen consumption and electrophysiological potassium currents at the cell plasma membrane. These data support the view that the physical structure of the graphene particles has an impact on human macrophage responses, paving the way for the development of new mechanisms to modulate the activity of the immune system.
Collapse
Affiliation(s)
- Adrián Povo-Retana
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, Madrid, 28029, Spain
| | - Marina Mojena
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, Madrid, 28029, Spain
| | - Alberto Boscá
- Instituto de Sistemas Optoelectrónicos y Microtecnología (ISOM) and Departamento de Ingeniería Electrónica, E.T.S.I. de Telecomunicación, Universidad Politécnica de Madrid, Madrid, 28040, Spain
| | - Jorge Pedrós
- Instituto de Sistemas Optoelectrónicos y Microtecnología (ISOM) and Departamento de Ingeniería Electrónica, E.T.S.I. de Telecomunicación, Universidad Politécnica de Madrid, Madrid, 28040, Spain
| | - Diego Alberto Peraza
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, Madrid, 28029, Spain
| | - Carmen Valenzuela
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, Madrid, 28029, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares, CIBERCV, Melchor Fernández Almagro, Madrid, 28029, Spain
| | - José Moisés Laparra
- J. M. Laparra, Madrid Institute for Advanced studies in Food (IMDEA Food), Ctra. Cantoblanco 8, Madrid, 28049, Spain
| | - Fernando Calle
- Instituto de Sistemas Optoelectrónicos y Microtecnología (ISOM) and Departamento de Ingeniería Electrónica, E.T.S.I. de Telecomunicación, Universidad Politécnica de Madrid, Madrid, 28040, Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, Madrid, 28029, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares, CIBERCV, Melchor Fernández Almagro, Madrid, 28029, Spain
| |
Collapse
|
60
|
Zamorina SA, Khramtsov PV, Rayev MB, Timganova VP, Bochkova MS, Nechaev AI, Shunkin EO, Khaziakhmatova OG, Malaschenko VV, Litvinova LS. Graphene Oxide Nanoparticels Interaction with Jurkat Cell Line in Cell-IQ System. DOKL BIOCHEM BIOPHYS 2021; 501:438-443. [PMID: 34966968 DOI: 10.1134/s1607672921060089] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 11/23/2022]
Abstract
In recent years, materials based on graphene oxide (GO) have been actively studied for their use in biomedicine. The aim of our study was to investigate the increase in cell mass and viability of Jurkat tumor line T cells during 24 h of contact with GO nanoparticles in the Cell-IQ system of intravital observation. We used nanoparticles of different sizes coated with linear or branched polyethylene glycol (PEG) at concentrations of 5 and 25 μg/mL. It was shown for the first time that direct contact with GO nanoparticles reduced the growth in cell mass at the visualization points by more than twofold, regardless of nanoparticle size and concentration. Moreover, the number of live cells in the culture decreased by 5-9% after 24 h of monitoring. Thus, PEG-coated GO nanoparticles were found to suppress the proliferation and viability of Jurkat cell line T lymphocytes.
Collapse
Affiliation(s)
- S A Zamorina
- Institute of Ecology and Genetics of Microorganisms, UB RAS Branch of the Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Perm, Russia.
| | - P V Khramtsov
- Institute of Ecology and Genetics of Microorganisms, UB RAS Branch of the Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Perm, Russia
| | - M B Rayev
- Institute of Ecology and Genetics of Microorganisms, UB RAS Branch of the Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Perm, Russia
| | - V P Timganova
- Institute of Ecology and Genetics of Microorganisms, UB RAS Branch of the Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Perm, Russia
| | - M S Bochkova
- Institute of Ecology and Genetics of Microorganisms, UB RAS Branch of the Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Perm, Russia
| | - A I Nechaev
- Institute of Technical Chemistry, UB RAS Branch of the Perm Federal Research, Center of the Ural Branch of the Russian Academy of Sciences, Perm, Russia
| | - E O Shunkin
- Kant Baltic Federal University, Kaliningrad, Russia
| | | | | | | |
Collapse
|
61
|
Li FH, Guo SW, Zhan TW, Mo HR, Chen X, Wang H, Wei LL, Feng PF, Wu JG, Li P. Integrating network pharmacology and experimental evidence to decipher the cardioprotective mechanism of Yiqihuoxue decoction in rats after myocardial infarction. JOURNAL OF ETHNOPHARMACOLOGY 2021; 279:114062. [PMID: 33771641 DOI: 10.1016/j.jep.2021.114062] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 05/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE "Qi deficiency and blood stasis" syndrome is one of the most common syndromes treated with Traditional Chinese Medicine among ischemic heart disease (IHD) patients in clinic. As a Chinese herbal formula with the function of tonifying Qi and activating blood, Yiqihuoxue Decoction (YQHX) has been frequently proven to be effective in the clinical treatment of IHD. AIM OF THE STUDY The cardioprotective mechanisms of YQHX in treating ischemic heart disease were investigated, with emphasis on the key targets and pathways. MATERIALS AND METHODS In the present study, the potential targets of compounds identified in YQHX were predicted using PharmMapper, Symmap, and STITCH databases, and a "herb-compound-target" network was constructed using Cytoscape. Subsequently, the GO and KEGG functional enrichment analyses were analyzed using the DAVID database. Furthermore, a protein-protein interaction network was constructed using STRING to obtain the key target information. Besides, we used a myocardial ischemia rat model to investigate the cardioprotective effects of YQHX. Transmission electron microscopy and Western blotting were used to observe apoptotic bodies and confirm protein expressions of key candidate targets, respectively. RESULTS Network pharmacology showed that a total of 141 potential targets were obtained from these databases. The functional analysis results revealed that the targets of YQHX were largely associated with apoptosis, and the PI3K-AKT and MAPK pathways might represent key functional pathways. The hub genes of network include ALB, TP53, AKT1, TNF, VEGFA, EGFR, MAPK1, CASP3, JUN, FN1, MMP9, and MAPK8. In vivo, YQHX significantly improved cardiac function and suppressed apoptosis in ischemic rat myocardium. Furthermore, YQHX could significantly upregulate Nrf2 and HO-1 expression, and inhibit JNK phosphorylation. CONCLUSIONS Based on network pharmacology and experimental evidence, this study proves that the cardioprotective effects and mechanisms of YQHX depend on multi-component, multi-target, and multi-pathway. In particular, YQHX exerts anti-apoptotic effects potentially by regulating the Nrf2/HO-1 and JNK-MAPK pathways.
Collapse
Affiliation(s)
- Fang-He Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China; The 3rd Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Shu-Wen Guo
- Fangshan Hospital, Beijing University of Chinese Medicine, Beijing, 102400, PR China.
| | - Tian-Wei Zhan
- Science and Technology College of Jiangxi University of Traditional Chinese Medicine, Nanchang, 330000, PR China
| | - Han-Rong Mo
- Beijing Friendship Hospital, Capital Medical University, Beijing, 100029, PR China
| | - Xi Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Hui Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Lu-Lu Wei
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Peng-Fei Feng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Jian-Gong Wu
- Department of Traditional Chinese Medicine, Peking University People's Hospital, Beijing, 100029, PR China
| | - Ping Li
- The 3rd Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
62
|
Tirumala MG, Anchi P, Raja S, Rachamalla M, Godugu C. Novel Methods and Approaches for Safety Evaluation of Nanoparticle Formulations: A Focus Towards In Vitro Models and Adverse Outcome Pathways. Front Pharmacol 2021; 12:612659. [PMID: 34566630 PMCID: PMC8458898 DOI: 10.3389/fphar.2021.612659] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 08/05/2021] [Indexed: 12/18/2022] Open
Abstract
Nanotoxicology is an emerging field employed in the assessment of unintentional hazardous effects produced by nanoparticles (NPs) impacting human health and the environment. The nanotoxicity affects the range between induction of cellular stress and cytotoxicity. The reasons so far reported for these toxicological effects are due to their variable sizes with high surface areas, shape, charge, and physicochemical properties, which upon interaction with the biological components may influence their functioning and result in adverse outcomes (AO). Thus, understanding the risk produced by these materials now is an important safety concern for the development of nanotechnology and nanomedicine. Since the time nanotoxicology has evolved, the methods employed have been majorly relied on in vitro cell-based evaluations, while these simple methods may not predict the complexity involved in preclinical and clinical conditions concerning pharmacokinetics, organ toxicity, and toxicities evidenced through multiple cellular levels. The safety profiles of nanoscale nanomaterials and nanoformulations in the delivery of drugs and therapeutic applications are of considerable concern. In addition, the safety assessment for new nanomedicine formulas lacks regulatory standards. Though the in vivo studies are greatly needed, the end parameters used for risk assessment are not predicting the possible toxic effects produced by various nanoformulations. On the other side, due to increased restrictions on animal usage and demand for the need for high-throughput assays, there is a need for developing and exploring novel methods to evaluate NPs safety concerns. The progress made in molecular biology and the availability of several modern techniques may offer novel and innovative methods to evaluate the toxicological behavior of different NPs by using single cells, cell population, and whole organisms. This review highlights the recent novel methods developed for the evaluation of the safety impacts of NPs and attempts to solve the problems that come with risk assessment. The relevance of investigating adverse outcome pathways (AOPs) in nanotoxicology has been stressed in particular.
Collapse
Affiliation(s)
- Mounika Gayathri Tirumala
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Pratibha Anchi
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Susmitha Raja
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Mahesh Rachamalla
- Department of Biology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Chandraiah Godugu
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| |
Collapse
|
63
|
Achawi S, Pourchez J, Feneon B, Forest V. Graphene-Based Materials In Vitro Toxicity and Their Structure-Activity Relationships: A Systematic Literature Review. Chem Res Toxicol 2021; 34:2003-2018. [PMID: 34424669 DOI: 10.1021/acs.chemrestox.1c00243] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The unique properties of graphene-based materials (GBMs) placed them among the most exciting nanomaterials of the past decade. Scientists and industry are looking forward to working with not only efficient but also safe, sustainable GBMs. Designing a safer-by-design GBM implies to acquire the knowledge of which physicochemical characteristics (PCCs) can increase toxicity. In this systematic review, we extracted data from the literature to provide the available information about the structure-activity relationship of GBMs. 93 papers studying a total of 185 GBMs are included. Graphene oxides (GOs) and few-layer graphenes (FLGs) are the most studied GBMs. While reduced graphene oxides were often classified as poorly oxidant and weakly cytotoxic, graphene quantum dots were mostly moderately or highly cytotoxic. FLGs demonstrated relationships between median size and oxidative stress, between lateral size and both cytotoxicity and oxidative stress, and between thickness and cytotoxicity. We also underline relationships between median size, lateral size, and thickness of GOs and oxidative stress. However, it appears difficult to highlight clear structure-activity relationships for most PCCs and biological end points because despite a large amount of available data, the GBMs are often too poorly characterized in terms of PCCs descriptors and the biological end points investigation is not standardized enough. There is an urgent need for a better standardization of the experimental investigation of both PCCs and biological end points to allow research teams to play a part in the collaborative work toward the construction of a safer-by-design GBM through a better understanding of their key toxicity drivers.
Collapse
Affiliation(s)
- Salma Achawi
- Manufacture Française des Pneumatiques Michelin, Place des Carmes Déchaux, 63040 Clermont-Ferrand, Cedex 9, France.,Mines Saint-Etienne, Université Lyon, Université Jean Monnet, INSERM, U1059 Sainbiose, Centre CIS, F-42023 Saint-Etienne, France
| | - Jérémie Pourchez
- Mines Saint-Etienne, Université Lyon, Université Jean Monnet, INSERM, U1059 Sainbiose, Centre CIS, F-42023 Saint-Etienne, France
| | - Bruno Feneon
- Manufacture Française des Pneumatiques Michelin, Place des Carmes Déchaux, 63040 Clermont-Ferrand, Cedex 9, France
| | - Valérie Forest
- Mines Saint-Etienne, Université Lyon, Université Jean Monnet, INSERM, U1059 Sainbiose, Centre CIS, F-42023 Saint-Etienne, France
| |
Collapse
|
64
|
Zhang X, Xu X, Lu L, Wan X, Qin Y, Ruan W, Lv C, He L, Guo X. A new Mfn-2 related synthetic peptide promotes vascular smooth muscle cell apoptosis via regulating the mitochondrial apoptotic pathway by inhibiting Akt signaling. J Transl Med 2021; 19:395. [PMID: 34538249 PMCID: PMC8451139 DOI: 10.1186/s12967-021-03064-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 09/02/2021] [Indexed: 01/11/2023] Open
Abstract
Background Restenosis after angioplasty is a major challenge for the treatment of coronary artery diseases. Facilitation of vascular smooth muscle cell (VSMC) apoptosis may be an attractive approach to decrease the incidence of restenosis. We synthesized a 16-amino acid mitofusin-2 (Mfn-2) gene related peptide (MRSP) based on the sequence of the p21ras signature motif, the smallest functional sequence of the Mfn-2 gene with proapoptotic properties in VSMC. We investigated whether MRSP enhanced apoptotic activities to inhibit VSMC accumulation and neointimal hyperplasia in rats with carotid balloon injury. Methods VSMCs were treated with different concentrations of MRSP, the PI3K agonist 740 Y-P and the inhibitor LY294002. Cell apoptosis and related pathway molecules were assessed. MRSP was also given to rats with carotid artery balloon injury. Neointimal hyperplasia and cell apoptotic pathways were detected. Results In vitro experiments revealed that MRSP treatment significantly increased VSMC apoptosis and induced increases in procaspase-9 cleavage, caspase-3 activation, cytochrome c release from mitochondria to the cytoplasm and the Bax/Bcl-2 ratio but not caspase-8 expression, indicating that the mitochondrial apoptotic cascade was activated by MRSP, which might be attributed to suppression of the PI3K/Akt signaling pathway. We further found that the PI3K agonist 740 Y-P prevented and that the inhibitor LY294002 strengthened the proapoptotic effects of MRSP. MRSP strongly inhibited neointimal hyperplasia and VSMC accumulation, but increased VSMC apoptosis in the vascular wall after balloon injury. Moreover, MRSP substantially enhanced Bax and cleaved caspase-3 expression and decreased Bcl-2 levels in intima, accompanied by decreased levels of phosphorylated Akt and PI3K in vivo. Conclusions Taken together, the present study showed that MRSP treatment results in a strong proapoptotic effect by activating the mitochondrial apoptotic cascade through suppression of the PI3K/Akt pathway. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-03064-1.
Collapse
Affiliation(s)
- Xinxin Zhang
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiangyu Xu
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Department of Cardiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Shandong, China
| | - Li Lu
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoning Wan
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yating Qin
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Weibin Ruan
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chao Lv
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lin He
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiaotong University, Shanghai, China
| | - Xiaomei Guo
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
65
|
Sun R, He X, Jiang X, Tao H. The new role of riluzole in the treatment of pancreatic cancer through the apoptosis and autophagy pathways. J Cell Biochem 2021; 122:934-944. [PMID: 31709624 DOI: 10.1002/jcb.29533] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 10/08/2019] [Indexed: 01/25/2023]
Abstract
Pancreatic cancer is always diagnosed at an advanced stage. Hence, chemotherapy becomes the best choice for patients. Therefore, new anticancer drugs for pancreatic cancer are needed. Riluzole (RIL) is mainly used to treat amyotrophic lateral sclerosis clinically, but many previous studies have shown that RIL could inhibit tumors. However, no report has explored the association between RIL and pancreatic cancer. To validate this association, we performed this study. Our data showed that RIL could induce cytotoxicity, block the cell cycle, and inhibit clone formation, apoptosis, and migration in pancreatic cancer cells. Moreover, we demonstrated that RIL could suppress autophagy. However, more experiments will be needed to validate the reliability of our conclusions. In summary, our data suggest that RIL might provide clues for the development of a treatment for human pancreatic cancer in the future.
Collapse
Affiliation(s)
- Rulin Sun
- Clinical Laboratory Center, Zhejiang Provincial People's Hospital, Zhejiang, P. R. China
- People's Hospital of Hangzhou Medical College, Hangzhou, P. R. China
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Zhejiang, P. R. China
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, Zhejiang, P. R. China
| | - Xujun He
- People's Hospital of Hangzhou Medical College, Hangzhou, P. R. China
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, Zhejiang, P. R. China
| | - Xiaoting Jiang
- Clinical Laboratory Center, Zhejiang Provincial People's Hospital, Zhejiang, P. R. China
- People's Hospital of Hangzhou Medical College, Hangzhou, P. R. China
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, Zhejiang, P. R. China
| | - Houquan Tao
- People's Hospital of Hangzhou Medical College, Hangzhou, P. R. China
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, Zhejiang, P. R. China
| |
Collapse
|
66
|
Yu YY, Liu QP, Li MT, An P, Chen YY, Luan X, Lv C, Zhang H. Hu-Zhang-Qing-Mai-Yin Inhibits Proliferation of Human Retinal Capillary Endothelial Cells Exposed to High Glucose. Front Pharmacol 2021; 12:732655. [PMID: 34421625 PMCID: PMC8377758 DOI: 10.3389/fphar.2021.732655] [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: 06/29/2021] [Accepted: 07/29/2021] [Indexed: 11/27/2022] Open
Abstract
Background: Diabetic retinopathy (DR) is one of the serious complications of diabetes and an important cause of blindness. Despite much research on the pathogenesis of DR, there is still a lack of safe and effective treatment methods. Hu-zhang-qing-mai-yin (HZQMY), a Chinese medicine formula, has been clinically used in the safe and effective treatment of DR for many years. However, the systematic pharmacological research is lacking. The aim of this study was to evaluate the anti-DR effects of HZQMY and explore the possible mechanism involved. Methods: The constituents of HZQMY were analyzed by LC-MS/MS. DR model was established by high glucose simulation on human retinal capillary endothelial cells (HRCECs) in vitro. The cell viability, cell proliferation, cell apoptosis, and tube formation were assessed. Subsequently the related mechanisms were analyzed by assays for JC-1 mitochondrial membrane potential (MMP), intracellular ROS, ATP, western blot and proteomics. Results: 27 main chemical components contained in HZQMY were identified. HZQMY significantly inhibited the viability and proliferation of HRCECs exposed to high glucose, and promoted the apoptosis. In addition, HZQMY also boosted the release of ROS and suppressed tube formation of HRCECs under high glucose exposure. Meanwhile, HRCECs treated with high glucose released more ROS than normal cells, which could be markedly inhibited by HZQMY in a dose-dependent manner. Additionally, western blot assay indicated that HZQMY increased the expression of proteins related to the P38 signaling pathway and inhibited nuclear factor kappa-B (NF-κB) pathway. Proteomic analysis predicted that HSPA4, MAPK3, ENO1, EEF2 and ERPS may be the candidate targets of HZQMY in HRCECs. Conclusions: HZQMY inhibited the proliferation and promoted the Mitochondria related apoptosis of HRCECs exposed to high glucose possibly through regulating P38 and NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yuan-Yuan Yu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qiu-Ping Liu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Meng-Ting Li
- Department of Neurology, Yueyang Integrated Traditional Chinese and Western Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Pei An
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu-Ying Chen
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin Luan
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chao Lv
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hong Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
67
|
Ristic B, Harhaji-Trajkovic L, Bosnjak M, Dakic I, Mijatovic S, Trajkovic V. Modulation of Cancer Cell Autophagic Responses by Graphene-Based Nanomaterials: Molecular Mechanisms and Therapeutic Implications. Cancers (Basel) 2021; 13:cancers13164145. [PMID: 34439299 PMCID: PMC8392723 DOI: 10.3390/cancers13164145] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/06/2021] [Accepted: 08/13/2021] [Indexed: 01/18/2023] Open
Abstract
Simple Summary Graphene-based nanomaterials (GNM) are one-to-several carbon atom-thick flakes of graphite with at least one lateral dimension <100 nm. The unique electronic structure, high surface-to-volume ratio, and relatively low toxicity make GNM potentially useful in cancer treatment. GNM such as graphene, graphene oxide, graphene quantum dots, and graphene nanofibers are able to induce autophagy in cancer cells. During autophagy the cell digests its own components in organelles called lysosomes, which can either kill cancer cells or promote their survival, as well as influence the immune response against the tumor. However, a deeper understanding of GNM-autophagy interaction at the mechanistic and functional level is needed before these findings could be exploited to increase GNM effectiveness as cancer therapeutics and drug delivery systems. In this review, we analyze molecular mechanisms of GNM-mediated autophagy modulation and its possible implications for the use of GNM in cancer therapy. Abstract Graphene-based nanomaterials (GNM) are plausible candidates for cancer therapeutics and drug delivery systems. Pure graphene and graphene oxide nanoparticles, as well as graphene quantum dots and graphene nanofibers, were all able to trigger autophagy in cancer cells through both transcriptional and post-transcriptional mechanisms involving oxidative/endoplasmic reticulum stress, AMP-activated protein kinase, mechanistic target of rapamycin, mitogen-activated protein kinase, and Toll-like receptor signaling. This was often coupled with lysosomal dysfunction and subsequent blockade of autophagic flux, which additionally increased the accumulation of autophagy mediators that participated in apoptotic, necrotic, or necroptotic death of cancer cells and influenced the immune response against the tumor. In this review, we analyze molecular mechanisms and structure–activity relationships of GNM-mediated autophagy modulation, its consequences for cancer cell survival/death and anti-tumor immune response, and the possible implications for the use of GNM in cancer therapy.
Collapse
Affiliation(s)
- Biljana Ristic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (B.R.); (I.D.)
| | - Ljubica Harhaji-Trajkovic
- Department of Neurophysiology, Institute for Biological Research “Sinisa Stankovic”, National Institute of Republic of Serbia, University of Belgrade, 11060 Belgrade, Serbia;
| | - Mihajlo Bosnjak
- Institute of Histology and Embryology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Ivana Dakic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (B.R.); (I.D.)
| | - Srdjan Mijatovic
- Clinic for Emergency Surgery, Clinical Centre of Serbia, 11000 Belgrade, Serbia;
| | - Vladimir Trajkovic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (B.R.); (I.D.)
- Correspondence:
| |
Collapse
|
68
|
Lan HY, An P, Liu QP, Chen YY, Yu YY, Luan X, Tang JY, Zhang H. Aidi injection induces apoptosis of hepatocellular carcinoma cells through the mitochondrial pathway. JOURNAL OF ETHNOPHARMACOLOGY 2021; 274:114073. [PMID: 33794335 DOI: 10.1016/j.jep.2021.114073] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/04/2021] [Accepted: 03/23/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The incidence and mortality rates of hepatocellular carcinoma are very high all over the world, which seriously threatens human life and health. Aidi injection as a Chinese medicine preparation has a positive curative effect on hepatocellular carcinoma, but its mechanism remains unclear. AIM OF THE STUDY The purpose of this study is to evaluate the anti-hepatocellular carcinoma effects of Aidi injection and explore its mechanism of action vitro and vivo. MATERIALS AND METHODS The main components of Aidi injection were determined by LC-MS/MS. The effects of Aidi injection on the viability of HepG2 and PLC/PRF/5 cells were detected via CCK-8 analysis and Calcein AM/PI staining. DAPI staining and flow cytometry were applied to analyze the apoptosis-induced effects of Aidi injection on hepatocellular carcinoma cells (HCCs). The growth inhibition of Aidi injection on hepatocellular carcinoma was observed in nude mice bearing PLC/PRF/5 cells. The related signal transduction and apoptosis pathways were investigated through assays for JC-1 mitochondrial membrane potential (MMP), RNA-seq, KEGG, PPI and WB. RESULTS There were 12 main chemical components contained in Aidi injection, viz. cantharidin, syringin, calycosin-7-o-β-Dglucoside, isozinpidine, ginsenosides Rd, Rc, Rb1, Re, and Rg1, astragalosides II and IV, and eleutheroside E. Aidi injection significantly inhibited the proliferation of HepG2 and PLC/PLF/5 cells with IC50 of 20.66 mg/ml and 27.5 mg/ml at 48h, respectively, increased the proportion of dead cells, induced cell apoptosis, suppressed the tumor growth of nude mice bearing PLC/PLF/5 cells, reduced MMP, activated PI3K/Akt and MAPK signal transduction pathways, down-regulated the expression of p-PI3K and Bcl-xL, and up-regulated the expression of p-JNK, p-p38 and Bim. CONCLUSION Aidi injection inhibits the growth of liver cancer probably through regulating PI3K/Akt and MAPK signal transduction pathways, inducing MMP collapse to activate the mitochondrial apoptosis pathway, and then eliciting apoptosis of HCCs.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Phytogenic/chemistry
- Antineoplastic Agents, Phytogenic/pharmacology
- Antineoplastic Agents, Phytogenic/therapeutic use
- Apoptosis/drug effects
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Cell Line, Tumor
- Cell Survival/drug effects
- Drugs, Chinese Herbal/chemistry
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/therapeutic use
- Gene Expression Profiling
- Humans
- Injections
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Male
- Membrane Potential, Mitochondrial/drug effects
- Mice, Inbred BALB C
- Mice, Nude
- Mitochondria/drug effects
- Mitochondria/physiology
- Mitogen-Activated Protein Kinases/metabolism
- Phosphatidylinositol 3-Kinases/metabolism
- Phytochemicals/analysis
- Phytochemicals/pharmacology
- Phytochemicals/therapeutic use
- Protein Interaction Maps
- Proto-Oncogene Proteins c-akt/metabolism
- Signal Transduction/drug effects
- Mice
Collapse
Affiliation(s)
- Hai-Yue Lan
- Institute of Interdisciplinary Integrative Medicine Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Pei An
- Institute of Interdisciplinary Integrative Medicine Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Qiu-Ping Liu
- Institute of Interdisciplinary Integrative Medicine Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yu-Ying Chen
- Institute of Interdisciplinary Integrative Medicine Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yuan-Yuan Yu
- Institute of Interdisciplinary Integrative Medicine Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xin Luan
- Institute of Interdisciplinary Integrative Medicine Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Jian-Yuan Tang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| | - Hong Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
69
|
Karthika C, Hari B, Mano V, Radhakrishnan A, Janani SK, Akter R, Kaushik D, Rahman MH. Curcumin as a great contributor for the treatment and mitigation of colorectal cancer. Exp Gerontol 2021; 152:111438. [PMID: 34098006 DOI: 10.1016/j.exger.2021.111438] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 05/30/2021] [Accepted: 06/01/2021] [Indexed: 12/13/2022]
Abstract
Cancer is one of the life-taking diseases worldwide and among cancer-related death; colorectal cancer is the third most. Though conventional methods of treatment are available, multidrug resistance and side effects are predominant. Physicians and scientists are working side by side to develop an effective medicament, which is safe and cost-effective. However, most failures are obtained when focused on the clinical perspective. This review mainly brings out the correlation between the curcumin and its use for the mitigation of colorectal cancer, the use of curcumin as a chemotherapeutic agent, chemosensitizer, and in a combination and synergistic approach. The pharmacokinetics and pharmacodynamics properties of curcumin and its formulation approach helps in giving an idea to develop new approaches for the treatment of colorectal cancer using curcumin.
Collapse
Affiliation(s)
- Chenmala Karthika
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India.
| | - Balaji Hari
- TIFAC CORE in Herbal Drugs, Department of Pharmacognosy, JSS Academy of Higher Education & Research, Ooty-643001, The Nilgiris, Tamil Nadu, India
| | - Vignesh Mano
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Arun Radhakrishnan
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - S K Janani
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Rokeya Akter
- Department of Pharmacy, Jagannath University, Sadarghat, Dhaka 1100, Bangladesh
| | - Deepak Kaushik
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India
| | - Md Habibur Rahman
- Department of Pharmacy, Southeast University, Banani, Dhaka 1213, Bangladesh; Department of Global Medical Science, Yonsei University Wonju College of Medicine, Yonsei University,Wonju 26426, Gangwon-do, Korea..
| |
Collapse
|
70
|
Jia C, Zhuge Y, Zhang S, Ni C, Wang L, Wu R, Niu C, Wen Z, Rong X, Qiu H, Chu M. IL-37b alleviates endothelial cell apoptosis and inflammation in Kawasaki disease through IL-1R8 pathway. Cell Death Dis 2021; 12:575. [PMID: 34083516 PMCID: PMC8174541 DOI: 10.1038/s41419-021-03852-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/11/2022]
Abstract
Kawasaki disease (KD) is an acute vasculitis of pediatric populations that may develop coronary artery aneurysms if untreated. It has been regarded as the principal cause of acquired heart disease in children of the developed countries. Interleukin (IL)-37, as one of the IL-1 family members, is a natural suppressor of inflammation that is caused by activation of innate and adaptive immunity. However, detailed roles of IL-37 in KD are largely unclear. Sera from patients with KD displayed that IL-37 level was significantly decreased compared with healthy controls (HCs). QRT-PCR and western blot analyses showed that the expression level of IL-37 variant, IL-37b, was remarkably downregulated in human umbilical vein endothelial cells (HUVECs) exposed to KD sera-treated THP1 cells. Therefore, we researched the role of IL-37b in the context of KD and hypothesized that IL-37b may have a powerful protective effect in KD patients. We first observed and substantiated the protective role of IL-37b in a mouse model of KD induced by Candida albicans cell wall extracts (CAWS). In vitro experiments demonstrated that IL-37b alleviated endothelial cell apoptosis and inflammation via IL-1R8 receptor by inhibiting ERK and NFκB activation, which were also recapitulated in the KD mouse model. Together, our findings suggest that IL-37b play an effective protective role in coronary endothelial damage in KD, providing new evidence that IL-37b is a potential candidate drug to treat KD.
Collapse
Affiliation(s)
- Chang Jia
- Pediatric Research Institute, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 325027, Wenzhou, China
| | - Yingzhi Zhuge
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 325027, Wenzhou, China
| | - Shuchi Zhang
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 325027, Wenzhou, China
| | - Chao Ni
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 325027, Wenzhou, China
| | - Linlin Wang
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 325027, Wenzhou, China
| | - Rongzhou Wu
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 325027, Wenzhou, China
| | - Chao Niu
- Pediatric Research Institute, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 325027, Wenzhou, China
| | - Zhengwang Wen
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 325027, Wenzhou, China
| | - Xing Rong
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 325027, Wenzhou, China
| | - Huixian Qiu
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 325027, Wenzhou, China.
| | - Maoping Chu
- Pediatric Research Institute, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 325027, Wenzhou, China. .,Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 325027, Wenzhou, China.
| |
Collapse
|
71
|
Zhao X, Zhang N, Huang Y, Dou X, Peng X, Wang W, Zhang Z, Wang R, Qiu Y, Jin M, Kong D. Lansoprazole Alone or in Combination With Gefitinib Shows Antitumor Activity Against Non-small Cell Lung Cancer A549 Cells in vitro and in vivo. Front Cell Dev Biol 2021; 9:655559. [PMID: 33959611 PMCID: PMC8093516 DOI: 10.3389/fcell.2021.655559] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/29/2021] [Indexed: 12/21/2022] Open
Abstract
Lansoprazole (Lpz) is an FDA-approved proton pump inhibitor (PPI) drug for the therapy of acid-related diseases. Aiming to explore the new application of old drugs, we recently investigated the antitumor effect of Lpz. We demonstrated that the PPI Lpz played a tumor suppressive role in non-small cell lung cancer (NSCLC) A549 cells. Mechanistically, Lpz induced apoptosis and G0/G1 cell cycle arrest by inhibiting the activation of signal transducer and activator of transcription (Stat) 3 and the phosphoinositide 3-kinase (PI3K)/Akt and Raf/ERK pathways. In addition, Lpz inhibited autophagy by blocking the fusion of autophagosomes with lysosomes. Furthermore, Lpz in combination with gefitinib (Gef) showed a synergistic antitumor effect on A549 cells, with enhanced G0/G1 cell cycle arrest and apoptosis. The combination inhibited Stat3 phosphorylation, PI3K/Akt and Raf/ERK signaling, affecting cell cycle-related proteins such as p-Rb, cyclin D1 and p27, as well as apoptotic proteins such as Bax, Bcl-2, caspase-3, and poly (ADP-ribose) polymerase (PARP). In vivo, coadministration with Lpz and Gef significantly attenuated the growth of A549 nude mouse xenograft models. These findings suggest that Lpz might be applied in combination with Gef for NSCLC therapy, but further evidence is required.
Collapse
Affiliation(s)
- Xiaoxia Zhao
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Ning Zhang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Yingying Huang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Xiaojing Dou
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Xiaolin Peng
- Department of Otorhinolaryngology Head and Neck, Institute of Otorhinolaryngology, Tianjin First Central Hospital, Tianjin, China
| | - Wei Wang
- Department of Otorhinolaryngology Head and Neck, Institute of Otorhinolaryngology, Tianjin First Central Hospital, Tianjin, China
| | - Zhe Zhang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Ran Wang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Yuling Qiu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Meihua Jin
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Dexin Kong
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China.,School of Medicine, Tianjin Tianshi College, Tianyuan University, Tianjin, China
| |
Collapse
|
72
|
Cao S, Tang J, Huang Y, Li G, Li Z, Cai W, Yuan Y, Liu J, Huang X, Zhang H. The Road of Solid Tumor Survival: From Drug-Induced Endoplasmic Reticulum Stress to Drug Resistance. Front Mol Biosci 2021; 8:620514. [PMID: 33928116 PMCID: PMC8076597 DOI: 10.3389/fmolb.2021.620514] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 02/12/2021] [Indexed: 12/24/2022] Open
Abstract
Endoplasmic reticulum stress (ERS), which refers to a series of adaptive responses to the disruption of endoplasmic reticulum (ER) homeostasis, occurs when cells are treated by drugs or undergo microenvironmental changes that cause the accumulation of unfolded/misfolded proteins. ERS is one of the key responses during the drug treatment of solid tumors. Drugs induce ERS by reactive oxygen species (ROS) accumulation and Ca2+ overload. The unfolded protein response (UPR) is one of ERS. Studies have indicated that the mechanism of ERS-mediated drug resistance is primarily associated with UPR, which has three main sensors (PERK, IRE1α, and ATF6). ERS-mediated drug resistance in solid tumor cells is both intrinsic and extrinsic. Intrinsic ERS in the solid tumor cells, the signal pathway of UPR-mediated drug resistance, includes apoptosis inhibition signal pathway, protective autophagy signal pathway, ABC transporter signal pathway, Wnt/β-Catenin signal pathway, and noncoding RNA. Among them, apoptosis inhibition is one of the major causes of drug resistance. Drugs activate ERS and its downstream antiapoptotic proteins, which leads to drug resistance. Protective autophagy promotes the survival of solid tumor cells by devouring the damaged organelles and other materials and providing new energy for the cells. ERS induces protective autophagy by promoting the expression of autophagy-related genes, such as Beclin-1 and ATG5–ATG12. ABC transporters pump drugs out of the cell, which reduces the drug-induced apoptosis effect and leads to drug resistance. In addition, the Wnt/β-catenin signal pathway is also involved in the drug resistance of solid tumor cells. Furthermore, noncoding RNA regulates the ERS-mediated survival and death of solid tumor cells. Extrinsic ERS in the solid tumor cells, such as ERS in immune cells of the tumor microenvironment (TME), also plays a crucial role in drug resistance by triggering immunosuppression. In immune system cells, ERS in dendritic cells (DCs) and myeloid-derived suppressor cells (MDSCs) influences the antitumor function of normal T cells, which results in immunosuppression. Meanwhile, ERS in T cells can also cause impaired functioning and apoptosis, leading to immunosuppression. In this review, we highlight the core molecular mechanism of drug-induced ERS involved in drug resistance, thereby providing a new strategy for solid tumor treatment.
Collapse
Affiliation(s)
- Shulong Cao
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Jingyi Tang
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Yichun Huang
- Clinical Medical College, Hubei University of Science and Technology, Xianning, China
| | - Gaofeng Li
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Zhuoya Li
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Wenqi Cai
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Yuning Yuan
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Junlong Liu
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Xuqun Huang
- Edong Healthcare Group, Department of Medical Oncology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, China
| | - Haiyuan Zhang
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| |
Collapse
|
73
|
Amit C, Sathe G, Shunmugam A, Athyala PK, Ghose V, Chitipothu S, Janakiraman N, Sundara R, Elchuri SV. Graphitic Carbon Nitride Causes Widespread Global Molecular Changes in Epithelial and Fibroblast Cells. ACS OMEGA 2021; 6:9368-9380. [PMID: 33869917 PMCID: PMC8047657 DOI: 10.1021/acsomega.0c05513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/23/2021] [Indexed: 06/12/2023]
Abstract
For scaffold and imaging applications, nanomaterials such as graphene and its derivatives have been widely used. Graphitic carbon nitride (g-C3N4) is among one such derivative of graphenes, which draws strong consideration due to its physicochemical properties and photocatalytic activity. To use g-C3N4 for biological applications, such as molecular imaging or drug delivery, it must interact with the epithelium, cross the epithelial barrier, and then come in contact with the extracellular matrix of the fibroblast cells. Thus, it becomes essential to understand its molecular mechanism of action. Hence, in this study, to understand the molecular reprogramming associated with g-C3N4, global gene expression using DNA microarrays and proteomics using tandem mass tag (TMT) labeling and mass spectrometry were performed in epithelial and fibroblast cells, respectively. Our results showed that g-C3N4 can cross the epithelial barrier by regulating the adherens junction proteins. Further, using g-C3N4-PDMS scaffolds as a mimic of the extracellular matrix for fibroblast cells, the common signaling pathways were identified between the epithelium and fibroblast cells. These pathways include Wnt signaling, integrin signaling, TGF-β signaling, cadherin signaling, oxidative stress response, ubiquitin proteasome pathway, and EGF receptor signaling pathways. These altered signature pathways identified could play a prominent role in g-C3N4-mediated cellular interactions in both epithelial and fibroblast cells. Additionally, β catenin, EGFR, and MAP2K2 protein-protein interaction networks could play a prominent role in fibroblast cell proliferation. The findings could further our knowledge on g-C3N4-mediated alterations in cellular molecular signatures, enabling the potential use of these materials for biological applications such as molecular imaging and drug delivery.
Collapse
Affiliation(s)
- Chatterjee Amit
- Department
of Nanobiotechnology, Vision Research Foundation, Chennai 600006, India
| | - Gajanan Sathe
- Institute
of Bioinformatics, Bangalore 560066, Karnataka, India
- Manipal
Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - Abinaya Shunmugam
- Department
of Physics, Indian Institute of Technology,
Madras, Chennai 600036, India
| | | | - Vivek Ghose
- Institute
of Bioinformatics, Bangalore 560066, Karnataka, India
| | - Srujana Chitipothu
- Central
Research Instrumentation Facility, Core Lab, Vision Research Foundation, Chennai 600006, India
| | | | - Ramaprabhu Sundara
- Department
of Physics, Indian Institute of Technology,
Madras, Chennai 600036, India
| | - Sailaja V. Elchuri
- Department
of Nanobiotechnology, Vision Research Foundation, Chennai 600006, India
| |
Collapse
|
74
|
Li J, Wang X, Mei KC, Chang CH, Jiang J, Liu X, Liu Q, Guiney LM, Hersam MC, Liao YP, Meng H, Xia T. Lateral size of graphene oxide determines differential cellular uptake and cell death pathways in Kupffer cells, LSECs, and hepatocytes. NANO TODAY 2021; 37:101061. [PMID: 34055032 PMCID: PMC8153408 DOI: 10.1016/j.nantod.2020.101061] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
As a representative two-dimensional (2D) nanomaterial, graphene oxide (GO) has shown high potential in many applications due to its large surface area, high flexibility, and excellent dispersibility in aqueous solutions. These properties make GO an ideal candidate for bio-imaging, drug delivery, and cancer therapy. When delivered to the body, GO has been shown to accumulate in the liver, the primary accumulation site of systemic delivery or secondary spread from other uptake sites, and induce liver toxicity. However, the contribution of the GO physicochemical properties and individual liver cell types to this toxicity is unclear due to property variations and diverse cell types in the liver. Herein, we compare the effects of GOs with small (GO-S) and large (GO-L) lateral sizes in three major cell types in liver, Kupffer cells (KCs), liver sinusoidal endothelial cells (LSECs), and hepatocytes. While GOs induced cytotoxicity in KCs, they induced significantly less toxicity in LSECs and hepatocytes. For KCs, we found that GOs were phagocytosed that triggered NADPH oxidase mediated plasma membrane lipid peroxidation, which leads to PLC activation, calcium flux, mitochondrial ROS generation, and NLRP3 inflammasome activation. The subsequent caspase-1 activation induced IL-1β production and GSDMD-mediated pyroptosis. These effects were lateral size-dependent with GO-L showing stronger effects than GO-S. Amongst the liver cell types, decreased cell association and the absence of lipid peroxidation resulted in low cytotoxicity in LSECs and hepatocytes. Using additional GO samples with different lateral sizes, surface functionalities, or thickness, we further confirmed the differential cytotoxic effects in liver cells and the major role of GO lateral size in KUP5 pyroptosis by correlation studies. These findings delineated the GO effects on cellular uptake and cell death pathways in liver cells, and provide valuable information to further evaluate GO effects on the liver for biomedical applications.
Collapse
Affiliation(s)
- Jiulong Li
- Center of Environmental Implications of Nanotechnology (UC CEIN), University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Xiang Wang
- Center of Environmental Implications of Nanotechnology (UC CEIN), University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Kuo-Ching Mei
- Center of Environmental Implications of Nanotechnology (UC CEIN), University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Chong Hyun Chang
- Center of Environmental Implications of Nanotechnology (UC CEIN), University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Jinhong Jiang
- Center of Environmental Implications of Nanotechnology (UC CEIN), University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Xiangsheng Liu
- Center of Environmental Implications of Nanotechnology (UC CEIN), University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Qi Liu
- Center of Environmental Implications of Nanotechnology (UC CEIN), University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Linda M. Guiney
- Departments of Materials Science and Engineering, Chemistry, and Medicine, Northwestern University, Evanston, IL 60208, USA
| | - Mark C. Hersam
- Departments of Materials Science and Engineering, Chemistry, and Medicine, Northwestern University, Evanston, IL 60208, USA
| | - Yu-Pei Liao
- Center of Environmental Implications of Nanotechnology (UC CEIN), University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Huan Meng
- Center of Environmental Implications of Nanotechnology (UC CEIN), University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
- Division of NanoMedicine, Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Tian Xia
- Center of Environmental Implications of Nanotechnology (UC CEIN), University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
- Division of NanoMedicine, Department of Medicine, University of California, Los Angeles, CA 90095, USA
- Corresponding Author: Tian Xia, M.D./Ph.D., Department of Medicine, Division of NanoMedicine, UCLA School of Medicine, 52-175 CHS, 10833 Le Conte Ave, Los Angeles, CA 90095-1680.
| |
Collapse
|
75
|
Zhong G, Wan F, Ning Z, Wu S, Jiang X, Tang Z, Huang R, Hu L. The protective role of autophagy against arsenic trioxide-induced cytotoxicity and ROS-dependent pyroptosis in NCTC-1469 cells. J Inorg Biochem 2021; 217:111396. [PMID: 33610032 DOI: 10.1016/j.jinorgbio.2021.111396] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 02/07/2021] [Accepted: 02/07/2021] [Indexed: 12/30/2022]
Abstract
Arsenic trioxide (As2O3) is widely used in traditional Chinese medicine to treat tumors. This study investigated the effect of As(III) on pyroptosis in murine hepatocytes in vitro and how this relates to autophagy. NCTC1469-cells were treated with As(III) alone (6, 12 and 18 μM) or in combination with N-acetylcysteine (NAC,1 mM), 3-methyladenine (3-MA, 5 mM) or rapamycin (Rapa,100 nM) for 24 h. The results showed that As(III)-treatment reduced cell viability in a dose-dependent manner, but induced lactic dehydrogenase (LDH) activity. As(III)-treatment also resulted in increased intracellular reactive oxygen species (ROS) levels and decreased mitochondrial membrane potential (MMP), therefore promoting pyroptosis. Moreover, As(III)-treatment upregulated the expression of autophagy and pyroptosis-related genes (LC3-A, LC3-B, P62, Beclin-1, Atg5, Caspase-1, Gasdermin D, IL-18, IL-1β) and downregulated the expression of m-TOR, NLRP3, ASC genes. Meanwhile the accumulation of light chain 3-B/A (LC3B/LC3A), autophagy-related gene 5 (Atg-5), Bcl-2-interacting protein (Beclin-1), Caspase-1, Gasdermin D, interleukin-1β (IL-1β), IL-18 and poptosis-associated speck-like protein (ASC) proteins were upregulated while nucleotide binding and oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) was downregulated in all As(III)-treatment groups. Furthermore, the inhibition of autophagy by 3-MA aggravated AsIII-induced pyroptosis and cytotoxicity. However, NAC or Rapa markedly alleviated the abovementioned phenomenon under As(III) stress. In addition, we speculate that the protective mechanism of NAC on As(III)-induced pyroptosis in hepatocytes mainly include the elimination of ROS because of the chelation of As(III) in the culture medium. In conclusion, these results provide new insight into the mechanisms underlying AsIII-induced cytotoxicity and pyroptosis in hepatocytes in vitro.
Collapse
Affiliation(s)
- Gaolong Zhong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Fang Wan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Zhijun Ning
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Shaofeng Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Xuanxuan Jiang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zhaoxin Tang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Riming Huang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| | - Lianmei Hu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
76
|
Xiong J, Qiang H, Li T, Zhao J, Wang Z, Li F, Xu J. Human adipose-derived stem cells promote seawater-immersed wound healing via proangiogenic effects. Aging (Albany NY) 2021; 13:17118-17136. [PMID: 33819183 PMCID: PMC8312430 DOI: 10.18632/aging.202773] [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: 12/14/2020] [Accepted: 02/18/2021] [Indexed: 11/25/2022]
Abstract
Seawater immersion can increase the damage to skin wounds and produce chronic wounds, and the application of human adipose-derived stem cells can significantly promote healing. However, the mechanism underlying angiogenesis is currently unclear. In this study, we investigated the vascularization effect of human adipose-derived stem cells on the repair of seawater-treated skin wounds and explored the underlying mechanisms using bioinformatics. The results showed that human adipose-derived stem cells differentiated into vascular endothelial cells and promoted seawater-immersed wound vascularization by promoting vascular endothelial cell proliferation and migration. The differentially expressed genes between human adipose-derived stem cells and fibroblasts were identified and analyzed (including via gene ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment, protein–protein interaction network, and correlation analyses). The genes may promote wound healing by regulating the mechanisms of extracellular matrix remodeling, programmed cell death, inflammation, and vascularization. In conclusion, this study provides novel insights into the use of human adipose-derived stem cells in the regeneration of seawater-immersed skin wounds and chronic wounds.
Collapse
Affiliation(s)
- Jiachao Xiong
- Department of Plastic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Hong Qiang
- Department of Nursing, Shanghai Yangpu Shidong Hospital, Shanghai 200438, China
| | - Ting Li
- Department of Nursing, Shanghai Yangpu Shidong Hospital, Shanghai 200438, China
| | - Jiayi Zhao
- Department of General Practice, Changhai Hospital, Naval Military Medical University, Shanghai 200433, China
| | - Ziyu Wang
- Department of General Practice, Changhai Hospital, Naval Military Medical University, Shanghai 200433, China
| | - Fei Li
- Department of Neurology, Shanghai Yangpu Shidong Hospital, Shanghai 200438, China
| | - Jianwen Xu
- Department of Wound Stoma Clinic, Shanghai Yangpu Shidong Hospital, Shanghai 200438, China
| |
Collapse
|
77
|
Green synthesized selenium nanoparticles for ovarian cancer cell apoptosis. RESEARCH ON CHEMICAL INTERMEDIATES 2021. [DOI: 10.1007/s11164-021-04424-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
78
|
Qianru C, Xueyuan H, Bing Z, Qing Z, Kaixin Z, Shu L. Regulation of H 2S-induced necroptosis and inflammation in broiler bursa of Fabricius by the miR-15b-5p/TGFBR3 axis and the involvement of oxidative stress in this process. JOURNAL OF HAZARDOUS MATERIALS 2021; 406:124682. [PMID: 33307448 DOI: 10.1016/j.jhazmat.2020.124682] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/18/2020] [Accepted: 11/23/2020] [Indexed: 06/12/2023]
Abstract
Hydrogen sulfide (H2S) is an air pollutant, having toxic effects on immune system. Necroptosis has been discussed as a new form of cell death and plays an important role in inflammation. To investigate the mechanism of H2S-induced immune injury, and the role of microRNAs (miRNAs) in this process, based on the results of high-throughput sequencing, we selected the most significantly changed miR-15b-5p for subsequent experiments. We further predicted and determined the targeting relationship between miR-15b-5p and TGFBR3 in HD11 through miRDB, Targetscan and dual-luciferase, and found that miR-15b-5p is highly expressed in H2S-induced necroptosis and inflammation. To understand whether miR-15b-5p/TGFBR3 axis could involve in the process of necroptosis and inflammation, we further revealed that the high expression of miR-15b-5p and the knockdown of TGFBR3 can induce necroptosis. Nec-1 treatment enhanced the survival rate of cells. Notably, H2S exposure induces oxidative stress and activates the TGF-β pathway, which are collectively regulated by the miR-15b-5p/TGFBR3 axis. Our present study provides a new perspective for necroptosis regulated by the miR-15b-5p/TGFBR3 axis and reveals a new form of inflammation regulation in immune diseases.
Collapse
Affiliation(s)
- Chi Qianru
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Hu Xueyuan
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, China
| | - Zhao Bing
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Zhang Qing
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Zhang Kaixin
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Li Shu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
79
|
Guvenc D, Inal S, Kuruca N, Gokmen S, Guvenc T. Synthetic pyrethroids common metabolite 3-phenoxybenzoic acid induces caspase-3 and Bcl-2 mediated apoptosis in human hepatocyte cells. Drug Chem Toxicol 2021; 45:1971-1977. [PMID: 33706615 DOI: 10.1080/01480545.2021.1894720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Synthetic pyrethroids are a group of insecticides frequently used in public health and agriculture, and 3-PBA is a common metabolite of them. Although the liver is the primary organ responsible for metabolizing many compounds including pesticides, to the authors' knowledge there have been no studies on the direct hepatotoxic effects of 3-PBA. Therefore, this study aimed to investigate the possible hepatotoxic effects of 3-PBA on a Human Hepatoma Cell Line (HepG2) and the underlying apoptotic mechanisms. Firstly, an LC50 of 1041.242 µM was calculated for 3-PBA by using the WST-1 test with concentrations ranging between 1 µM and 10 mM. Following that, the HepG2 cells in the experimental group were exposed to 3 different concentrations of 3-PBA (1/5 LC50, 1/10 LC50 and 1/20 LC50) for 24 hours. The apoptotic mechanism was evaluated by using flow cytometry, and immunofluorescence assays for Caspase 3 and Bcl-2. In the flow cytometry assay, the total number of apoptotic cells increased in a dose dependent manner (p < 0.05). In the immunofluorescence assay, the Caspase 3 protein showed strong immunoreactivity in the experimental groups, while the reaction to the Bcl-2 protein was minimal. These results demonstrated that 3-PBA has a significant hepatotoxic effect on HepG2 cells and induces apoptosis via the regulation of Caspase-3 and Bcl-2. Furthermore, our results could further the understanding of the fundamental molecular mechanisms of 3-PBA hepatotoxicity. More studies are needed to determine the effects of long-term exposure to 3-PBA and also the molecular mechanisms underlying hepatotoxicity.
Collapse
Affiliation(s)
- Dilek Guvenc
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ondokuz Mayis, Samsun, Turkey
| | - Sinem Inal
- Department of Pathology, Faculty of Veterinary Medicine, University of Ondokuz Mayis, Samsun, Turkey
| | - Nilufer Kuruca
- Department of Pathology, Faculty of Veterinary Medicine, University of Ondokuz Mayis, Samsun, Turkey
| | - Sedat Gokmen
- Department of Laborant and Veterinary Health, Suluova Vocational School, Amasya University, Amasya, Turkey
| | - Tolga Guvenc
- Department of Pathology, Faculty of Veterinary Medicine, University of Ondokuz Mayis, Samsun, Turkey
| |
Collapse
|
80
|
Bone marrow-derived mesenchymal stem cells modulate autophagy in RAW264.7 macrophages via the phosphoinositide 3-kinase/protein kinase B/heme oxygenase-1 signaling pathway under oxygen-glucose deprivation/restoration conditions. Chin Med J (Engl) 2021; 134:699-707. [PMID: 33605598 PMCID: PMC7989993 DOI: 10.1097/cm9.0000000000001133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Background Autophagy of alveolar macrophages is a crucial process in ischemia/reperfusion injury-induced acute lung injury (ALI). Bone marrow-derived mesenchymal stem cells (BM-MSCs) are multipotent cells with the potential for repairing injured sites and regulating autophagy. This study was to investigate the influence of BM-MSCs on autophagy of macrophages in the oxygen-glucose deprivation/restoration (OGD/R) microenvironment and to explore the potential mechanism. Methods We established a co-culture system of macrophages (RAW264.7) with BM-MSCs under OGD/R conditions in vitro. RAW264.7 cells were transfected with recombinant adenovirus (Ad-mCherry-GFP-LC3B) and autophagic status of RAW264.7 cells was observed under a fluorescence microscope. Autophagy-related proteins light chain 3 (LC3)-I, LC3-II, and p62 in RAW264.7 cells were detected by Western blotting. We used microarray expression analysis to identify the differently expressed genes between OGD/R treated macrophages and macrophages co-culture with BM-MSCs. We investigated the gene heme oxygenase-1 (HO-1), which is downstream of the phosphoinositide 3-kinase/protein kinase B (PI3K/Akt) signaling pathway. Results The ratio of LC3-II/LC3-I of OGD/R treated RAW264.7 cells was increased (1.27 ± 0.20 vs. 0.44 ± 0.08, t = 6.67, P < 0.05), while the expression of p62 was decreased (0.77 ± 0.04 vs. 0.95 ± 0.10, t = 2.90, P < 0.05), and PI3K (0.40 ± 0.06 vs. 0.63 ± 0.10, t = 3.42, P < 0.05) and p-Akt/Akt ratio was also decreased (0.39 ± 0.02 vs. 0.58 ± 0.03, t = 9.13, P < 0.05). BM-MSCs reduced the LC3-II/LC3-I ratio of OGD/R treated RAW264.7 cells (0.68 ± 0.14 vs. 1.27 ± 0.20, t = 4.12, P < 0.05), up-regulated p62 expression (1.10 ± 0.20 vs. 0.77 ± 0.04, t = 2.80, P < 0.05), and up-regulated PI3K (0.54 ± 0.05 vs. 0.40 ± 0.06, t = 3.11, P < 0.05) and p-Akt/Akt ratios (0.52 ± 0.05 vs. 0.39 ± 0.02, t = 9.13, P < 0.05). A whole-genome microarray assay screened the differentially expressed gene HO-1, which is downstream of the PI3K/Akt signaling pathway, and the alteration of HO-1 mRNA and protein expression was consistent with the data on PI3K/Akt pathway. Conclusions Our results suggest the existence of the PI3K/Akt/HO-1 signaling pathway in RAW264.7 cells under OGD/R circumstances in vitro, revealing the mechanism underlying BM-MSC-mediated regulation of autophagy and enriching the understanding of potential therapeutic targets for the treatment of ALI.
Collapse
|
81
|
Zhang Y, Zhang Y, Wu J, Liu J, Kang Y, Hu C, Feng X, Liu W, Luo H, Chen A, Chen L, Shao L. Effects of carbon-based nanomaterials on vascular endothelia under physiological and pathological conditions: interactions, mechanisms and potential therapeutic applications. J Control Release 2021; 330:945-962. [DOI: 10.1016/j.jconrel.2020.10.067] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 10/31/2020] [Accepted: 10/31/2020] [Indexed: 12/11/2022]
|
82
|
Iyappan P, Bala MD, Sureshkumar M, Veeraraghavan VP, Palanisamy A. D-carvone induced ROS mediated apoptotic cell death in human leukemic cell lines (Molt-4). Bioinformation 2021; 17:171-180. [PMID: 34393434 PMCID: PMC8340696 DOI: 10.6026/97320630017171] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 12/31/2020] [Accepted: 01/26/2021] [Indexed: 12/21/2022] Open
Abstract
The immature lymphoid cells with chromosomal structural and numerical abnormalities cause the acute lymphoblastic leukemia (ALL). This hematologic disorder constitutes about 25% of cancer prognosis among children and adolescents. D-Carvone, a monocyclic monoterpene obtained from the essential oils extracted from plants is reported to possess the various biological activities. The present study was aimed to investigate the anticancer potential of D-Carvone against the human leukemic Molt-4 cells. The cytotoxicity of DCarvone was analyzed by MTT assay. The level of lipid peroxidation and antioxidants were determined. The intracellular ROS, MMP and apoptosis were demonstrated by fluorescent staining techniques. The MTT assay revealed that the D-Carvone treatment suppressed the viability of Molt-4 cells and the IC50 was determined at 20 µM/ml. The D-Carvone treatment was increased the oxidative stress and reduced the level of antioxidants in the Molt-4 cell lines. The increased intracellular ROS, apoptotic cell death, and diminished MMP was noted in the D-Carvone treatment. In the Molt-4 cells, D-carvone induced the apoptosis in a time and dose dependent manner by the activation of caspases-8, -9 and -3. Thus, data provide insights for the clinical application of D-Carvone in the treatment of blood cancer Molt-4 cells. Our study suggests the therapeutic potential D-Carvone for the treatment of leukemia in future.
Collapse
Affiliation(s)
- Petchi Iyappan
- Senior Lecturer, Faculty of Medicine, Bioscience and Nursing, School of Bioscience, Mahsa University, Saujana Putra Campus, Jalan SP2, Bandar Saujana Putra, 42610, Jenjarom, Selangor, Malaysia
| | - M Devi Bala
- Research Scholar, Muthayammal College of Arts & Science (A Unit of VANETRA Group), Rasipuram, 637408, Namakkal, Tamilnadu, India
| | - M Sureshkumar
- Department of Zoology & Biotechnology, Muthayammal College of Arts & Science (A Unit of VANETRA Group), Rasipuram, 637408, Namakkal, Tamilnadu, India
| | - Vishnu Priya Veeraraghavan
- Department of Biochemistry, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai - 600 077
| | - Arulselvan Palanisamy
- Adjunct Associate Professor,Muthayammal Centre for Advanced Research (MCAR), Muthayammal College of Arts & Science (A Unit of VANETRA Group),Rasipuram, 637408, Namakkal, Tamilnadu, India
| |
Collapse
|
83
|
Huang Y, Yuan K, Tang M, Yue J, Bao L, Wu S, Zhang Y, Li Y, Wang Y, Ou X, Gou J, Zhao Q, Yuan L. Melatonin inhibiting the survival of human gastric cancer cells under ER stress involving autophagy and Ras-Raf-MAPK signalling. J Cell Mol Med 2020; 25:1480-1492. [PMID: 33369155 PMCID: PMC7875909 DOI: 10.1111/jcmm.16237] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/24/2020] [Accepted: 12/14/2020] [Indexed: 12/18/2022] Open
Abstract
Melatonin exhibits antitumour activities in the treatment of many human cancers. In the present study, we aimed to improve the therapeutic potential of melatonin in gastric cancer. Our results confirmed that melatonin dose‐dependently suppressed the proliferation and necrosis, and increased G0/G1 phase arrest, apoptosis, autophagy and endoplasmic reticulum (ER) stress. The Ras‐Raf‐MAPK signalling pathway was activated in cells after melatonin treatment. RNA‐seq was performed and GSEA analysis further confirmed that many down‐regulated genes in melatonin‐treated cells were associated with proliferation. However, GSEA analysis also indicated that many pathways related to metastasis were increased after melatonin treatment. Subsequently, combinatorial treatment was conducted to further investigate the therapeutic outcomes of melatonin. A combination of melatonin and thapsigargin increased the apoptotic rate and G0/G1 cell cycle arrest when compared to treatment with melatonin alone. Melatonin in combination with thapsigargin triggered the increased expression of Bip, LC3‐II, phospho‐Erk1/2 and phospho‐p38 MAPK. In addition, STF‐083010, an IRE1a inhibitor, further exacerbated the decrease in survival rate induced by combinatorial treatment with melatonin and thapsigargin. Collectively, melatonin was effective in gastric cancer treatment by modifying ER stress.
Collapse
Affiliation(s)
- Yongye Huang
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Kexun Yuan
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Meifang Tang
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Jiaming Yue
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Lijun Bao
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Shuang Wu
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Yanxin Zhang
- National Academy of Innovation Strategy, Beijing, China
| | - Yin Li
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Yihang Wang
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Xu Ou
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Jiaxin Gou
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Qi Zhao
- School of Computer Science and Software Engineering, University of Science and Technology Liaoning, Anshan, China
| | - Lin Yuan
- Institute of Health Science, China Medical University, Shenyang, China
| |
Collapse
|
84
|
Synergistic Utilization of Necrostatin-1 and Z-VAD-FMK Efficiently Promotes the Survival of Compression-Induced Nucleus Pulposus Cells via Alleviating Mitochondrial Dysfunction. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6976317. [PMID: 33376733 PMCID: PMC7738793 DOI: 10.1155/2020/6976317] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/23/2020] [Accepted: 11/19/2020] [Indexed: 01/10/2023]
Abstract
We recently reported that necroptosis contributed to compression-induced nucleus pulposus (NP) cells death. In the current study, we investigated the regulative effect of necroptosis inhibitor Necrostatin-1 on NP cells apoptosis and autophagy. Necrostatin-1, autophagy inhibitor 3-Methyladenine and apoptosis inhibitor Z-VAD-FMK were employed, and NP cells were exposed to 1.0 MPa compression for 0, 24 and 36 h. Necroptosis-associated molecules were measured by Western blot and RT-PCR. Autophagy and apoptosis levels were evaluated by Western blot and quantified by flow cytometry after monodansylcadaverine and Annexin V-FITC/propidium iodide staining, respectively. The cell viability and cell death were also examined. Furthermore, we measured mitochondrial membrane potential (MMP), mitochondrial permeability transition pore (MPTP) and indices of oxidative stress to assess mitochondrial dysfunction. The results established that Necrostatin-1 blocked NP cells autophagy, and 3-Methyladenine had little influence on NP cells necroptosis. The Necrostatin-1+3-Methyladenine treatment exerted almost the same role as Necrostatin-1 in reducing NP cells death. Necrostatin-1 restrained NP cells apoptosis, while Z-VAD-FMK enhanced NP cells necroptosis. The Necrostatin-1+Z-VAD-FMK treatment provided more prominent role in blocking NP cells death compared with Necrostatin-1, consistent with increased MMP, reduced opening of MPTP and oxidative stress. In summary, the synergistic utilization of Necrostatin-1 and Z-VAD-FMK is a very worthwhile solution in preventing compression-mediated NP cells death, which might be largely attributed to restored mitochondrial function.
Collapse
|
85
|
Luo Y, Wang X, Cao Y. Transcriptomic analysis suggested the involvement of impaired lipid droplet biogenesis in graphene oxide-induced cytotoxicity in human umbilical vein endothelial cells. Chem Biol Interact 2020; 333:109325. [PMID: 33221320 DOI: 10.1016/j.cbi.2020.109325] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/07/2020] [Accepted: 11/13/2020] [Indexed: 12/17/2022]
Abstract
Previous studies revealed that direct contact with graphene oxide (GO) induced cytotoxic effects, but the importance of involvement of metabolic pathways, in particular lipid metabolism pathways, might be overlooked. In this study, human umbilical vein endothelial cells (HUVECs) were exposed to GO with large size (denoted as GO-L) or small size (denoted as GO-S), and transcriptomics were used to understand the mechanisms of cytotoxicity of GO at systemic levels. It was shown that GO-L more significantly induced cytotoxicity compared with GO-S. Transcriptomic analysis revealed that compared with GO-S, GO-L had larger impact on gene ontology terms related with mitochondrial function as well as Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways related with cell death and growth. But GO-S showed greater influence on KEGG pathways related with lipid metabolism. Both types of GO showed minimal impact on oxidative stress but increased de novo lipogenesis protein fatty acid synthase (FASN). However, only GO-S significantly promoted acyl-CoA synthetase 3 (ACSL3), a key enzyme responsible for esterification of free fatty acids and lipid droplet biogenesis. Not surprisingly, GO-L but not GO-S impaired lipid droplet biogenesis, and increasing lipid levels by oleic acid or α-linolenic acid reduced the cytotoxicity of GO-L to HUVECs. Combined, the results from this study suggested that impaired lipid droplet biogenesis was involved in GO-induced cytotoxicity in HUVECs, and inducing lipid droplet biogenesis could prevent the cytotoxicity of GO.
Collapse
Affiliation(s)
- Yingmei Luo
- Key Laboratory of Environment-Friendly Chemistry and Application of Ministry of Education, Laboratory of Biochemistry, College of Chemistry, Xiangtan University, Xiangtan, 411105, China
| | - Xuefeng Wang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, 510632, China
| | - Yi Cao
- Key Laboratory of Environment-Friendly Chemistry and Application of Ministry of Education, Laboratory of Biochemistry, College of Chemistry, Xiangtan University, Xiangtan, 411105, China.
| |
Collapse
|
86
|
Gao Y, Chen S, Sun J, Su S, Yang D, Xiang L, Meng X. Traditional Chinese medicine may be further explored as candidate drugs for pancreatic cancer: A review. Phytother Res 2020; 35:603-628. [PMID: 32965773 DOI: 10.1002/ptr.6847] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/19/2020] [Accepted: 07/20/2020] [Indexed: 12/15/2022]
Abstract
Pancreatic cancer is a disease with a high mortality rate. Although survival rates for different types of cancers have improved in recent years, the five-year survival rate of pancreatic cancer stands at 8%. Moreover, the current first-line therapy, gemcitabine, results in low remission rates and is associated with drug resistance problems. Alternative treatments for pancreatic cancer such as surgery, chemotherapy and radiation therapy provide marginal remission and survival rates. This calls for the search of more effective drugs or treatments. Traditional Chinese medicine contains numerous bioactive ingredients some of which show activity against pancreatic cancer. In this review, we summarize the mechanisms of five types of traditional Chinese medicine monomers. In so-doing, we provide new potential drug candidates for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Yue Gao
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shiyu Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiayi Sun
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Siyu Su
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dong Yang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Xiang
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xianli Meng
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
87
|
Patel TN, R P, Vashi Y, Bhattacharya P. Toxic impacts and industrial potential of graphene. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, TOXICOLOGY AND CARCINOGENESIS 2020; 38:269-297. [PMID: 32897810 DOI: 10.1080/26896583.2020.1812335] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Advancement in the field of nanotechnology has increased the synthesis and exploitation of graphene-like nanomaterials. Graphene is a two-dimensional planar and hexagonal array of carbon atoms. Due to its flexible nature graphene and its derivatives have several significant prospects extending from electronics to life sciences and drug delivery systems. In this review, we enlist some of the toxic effects of graphene family nanomaterials (GFNs) in various aspects of biosystems viz., in vitro, in vivo, microbial, molecular and environmental. We also appreciate their extensive and promising applications though with some underlying challenges. This review also draws attention toward current and future prospect of global graphene market for wide-range commercialization.
Collapse
Affiliation(s)
- Trupti N Patel
- Department of Integrative Biology, Vellore Institute of Technology, Vellore, India
| | - Priyanka R
- College of Veterinary Medicine, Jeju National University, Jeju, Republic of Korea
| | - Yash Vashi
- Operations and Product Development Department, University of Southern California, Los Angeles, California, USA
| | | |
Collapse
|
88
|
Oh J, Yoon S, Ham SW. Determination of Binding Affinities Between
NF‐κB
and Importins Using
Single‐Molecule
Binding Assays. B KOREAN CHEM SOC 2020. [DOI: 10.1002/bkcs.12082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Jiwon Oh
- Department of Chemistry Chung‐Ang University Seoul 06974 Republic of Korea
| | - Seungil Yoon
- Department of Chemistry Chung‐Ang University Seoul 06974 Republic of Korea
| | - Seung Wook Ham
- Department of Chemistry Chung‐Ang University Seoul 06974 Republic of Korea
| |
Collapse
|
89
|
Wu Y, Ma J, Sun Y, Tang M, Kong L. Effect and mechanism of PI3K/AKT/mTOR signaling pathway in the apoptosis of GC-1 cells induced by nickel nanoparticles. CHEMOSPHERE 2020; 255:126913. [PMID: 32402875 DOI: 10.1016/j.chemosphere.2020.126913] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/21/2020] [Accepted: 04/26/2020] [Indexed: 05/28/2023]
Abstract
Nickel nanoparticles (Ni NPs) have a wide range of application prospects, but there is still a lack of their safety evaluation for the reproductive system. Nowadays, male reproductive health has been widely concerned because of the increasing incidence of male infertility. Studies have shown that Ni NPs can cause male reproductive toxicity. The purpose of this study was to investigate the toxicity of Ni NPs on GC-1 cells, a mouse spermatogonia cell line, and to explore the possible mechanism underlying the induction of apoptosis via PI3K/AKT/mTOR signaling pathway. The cell ultrastructure was firstly observed under a transmission electron microscope. Then, cell proliferation, cycle and apoptosis were detected by CCK-8 and flow cytometry, respectively. Furthermore, the expression levels of related proteins and genes were determined by Western blot and Reverse transcription-polymerase chain reaction, respectively. The results showed that Ni NPs could not only cause changes in cell ultrastructure, decreased survival rate and arrested G1 phase cell cycle, but also activated apoptosis pathway by inhibiting the PI3K/AKT/mTOR signaling pathway. The results of this study provide novel insights to explore the mechanisms of reproductive toxicity of Ni NPs and are of great significance to develop safety evaluation criteria for Ni NPs.
Collapse
Affiliation(s)
- Yongya Wu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Jun Ma
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Yufei Sun
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Meng Tang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Lu Kong
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
90
|
Bu H, Liu D, Zhang G, Chen L, Song Z. AMPK/mTOR/ULK1 Axis-Mediated Pathway Participates in Apoptosis and Autophagy Induction by Oridonin in Colon Cancer DLD-1 Cells. Onco Targets Ther 2020; 13:8533-8545. [PMID: 32904616 PMCID: PMC7457577 DOI: 10.2147/ott.s262022] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 08/05/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Oridonin has been demonstrated to exert strong antitumor activities in various types of human cancers. Our previous study established that oridonin induced the apoptosis of and exerted an inhibitory effect on colon cancer cells in vitro and in vivo. However, the mechanisms behind the antitumor effects of oridonin on colorectal cancer are not clearly known. This study explored whether autophagy was involved in antitumorigenesis effects caused by the usage of oridonin in colon cancer and examined whether the AMPK/mTOR/ULK1 signaling pathway was involved in this process. METHODS Cell viability was determined using CCK-8 assay. The distribution of cell apoptosis was evaluated using flow cytometry. RT-PCR and Western blotting analysis were conducted to identify the key target genes and proteins involved in the AMPK/mTOR cascade. AMPK siRNA was used to disturb AMPK expression. A DLD-1 cell orthotopic transplantation tumor model was established to explore the anti-cancer effects in vivo. RESULTS Oridonin exhibited a suppressive effect on DLD-1 cells in a concentration- and time-dependent manner. Additionally, in a dose-dependent manner, oridonin induced cell apoptosis via inducing the protein expression levels of cleaved caspase-3, cleaved PARP and stimulated autophagy by increasing protein expression levels of Becin1, LC3-II, decreasing protein expression levels of LC3-I, p62, which were respectively attenuated and elevated by autophagy inhibitor 3-MA. Furthermore, oridonin upregulated the expression level of p-AMPK and downregulated the expression levels of p-mTOR, p-ULK1 in the DLD-1 cells in a dose-dependent manner. Moreover, knockdown of AMPK by a specific siRNA reversed the expression levels of proteins involved in the AMPK/mTOR pathway, autophagy and apoptosis. In addition, outcomes from the in vivo experiments also showed that oridonin treatment significantly repressed tumorigenic growth of DLD-1 cells without any side effects, which was accompanied by the upregulation of p-AMPK, LC3-II, active caspase-3 protein expression levels and the downregulation of p-mTOR and p-ULK1 protein expression levels. CONCLUSION This study demonstrated that oridonin induced apoptosis and autophagy of colon cancer DLD-1 cells via regulating the AMPK/mTOR/ULK1 pathway, which indicated that oridonin may be used as a novel therapeutic intervention for patients with colorectal cancer.
Collapse
Affiliation(s)
- Heqi Bu
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou310016, People’s Republic of China
- Department of Coloproctology Surgery, Tongde Hospital of Zhejiang Province, Hangzhou310012, People’s Republic of China
| | - Dianlei Liu
- Department of Surgery, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou310006, People’s Republic of China
| | - Guolin Zhang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou310016, People’s Republic of China
| | - Li Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou310016, People’s Republic of China
| | - Zhangfa Song
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou310016, People’s Republic of China
| |
Collapse
|
91
|
Jastrzębska AM, Scheibe B, Szuplewska A, Rozmysłowska-Wojciechowska A, Chudy M, Aparicio C, Scheibe M, Janica I, Ciesielski A, Otyepka M, Barsoum MW. On the rapid in situ oxidation of two-dimensional V 2CT z MXene in culture cell media and their cytotoxicity. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 119:111431. [PMID: 33321581 DOI: 10.1016/j.msec.2020.111431] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/24/2020] [Accepted: 08/20/2020] [Indexed: 01/11/2023]
Abstract
The plethora of emerging two-dimensional (2D) materials exhibit wide potential application in novel technologies and advanced devices. However, their stability in environmental conditions could be an issue, affecting their application possibilities and posing health risks. Moreover, their decomposed leftovers can also induce a negative influence on human health. In particular, transition metal carbides commonly referred to as MXenes are susceptible to environmental oxidation being decomposed toward transition metal oxides and carbide-derived carbon. In this study we focused on the oxidation-state-related in vitro cytotoxicity of delaminated V2CTz onto immortalized keratinocytes (HaCaT) and malignant melanoma (A375) human cell lines. Due to the fact, that the V2CTx MXenes are least stable from all known obtained MXenes up to date, the vanadium ones were a practical choice to visualize the oxidation-cytotoxic correlation keeping the standards of 24-48 h of cell culturing. We found that the oxidation of V2CTz highly increases their cytotoxicity toward human cells, which is also time and dose dependent. The identified mode of action relates to the cell cycle as well as cellular membrane disintegration through direct physicochemical interactions.
Collapse
Affiliation(s)
- A M Jastrzębska
- Warsaw University of Technology, Faculty of Materials Science and Engineering, 02-507 Warsaw, Wołoska 141, Poland.
| | - B Scheibe
- Palacký University, Regional Centre of Advanced Technologies and Materials, 78371 Olomouc, Šlechtitelů 27, Czech Republic; Adam Mickiewicz University, NanoBioMedical Centre, 61-614 Poznań, Wszechnicy Piastowskiej 3, Poland.
| | - A Szuplewska
- Warsaw University of Technology, Faculty of Chemistry, 00-664 Warsaw, Noakowskiego 3, Poland.
| | - A Rozmysłowska-Wojciechowska
- Warsaw University of Technology, Faculty of Materials Science and Engineering, 02-507 Warsaw, Wołoska 141, Poland
| | - M Chudy
- Warsaw University of Technology, Faculty of Chemistry, 00-664 Warsaw, Noakowskiego 3, Poland.
| | - C Aparicio
- Palacký University, Regional Centre of Advanced Technologies and Materials, 78371 Olomouc, Šlechtitelů 27, Czech Republic.
| | - M Scheibe
- Palacký University, Regional Centre of Advanced Technologies and Materials, 78371 Olomouc, Šlechtitelů 27, Czech Republic.
| | - I Janica
- Adam Mickiewicz University, Faculty of Chemistry, 61-614 Poznań, Uniwersytetu Poznańskiego 8, Poland; Centre for Advanced Technologies, Adam Mickiewicz University, 61-614 Poznań, Uniwersytetu Poznańskiego 10, Poland.
| | - A Ciesielski
- Centre for Advanced Technologies, Adam Mickiewicz University, 61-614 Poznań, Uniwersytetu Poznańskiego 10, Poland; Université de Strasbourg, CNRS, ISIS, 67000 Strasbourg, 8 allée Gaspard Monge, France.
| | - M Otyepka
- Palacký University, Regional Centre of Advanced Technologies and Materials, 78371 Olomouc, Šlechtitelů 27, Czech Republic.
| | - M W Barsoum
- Drexel University, Department of Materials Science and Engineering, Philadelphia, Chestnut Street 3141, PA 19104, USA.
| |
Collapse
|
92
|
Combined Extracts of Epimedii Folium and Ligustri Lucidi Fructus with Budesonide Attenuate Airway Remodeling in the Asthmatic Rats by Regulating Apoptosis and Autophagy. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:2319409. [PMID: 32831860 PMCID: PMC7426755 DOI: 10.1155/2020/2319409] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 12/16/2022]
Abstract
This study aimed to investigate the effects of the coadministration of budesonide (Bud) and the extracts of Epimedii Folium and Ligustri Lucidi Fructus (EEL) on regulating apoptosis and autophagy in asthmatic rats. Forty Sprague-Dawley rats were divided randomly into five groups (8 rats in each group): normal control (control), asthma model (asthma), Bud (1 mg Bud suspension in 50 ml sterile physiological saline for 30 min), EEL (100 mg/kg EEL), and group of coadministration of Bud and EEL (Bud&EEL, 100 mg/kg EEL plus Bud by nebulized inhalation for 30 min). Rats were sensitized and challenged with ovalbumin for 7 weeks and treated with corresponding drug for 4 weeks. We anesthetized all rats with 25% ethyl carbamate (4 ml/kg) and took lung tissues and BALF after final ovalbumin challenge to observe the lung histopathology and morphometry; apoptosis in BALF and lung tissue; protein expressions of Ki-67, α-SMA, cleaved Caspase-3, p-mTOR, and LC3; and protein and mRNA expressions of Bax, Bcl-2, Caspase-3, P53, mTOR, and Beclin-1. Results showed that Bud&EEL could alleviate airway remodeling, inhibit cell proliferation and autophagy in lung tissue, and promote apoptosis in BALF and lung tissue in ovalbumin-induced asthma rats through downregulating the protein expressions of α-SMA and Ki-67, the protein ratio of LC3-II/LC3-I and Bcl-2/Bax, and the protein and mRNA expressions of Bcl-2 and Beclin-1, while upregulating the protein expressions of cleaved Caspase-3 and p-mTOR, and the protein and mRNA expressions of Bax, Caspase-3, P53, and mTOR. Bud&EEL had better effects than single-use Bud on improving airway remodeling, promoting apoptosis, and regulating the expressions of autophagy- and apoptosis-related proteins. This study suggested that the effects of coadministration of EEL and Bud on regulating apoptosis and autophagy were better than those of single-use Bud treatment, and that might be the mechanism of attenuating airway remodeling, providing an alternative therapy for asthma.
Collapse
|
93
|
Wu T, Liang X, Liu X, Li Y, Wang Y, Kong L, Tang M. Induction of ferroptosis in response to graphene quantum dots through mitochondrial oxidative stress in microglia. Part Fibre Toxicol 2020; 17:30. [PMID: 32652997 PMCID: PMC7353734 DOI: 10.1186/s12989-020-00363-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 07/06/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Graphene quantum dots (GQDs) provide a bright prospect in the biomedical application because they contain low-toxic compounds and promise imaging of deep tissues and tiny vascular structures. However, the biosafety of this novel QDs has not been thoroughly evaluated, especially in the central nervous system (CNS). The microarray analysis provides a hint that nitrogen-doped GQDs (N-GQDs) exposure could cause ferroptosis in microglia, which is a novel form of cell death dependent on iron overload and lipid peroxidation. RESULTS The cytosolic iron overload, glutathione (GSH) depletion, excessive reactive oxygen species (ROS) production and lipid peroxidation (LPO) were observed in microglial BV2 cells treated with N-GQDs, which indicated that N-GQDs could damage the iron metabolism and redox balance in microglia. The pre-treatments of a specific ferroptosis inhibitor Ferrostatin-1 (Fer-1) and an iron chelater Deferoxamine mesylate (DFO) not only inhibited cell death, but also alleviated iron overload, LPO and alternations in ferroptosis biomarkers in microglia, which were caused by N-GQDs. When assessing the potential mechanisms of N-GQDs causing ferroptosis in microglia, we found that the iron content, ROS generation and LPO level in mitochondria of BV2 cells all enhanced after N-GQDs exposure. When the antioxidant ability of mitochondria was increased by the pre-treatment of a mitochondria targeted ROS scavenger MitoTEMPO, the ferroptotic biological changes were effectively reversed in BV2 cells treated with N-GQDs, which indicated that the N-GQDs-induced ferroptosis in microglia could be attributed to the mitochondrial oxidative stress. Additionally, amino functionalized GQDs (A-GQDs) elicited milder redox imbalance in mitochondria and resulted in less ferroptotic effects than N-GQDs in microglia, which suggested a slight protection of amino group functionalization in GQDs causing ferroptosis. CONCLUSION N-GQDs exposure caused ferroptosis in microglia via inducing mitochondrial oxidative stress, and the ferroptotic effects induced by A-GQDs were milder than N-GQDs when the exposure method is same. This study will not only provide new insights in the GQDs-induced cell damage performed in multiple types of cell death, but also in the influence of chemical modification on the toxicity of GQDs.
Collapse
Affiliation(s)
- Tianshu Wu
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education; School of Public Health, Southeast University, Nanjing, 210009, P. R. China.
| | - Xue Liang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education; School of Public Health, Southeast University, Nanjing, 210009, P. R. China
| | - Xi Liu
- School of Medicine, Southeast University, Nanjing, 210009, P. R. China
| | - Yimeng Li
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education; School of Public Health, Southeast University, Nanjing, 210009, P. R. China
| | - Yutong Wang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education; School of Public Health, Southeast University, Nanjing, 210009, P. R. China
| | - Lu Kong
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education; School of Public Health, Southeast University, Nanjing, 210009, P. R. China
| | - Meng Tang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education; School of Public Health, Southeast University, Nanjing, 210009, P. R. China.
| |
Collapse
|
94
|
Cao Y, Kong S, Xin Y, Meng Y, Shang S, Qi Y. Lestaurtinib potentiates TRAIL-induced apoptosis in glioma via CHOP-dependent DR5 induction. J Cell Mol Med 2020; 24:7829-7840. [PMID: 32441887 PMCID: PMC7348155 DOI: 10.1111/jcmm.15415] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 04/19/2020] [Accepted: 05/03/2020] [Indexed: 12/22/2022] Open
Abstract
Lestaurtinib, also called CEP-701, is an inhibitor of tyrosine kinase, causes haematological remission in patients with AML possessing FLT3-ITD (FLT3 gene) internal tandem duplication and strongly inhibits tyrosine kinase FLT3. Treatment with lestaurtinib modulates various signalling pathways and leads to cell growth arrest and programmed cell death in several tumour types. However, the effect of lestaurtinib on glioma remains unclear. In this study, we examined lestaurtinib and TRAIL interactions in glioma cells and observed their synergistic activity on glioma cell apoptosis. While U87 and U251 cells showed resistance to TRAIL single treatment, they were sensitized to apoptosis induced by TRAIL in the presence of lestaurtinib because of increased death receptor 5 (DR5) levels through CHOP-dependent manner. We also demonstrated using a xenograft model of mouse that the tumour growth was absolutely suppressed because of the combined treatment compared to TRAIL or lestaurtinib treatment carried out singly. Our findings reveal a potential new strategy to improve antitumour activity induced by TRAIL in glioma cells using lestaurtinib through a mechanism dependent on CHOP.
Collapse
Affiliation(s)
- Yingxiao Cao
- Department of NeurosurgeryXingtai People’s HospitalXingtaiChina
| | - Shiqi Kong
- Department of NeurosurgeryXingtai People’s HospitalXingtaiChina
| | - Yuling Xin
- Department of NeurosurgeryXingtai People’s HospitalXingtaiChina
| | - Yan Meng
- Department of Operating RoomXingtai People’s HospitalXingtaiChina
| | - Shuling Shang
- Department of Operating RoomXingtai People’s HospitalXingtaiChina
| | - Yanhui Qi
- Department of Intensive Care UnitXingtai People’s HospitalXingtaiChina
| |
Collapse
|
95
|
Graphene oxide enhances β-amyloid clearance by inducing autophagy of microglia and neurons. Chem Biol Interact 2020; 325:109126. [DOI: 10.1016/j.cbi.2020.109126] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/29/2020] [Accepted: 05/03/2020] [Indexed: 12/15/2022]
|
96
|
Burnett M, Abuetabh Y, Wronski A, Shen F, Persad S, Leng R, Eisenstat D, Sergi C. Graphene Oxide Nanoparticles Induce Apoptosis in wild-type and CRISPR/Cas9-IGF/IGFBP3 knocked-out Osteosarcoma Cells. J Cancer 2020; 11:5007-5023. [PMID: 32742448 PMCID: PMC7378933 DOI: 10.7150/jca.46464] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 06/09/2020] [Indexed: 12/23/2022] Open
Abstract
Osteosarcoma affects both adolescents and adults, and some improvement in the survival rate for affected patients has been reached in the last decade. Still, non-specificity and systemic toxicity may limit traditional therapeutic approaches to some extent. The insulin growth factor 1 (IGF1) and its binding protein (IGFBP3) have been implicated in the tumorigenesis. Nanoparticles, such as graphene oxide (GO), can provide an effective treatment for cancer as they can specifically target cancer cells while reducing undesired side effects. This study aimed to evaluate the toxicity of GO on osteosarcoma in vitro using tumor cell lines with and without knocking out the IGF and IGFBP3 genes. Human osteosarcoma cell lines, U2OS and SAOS2, and the normal osteoblast cell line hFOB1.19 were used. The IGF1 and IGFBP3 genes were eliminated using CRISPR/Cas9. Tumor cells were cultured and treated with GO. Apoptosis and reactive oxygen species (ROS) were analyzed by Annexin V-FITC and ROS assays. The nuclear factor erythroid 2-related factor 2 (NRF2), which is a crucial regulator of cellular resistance to oxidants, was investigated by Western blotting. We found a significantly higher rate of apoptosis in the OS than hFOB1.19, especially in U2OS cells in which IGF1 and IGFBP3 were knocked out. ROS increase due to GO exposure was remarkably time and concentration-dependent. Based on the rate of apoptosis, ROS, Nrf-2 decrease, and cytomorphological changes, GO has a significant cytotoxic effect against OS. Targeting the IGF1 and IGFBP3 signaling pathway may strengthen GO-related cytotoxicity with the potential to increase the survival of patients affected by this tumor.
Collapse
Affiliation(s)
- Mervin Burnett
- Department of Laboratory Medicine and Pathology, Stollery Children's Hospital, University of Alberta, Edmonton, Alberta, Canada
| | - Yasser Abuetabh
- Department of Laboratory Medicine and Pathology, Heritage Medical Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | | | - Fan Shen
- Department of Laboratory Medicine and Pathology, Stollery Children's Hospital, University of Alberta, Edmonton, Alberta, Canada
| | - Sujata Persad
- Department of Paediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Roger Leng
- Department of Laboratory Medicine and Pathology, Heritage Medical Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - David Eisenstat
- Department of Paediatrics, University of Alberta, Edmonton, Alberta, Canada.,Department of Medical Genetics, University of Alberta, Edmonton, Alberta, Canada.,Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Consolato Sergi
- Department of Laboratory Medicine and Pathology, Stollery Children's Hospital, University of Alberta, Edmonton, Alberta, Canada.,Department of Paediatrics, University of Alberta, Edmonton, Alberta, Canada.,National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, P.R. China.,Department of Orthopedics, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, Hubei, P.R. China
| |
Collapse
|
97
|
Wang G, Wang Q, Liang N, Xue H, Yang T, Chen X, Qiu Z, Zeng C, Sun T, Yuan W, Liu C, Chen Z, He X. Oncogenic driver genes and tumor microenvironment determine the type of liver cancer. Cell Death Dis 2020; 11:313. [PMID: 32366840 PMCID: PMC7198508 DOI: 10.1038/s41419-020-2509-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/10/2020] [Accepted: 04/14/2020] [Indexed: 02/06/2023]
Abstract
Primary liver cancer (PLC) may be mainly classified as the following four types: hepatocellular carcinoma (HCC), intrahepatic cholangiocarcinoma (ICC), hepatoblastoma (HB), and combined hepatocellular carcinoma and intrahepatic cholangiocarcinoma (cHCC-ICC). The majority of PLC develops in the background of tumor microenvironment, such as inflammatory microenvironments caused by viral hepatitis, alcoholic or nonalcoholic steatohepatitis, carbon tetrachloride (CCl4), 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC), and necroptosis-associated hepatic cytokine microenvironment caused by necroptosis of hepatocytes. However, the impact of different types of microenvironments on the phenotypes of PLC generated by distinct oncogenes is still unclear. In addition, the cell origin of different liver cancers have not been clarified, as far as we know. Recent researches show that mature hepatocytes retain phenotypic plasticity to differentiate into cholangiocytes. More importantly, our results initially demonstrated that HCC, ICC, and cHCC-ICC could originate from mature hepatocytes rather than liver progenitor cells (LPCs), hepatic stellate cells (HSCs) and cholangiocytes in AKT-driven, AKT/NICD-driven and AKT/CAT-driven mouse PLC models respectively by using hydrodynamic transfection methodology. Therefore, liver tumors originated from mature hepatocytes embody a wide spectrum of phenotypes from HCC to CC, possibly including cHCC-ICC and HB. However, the underlying mechanism determining the cancer phenotype of liver tumors has yet to be delineated. In this review, we will provide a summary of the possible mechanisms for directing the cancer phenotype of liver tumors (i.e., ICC, HCC, and cHCC-ICC) in terms of oncogenic driver genes and tumor microenvironment. Moreover, this study initially revealed the cell origin of different types of liver cancer.
Collapse
Affiliation(s)
- Gang Wang
- Department of General Surgery, The 74th Group Army Hospital, Guangzhou, 510220, China.,Department of General Surgery, Tangdu Hospital, Air Force Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Qian Wang
- Department of General Surgery, Tangdu Hospital, Air Force Military Medical University, Xi'an, 710032, Shaanxi, China.,Department of Anorectal Surgery, First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China
| | - Ning Liang
- Department of General Surgery, The 75th Group Army Hospital, Dali, 671000, China
| | - Hongyuan Xue
- Department of General Surgery, Huashan North Hospital, Fudan University, Shanghai, 201907, China
| | - Tao Yang
- Department of Pain Treatment, Tangdu Hospital, Air Force Military Medical University, Xi'an, 710032, Shanxi, China
| | - Xuguang Chen
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, 510091, China
| | - Zhaoyan Qiu
- Department of General Surgery, Chinese PLA General Hospital, Beijing, China
| | - Chao Zeng
- Department of Cardiology, The 74th Group Army Hospital, Guangzhou, 510318, China
| | - Tao Sun
- Departmentof Neurosurgery, First Affiliated Hospital, Zhengzhou University, Zheng zhou, 450052, China
| | - Weitang Yuan
- Department of Anorectal Surgery, First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China
| | - Chaoxu Liu
- Department of General Surgery, Huashan North Hospital, Fudan University, Shanghai, 201907, China. .,Department of Anorectal Surgery, The First Affiliated Hospital of Zhejiang University, Hangzhou, 310003, China.
| | - Zhangqian Chen
- Department of Infectious Diseases, Xijing Hospital, Air Force Military Medical University, Xi'an, 710032, Shaanxi, China. .,State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, 710032, Shaanxi, China.
| | - Xianli He
- Department of General Surgery, Tangdu Hospital, Air Force Military Medical University, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
98
|
Wang SY, Ni X, Hu KQ, Meng FL, Li M, Ma XL, Meng TT, Wu HH, Ge D, Zhao J, Li Y, Su GH. Cilostazol alleviate nicotine induced cardiomyocytes hypertrophy through modulation of autophagy by CTSB/ROS/p38MAPK/JNK feedback loop. Int J Biol Sci 2020; 16:2001-2013. [PMID: 32398966 PMCID: PMC7211170 DOI: 10.7150/ijbs.43825] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 04/06/2020] [Indexed: 12/21/2022] Open
Abstract
Nicotine is proved to be an important factor for cardiac hypertrophy. Autophagy is important cell recycling system involved in the regulation of cardiac hypertrophy. Cilostazol, which is often used in the management of peripheral vascular disease. However, the effects of cilostazol on nicotine induced autophagy and cardiac hypertrophy are unclear. Here, we aim to determine the role and molecular mechanism of cilostazol in alleviating nicotine-induced cardiomyocytes hypertrophy through modulating autophagy and the underlying mechanisms. Our results clarified that nicotine stimulation caused cardiomyocytes hypertrophy and autophagy flux impairment significantly in neonatal rat ventricular myocytes (NRVMs), which were evidenced by augments of LC3-II and p62 levels, and impaired autophagosomes clearance. Interestingly, cathepsin B (CTSB) activity decreased dramatically after stimulation with nicotine in NRVMs, which was crucial for substrate degradation in the late stage of autophagy process, and cilostazol could reverse this effect dramatically. Intracellular ROS levels were increased significantly after nicotine exposure. Meanwhile, p38MAPK and JNK were activated after nicotine treatment. By using ROS scavenger N-acetyl-cysteine (NAC) could reverse the effects of nicotine by down-regulation the phosphorylation of p38MAPK and JNK pathways, and pretreatment of specific inhibitors of p38MAPK and JNK could restore the autophagy impairment and cardiomyocytes hypertrophy induced by nicotine. Moreover, CTSB activity of lysosome regained after the treatment with cilostazol. Cilostazol also inhibited the ROS accumulation and the activation of p38MAPK and JNK, which providing novel connection between lysosome CTSB and ROS/p38MAPK/JNK related oxidative stress pathway. This is the first demonstration that cilostazol could alleviate nicotine induced cardiomyocytes hypertrophy through restoration of autophagy flux by activation of CTSB and inhibiting ROS/p38/JNK pathway, exhibiting a feedback loop on regulation of autophagy and cardiomyocytes hypertrophy.
Collapse
Affiliation(s)
- Shu-Ya Wang
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Xi Ni
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China.,Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ke-Qing Hu
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China.,Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Fan-Liang Meng
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Min Li
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Xiao-Li Ma
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China.,Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ting-Ting Meng
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Hui-Hui Wu
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Di Ge
- School of Biological Science and Technology, University of Jinan, China
| | - Jing Zhao
- Development Biology, School of Life Science, Shandong University, Jinan, China
| | - Ying Li
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China.,Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Guo-Hai Su
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China.,Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
99
|
Xi M, He W, Li B, Zhou J, Xu Z, Wu H, Zhang Y, Song D, Hu L, Lu Y, Bu W, Kong Y, Chen G, Chang S, Shi J, Zhu W. Novel cyclophosphamide of natural products osalmide and pterostilbene induces cytotoxicity and cell cycle arrest in diffuse large B-cell lymphoma cells. Acta Biochim Biophys Sin (Shanghai) 2020; 52:401-410. [PMID: 32259210 DOI: 10.1093/abbs/gmaa009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/27/2019] [Accepted: 11/28/2019] [Indexed: 02/07/2023] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common category and disease entity of non-Hodgkin lymphoma. Osalmide and pterostilbene are natural products with anticancer activities via different mechanism. In this study, using a new synthetic strategy for the two natural products, we obtained the compound DCZ0801, which was previously found to have anti-multiple myeloma activity. We performed both in vitro and in vivo assays to investigate its bioactivity and explore its underlying mechanism against DLBCL cells. The results showed that DCZ0801 treatment gave rise to a dose- and time-dependent inhibition of cell viability as determined by CCK-8 assay and flow cytometry assay. Western blot analysis results showed that the expression of caspase-3, caspase-8, caspase-9 and Bax was increased, while BCL-2 and BCL-XL levels were decreased, which suggested that DCZ0801 inhibited cell proliferation and promoted intrinsic apoptosis. In addition, DCZ0801 induced G0/G1 phase arrest by downregulating the protein expression levels of CDK4, CDK6 and cyclin D1. Furthermore, DCZ0801 exerted an anti-tumor effect by down-regulating the expressions of p-PI3K and p-AKT. There also existed a trend that the expression of p-JNK and p-P38 was restrained. Intraperitoneal injection of DCZ0801 suppressed tumor development in xenograft mouse models. The preliminary metabolic study showed that DCZ0801 displayed a rapid metabolism within 30 min. These results demonstrated that DCZ0801 may be a new potential anti-DLBCL agent in DLBCL therapy.
Collapse
Affiliation(s)
- Mengyu Xi
- Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wan He
- Department of Hematology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Bo Li
- Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinfeng Zhou
- Department of Hematology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Zhijian Xu
- Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huiqun Wu
- Department of Hematology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Yong Zhang
- Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dongliang Song
- Department of Hematology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Liangning Hu
- Department of Hematology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Ye Lu
- Department of Hematology and Oncology, Soochow University Affiliated Taicang Hospital (The First Peoples Hospital of Taicang), Jiangsu 215400, China
| | - Wenxuan Bu
- Department of Hematology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Yuanyuan Kong
- Department of Hematology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Gege Chen
- Department of Hematology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Shuaikang Chang
- Department of Hematology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jumei Shi
- Department of Hematology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Weiliang Zhu
- Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
100
|
Di Cristo L, Grimaldi B, Catelani T, Vázquez E, Pompa PP, Sabella S. Repeated exposure to aerosolized graphene oxide mediates autophagy inhibition and inflammation in a three-dimensional human airway model. Mater Today Bio 2020; 6:100050. [PMID: 32322818 PMCID: PMC7171197 DOI: 10.1016/j.mtbio.2020.100050] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/08/2020] [Accepted: 03/10/2020] [Indexed: 12/20/2022] Open
Abstract
Hazard evaluation of engineered nanomaterials (ENMs) using real-world exposure scenario could provide better interpretation of toxicity end points for their use in the assessment of human safety and for their implications in many fields such as toxicology, nanomedicine, and so forth. However, most of the current studies, both in vivo and in vitro, do not reflect realistic conditions of human exposure to ENMs, due to the high doses implemented. Moreover, the use of cellular models cultured under submerged conditions limits their physiological relevance for lung exposure, where cells are primarily cultured at the air-liquid interface. Addressing such issues is even more challenging for emergent nanomaterials, such as graphene oxide (GO), for which little or no information on exposure is available. In this work, we studied the impact of repeated exposure of GO on a three-dimensional (3D) reconstruct of human bronchial tissue, using a nebulizer system focusing on short-term effects. The selected doses (reaching a maximum of ca. 20 μg/cm2 for a period of 4 weeks of exposure) were extrapolated from alveolar mass deposition values of a broader class of carbon-based nanomaterials, reflecting a full working lifetime of human exposure. Experimental results did not show strong toxic effects of GO in terms of viability and integrity of the lung tissue. However, since 2 weeks of treatment, repeated GO exposure elicited a proinflammatory response, moderate barrier impairment, and autophagosome accumulation, a process resulting from blockade of autophagy flux. Interestingly, the 3D airway model could recover such an effect by restoring autophagy flux at longer exposure (30 days). These findings indicate that prolonged exposure to GO produces a time window (during the 30 days of treatment set for this study) for which GO-mediated autophagy inhibition along with inflammation may potentially increase the susceptibility of exposed humans to pulmonary infections and/or lung diseases. This study also highlights the importance of using physiologically relevant in vitro models and doses derived from real-world exposure to obtain focused data for the assessment of human safety.
Collapse
Affiliation(s)
- L Di Cristo
- Drug Discovery and Development Department, Istituto Italiano di Tecnologia, Via Morego 30, Genova, 16136, Italy
| | - B Grimaldi
- Drug Discovery and Development Department, Istituto Italiano di Tecnologia, Via Morego 30, Genova, 16136, Italy
| | - T Catelani
- Electron Microscopy Facility, Istituto Italiano di Tecnologia, Via Morego 30, Genova, 16163, Italy
| | - E Vázquez
- Departamento de Química Orgánica, Facultad de Ciencias y Tecnologías Químicas-IRICA, Universidad de Castilla-La Mancha, Ciudad Real, 13071, Spain
| | - P P Pompa
- Nanobiointeractions & Nanodiagnostics, Istituto Italiano di Tecnologia (IIT), Via Morego 30, Genova, 16163, Italy
| | - S Sabella
- Drug Discovery and Development Department, Istituto Italiano di Tecnologia, Via Morego 30, Genova, 16136, Italy
| |
Collapse
|