1
|
Dey R, Shaw S, Yadav R, Patel BD, Bhatt HG, Natesan G, Jha AB, Chaube U. Morpholine-Substituted Tetrahydroquinoline Derivatives as Potential mTOR Inhibitors: Synthesis, Computational Insights, and Cellular Analysis. Cancers (Basel) 2025; 17:759. [PMID: 40075606 PMCID: PMC11898650 DOI: 10.3390/cancers17050759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Backgrounds: This study explores the design of substituted tetrahydroquinoline (THQ) derivatives and their synthesis as possible inhibitors of mTOR inhibitors for targeted cancer therapy. Methods: Inspired by the structural characteristics of known mTOR inhibitors, eight novel derivatives were synthesized, characterized using mass spectroscopy, 1H, and 13C NMR, and evaluated for anticancer activity. Results: Computational studies, including molecular docking and molecular dynamics (MD) simulations, highlighted the derivative's strong binding interaction and stability within the mTOR active site. Assays for in vitro cytotoxicity showed strong and specific anticancer action against cell lines of triple-negative breast cancer, lung cancer, and breast cancer while causing negligible impact on healthy cells. Conclusions: Compound 10e emerged as the most promising candidate, displaying exceptional activity against A549 cells (IC50 = 0.033 µM) and inducing apoptosis in a dose-dependent manner, surpassing standard agents, like Everolimus and 5-flurouracil. Structure-activity relationship analysis revealed that incorporating trifluoromethyl and morpholine moieties significantly enhanced selectivity and potency. MD simulations further validated these findings, confirming stable protein-ligand interactions and favorable dynamics over a 100-ns simulation period. Collectively, this study underscores the therapeutic potential of THQ derivatives, particularly compound 10e, as promising mTOR inhibitors with potential applications in lung cancer treatment.
Collapse
Affiliation(s)
- Rajdeep Dey
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad 382481, Gujarat, India; (R.D.); (S.S.); (B.D.P.); (H.G.B.); (G.N.)
| | - Suman Shaw
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad 382481, Gujarat, India; (R.D.); (S.S.); (B.D.P.); (H.G.B.); (G.N.)
| | - Ruchi Yadav
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad 382481, Gujarat, India;
| | - Bhumika D. Patel
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad 382481, Gujarat, India; (R.D.); (S.S.); (B.D.P.); (H.G.B.); (G.N.)
| | - Hardik G. Bhatt
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad 382481, Gujarat, India; (R.D.); (S.S.); (B.D.P.); (H.G.B.); (G.N.)
| | - Gopal Natesan
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad 382481, Gujarat, India; (R.D.); (S.S.); (B.D.P.); (H.G.B.); (G.N.)
| | - Abhishek B. Jha
- Department of Internal Medicine, Roy J.and Lucille A. Carver College of Medicine, University of IOWA, Iowa City, IA 52242, USA
| | - Udit Chaube
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad 382481, Gujarat, India; (R.D.); (S.S.); (B.D.P.); (H.G.B.); (G.N.)
| |
Collapse
|
2
|
Li Y, Liu H, Fang R, Jin J, Yang F, Chen J, Zhang J. Designing novel Au(III) complexes based on the structure of diazepam: Achieving a multiaction mechanism against glioma. Eur J Med Chem 2025; 283:117171. [PMID: 39705733 DOI: 10.1016/j.ejmech.2024.117171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/01/2024] [Accepted: 12/12/2024] [Indexed: 12/22/2024]
Abstract
Metal-based drugs have been used in the clinical treatment of tumors for over 30 years. However, no metal-based drugs have been clinically approved to treat glioma. Although metal complexes have excellent cytotoxicity, their most critical problem is crossing the blood-brain barrier. Therefore, to enable metal complexes to cross blood-brain barrier and target glioma therapy, herein, we propose to rationally used the basic structure of diazepam (5-chlorobenzophenone) and thiosemicarbazide to synthesize gold (Au) complexes C1, C2 and C3 with antiglioma activity. The C3 complex with two methyl groups attached to the N3 of thiosemicarbazone exhibited excellent cytotoxicity to glioma cells through its multiaction mechanism against glioma, inducing apoptosis, autophagy death, and deoxyribonucleic acid damage. In addition, the synthesized C3 complex can effectively cross the blood-brain barrier and accumulate in glioma, considerably decreasing the untoward reaction in vivo. Our findings provide a novel strategy for designing metal-based complexes for the treatment of glioma.
Collapse
Affiliation(s)
- Yanping Li
- Mental Health Education Center of College Student, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China
| | - Haoran Liu
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China
| | - Ronghao Fang
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China
| | - Jiamin Jin
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi, 541004, PR China
| | - Jian Chen
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China.
| | - Juzheng Zhang
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China.
| |
Collapse
|
3
|
Peng X, Tan X, Dai L, Xia W, Wu Z. Exploring the impact of Apelin and Reactive Oxygen Species on autophagy and cell senescence in pre-eclampsia. Free Radic Res 2025; 59:23-48. [PMID: 39714262 DOI: 10.1080/10715762.2024.2446337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 12/24/2024]
Abstract
This research investigates the interplay between Reactive Oxygen Species (ROS) and Apelin (APLN) in regulating autophagy, with implications for placental cell senescence and apoptosis in pre-eclampsia (PE). We manipulated APLN expression using sgRNA to study its effects on ROS levels and subsequent cellular responses. Our findings reveal that APLN overexpression elevates ROS production, accelerating cellular senescence and apoptosis. In contrast, silencing APLN enhances autophagy, thereby diminishing cellular aging and apoptosis. These outcomes were confirmed in vitro and in vivo experiments, establishing a causative relationship between ROS-mediated APLN modulation and altered placental cell dynamics in PE. The results suggest potential therapeutic targets within the ROS and APLN pathways to alleviate detrimental changes in the placenta, offering new strategies for the clinical management of PE. This study emphasizes the crucial role of autophagy in placental health and sets the stage for future investigations into therapeutic interventions for pregnancy-related complications.
Collapse
Affiliation(s)
- Xue Peng
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Xi Tan
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Li Dai
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Wei Xia
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Zhao Wu
- Department of Obstetrics and Gynecology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
4
|
Dorairaj DP, Kumar P, Rajasekaran H, Bhuvanesh N, Hsu SCN, Karvembu R. Copper(II) complexes containing hydrazone and bipyridine/phenanthroline ligands for anticancer application against breast cancer cells. J Inorg Biochem 2025; 262:112759. [PMID: 39426333 DOI: 10.1016/j.jinorgbio.2024.112759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/29/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024]
Abstract
In this work, mixed ligand Cu(II) complexes containing hydrazone and bipyridine ligands (CB1-CB5), or hydrazone and phenanthroline ligands (CP1-CP5) have been synthesized and characterized by spectroscopic and analytical techniques. Single crystal X-ray structure of complex CB1 revealed that two nitrogen atoms from bipyridine, one carbonyl oxygen, one azomethine nitrogen and one hydroxyl oxygen from the hydrazone ligand coordinated to Cu(II) ion, adopting a distorted square pyramidal geometry. Interaction of these complexes with calf thymus (CT) DNA and bovine serum albumin (BSA) was analyzed by absorption and emission studies. Further, the in vitro anticancer activity of the complexes was investigated exclusively against the breast cancer cells namely MCF7, T47D and MDA MB 231, and a normal breast MCF 10a cell line. The phenanthroline bearing complexes (CP1-CP5) displayed better activity than their bipyridine counterparts as seen from the IC50 values. In addition, the most active complex CP1 having an IC50 value of 5.8 ± 0.3 μM against T47D cancer cells was investigated for its mode of cell death through acridine orange/ethidium bromide(AO/EB), 4',6-diamidino-2-phenylindole (DAPI) and Annexin-V fluorescein isothiocyanate (FITC) staining assays which revealed apoptosis. Lastly, the cell cycle analysis revealed that complex CP1 induced cell death in T47D cancer cells at the G0/G1 phase.
Collapse
Affiliation(s)
| | - Prashant Kumar
- Department of Chemistry, National Institute of Technology, Tiruchirappalli 620015, India
| | - Haritha Rajasekaran
- Department of Chemistry, National Institute of Technology, Tiruchirappalli 620015, India
| | - Nattamai Bhuvanesh
- Department of Chemistry, Texas A & M University, College Station, TX 77842, USA
| | - Sodio C N Hsu
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ramasamy Karvembu
- Department of Chemistry, National Institute of Technology, Tiruchirappalli 620015, India.
| |
Collapse
|
5
|
Chen P, Ji XY, Feng JT, Wang XQ, Zhang B. The Synergistic Mechanism of Chelidonium majus Alkaloids on Melanoma Treatment via a Multi-Strategy Insight. Molecules 2024; 29:5412. [PMID: 39598801 PMCID: PMC11597347 DOI: 10.3390/molecules29225412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/08/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024] Open
Abstract
Melanoma represents a formidable challenge in dermatological oncology due to its resistance to conventional treatments. The Celandine Alkali Injection Formula (CAIF) offers benefits on clinical internal medicine treatments, within which chelidonine and tetrandrine are recognized as potential quality markers. However, their synergistic mechanisms facilitating their anti-melanoma action remain unveiled. This study embarked on an exploration of CAIF's therapeutic potential through a multifaceted research design, integrating system pharmacological predictions with empirical molecular biological evaluations. The dual application of chelidonine and tetrandrine within CAIF exhibited a pronounced inhibitory effect on the proliferation of B16F10 cells, surpassing the effectiveness of individual compound administration. Computational predictions identified the top 50 targets, involved in key signaling pathways including cell cycle regulation, and melanogenesis. RNA sequencing further elucidated that the combinatory treatment modulated a broader spectrum of differentially expressed genes, implicating crucial biological processes including cell differentiation, and tyrosinase metabolism. The combination markedly enhanced melanogenesis and apoptotic indices, arrested cell cycle progression, and fostered cellular differentiation. Notably, chelidonine additionally curtailed the migratory capacity of B16F10 cells. Our findings underscore the therapeutic potential of chelidonine and tetrandrine, key components of CAIF, in effectively combating melanoma by targeting cell proliferation, migration, differentiation, and melanogenesis.
Collapse
Affiliation(s)
- Peng Chen
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China; (P.C.); (X.-Q.W.)
- Key Laboratory of Xinjiang Phytomedicine Resources and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi 832003, China; (X.-Y.J.); (J.-T.F.)
| | - Xin-Ye Ji
- Key Laboratory of Xinjiang Phytomedicine Resources and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi 832003, China; (X.-Y.J.); (J.-T.F.)
| | - Jian-Ting Feng
- Key Laboratory of Xinjiang Phytomedicine Resources and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi 832003, China; (X.-Y.J.); (J.-T.F.)
| | - Xiao-Qin Wang
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China; (P.C.); (X.-Q.W.)
- Key Laboratory of Xinjiang Phytomedicine Resources and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi 832003, China; (X.-Y.J.); (J.-T.F.)
| | - Bo Zhang
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China; (P.C.); (X.-Q.W.)
- Key Laboratory of Xinjiang Phytomedicine Resources and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi 832003, China; (X.-Y.J.); (J.-T.F.)
| |
Collapse
|
6
|
Xu Y, Shao L, Zhou Z, Zhao L, Wan S, Sun W, Wanyan W, Yuan Y. ARG2 knockdown promotes G0/G1 cell cycle arrest and mitochondrial dysfunction in adenomyosis via regulation NF-κB and Wnt/Β-catenin signaling cascades. Int Immunopharmacol 2024; 140:112817. [PMID: 39116499 DOI: 10.1016/j.intimp.2024.112817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/28/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND Adenomyosis is a common gynecological disease, characterized by overgrowth of endometrial glands and stroma in the myometrium, however its exact pathophysiology still remains uncertain. Emerging evidence has demonstrated the elevated level of arginase 2 (ARG2) in endometriosis and adenomyosis. This study aimed to determine whether ARG2 involved in mitochondrial function and epithelial to mesenchymal transition (EMT) in adenomyosis and its potential underlying mechanisms. MATERIALS AND METHODS RNA interference was used to inhibit ARG2 gene, and then Cell Counting Kit (CCK-8) assay and flow cytometery were performed to detect the cell proliferation capacity, cell cycle, and apoptosis progression, respectively. The mouse adenomyosis model was established and RT-PCR, Western blot analysis, mitochondrial membrane potential (Δψm) detection and mPTP opening evaluation were conducted. RESULTS Silencing ARG2 effectively down-regulated its expression at the mRNA and protein levels in endometrial cells, leading to decreased enzyme activity and inhibition of cell viability. Additionally, ARG2 knockdown induced G0/G1 cell cycle arrest, promoted apoptosis, and modulated the expression of cell cycle- and apoptosis-related regulators. Notably, the interference with ARG2 induces apoptosis by mitochondrial dysfunction, ROS production, ATP depletion, decreasing the Bcl-2/Bax ratio, releasing Cytochrome c, and increasing the expression of Caspase-9/-3 and PARP. In vivo study in a mouse model of adenomyosis demonstrated also elevated levels of ARG2 and EMT markers, while siARG2 treatment reversed EMT and modulated inflammatory cytokines. Furthermore, ARG2 knockdown was found to modulate the NF-κB and Wnt/β-catenin signaling pathways in mouse adenomyosis. CONCLUSION Consequently, ARG2 silencing could induce apoptosis through a mitochondria-dependent pathway mediated by ROS, and G0/G1 cell cycle arrest via suppressing NF-κB and Wnt/β-catenin signaling pathways in Ishikawa cells. These findings collectively suggest that ARG2 plays a crucial role in the pathogenesis of adenomyosis and may serve as a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Yaping Xu
- Department of Gynaecology, Shandong Provincial Third Hospital, Shandong University, No.11 Wuyingshan Middle Road, Tianqiao District, Jinan, Shandong 250031, China; State Key Laboratory of Ultrasound in Medicine and Engineering, No.1 Medical College Road, Yuzhong District, Chongqing, China
| | - Lin Shao
- Department of Gynaecology, Shandong Provincial Third Hospital, Shandong University, No.11 Wuyingshan Middle Road, Tianqiao District, Jinan, Shandong 250031, China; State Key Laboratory of Ultrasound in Medicine and Engineering, No.1 Medical College Road, Yuzhong District, Chongqing, China
| | - Zhan Zhou
- Department of Gynaecology, Shandong Provincial Third Hospital, Shandong University, No.11 Wuyingshan Middle Road, Tianqiao District, Jinan, Shandong 250031, China
| | - Liying Zhao
- Department of Gynaecology, Shandong Provincial Third Hospital, Shandong University, No.11 Wuyingshan Middle Road, Tianqiao District, Jinan, Shandong 250031, China
| | - Shuquan Wan
- Department of Gynaecology, Shandong Provincial Third Hospital, Shandong University, No.11 Wuyingshan Middle Road, Tianqiao District, Jinan, Shandong 250031, China
| | - Wenjing Sun
- Department of Gynaecology, Shandong Provincial Third Hospital, Shandong University, No.11 Wuyingshan Middle Road, Tianqiao District, Jinan, Shandong 250031, China
| | - Wenya Wanyan
- Department of Gynaecology, Shandong Provincial Third Hospital, Shandong University, No.11 Wuyingshan Middle Road, Tianqiao District, Jinan, Shandong 250031, China
| | - Yinping Yuan
- State Key Laboratory of Ultrasound in Medicine and Engineering, No.1 Medical College Road, Yuzhong District, Chongqing, China; Department of Pathology, Shandong Cancer Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250031, China.
| |
Collapse
|
7
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Overview of pyroptosis mechanism and in-depth analysis of cardiomyocyte pyroptosis mediated by NF-κB pathway in heart failure. Biomed Pharmacother 2024; 179:117367. [PMID: 39214011 DOI: 10.1016/j.biopha.2024.117367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
The pyroptosis of cardiomyocytes has become an essential topic in heart failure research. The abnormal accumulation of these biological factors, including angiotensin II, advanced glycation end products, and various growth factors (such as connective tissue growth factor, vascular endothelial growth factor, transforming growth factor beta, among others), activates the nuclear factor-κB (NF-κB) signaling pathway in cardiovascular diseases, ultimately leading to pyroptosis of cardiomyocytes. Therefore, exploring the underlying molecular biological mechanisms is essential for developing novel drugs and therapeutic strategies. However, our current understanding of the precise regulatory mechanism of this complex signaling pathway in cardiomyocyte pyroptosis is still limited. Given this, this study reviews the milestone discoveries in the field of pyroptosis research since 1986, analyzes in detail the similarities, differences, and interactions between pyroptosis and other cell death modes (such as apoptosis, necroptosis, autophagy, and ferroptosis), and explores the deep connection between pyroptosis and heart failure. At the same time, it depicts in detail the complete pathway of the activation, transmission, and eventual cardiomyocyte pyroptosis of the NF-κB signaling pathway in the process of heart failure. In addition, the study also systematically summarizes various therapeutic approaches that can inhibit NF-κB to reduce cardiomyocyte pyroptosis, including drugs, natural compounds, small molecule inhibitors, gene editing, and other cutting-edge technologies, aiming to provide solid scientific support and new research perspectives for the prevention and treatment of heart failure.
Collapse
Affiliation(s)
- Zeyu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhihua Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shuai Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Xianliang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Jingyuan Mao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
8
|
Abiri E, Mirzaii M, Moghbeli M, Atashi A, Harati AA. Investigating the relationship between lymphocyte cells apoptosis and DNA damage and oxidative stress and therapeutic and clinical outcomes of COVID-19 elderly patients. BMC Infect Dis 2024; 24:940. [PMID: 39251946 PMCID: PMC11385507 DOI: 10.1186/s12879-024-09734-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/07/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND While COVID-19 has been controlled and deaths have decreased, the long-term consequences of COVID-19 remain a challenge we face today. This study was conducted to determine the relationship between the apoptosis of lymphocyte cells with DNA damage and oxidative stress and the therapeutic and clinical outcomes of elderly patients with COVID-19. METHODS This study was conducted from April 2020 to May 2021 (the period of severe attacks of the epidemic peak of COVID-19) and September 2022 (the post-COVID-19 period). The study groups included elderly patients with COVID-19 hospitalized in the ICU and normal wards of the hospital as well as elderly patients with influenza. A polymerase chain reaction was used to check the validity of the studied diseases. The Annexin V/Propidium Iodide method was used to evaluate the level of apoptosis. Genotoxic effects and DNA damage were assessed by the comet assay method. Total antioxidant status (TAS), total oxidant status (TOS), and myeloperoxidase activity (MPO) were measured by photometric methods. RESULTS The highest level of apoptosis in peripheral blood lymphocytes and the highest level of DNA damage were observed at both times in the intubated-ICU and non-intubated-ICU groups. In all groups, there was a significant increase in peripheral blood lymphocyte apoptosis levels and DNA damage levels compared to the healthy control group (p < 0.01). The level of apoptosis and DNA damage decreased significantly in the post-COVID-19 period (p < 0.01). In the investigation of oxidative stress biomarkers, the oxidative stress index, including TOS and MPO levels, increased in patients (p < 0.01), and the TAS level decreased (p < 0.01). CONCLUSION It shows that the apoptosis of lymphocyte cells, DNA damage, and oxidative stress can be effective in prognostic decisions and is a suitable predictor for diagnosing the condition of patients with viral infections such as COVID-19 and influenza.
Collapse
Affiliation(s)
- Elaheh Abiri
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Mehdi Mirzaii
- Department of Biology, Haftham Tir Square - Shahroud University of Medical Sciences and Health Services, Shahroud, Iran.
| | - Majid Moghbeli
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Amir Atashi
- Department of Biology, Haftham Tir Square - Shahroud University of Medical Sciences and Health Services, Shahroud, Iran
| | - Ahad Ali Harati
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| |
Collapse
|
9
|
Wu H, Wang Y, Tan P, Ran Y, Guan Y, Qian S, Feng X, Jiang Y, Peng Y, Sheng K, Xi H, Ji W, Guo X. Ferulic acid suppresses the inflammation and apoptosis in Kawasaki disease through activating the AMPK/mTOR/NF-κB pathway. Front Pharmacol 2024; 15:1420602. [PMID: 39268468 PMCID: PMC11390509 DOI: 10.3389/fphar.2024.1420602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 08/14/2024] [Indexed: 09/15/2024] Open
Abstract
Background Kawasaki disease (KD) is a self-limiting and acute systemic vasculitis of unknown etiology, mainly affecting children. Ferulic acid (FA), a natural phenolic substance, has multiple pharmacological properties, including anti-inflammatory, anti-apoptosis, and anti-fibrosis, and so on. So far, the protective effects of FA on KD have not been explored. Methods In this study, we established Candida albicans water soluble fraction (CAWS)-induced mouse coronary artery vasculitis of KD model and the tumor necrosis factor α (TNF-α)-induced human umbilical vein endothelial cells (HUVECs) injury model to investigate the anti-inflammatory and anti-apoptosis effects of FA on KD, and try to elucidate the underlying mechanism. Results Our in vivo results demonstrated that FA exerted anti-inflammatory effects on KD by inhibiting the infiltration of CD45-positive leukocytes and fibrosis around the coronary artery. Additionally, FA downregulated the levels of inflammatory and chemotactic cytokines, alleviated splenomegaly, and exhibited anti-apoptotic effects on KD by reducing TUNEL-positive cells, downregulating BAX expression, and upregulating BCL-2 expression. In addition, Our in vitro findings showed that FA could effectively inhibit TNF-α-induced HUVEC inflammation like NF-κB inhibitor QNZ by downregulating the expression of pro-inflammatory cytokines as well as attenuated TNF-α-induced HUVEC apoptosis by reducing apoptotic cell numbers and the BAX/BCL-2 ratio, which could be reversed by the AMPK inhibitor compound c (CC). The further mechanistic study demonstrated that FA could restrain vascular endothelial cell inflammation and apoptosis in KD through activating the AMPK/mTOR/NF-κB pathway. However, FA alone is hard to completely restore KD into normal condition. Conclusion In conclusion, FA has potential protective effects on KD, suggesting its promising role as an adjuvant for KD therapy in the future.
Collapse
Affiliation(s)
- Huilan Wu
- Basic Medical Research Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yijia Wang
- Basic Medical Research Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Pingping Tan
- Basic Medical Research Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuqing Ran
- Basic Medical Research Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuting Guan
- Basic Medical Research Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Songwei Qian
- Department of General Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, China
| | - Xing Feng
- Basic Medical Research Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yalan Jiang
- Basic Medical Research Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yongmiao Peng
- Basic Medical Research Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ke Sheng
- Basic Medical Research Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Haitao Xi
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weiping Ji
- Department of General Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, China
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoling Guo
- Basic Medical Research Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Scientific Research Department, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
10
|
Wang S, Shao Z, Chen G, Lin B, Li D, Chen J. Assessment of chlorine and hydrogen peroxide on airborne bacteria: Disinfection efficiency and induction of antibiotic resistance. JOURNAL OF HAZARDOUS MATERIALS 2024; 474:134697. [PMID: 38823102 DOI: 10.1016/j.jhazmat.2024.134697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/23/2024] [Accepted: 05/21/2024] [Indexed: 06/03/2024]
Abstract
Airborne pathogens severely threaten public health worldwide. Air disinfection is essential to ensure public health. However, excessive use of disinfectants may endanger environmental and ecological security due to the residual disinfectants and their by-products. This study systematically evaluated disinfection efficiency, induction of multidrug resistance, and the underlying mechanisms of disinfectants (NaClO and H2O2) on airborne bacteria. The results showed that airborne bacteria were effectively inactivated by atomized NaClO (>160 μg/L) and H2O2 (>320 μg/L) after 15 min. However, some bacteria still survived after disinfection by atomized NaClO (0-80 μg/L) and H2O2 (0-160 μg/L), and they exhibited significant increases in antibiotic resistance. The whole-genome sequencing of the resistant bacteria revealed distinct mutations that were responsible for both antibiotic resistance and virulence. This study also provided evidences and insights into possible mechanisms underlying the induction of antibiotic resistance by air disinfection, which involved intracellular reactive oxygen species formation, oxidative stress responses, alterations in bacterial membranes, activation of efflux pumps, and the thickening of biofilms. The present results also shed light on the role of air disinfection in inducing antibiotic resistance, which could be a crucial factor contributing to the global spread of antibiotic resistance through the air.
Collapse
Affiliation(s)
- Siyi Wang
- Shanghai Key Laboratory of Atmospheric Particle Pollution and Prevention (LAP3), Department of Environmental Science and Engineering, Fudan University, Shanghai 200433, China
| | - Zhiwei Shao
- Shanghai Key Laboratory of Atmospheric Particle Pollution and Prevention (LAP3), Department of Environmental Science and Engineering, Fudan University, Shanghai 200433, China
| | - Guang Chen
- Shanghai Chengtou Sewage Treatment Co., LtD., Shanghai 201203, China
| | - Bingjie Lin
- Shanghai Chengtou Sewage Treatment Co., LtD., Shanghai 201203, China
| | - Dan Li
- Shanghai Key Laboratory of Atmospheric Particle Pollution and Prevention (LAP3), Department of Environmental Science and Engineering, Fudan University, Shanghai 200433, China.
| | - Jianmin Chen
- Shanghai Key Laboratory of Atmospheric Particle Pollution and Prevention (LAP3), Department of Environmental Science and Engineering, Fudan University, Shanghai 200433, China
| |
Collapse
|
11
|
Shi H, Yang J, Lin J, Hong X, Zhou Z, Zhao J, Li Y, Li J, Wu C, Yan J, Wong NK, Gao L. A facile fluorescence-coupling approach to visualizing leonurine uptake and distribution in living cells and Caenorhabditis elegans. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155737. [PMID: 38772183 DOI: 10.1016/j.phymed.2024.155737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 04/16/2024] [Accepted: 05/13/2024] [Indexed: 05/23/2024]
Abstract
BACKGROUND Caenorhabditis elegans (C. elegans) has been recognized for being a useful model organism in small-molecule drug screens and drug efficacy investigation. However, there remain bottlenecks in evaluating such processes as drug uptake and distribution due to a lack of appropriate chemical tools. PURPOSE This study aims to prepare fluorescence-labeled leonurine as an example to monitor drug uptake and distribution of small molecule in C. elegans and living cells. METHODS FITC-conjugated leonurine (leonurine-P) was synthesized and characterized by LC/MS, NMR, UV absorption and fluorescence intensity. Leonurine-P was used to stain C. elegans and various mammalian cell lines. Different concentrations of leonurine were tested in conjunction with a competing parent molecule to determine whether leonurine-P and leonurine shared the same biological targets. Drug distribution was analyzed by imaging. Fluorometry in microplates and flow cytometry were performed for quantitative measurements of drug uptake. RESULTS The UV absorption peak of leonurine-P was 490∼495 nm and emission peak was 520 nm. Leonurine-P specifically bound to endogenous protein targets in C. elegans and mammalian cells, which was competitively blocked by leonurine. The highest enrichment levels of leonurine-P were observed around 72 h following exposure in C. elegans. Leonurine-P can be used in a variety of cells to observe drug distribution dynamics. Flow cytometry of stained cells can be facilely carried out to quantitatively detect probe signals. CONCLUSIONS The strategy of fluorescein-labeled drugs reported herein allows quantification of drug enrichment and visualization of drug distribution, thus illustrates a convenient approach to study phytodrugs in pharmacological contexts.
Collapse
Affiliation(s)
- Hao Shi
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jinrong Yang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Jiajie Lin
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Xiaobing Hong
- Department of Pharmacy, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Ziyuan Zhou
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
| | - Jiamin Zhao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yiwen Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Junjie Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Chaofeng Wu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jinwu Yan
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China.
| | - Nai-Kei Wong
- Clinical Pharmacology Section, Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong 515041, China.
| | - Lei Gao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, Guangdong 510515, China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
12
|
Chen J, Chen W, Li X, Ye Y, Huang W, Gao L, Zhang M. CBC-1 as a Cynanbungeigenin C derivative inhibits the growth of colorectal cancer through targeting Hedgehog pathway component GLI 1. Steroids 2024; 206:109421. [PMID: 38614233 DOI: 10.1016/j.steroids.2024.109421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 03/28/2024] [Accepted: 04/06/2024] [Indexed: 04/15/2024]
Abstract
Colorectal cancer (CRC) is one of the most common gastrointestinal cancers that results in death in worldwide. The Hedgehog (HH) signalling pathway regulates the initiation and progression of CRC. Inhibiting the HH pathway has been presented as a potential treatment strategy in recent years. Cynanbungeigenin C (CBC) is a new type of C21 steroid that has been previously reported for the treatment of medulloblastoma. However, its further investigation was limited by its poor water solubility. In this study, six new CBC derivatives were synthesized through the structural modification of CBC, and four of them showed better water solubility than CBC. Moreover, their antiproliferative activities on CRC were evaluated. It was found that CBC-1 presented the best inhibitory effect on three types of CRC cell lines, and this effect was superior to that of CBC. Mechanistically, CBC-1 inhibited the proliferation of CRC cells through regulation of mRNA and proteins of the HH pathway according to qRT-PCR and Western blotting analysis. Furthermore, Cellular Thermal Shift Assay results indicated that CBC-1 regulated this signalling pathway by targeting glioma‑associated oncogene (GLI 1).In addition, cell apoptosis was induced increasingly by transfection with GLI 1 siRNA or treatment with CBC-1 to downregulate GLI 1. Last, the in vivo results demonstrated that CBC-1 significantly reduced tumour size and downregulated GLI 1 in CRC. Therefore, this study suggests that CBC-1, a new GLI 1 inhibitor derived from natural products, may be developed as a potential antitumour candidate for CRC treatment.
Collapse
Affiliation(s)
- Jinwen Chen
- Department of Pharmacy, The First People's Hospital of Xiaoshan District, Hangzhou, Zhejiang 311200, China
| | - Wei Chen
- Department of Pharmacy, The First People's Hospital of Xiaoshan District, Hangzhou, Zhejiang 311200, China
| | - Xiaoyu Li
- Department of Pharmacy, The First People's Hospital of Xiaoshan District, Hangzhou, Zhejiang 311200, China.
| | - Yiping Ye
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Wenkang Huang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Lijuan Gao
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Meng Zhang
- Department of Pharmacy, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
| |
Collapse
|
13
|
Duan X, Xu M, Wang Y, Liu N, Wang X, Liu Y, Zhang W, Ma W, Ma L, Fan Y. Effect of miR-17 on Polygonum Cillinerve polysaccharide against transmissible gastroenteritis virus. Front Vet Sci 2024; 11:1360102. [PMID: 38444776 PMCID: PMC10912159 DOI: 10.3389/fvets.2024.1360102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/05/2024] [Indexed: 03/07/2024] Open
Abstract
Transmissible gastroenteritis virus (TGEV) could cause diarrhea, vomiting, dehydration and even death in piglets, miRNA played an important role in the interaction between virus and cell. The study aimed to investigate the impact of miR-17 on the polysaccharide of Polygonum Cillinerve (PCP) in combating TGEV. miR-17 was screened and transfection validation was performed by Real-time PCR. The function of miR-17 on PK15 cells infected with TGEV and treated with PCP was investigated by DCFH-DA loading probe, JC-1 staining and Hoechst fluorescence staining. Furthermore, the effect of miR-17 on PCP inhibiting TGEV replication and apoptosis signaling pathways during PCP against TGEV infection was measured through Real-time PCR and Western blot. The results showed that miR-17 mimic and inhibitor could be transferred into PK15 cells and the expression of miR-17 significantly increased and decreased respectively compared with miR-17 mimic and inhibitor (P < 0.05). A total 250 μg/mL of PCP could inhibit cells apoptosis after transfection with miR-17. PCP (250 μg/mL and 125 μg/mL) significantly inhibited the decrease in mitochondrial membrane potential induced by TGEV after transfection with miR-17 (P < 0.05). After transfection of miR-17 mimic, PCP at concentrations of 250 μg/mL and 125 μg/mL significantly promoted the mRNA expression of P53, cyt C and caspase 9 (P < 0.05). Compared with the control group, the replication of TGEV gRNA and gene N was significantly inhibited by PCP at concentrations of 250 μg/mL and 125 μg/mL after transfection of both miR-17 mimic and inhibitor (P < 0.05). PCP at 62.5 μg/mL significantly inhibited the replication of gene S following transfection with miR-17 inhibitor (P < 0.05). These results suggested that PCP could inhibit the replication of TGEV and apoptosis induced by TGEV by regulating miR-17.
Collapse
Affiliation(s)
- Xueqin Duan
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Mengxin Xu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yunying Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Nishang Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Xingchen Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yingqiu Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Weimin Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Wuren Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Lin Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yunpeng Fan
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| |
Collapse
|
14
|
Megha KB, Mohanan PV. Cellular consequences triggered by ketamine on exposure to human glioblastoma epithelial (LN-229) cells. J Biochem Mol Toxicol 2023; 37:e23484. [PMID: 37515540 DOI: 10.1002/jbt.23484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 06/09/2023] [Accepted: 07/20/2023] [Indexed: 07/31/2023]
Abstract
Ketamine is generally a noncompetitive N-methyl-D-aspartate (NMDA) receptor antagonist that interrelates with various other receptors, contributing to a wide range of actions. They are mainly approved as a general anesthetic, but a low dose of ketamine is applied for pain management, depression, and as analgesics. However, there is a significant concern regarding the long-term usage as antidepressants and as an abused drug. The study mainly aims to exhibit the possible long-term side effects of ketamine as an antidepressant and in recreational users. The study explores the in vitro cytotoxicity revealed on LN-229 cells in a dose-dependent manner. According to the cell viability assays, there is a dose-dependent response toward ketamine. Morphological and nuclear integrity was changed on exposure and assessed using Giemsa, Rhodamine phalloidin, 4',6-diamidino-2-phenylindole (DAPI), and Acridine orange staining. The apoptotic cell death marked by nuclear condensation, Lactate dehydrogenase leakage, pro-inflammatory cytokine (interleukin [IL]-β) release, and inhibition of cell migration was observed. The study highlights the importance of nonanesthetic usage of ketamine, which can lead to severe adverse side effects on long-term exposure rather than a single exposure as an anesthetic agent.
Collapse
Affiliation(s)
- Kizhakkepurakkal B Megha
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, Kerala, India
| | - Parayanthala V Mohanan
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, Kerala, India
| |
Collapse
|
15
|
Chen G, Mao D, Wang X, Chen J, Gu C, Huang S, Yang Y, Zhang F, Tan W. Aptamer-based self-assembled nanomicelle enables efficient and targeted drug delivery. J Nanobiotechnology 2023; 21:415. [PMID: 37946192 PMCID: PMC10634091 DOI: 10.1186/s12951-023-02164-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 10/16/2023] [Indexed: 11/12/2023] Open
Abstract
Nucleic acid aptamer-based nanomicelles have great potential for nanomedicine and nanotechnology applications. However, amphiphilic aptamer micelles are known to be inherently unstable upon interaction with cell membranes in the physiological environment, thus potentially compromising their specific targeting against cancer cells. This flaw is addressed in the present work which reports a superstable micellar nanodelivery system as an amphiphilic copolymer self-assembled micelle composed of nucleic acid aptamer and polyvalent hydrophobic poly(maleic anhydride-alt-1-octadecene) (C18PMH). Using Ce6 as a drug model, these C18-aptamer micelles exhibit efficient tumor-targeting and -binding ability, facilitating the entry of Ce6 into targeted cells for photodynamic therapy. In addition, they can be loaded with other hydrophobic drugs and still demonstrate favorable therapeutic effects. As such, these C18-aptamer micelles can serve as a universal platform for loading multiple drugs, providing a safer and more effective solution for treating cancer.
Collapse
Affiliation(s)
- Ganghui Chen
- Institute of Molecular Medicine (IMM), Shanghai Jiao Tong University School of Medicine, Renji Hospital, College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, People's Republic of China
| | - Dongsheng Mao
- Institute of Molecular Medicine (IMM), Shanghai Jiao Tong University School of Medicine, Renji Hospital, College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xuan Wang
- Institute of Molecular Medicine (IMM), Shanghai Jiao Tong University School of Medicine, Renji Hospital, College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jingqi Chen
- Institute of Molecular Medicine (IMM), Shanghai Jiao Tong University School of Medicine, Renji Hospital, College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Chao Gu
- Institute of Molecular Medicine (IMM), Shanghai Jiao Tong University School of Medicine, Renji Hospital, College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Shuqin Huang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, People's Republic of China
| | - Yu Yang
- Institute of Molecular Medicine (IMM), Shanghai Jiao Tong University School of Medicine, Renji Hospital, College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Fang Zhang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, People's Republic of China.
| | - Weihong Tan
- Institute of Molecular Medicine (IMM), Shanghai Jiao Tong University School of Medicine, Renji Hospital, College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
16
|
Zhang W, Fan Y, Zhang J, Shi D, Yuan J, Ashrafizadeh M, Li W, Hu M, Abd El-Aty AM, Hacimuftuoglu A, Linnebacher M, Cheng Y, Li W, Fang S, Gong P, Zhang X. Cell membrane-camouflaged bufalin targets NOD2 and overcomes multidrug resistance in pancreatic cancer. Drug Resist Updat 2023; 71:101005. [PMID: 37647746 DOI: 10.1016/j.drup.2023.101005] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 08/14/2023] [Accepted: 08/19/2023] [Indexed: 09/01/2023]
Abstract
AIMS Multidrug resistance in pancreatic cancer poses a significant challenge in clinical treatment. Bufalin (BA), a compound found in secretions from the glands of toads, may help overcome this problem. However, severe cardiotoxicity thus far has hindered its clinical application. Hence, the present study aimed to develop a cell membrane-camouflaged and BA-loaded polylactic-co-glycolic acid nanoparticle (CBAP) and assess its potential to counter chemoresistance in pancreatic cancer. METHODS The toxicity of CBAP was evaluated by electrocardiogram, body weight, distress score, and nesting behavior of mice. In addition, the anticarcinoma activity and underlying mechanism were investigated both in vitro and in vivo. RESULTS CBAP significantly mitigated BA-mediated acute cardiotoxicity and enhanced the sensitivity of pancreatic cancer to several clinical drugs, such as gemcitabine, 5-fluorouracil, and FOLFIRINOX. Mechanistically, CBAP directly bound to nucleotide-binding and oligomerization domain containing protein 2 (NOD2) and inhibited the expression of nuclear factor kappa-light-chain-enhancer of activated B cells. This inhibits the expression of ATP-binding cassette transporters, which are responsible for chemoresistance in cancer cells. CONCLUSIONS Our findings indicate that CBAP directly inhibits NOD2. Combining CBAP with standard-of-care chemotherapeutics represents a safe and efficient strategy for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Wei Zhang
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518055, China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, Guangdong 518060, China; International Association for Diagnosis and Treatment of Cancer, Shenzhen, Guangdong 518055, China
| | - Yibao Fan
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518055, China; School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Jinze Zhang
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Dan Shi
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Jiahui Yuan
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Milad Ashrafizadeh
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Wei Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, China
| | - Man Hu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China
| | - A M Abd El-Aty
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, 12211 Giza, Egypt; Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum 25070, Turkey
| | - Ahmet Hacimuftuoglu
- Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum 25070, Turkey
| | - Michael Linnebacher
- Clinic of General Surgery, Molecular Oncology and Immunotherapy, Rostock University Medical Center, Rostock 18059, Germany
| | - Yongxian Cheng
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Weiguang Li
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, 999077, China.
| | - Shuo Fang
- Department of Oncology, The Seventh Affiliated Hospital Sun Yat-sen University, Shenzhen, Guangdong 518107, China.
| | - Peng Gong
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518055, China; International Association for Diagnosis and Treatment of Cancer, Shenzhen, Guangdong 518055, China.
| | - Xianbin Zhang
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518055, China; International Association for Diagnosis and Treatment of Cancer, Shenzhen, Guangdong 518055, China.
| |
Collapse
|
17
|
Lei W, Yang J, Wang J, Xiao Z, Zhou P, Zheng S, Zhu P. Synergetic EGCG and coenzyme Q10 DSPC liposome nanoparticles protect against myocardial infarction. Biomater Sci 2023; 11:6862-6870. [PMID: 37646313 DOI: 10.1039/d3bm00857f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
At the site of myocardial infarction (MI), various phenomena such as oxidative stress and myocardial apoptosis can be observed. Both epigallocatechin gallate (EGCG) and coenzyme Q10 (CoQ10) exhibit antioxidant and anti-inflammatory effects. Macrophages have demonstrated a higher internalization rate of cationic liposomes, thereby increasing their bioavailability. This study utilized EGCG in synergy with CoQ10 as an antioxidant agent and distearyl phosphatidylcholine (DSPC) as the carrier, to create liposome nanoparticles known as CE-LNPs. The CE-LNPs exhibited favorable biocompatibility and were effectively engulfed by macrophages in vitro. In addition, the CE-LNPs effectively eradicated reactive oxygen species (ROS) in hypoxic cardiomyocytes, mitigated myocardial cell apoptosis, and sustained the functionality and proliferation of myocardial cells. The anti-apoptotic effect of the CE-LNPs was further validated through TUNEL and Annexin V FITC/PI experiments. The therapeutic efficacy of CE-LNPs was evaluated in a murine model of MI. CE-LNPs demonstrated a significant reduction in scar area in vivo, facilitating cardiac repair and improving cardiac function. These findings provide evidence that EGCG synergistically combined with CoQ10 in DSPC liposome nanoparticles offers protection against MI.
Collapse
Affiliation(s)
- Wenrui Lei
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Jie Yang
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Junwei Wang
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Zezhou Xiao
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Pengyu Zhou
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Shaoyi Zheng
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Peng Zhu
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
18
|
Tigu AB, Constantinescu CS, Teodorescu P, Kegyes D, Munteanu R, Feder R, Peters M, Pralea I, Iuga C, Cenariu D, Marcu A, Tanase A, Colita A, Drula R, Bergthorsson JT, Greiff V, Dima D, Selicean C, Rus I, Zdrenghea M, Gulei D, Ghiaur G, Tomuleasa C. Design and preclinical testing of an anti-CD41 CAR T cell for the treatment of acute megakaryoblastic leukaemia. J Cell Mol Med 2023; 27:2864-2875. [PMID: 37667538 PMCID: PMC10538266 DOI: 10.1111/jcmm.17810] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 04/28/2023] [Accepted: 05/26/2023] [Indexed: 09/06/2023] Open
Abstract
Acute megakaryoblastic leukaemia (AMkL) is a rare subtype of acute myeloid leukaemia (AML) representing 5% of all reported cases, and frequently diagnosed in children with Down syndrome. Patients diagnosed with AMkL have low overall survival and have poor outcome to treatment, thus novel therapies such as CAR T cell therapy could represent an alternative in treating AMkL. We investigated the effect of a new CAR T cell which targets CD41, a specific surface antigen for M7-AMkL, against an in vitro model for AMkL, DAMI Luc2 cell line. The performed flow cytometry evaluation highlighted a percentage of 93.8% CAR T cells eGFP-positive and a limited acute effect on lowering the target cell population. However, the interaction between effector and target (E:T) cells, at a low ratio, lowered the cell membrane integrity, and reduced the M7-AMkL cell population after 24 h of co-culture, while the cytotoxic effect was not significant in groups with higher E:T ratio. Our findings suggest that the anti-CD41 CAR T cells are efficient for a limited time spawn and the cytotoxic effect is visible in all experimental groups with low E:T ratio.
Collapse
Affiliation(s)
- Adrian Bogdan Tigu
- Medfuture Research Center for Advanced MedicineIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
| | - Catalin Sorin Constantinescu
- Medfuture Research Center for Advanced MedicineIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
- Department of HematologyIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
- Intensive Care UnitEmergency Clinical HospitalCluj‐NapocaRomania
| | - Patric Teodorescu
- Department of HematologyIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
- Department of Leukemia, Sidney Kimmel Cancer Center at Johns HopkinsJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - David Kegyes
- Medfuture Research Center for Advanced MedicineIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
- Department of HematologyIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
| | - Raluca Munteanu
- Medfuture Research Center for Advanced MedicineIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
| | - Richard Feder
- Medfuture Research Center for Advanced MedicineIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
| | - Mareike Peters
- Medfuture Research Center for Advanced MedicineIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
- Department of HematologyIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
| | - Ioana Pralea
- Medfuture Research Center for Advanced MedicineIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
| | - Cristina Iuga
- Medfuture Research Center for Advanced MedicineIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
- Department of Drug AnalysisSchool of PharmacyIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
| | - Diana Cenariu
- Medfuture Research Center for Advanced MedicineIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
| | - Andra Marcu
- Department of PediatricsCarol Davila University of Medicine and PharmacyBucharestRomania
- Department of Stem Cell TransplantationFundeni Clinical InstituteBucharestRomania
| | - Alina Tanase
- Department of PediatricsCarol Davila University of Medicine and PharmacyBucharestRomania
- Department of Stem Cell TransplantationFundeni Clinical InstituteBucharestRomania
| | - Anca Colita
- Department of PediatricsCarol Davila University of Medicine and PharmacyBucharestRomania
- Department of Stem Cell TransplantationFundeni Clinical InstituteBucharestRomania
| | - Rares Drula
- Medfuture Research Center for Advanced MedicineIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
| | - Jon Thor Bergthorsson
- Stem Cell Research Unit, Biomedical Center, School of Health SciencesUniversity of IcelandReykjavíkIceland
- Department of Laboratory HematologyLandspitali University HospitalReykjavíkIceland
| | - Victor Greiff
- Department of ImmunologyUniversity of Oslo and Oslo University HospitalOsloNorway
| | - Delia Dima
- Department of HematologyIon Chiricuta Clinical Cancer CenterCluj NapocaRomania
| | - Cristina Selicean
- Department of HematologyIon Chiricuta Clinical Cancer CenterCluj NapocaRomania
| | - Ioana Rus
- Department of HematologyIon Chiricuta Clinical Cancer CenterCluj NapocaRomania
| | - Mihnea Zdrenghea
- Department of HematologyIon Chiricuta Clinical Cancer CenterCluj NapocaRomania
| | - Diana Gulei
- Medfuture Research Center for Advanced MedicineIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
| | - Gabriel Ghiaur
- Medfuture Research Center for Advanced MedicineIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
- Department of Leukemia, Sidney Kimmel Cancer Center at Johns HopkinsJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Ciprian Tomuleasa
- Medfuture Research Center for Advanced MedicineIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
- Department of HematologyIuliu Hatieganu University of Medicine and PharmacyCluj‐NapocaRomania
- Department of HematologyIon Chiricuta Clinical Cancer CenterCluj NapocaRomania
| |
Collapse
|
19
|
Balachandran C, Hirose M, Tanaka T, Zhu JJ, Yokoi K, Hisamatsu Y, Yamada Y, Aoki S. Design and Synthesis of Poly(2,2'-Bipyridyl) Ligands for Induction of Cell Death in Cancer Cells: Control of Anticancer Activity by Complexation/Decomplexation with Biorelevant Metal Cations. Inorg Chem 2023; 62:14615-14631. [PMID: 37642721 PMCID: PMC10498496 DOI: 10.1021/acs.inorgchem.3c01738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Indexed: 08/31/2023]
Abstract
Chelation therapy is a medical procedure for removing toxic metals from human organs and tissues and for the treatment of diseases by using metal-chelating agents. For example, iron chelation therapy is designed not only for the treatment of metal poisoning but also for some diseases that are induced by iron overload, cancer chemotherapy, and related diseases. However, the use of such metal chelators needs to be generally carried out very carefully, because of the side effects possibly due to the non-specific complexation with intracellular metal cations. Herein, we report on the preparation and characterization of some new poly(bpy) ligands (bpy: 2,2'-bipyridyl) that contain one-three bpy ligand moieties and their anticancer activity against Jurkat, MOLT-4, U937, HeLa S3, and A549 cell lines. The results of MTT assays revealed that the tris(bpy) and bis(bpy) ligands exhibit potent activity for inducing the cell death in cancer cells. Mechanistic studies suggest that the main pathway responsible for the cell death by these poly(bpy) ligands is apoptotic cell death. It was also found that the anticancer activity of the poly(bpy) ligands could be controlled by the complexation (anticancer activity is turned OFF) and decomplexation (anticancer activity is turned ON) with biorelevant metal cations. In this paper, these results will be described.
Collapse
Affiliation(s)
- Chandrasekar Balachandran
- Faculty
of Pharmaceutical Sciences, Tokyo University
of Science, 2641 Yamazaki, Noda 278-8510, Japan
- Research
Institute for Biomedical Sciences, Tokyo
University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Masumi Hirose
- Faculty
of Pharmaceutical Sciences, Tokyo University
of Science, 2641 Yamazaki, Noda 278-8510, Japan
| | - Tomohiro Tanaka
- Faculty
of Pharmaceutical Sciences, Tokyo University
of Science, 2641 Yamazaki, Noda 278-8510, Japan
| | - Jun Jie Zhu
- Faculty
of Pharmaceutical Sciences, Tokyo University
of Science, 2641 Yamazaki, Noda 278-8510, Japan
| | - Kenta Yokoi
- Faculty
of Pharmaceutical Sciences, Tokyo University
of Science, 2641 Yamazaki, Noda 278-8510, Japan
| | - Yosuke Hisamatsu
- Faculty
of Pharmaceutical Sciences, Tokyo University
of Science, 2641 Yamazaki, Noda 278-8510, Japan
- Graduate
School of Pharmaceutical Sciences, Nagoya
City University, 3-1
Tanabe-dori, Nagoya, Aichi 467-8603, Japan
| | - Yasuyuki Yamada
- Department
of Chemistry, Graduate School of Science, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8602, Japan
- Research
Center for Materials Science, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8602, Japan
| | - Shin Aoki
- Faculty
of Pharmaceutical Sciences, Tokyo University
of Science, 2641 Yamazaki, Noda 278-8510, Japan
- Research
Institute for Biomedical Sciences, Tokyo
University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
- Research
Institute for Science and Technology, Tokyo
University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| |
Collapse
|
20
|
Chen S, Fan L, Lin Y, Qi Y, Xu C, Ge Q, Zhang Y, Wang Q, Jia D, Wang L, Si J, Wang L. Bifidobacterium adolescentis orchestrates CD143 + cancer-associated fibroblasts to suppress colorectal tumorigenesis by Wnt signaling-regulated GAS1. Cancer Commun (Lond) 2023; 43:1027-1047. [PMID: 37533188 PMCID: PMC10508156 DOI: 10.1002/cac2.12469] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/16/2023] [Accepted: 07/13/2023] [Indexed: 08/04/2023] Open
Abstract
BACKGROUND The interplay between gut microbiota and tumor microenvironment (TME) in the pathogenesis of colorectal cancer (CRC) is not well explored. Here, we elucidated the functional role of Bifidobacterium adolescentis (B.a) on CRC and investigated its possible mechanism on the manipulation of cancer-associated fibroblasts (CAFs) in CRC. METHODS Different CRC animal models and various cell line models were established to explore the function of B.a on CRC. The single-cell RNA sequencing (scRNA-seq) or flow cytometry was used to detect the cell subsets in the TME of CRC. Western blot, quantitative real-time polymerase chain reaction (qRT-PCR), or immunofluorescence staining were performed to examine the activation of Wnt signaling and growth arrest specific 1 (GAS1) on CD143+ CAFs. Chromatin immunoprecipitation quantitative real-time PCR (CHIP-qPCR) was performed to investigate the regulation of transcription factor 4 (TCF4) on GAS1. Multi-immunofluorescence assay examined the expression level of CD143 and GAS1 on tissue microarray. RESULTS We found that B.a abundance was significantly reduced in CRC patients from two independent cohorts and the bacteria database of GMrepo. Supplementation with B.a suppressed ApcMin/+ spontaneous or AOM/DSS-induced tumorigenesis in mice. scRNA-seq revealed that B.a facilitated a subset of CD143+ CAFs by inhibiting the infiltration of Th2 cells, while promoting the TNF-alpha+ B cells in TME. CD143+ CAFs highly expressed GAS1 and exhibited tumor suppressive effect. Mechanistically, GAS1 was activated by the Wnt/β-catenin signaling in CD143+ CAFs. B.a abundance was correlated with the expression level of CD143 and GAS1. The level of CD143+ CAFs predicted the better survival outcome in CRC patients. CONCLUSIONS These results highlighted that B.a induced a new subset of CD143+ CAFs by Wnt signaling-regulated GAS1 to suppress tumorigenesis and provided a novel therapeutic target for probiotic-based modulation of TME in CRC.
Collapse
Affiliation(s)
- Shujie Chen
- Department of GastroenterologySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Institute of GastroenterologyZhejiang UniversityHangzhouZhejiangP. R. China
- Cancer CenterZhejiang UniversityHangzhouZhejiangP. R. China
- Research Center of Prevention and Treatment of Senescent DiseaseSchool of Medicine Zhejiang UniversityHangzhouZhejiangP. R. China
| | - Lina Fan
- Institute of GastroenterologyZhejiang UniversityHangzhouZhejiangP. R. China
- Department of GastroenterologySecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouZhejiangP. R. China
| | - Yifeng Lin
- Institute of GastroenterologyZhejiang UniversityHangzhouZhejiangP. R. China
- Department of GastroenterologySecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouZhejiangP. R. China
| | - Yadong Qi
- Department of GastroenterologySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Institute of GastroenterologyZhejiang UniversityHangzhouZhejiangP. R. China
| | - Chaochao Xu
- Institute of GastroenterologyZhejiang UniversityHangzhouZhejiangP. R. China
- Department of GastroenterologySecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouZhejiangP. R. China
| | - Qiwei Ge
- Institute of GastroenterologyZhejiang UniversityHangzhouZhejiangP. R. China
- Department of GastroenterologySecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouZhejiangP. R. China
| | - Ying Zhang
- Institute of GastroenterologyZhejiang UniversityHangzhouZhejiangP. R. China
- Department of GastroenterologySecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouZhejiangP. R. China
| | - Qiwen Wang
- Department of GastroenterologySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Institute of GastroenterologyZhejiang UniversityHangzhouZhejiangP. R. China
| | - Dingjiacheng Jia
- Institute of GastroenterologyZhejiang UniversityHangzhouZhejiangP. R. China
- Department of GastroenterologySecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouZhejiangP. R. China
| | - Lan Wang
- Department of GastroenterologySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Institute of GastroenterologyZhejiang UniversityHangzhouZhejiangP. R. China
| | - Jianmin Si
- Department of GastroenterologySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Institute of GastroenterologyZhejiang UniversityHangzhouZhejiangP. R. China
- Cancer CenterZhejiang UniversityHangzhouZhejiangP. R. China
- Research Center of Prevention and Treatment of Senescent DiseaseSchool of Medicine Zhejiang UniversityHangzhouZhejiangP. R. China
| | - Liangjing Wang
- Institute of GastroenterologyZhejiang UniversityHangzhouZhejiangP. R. China
- Cancer CenterZhejiang UniversityHangzhouZhejiangP. R. China
- Research Center of Prevention and Treatment of Senescent DiseaseSchool of Medicine Zhejiang UniversityHangzhouZhejiangP. R. China
- Department of GastroenterologySecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouZhejiangP. R. China
| |
Collapse
|
21
|
Zhang YH, Li CY, Zou GJ, Xian JY, Zhang Q, Yu BX, Huang LH, Liu HX, Sun XY. Corn Silk Polysaccharides with Different Carboxyl Contents Reduce the Oxidative Damage of Renal Epithelial Cells by Inhibiting Endocytosis of Nano-calcium Oxalate Crystals. ACS OMEGA 2023; 8:25839-25849. [PMID: 37521646 PMCID: PMC10373179 DOI: 10.1021/acsomega.3c01306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 06/29/2023] [Indexed: 08/01/2023]
Abstract
OBJECTIVE Renal epithelial cell injury and cell-crystal interaction are closely related to kidney stone formation. METHODS This study aims to explore the inhibition of endocytosis of nano-sized calcium oxalate monohydrate (nano-COM) crystals and the cell protection of corn silk polysaccharides (CCSPs) with different carboxyl contents (3.92, 7.75, 12.90, and 16.38%). The nano-COM crystals protected or unprotected by CCSPs were co-cultured with human renal proximal tubular epithelial cells (HK-2), and then the changes in the endocytosis of nano-COM and cell biochemical indicators were detected. RESULTS CCSPs could inhibit the endocytosis of nano-COM by HK-2 cells and reduce the accumulation of nano-COM in the cells. Under the protection of CCSPs, cell morphology is restored, intracellular superoxide dismutase levels are increased, lipid peroxidation product malondialdehyde release is decreased, and mitochondrial membrane potential and lysosomal integrity are increased. The release of Ca2+ ions in the cell, the level of cell autophagy, and the rate of cell apoptosis and necrosis are also reduced. CCSPs with higher carboxyl content have better cell protection abilities. CONCLUSION CCSPs could inhibit the endocytosis of nano-COM crystals and reduce cell oxidative damage. CCSP3, with the highest carboxyl content, shows the best biological activity.
Collapse
Affiliation(s)
- Yi-Han Zhang
- Department
of Urology, Guangzhou Institute of Urology, Guangdong Key Laboratory
of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510230, China
| | - Chun-Yao Li
- Department
of Urology, Guangzhou Institute of Urology, Guangdong Key Laboratory
of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510230, China
| | - Guo-Jun Zou
- Department
of Chemistry, Institute of Biomineralization and Lithiasis Research, Jinan University, Guangzhou, Guangdong 510632, China
| | - Jun-Yi Xian
- Department
of Urology, Guangzhou Institute of Urology, Guangdong Key Laboratory
of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510230, China
| | - Quan Zhang
- Department
of Urology, Guangzhou Institute of Urology, Guangdong Key Laboratory
of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510230, China
| | - Bang-Xian Yu
- Department
of Urology, Guangzhou Institute of Urology, Guangdong Key Laboratory
of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510230, China
| | - Ling-Hong Huang
- Department
of Urology, Guangzhou Institute of Urology, Guangdong Key Laboratory
of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510230, China
| | - Hong-Xing Liu
- Department
of Urology, Guangzhou Institute of Urology, Guangdong Key Laboratory
of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510230, China
| | - Xin-Yuan Sun
- Department
of Urology, Guangzhou Institute of Urology, Guangdong Key Laboratory
of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510230, China
| |
Collapse
|
22
|
Si Y, Hui C, Guo T, Liu M, Chen X, Dong C, Feng S. Phellodendronoside A Exerts Anticancer Effects Depending on Inducing Apoptosis Through ROS/Nrf2/Notch Pathway and Modulating Metabolite Profiles in Hepatocellular Carcinoma. J Hepatocell Carcinoma 2023; 10:935-948. [PMID: 37361906 PMCID: PMC10290457 DOI: 10.2147/jhc.s403630] [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: 03/06/2023] [Accepted: 05/15/2023] [Indexed: 06/28/2023] Open
Abstract
Purpose To reveal the potential mechanism of PDA on hepatocellular carcinoma SMMC-7721 cells in vitro. Methods The cytotoxic activity, colony formation, cell cycle distribution, apoptosis and their associated protein analysis, intracellular reactive oxygen species (ROS) and Ca2+ levels, proteins in Nrf2 and Ntoch pathways and metabolite profiles of PDA against hepatocellular carcinoma were investigated. Results PDA with cytotoxic activity inhibited cell proliferation and migration, increased intracellular ROS, Ca2+ levels and MCUR1 protein expression in a dose-dependent manner, caused cell cycle arrest in the S phase and induced apoptosis via adjusting the levels of Bcl-2, Bax, and Caspase 3 proteins, and inhibited the activation of Notch1, Jagged, Hes1, Nrf2 and HO-1 proteins. Metabonomics data showed that PDA significantly regulated 144 metabolite levels tend to be normal level, especially carnitine derivatives, bile acid metabolites associated with hepatocellular carcinoma, and mainly enriched in ABC transporter, arginine and proline metabolism, primary bile acid biosynthesis, Notch signaling pathway, etc, and proved that PDA markedly adjusted Notch signaling pathway. Conclusion PDA exhibited the proliferation inhibition of SMMC-7721 cells by inhibiting ROS/Nrf2/Notch signaling pathway and significantly affected the metabolic profile, suggesting PDA could be a potential therapeutic agent for patients with hepatocellular carcinoma.
Collapse
Affiliation(s)
- Yanpo Si
- Department of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
- Henan Engineering Research Center of Medicinal and Edible Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Chengcheng Hui
- Department of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Tao Guo
- Department of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
- Henan Engineering Research Center of Medicinal and Edible Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Mengqi Liu
- Department of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Xiaohui Chen
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Chunhong Dong
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
- Henan Key Laboratory of Chinese Medicine for Polysaccharides and Drugs Research, Zhengzhou, People’s Republic of China
| | - Shuying Feng
- Medical College, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| |
Collapse
|
23
|
Wang C, Wu W, Pang Z, Liu J, Qiu J, Luan T, Deng J, Fang Z. Polystyrene microplastics significantly facilitate influenza A virus infection of host cells. JOURNAL OF HAZARDOUS MATERIALS 2023; 446:130617. [PMID: 36623344 DOI: 10.1016/j.jhazmat.2022.130617] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 06/17/2023]
Abstract
Microplastics (MPs) are emerging pollutants which exist in various environments and pose a potential threat to human health. However, the effect of MP on respiratory pathogens-infected organisms is unknown. In order to explore the effect of MP on respiratory pathogen infection, we studied the effect of polystyrene microplastics (PS) on influenza A virus (IAV)-infected A549 cells. Western blot, qPCR, and viral plaque assay demonstrated that PS could promote IAV infection. Further study by bioluminescence imaging showed that a large number of IAV could be enriched on PS and entered cells through endocytosis. Meanwhile, the expression of IFITM3 in cells was significantly reduced. In addition, our results showed that PS down-regulated IRF3 and its active form P-IRF3 by down-regulating RIG-I and inhibiting TBK1 phosphorylation activation, which then significantly reduced IFN-β expression and affected the cellular innate antiviral immune system. Taken together, our results indicate the potential threat of MPs to respiratory diseases caused by IAV and provide new insights into human health protection.
Collapse
Affiliation(s)
- Chao Wang
- Guangdong Second Provincial General Hospital, 466 Middle Xingang Road, Guangzhou 510317, Guangdong, China
| | - Wenjiao Wu
- Guangdong Second Provincial General Hospital, 466 Middle Xingang Road, Guangzhou 510317, Guangdong, China
| | - Zefen Pang
- Guangdong Second Provincial General Hospital, 466 Middle Xingang Road, Guangzhou 510317, Guangdong, China; School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, 100 Waihuanxi Road, Guangzhou 510006, Guangdong, China
| | - Jiaxin Liu
- Guangdong Second Provincial General Hospital, 466 Middle Xingang Road, Guangzhou 510317, Guangdong, China
| | - Jianxiang Qiu
- Guangdong Second Provincial General Hospital, 466 Middle Xingang Road, Guangzhou 510317, Guangdong, China
| | - Tiangang Luan
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, 100 Waihuanxi Road, Guangzhou 510006, Guangdong, China; Jieyang Branch of Chemistry and Chemical Engineering Guangdong Laboratory 7 (Rongjiang Laboratory), Jieyang 515200, Guangdong, China
| | - Jiewei Deng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, 100 Waihuanxi Road, Guangzhou 510006, Guangdong, China.
| | - Zhixin Fang
- Guangdong Second Provincial General Hospital, 466 Middle Xingang Road, Guangzhou 510317, Guangdong, China.
| |
Collapse
|
24
|
Xie Y, Liang H, Jiang N, Liu D, Zhang N, Li Q, Zhang K, Sang X, Feng Y, Chen R, Zhang Y, Chen Q. Graphene quantum dots induce cascadic apoptosis via interaction with proteins associated with anti-oxidation after endocytosis by Trypanosoma brucei. Front Immunol 2022; 13:1022050. [PMID: 36561761 PMCID: PMC9763322 DOI: 10.3389/fimmu.2022.1022050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 11/08/2022] [Indexed: 12/12/2022] Open
Abstract
Trypanosoma brucei, the pathogen causing African sleeping sickness (trypanosomiasis) in humans, causes debilitating diseases in many regions of the world, but mainly in African countries with tropical and subtropical climates. Enormous efforts have been devoted to controlling trypanosomiasis, including expanding vector control programs, searching for novel anti-trypanosomial agents, and developing vaccines, but with limited success. In this study, we systematically investigated the effect of graphene quantum dots (GQDs) on trypanosomal parasites and their underlying mechanisms. Ultrasmall-sized GQDs can be efficiently endocytosed by T. brucei and with no toxicity to mammalian-derived cells, triggering a cascade of apoptotic reactions, including mitochondrial disorder, intracellular reactive oxygen species (ROS) elevation, Ca2+ accumulation, DNA fragmentation, adenosine triphosphate (ATP) synthesis impairment, and cell cycle arrest. All of these were caused by the direct interaction between GQDs and the proteins associated with cell apoptosis and anti-oxidation responses, such as trypanothione reductase (TryR), a key protein in anti-oxidation. GQDs specifically inhibited the enzymatic activity of TryR, leading to a reduction in the antioxidant capacity and, ultimately, parasite apoptotic death. These data, for the first time, provide a basis for the exploration of GQDs in the development of anti-trypanosomials.
Collapse
Affiliation(s)
- Yiwei Xie
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Hongrui Liang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Ning Jiang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Dingyuan Liu
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Naiwen Zhang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Qilong Li
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Kai Zhang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Xiaoyu Sang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Ying Feng
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Ran Chen
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Yiwei Zhang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Qijun Chen
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China,*Correspondence: Qijun Chen,
| |
Collapse
|
25
|
Wang Y, Wang G, Liu X, Yun D, Cui Q, Wu X, Lu W, Yang X, Zhang M. Inhibition of APLN suppresses cell proliferation and migration and promotes cell apoptosis in esophageal cancer cells <em>in vitro</em>, through activating PI3K/mTOR signaling pathway. Eur J Histochem 2022; 66. [PMID: 35920446 PMCID: PMC9422863 DOI: 10.4081/ejh.2022.3336] [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] [Received: 10/01/2021] [Accepted: 07/13/2022] [Indexed: 11/23/2022] Open
Abstract
Esophageal cancer is the sixth leading cause of cancer mortalities globally with a high incidence rate. Apelin (APLN) plays regulatory roles in different organs. However, its role in esophageal cancer remains unknown. Therefore, our study aims to explore the effect of APLN on esophageal cancer. One hundred and eighty-four (184) esophageal tumor tissues samples from patients with esophageal cancer, and 11 esophageal tissues samples from healthy volunteers were analyzed for the expression of APLN. APLN was highly expressed in the tumor of patients with esophageal cancer and esophageal cancer cells. Patients with high expressions of APLN had a lower survival rate than the ones with low to medium expressions of APLN. Human esophageal carcinoma cell lines, TE-1 and ECA-109 cells were transfected with APLN siRNA to knockdown APLN, or transfected with pcDNA-APLN to overexpress APLN. Inhibition of APLN by siRNA-APLN reduced proliferative, migrative, and invasive abilities of esophageal cancer cells and promoted cell apoptosis, which could be all restored by pcDNA-APLN. Moreover, knocking down APLN by siRNA-APLN suppressed the PI3K/mTOR signaling pathway. These findings identify that APLN inhibition might ameliorate esophageal cancer through activating the PI3K/mTOR signaling pathway, thus APLN could be a potential target for esophageal cancer.
Collapse
Affiliation(s)
- Yuhan Wang
- Department of Integrated Traditional Chinese and Western Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University.
| | - Gang Wang
- Pharmacy Department, Puai District of Huangshi Central Hospital. Wuhan.
| | - Xiaojun Liu
- Pharmacy Department, Puai District of Huangshi Central Hospital, Wuhan.
| | - Dong Yun
- Department of Integrated Traditional Chinese and Western Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University.
| | - Qing Cui
- Department of Integrated Traditional Chinese and Western Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University.
| | - Xiaoting Wu
- Shanghai Jiao Tong University School of Medicine.
| | - Wenfeng Lu
- Department of Integrated Traditional Chinese and Western Medicine, Zhongshan Hospital, Fudan University.
| | - Xiwen Yang
- Shanghai Literature Institute of Traditional Chinese Medicine.
| | - Ming Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University.
| |
Collapse
|
26
|
Tian L, Pei R, Zhang X, Li K, Zhong Y, Luo Y, Zhou SF, Chen L. Tumor Cell-Specific and Lipase-Responsive Delivery of Hydrogen Sulfide for Sensitizing Chemotherapy of Pancreatic Cancer. Front Bioeng Biotechnol 2022; 10:934151. [PMID: 35898641 PMCID: PMC9309817 DOI: 10.3389/fbioe.2022.934151] [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: 05/02/2022] [Accepted: 06/08/2022] [Indexed: 11/23/2022] Open
Abstract
The inability of small molecule drugs to diffuse into tumor interstitium is responsible for the relatively low effectiveness of chemotherapy. Herein, a hydrogen sulfide (H2S) gas-involved chemosensitization strategy is proposed for pancreatic cancer treatment by developing a tumor-specific lipase-responsive nanomedicine based on aptamer-conjugated DATS/Dox co-loaded PCL-b-PEO micelle (DA/D@Ms-A). After receptor-mediated endocytosis and subsequent digestion of PCL blocks by intracellular lipase, the nanomedicine releases Dox and DATS, which then react with intracellular glutathione to produce H2S. The cytotoxicity result indicates that H2S can enhance Dox chemotherapy efficiency owing to the synergetic therapeutic effect of Dox and H2S. Moreover, the nanomedicine is featured with well tumor penetration capability benefitting from the targeting ability of aptamers and high in vivo biocompatibility due to the high density of PEO and biodegradable PCL. The nanomedicine capable of synergetic gas-chemotherapy holds great potential for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Libing Tian
- College of Chemical Engineering, Huaqiao University, Xiamen, China
| | - Rui Pei
- College of Chemical Engineering, Huaqiao University, Xiamen, China
| | - Xiaojun Zhang
- College of Chemical Engineering, Huaqiao University, Xiamen, China
| | - Kun Li
- College of Chemical Engineering, Huaqiao University, Xiamen, China
| | - Yuting Zhong
- College of Chemical Engineering, Huaqiao University, Xiamen, China
| | - Yougen Luo
- Department of Basic Medical Science, Jiangsu Vocational College of Medicine, Yancheng, China
| | - Shu-Feng Zhou
- College of Chemical Engineering, Huaqiao University, Xiamen, China
| | - Lichan Chen
- College of Chemical Engineering, Huaqiao University, Xiamen, China
| |
Collapse
|
27
|
Li Y, Sun S, Wen C, Zhong J, Jiang Q. Effect of Enterococcus faecalis OG1RF on human calvarial osteoblast apoptosis. BMC Oral Health 2022; 22:279. [PMID: 35804353 PMCID: PMC9264677 DOI: 10.1186/s12903-022-02295-y] [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: 04/15/2022] [Accepted: 06/13/2022] [Indexed: 11/24/2022] Open
Abstract
Background Enterococcus faecalis is a dominant pathogen in the root canals of teeth with persistent apical periodontitis (PAP), and osteoblast apoptosis contributes to imbalanced bone remodelling in PAP. Here, we investigated the effect of E. faecalis OG1RF on apoptosis in primary human calvarial osteoblasts. Specifically, the expression of apoptosis-related genes and the role of anti-apoptotic and pro-apoptotic members of the BCL-2 family were examined. Methods Primary human calvarial osteoblasts were incubated with E. faecalis OG1RF at multiplicities of infection corresponding to infection time points. Flow cytometry, terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL) assay, caspase-3/-8/-9 activity assay, polymerase chain reaction (PCR) array, and quantitative real-time PCR were used to assess osteoblast apoptosis. Results E. faecalis infection increased the number of early- and late-phase apoptotic cells and TUNEL-positive cells, decreased the mitochondrial membrane potential (ΔΨm), and activated the caspase-3/-8/-9 pathway. Moreover, of all 84 apoptosis-related genes in the PCR array, the expression of 16 genes was upregulated and that of four genes was downregulated in the infected osteoblasts. Notably, the mRNA expression of anti-apoptotic BCL2 was downregulated, whereas that of the pro-apoptotic BCL2L11, HRK, BIK, BMF, NOXA, and BECN1 and anti-apoptotic BCL2A1 was upregulated. Conclusions E. faecalis OG1RF infection triggered apoptosis in human calvarial osteoblasts, and BCL-2 family members acted as regulators of osteoblast apoptosis. Therefore, BCL-2 family members may act as potential therapeutic targets for persistent apical periodontitis.
Collapse
Affiliation(s)
- Yang Li
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, 510182, China
| | - Shuyu Sun
- Department of Endodontics, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Cheng Wen
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, 510182, China
| | - Jialin Zhong
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, 510182, China
| | - Qianzhou Jiang
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, 510182, China.
| |
Collapse
|
28
|
Fang D, Liu Z, Jin H, Huang X, Shi Y, Ben S. Manganese-Based Prussian Blue Nanocatalysts Suppress Non-Small Cell Lung Cancer Growth and Metastasis via Photothermal and Chemodynamic Therapy. Front Bioeng Biotechnol 2022; 10:939158. [PMID: 35814022 PMCID: PMC9257087 DOI: 10.3389/fbioe.2022.939158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 05/27/2022] [Indexed: 12/18/2022] Open
Abstract
Based on the safety of prussian blue (PB) in biomedical application, we prepared manganese-based prussian blue (MnPB) nanocatalysts to achieve enhanced photothermal therapy and chemodynamic therapy. And we conducted a series of experiments to explore the therapeutic effects of MnPB nanoparticles (NPs) on non-small cell lung cancer (NSCLC) in vivo and in vitro. For in vitro experiments, the MnPB NPs suppressed growth of A549 cells by reactive oxygen species upregulation and near-infrared irradiation. Moreover, the MnPB NPs could inhibit lung cancer metastasis through downregulating the matrix metalloproteinase (MMP)-2 and MMP-9 expression in A549 cells. And for in vivo experiments, the MnPB NPs inhibited the growth of xenografted tumor effectively and were biologically safe. Meanwhile, Mn2+ as a T1-weighted agent could realize magnetic resonance imaging-guided diagnosis and treatment. To sum up, the results in this study clearly demonstrated that the MnPB NPs had remarkable effects for inhibiting the growth and metastasis of NSCLC and might serve as a promising multifunctional nanoplatform for NSCLC treatment.
Collapse
Affiliation(s)
- Danruo Fang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zeyu Liu
- Department of Respiratory and Critical Care Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Hansong Jin
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiulin Huang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongxin Shi
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Suqin Ben
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Suqin Ben,
| |
Collapse
|
29
|
Cao W, Sun Y, Liu L, Yu J, Ji J, Wang Y, Yang J. HOTAIR mediates cisplatin resistance in nasopharyngeal carcinoma by regulating miR-106a-5p/SOX4 axis. Bioengineered 2022; 13:6567-6578. [PMID: 35227173 PMCID: PMC8975274 DOI: 10.1080/21655979.2022.2038429] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
This study explored the function and mechanisms of HOX transcript antisense RNA (HOTAIR) in the drug resistance of nasopharyngeal carcinoma (NPC). Quantitative PCR, Western blotting, MTT assay, flow cytometry, Transwell assay, and luciferase assay were performed. HOTAIR expression levels were upregulated in cisplatin (DDP)-resistant NPC tissues and cells. Knockdown of HOTAIR in DDP-resistant NPC cells increased cell sensitivity of DDP, as well as decreased cell viability, expression of chemoresistance-related proteins, migration and invasion, increased cell apoptosis. In addition, downregulation of microRNA 106a-5p (miR-106a-5p) expression and upregulation of SRY-box transcription factor 4 (SOX4) expression were observed in DDP-resistant NPC tissues and cells. MiR-106a-5p targets HOTAIR and SOX4; thus, silencing of HOTAIR significantly increased miR-106a-5p expression. The overexpression of miR-106a-5p significantly reversed the increase in SOX4 expression induced by HOTAIR lentivirus (Lv-HOTAIR). Knockdown of SOX4 reduced the drug resistance of DDP caused by the silencing of miR-106a-5p expression. In summary, HOTAIR enhanced DDP resistance in NPC cells by regulating the miR-106a-5p/SOX4 axis.
Collapse
Affiliation(s)
- Wei Cao
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Hospital of Anhui Medical University, Hefei, China
| | - Yi Sun
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Hospital of Anhui Medical University, Hefei, China
| | - Long Liu
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Hospital of Anhui Medical University, Hefei, China
| | - Junwei Yu
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Hospital of Anhui Medical University, Hefei, China
| | - Jiabiao Ji
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Hospital of Anhui Medical University, Hefei, China
| | - Yatang Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Hospital of Anhui Medical University, Hefei, China
| | - Jianming Yang
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
30
|
Circumventing Doxorubicin Resistance Using Elastin-like Polypeptide Biopolymer-Mediated Drug Delivery. Int J Mol Sci 2022; 23:ijms23042301. [PMID: 35216417 PMCID: PMC8878013 DOI: 10.3390/ijms23042301] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 01/14/2022] [Accepted: 02/12/2022] [Indexed: 01/02/2023] Open
Abstract
Although doxorubicin (dox), an anthracycline antibiotic, is widely used and effective in treating cancer, its treatment efficiency is limited by low blood plasma solubility, poor pharmacokinetics, and adverse side effects, including irreversible cardiotoxicity. Moreover, cancer cells often develop drug resistance over time, which decreases the efficacy of anti-cancer drugs, including dox. In this study, we examine a macromolecular drug delivery system for its ability to specifically deliver doxorubicin to cancer cells with and without drug resistance. This drug delivery system consists of a multi-part macromolecule, which includes the following: elastin-like polypeptide (ELP), cell penetrating peptide (CPP), a cleavable linker (releasing at low pH), and a derivative of doxorubicin. ELP is thermally responsive and improves drug solubility, while the CPP mediates cellular uptake of macromolecules. We compared cytotoxicity of two doxorubicin derivatives, where one is cleavable (DOXO) and contains a pH-sensitive linker and releases dox in an acidic environment, and the other is non-cleavable (ncDox) doxorubicin. Cytotoxicity, apoptosis, cell cycle distribution and mechanism of action of these constructs were tested and compared between dox-responsive MCF-7 and dox-resistant NCI/ADR cell lines. Dox delivered by the ELP construct is comparably toxic to both sensitive and drug resistant cell lines, compared to unconjugated doxorubicin, and given the pharmacokinetic and targeting benefits conveyed by conjugation to ELP, these biopolymers have potential to overcome dox resistance in vivo.
Collapse
|
31
|
Nguyen P, Doan P, Murugesan A, Ramesh T, Rimpilainen T, Candeias NR, Yli-Harja O, Kandhavelu M. GPR17 signaling activation by CHBC agonist induced cell death via modulation of MAPK pathway in glioblastoma. Life Sci 2022; 291:120307. [PMID: 35016881 DOI: 10.1016/j.lfs.2022.120307] [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: 09/23/2021] [Revised: 12/17/2021] [Accepted: 01/04/2022] [Indexed: 01/10/2023]
Abstract
AIM Glioblastoma multiforme (GBM) is the most common and aggressive primary adult brain tumor. GBM is characterized by a heterogeneous population of cells that are resistant to chemotherapy. Recently, we have synthesized CHBC, a novel indole derivative targeted to GBM biomarker G-protein-coupled receptor 17 and inhibitor of GBM cells. In this study, CHBC was further investigated to characterize the efficiency of this agonist at the molecular level and its underlying mechanism in GBM cell death induction. MATERIALS AND METHODS The effect of CHBC and TMZ was determined using time dependent inhibitor assay in glioblastoma cells, LN229 and SNB19. Drug induced cell cycle arrest was measured using PI staining followed by image analysis. The induction of apoptosis and mechanism of action of CHBC was studied using apoptosis, caspase 3/7 and mitochondrial membrane permeability assays. Modulation of the key genes involved in MAPK signaling pathway was also measured using immunoblotting array. KEY FINDINGS The inhibitory kinetic study has revealed that CHBC inhibited SNB19 and LN229 cell growth in a time-dependent manner. Furthermore, CHBC with the IC50 of 85 μM, mediated cell death through an apoptosis mechanism in both studied cell lines. The study also has revealed that CHBC targets GPR17 leading to the induction of apoptosis via the activation of Caspase 3/7 and dysfunction of mitochondrial membrane potential. In addition, CHBC treatment led to marked G2/M cell cycle arrest. The protein array has confirmed the anticancer effect of CHBC by the disruption of the mitogen-activated protein kinase pathway (MAPK). SIGNIFICANCE Taken together, these results demonstrated that CHBC induced G2/M cell cycle arrest and apoptosis by disrupting MAPK signaling in human glioblastoma cells. This study concludes that CHBC represent a class of compounds for treating glioblastoma.
Collapse
Affiliation(s)
- Phung Nguyen
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, Tampere 33720, Finland; BioMeditech and Tays Cancer Center, Tampere University Hospital, P.O. Box 553, 33101 Tampere, Finland
| | - Phuong Doan
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, Tampere 33720, Finland; BioMeditech and Tays Cancer Center, Tampere University Hospital, P.O. Box 553, 33101 Tampere, Finland
| | - Akshaya Murugesan
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, Tampere 33720, Finland; Department of Biotechnology, Lady Doak College, Thallakulam, Madurai 625002, India
| | - Thiyagarajan Ramesh
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Tatu Rimpilainen
- Faculty of Engineering and Natural Sciences, Tampere University, 33101 Tampere, Finland
| | - Nuno R Candeias
- Faculty of Engineering and Natural Sciences, Tampere University, 33101 Tampere, Finland; LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Olli Yli-Harja
- Computational Systems Biology Group, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101 Tampere, Finland; Institute for Systems Biology, 1441N 34th Street, Seattle, WA 98103-8904, USA
| | - Meenakshisundaram Kandhavelu
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, Tampere 33720, Finland; BioMeditech and Tays Cancer Center, Tampere University Hospital, P.O. Box 553, 33101 Tampere, Finland.
| |
Collapse
|
32
|
All-Trans Retinoic Acid Attenuates Transmissible Gastroenteritis Virus-Induced Apoptosis in IPEC-J2 Cells via Inhibiting ROS-Mediated P38MAPK Signaling Pathway. Antioxidants (Basel) 2022; 11:antiox11020345. [PMID: 35204227 PMCID: PMC8868330 DOI: 10.3390/antiox11020345] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/29/2022] [Accepted: 01/31/2022] [Indexed: 02/06/2023] Open
Abstract
Transmissible gastroenteritis virus (TGEV) can cause diarrhea, dehydration, and high mortality in piglets, which is closely related to intestinal epithelial cell apoptosis caused by TGEV infection. All-trans retinoic acid (ATRA) is the active metabolite of vitamin A, which has antioxidant and anti-apoptotic properties. However, it is unknown whether ATRA can attenuate TGEV-induced IPEC-J2 cells apoptosis. Therefore, we investigated the protective effects of ATRA on TGEV-induced apoptosis of IPEC-J2 cells and explored the potential molecular mechanism. Our results indicated that TGEV infection caused IPEC-J2 cells damage and apoptosis. However, ATRA treatment attenuated TGEV-induced IPEC-J2 cells damage by upregulating the mRNA expressions of ZO-1, Occludin, and Mucin-1. ATRA treatment also attenuated TGEV-induced apoptosis in IPEC-J2 cells by downregulating the expression of Caspase-3, which is related to the inhibition of death receptor (Fas and Caspase-8) and mitochondrial (Bax, Bcl-2, and Caspase-9) pathways. Moreover, ATRA treatment prevented TGEV-induced ROS and MDA production and the upregulation of P38MAPK phosphorylation level, which is related to the increase in the activities of antioxidant enzymes (SOD, CAT, and T-AOC) and the mRNA abundance of antioxidant-related genes (GPX1, GPX2, SOD1, CAT, GCLC, and GCLM). In addition, treatment of TGEV-infected IPEC-J2 cells with the ROS inhibitors (NAC) significantly reduced the protein levels of p-P38MAPK, Fas, Bax, and Cleaved-caspase-3 and the percentage of apoptotic cells. Our results indicated that ATRA attenuated TGEV-induced apoptosis in IPEC-J2 cells via improving the antioxidant capacity, thereby inhibiting the cell damage. the mechanism of which is associated with the inhibition of ROS-mediated P38MAPK signaling pathway.
Collapse
|
33
|
Guo F, Zhang Y, Dong W, Guan Y, Shang D. Effect of hydrophobicity on distinct anticancer mechanism of antimicrobial peptide chensinin-1b and its lipoanalog PA-C1b in breast cancer cells. Int J Biochem Cell Biol 2022; 143:106156. [PMID: 34999227 DOI: 10.1016/j.biocel.2022.106156] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/13/2021] [Accepted: 01/05/2022] [Indexed: 12/21/2022]
Abstract
Chensinin-1b and its lipoanalogs demonstrate different anticancer activities against selected cancer cells, and the anticancer activity of PA-C1b is improved up to 3-fold compared with that of the parent peptide chensinin-1b. However, detailing the mechanism of action of these peptides is required to better understand the structure-function relationship. In this study, chensinin-1b and PA-C1b were selected as the representative peptides to investigate the mode of action in cancer cells. The results indicated that the boundary of the cell membrane was broken when the cells were treated with chensinin-1b, while that of cells treated with PA-C1b remained intact based on morphological changes. Apoptosis assays indicated that PA-C1b induced MCF-7 cancer cell apoptosis, while chensinin-1b mainly damaged the cell membrane. MCF-7 cancer cells treated with the peptides induced the loss of mitochondrial membrane potential, and cytochrome c was released from mitochondria, but PA-C1b enhanced ROS generation. Additionally, PA-C1b uptake occurred via an energy-dependent pathway and was inhibited by selected endocytosis inhibitors. Furthermore, treatment of MCF-7 cells with PA-C1b suppressed Bcl-2 mRNA levels and increased Bax mRNA levels, upregulated the expression of the proapoptotic protein Bax and downregulated the expression of the antiapoptotic protein Bcl-2. These results indicate that the anticancer mechanism of AMPs may be considerably affected by only a slight difference in the hydrophobicity of the two peptides; and such a study may facilitate the design of novel peptide-based anticancer agents.
Collapse
Affiliation(s)
- Feilu Guo
- School of Life Science, Liaoning Normal University, Dalian 116081, China
| | - Yao Zhang
- School of Life Science, Liaoning Normal University, Dalian 116081, China
| | - Weibing Dong
- School of Life Science, Liaoning Normal University, Dalian 116081, China; Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian 116081, China.
| | - Yue Guan
- School of Life Science, Liaoning Normal University, Dalian 116081, China; Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian 116081, China
| | - Dejing Shang
- School of Life Science, Liaoning Normal University, Dalian 116081, China; Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian 116081, China.
| |
Collapse
|
34
|
Zhang L, Li C, Fu L, Yu Z, Xu G, Zhou J, Shen M, Feng Z, Zhu H, Xie T, Zhou L, Zhou X. Protection of catalpol against triptolide-induced hepatotoxicity by inhibiting excessive autophagy via the PERK-ATF4-CHOP pathway. PeerJ 2022; 10:e12759. [PMID: 35036109 PMCID: PMC8742543 DOI: 10.7717/peerj.12759] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 12/16/2021] [Indexed: 01/07/2023] Open
Abstract
Catalpol significantly reduces triptolide-induced hepatotoxicity, which is closely related to autophagy. The aim of this study was to explore the unclear protective mechanism of catalpol against triptolide. The detoxification effect of catalpol on triptolide was investigated in HepaRG cell line. The detoxification effects were assessed by measuring cell viability, autophagy, and apoptosis, as well as the endoplasmic reticulum stress protein and mRNA expression levels. We found that 5-20 µg/L triptolide treatments increased the levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), and lactate dehydrogenase (LDH), as well as the expression of autophagy proteins including LC3 and Beclin1. The expression of P62 was downregulated and the production of autophagosomes was increased, as determined by transmission electron microscope and monodansylcadaverine staining. In contrast, 40 µg/L catalpol reversed these triptolide-induced changes in the liver function index, autophagy level, and apoptotic protein expression, including Cleaved-caspase3 and Cleaved-caspase9 by inhibiting excessive autophagy. Simultaneously, catalpol reversed endoplasmic reticulum stress, including the expression of PERK, which regulates autophagy. Moreover, we used the PERK inhibitor GSK2656157 to prove that the PERK-ATF4-CHOP pathway of the unfolded protein response is an important pathway that could induce autophagy. Catalpol inhibited excessive autophagy by suppressing the PERK pathway. Altogether, catalpol protects against triptolide-induced hepatotoxicity by inhibiting excessive autophagy via the PERK-ATF4-CHOP pathway. The results of this study are beneficial to clarify the detoxification mechanism of catalpol against triptolide-induced hepatotoxicity and to promote the application of triptolide.
Collapse
Affiliation(s)
- Linluo Zhang
- Department of First Clinical College, Nanjing University of Traditional Chinese Medicine, Nanjing City, Jiangsu, China
| | - Changqing Li
- Department of First Clinical College, Nanjing University of Traditional Chinese Medicine, Nanjing City, Jiangsu, China
| | - Ling Fu
- Department of First Clinical College, Nanjing University of Traditional Chinese Medicine, Nanjing City, Jiangsu, China,Department of Second Clinical College, Nanjing University of Traditional Chinese Medicine, Nanjing City, Jiangsu, China
| | - Zhichao Yu
- Department of First Clinical College, Nanjing University of Traditional Chinese Medicine, Nanjing City, Jiangsu, China
| | - Gengrui Xu
- Department of First Clinical College, Nanjing University of Traditional Chinese Medicine, Nanjing City, Jiangsu, China
| | - Jie Zhou
- Department of First Clinical College, Nanjing University of Traditional Chinese Medicine, Nanjing City, Jiangsu, China
| | - Meiyu Shen
- Department of Pharmacy, Nanjing University of Traditional Chinese Medicine, Nanjing City, Jiangsu, China
| | - Zhe Feng
- Department of First Clinical College, Nanjing University of Traditional Chinese Medicine, Nanjing City, Jiangsu, China
| | - Huaxu Zhu
- Department of Pharmacy, Nanjing University of Traditional Chinese Medicine, Nanjing City, Jiangsu, China
| | - Tong Xie
- Department of Pharmacy, Nanjing University of Traditional Chinese Medicine, Nanjing City, Jiangsu, China
| | - Lingling Zhou
- Department of Pharmacy, Nanjing University of Traditional Chinese Medicine, Nanjing City, Jiangsu, China
| | - Xueping Zhou
- Department of First Clinical College, Nanjing University of Traditional Chinese Medicine, Nanjing City, Jiangsu, China
| |
Collapse
|
35
|
Ma M, Cheng L, Wang L, Liang X, Yang L, Zhang A. Enhanced photodynamic therapy of TiO2/N-succinyl-chitosan composite for killing cancer cells. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e181116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Min Ma
- Shanxi Medical University, P. R. China
| | - Lu Cheng
- Shanxi Medical University, P. R. China
| | - Ling Wang
- Shanxi Medical University, P. R. China; Linfen Fourth People’s Hospital, P. R. China
| | | | | | | |
Collapse
|
36
|
Lang Y, Zhang H, Yu H, Li Y, Liu X, Li M. Long non-coding RNA myocardial infarction-associated transcript promotes 1-Methyl-4-phenylpyridinium ion-induced neuronal inflammation and oxidative stress in Parkinson's disease through regulating microRNA-221-3p/ transforming growth factor /nuclear factor E2-related factor 2 axis. Bioengineered 2021; 13:930-940. [PMID: 34967706 PMCID: PMC8805986 DOI: 10.1080/21655979.2021.2015527] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
This study attempted to evaluate the role of long non-coding RNA myocardial infarction-associated transcript (LncRNA MIAT) in Parkinson’s disease (PD). The mouse model was established through intraperitoneal injection with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), and in vitro model was induced by administrating cell with 1-Methyl-4-phenylpyridinium ion (MPP+). Rotarod test was conducted to evaluate the motor coordination of PD mice. In order to investigate the roles of LncRNA MIAT in neuronal inflammation and oxidative stress, MIAT shRNA (shMIAT) was transfected into MPP+-treated cells, and cell viability, cell apoptosis and oxidative stress response were evaluated. To evaluate the interactions between LncRNA MIAT and microRNA-221-3p (miR-221-3p)/TGF-β1/Nrf2, miR-221-3p mimic, miR-221-3p inhibitor, NC-inhibitor and transforming growth factor-β1 shRNA (shTGF-β1) were subsequently transfected into MPP+-treated cells. Dual-luciferase reporter gene assays were performed to determine the interaction of miR-221-3p with MIAT or TGFB receptor 1 (TGFBR1). The expressions of LncRNA MIAT, miR-221-3p, TGFBR1, transforming growth factor (TGF-β1) and nuclear factor E2-related factor 2 (Nrf2) were measured by quantitative reverse-transcription polymerase chain reaction (RT-qPCR) and immunoblotting. As a result, LncRNA MIAT was abundantly expressed in PD mice and cells, while downregulation of LncRNA MIAT promoted the survival of neurons, inhibited apoptosis and oxidative stress in neurons. LncRNA MIAT bound to miR-221-3p, and there was a negative correlation between miR-221-3p and LncRNA MIAT expression. In addition, miR-221-3p targeted TGFBR1 and suppressed TGF-β1 expression but increased Nrf2 expression. LncRNA MIAT promoted MPP+-induced neuronal injury in PD via regulating TGF-β1/Nrf2 axis through binding with miR-221-3p.
Collapse
Affiliation(s)
- Yue Lang
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian City, Liaoning Province, China
| | - Hui Zhang
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian City, Liaoning Province, China
| | - Haojia Yu
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian City, Liaoning Province, China
| | - Yu Li
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian City, Liaoning Province, China
| | - Xiao Liu
- Graduate School, Dalian Medical University, Dalian City, Liaoning Province, China
| | - Minjie Li
- Graduate School, Dalian Medical University, Dalian City, Liaoning Province, China
| |
Collapse
|
37
|
Amiri Rudbari H, Saadati A, Aryaeifar M, Blacque O, Cuevas-Vicario JV, Cabral R, Raposo LR, Fernandes AR. Platinum(II) and Copper(II) complexes of asymmetric halogen-substituted [NN'O] ligands: Synthesis, characterization, structural investigations and antiproliferative activity. Bioorg Chem 2021; 119:105556. [PMID: 34959175 DOI: 10.1016/j.bioorg.2021.105556] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/06/2021] [Accepted: 12/09/2021] [Indexed: 12/20/2022]
Abstract
In order to better understand the effect of structure, halogen substitution, metal ions and ligand flexibility on antiproliferative activity, eight Cu(II) complexes and eight Pt(II) complexes were obtained of 2,4-X1,X2-6-((pyridine-2-ylmethylamino)methyl)phenol and 2,4-X1,X2-6-((pyridine-2-ylmethylamino)ethyl)phenol (where X is Cl, Br, or I) ligands. The compounds were characterized with various techniques, such as FT-IR, NMR, elemental analysis and single-crystal X-ray diffraction (SCXRD). The X-ray structures showed that ligand acts as a bidentate and tridentate donor in Cu(II) and Pt(II) complexes, respectively. This difference in structures is due to the use or non-use of base in the preparation of complexes. Also, complexation of Cl2-H2L1 with CuCl2·2H2O gives two different types of structures: polymer (Cl2-H2L1-Cupolymer) and dimer (Cl2-H2L1-Cudimer), according to the crystal color. In addition, 1H NMR spectrum for platinum complexes display two set of signals that can be attributed to the presence of two isomers in solution. All complexes induced moderate to high reduction in A2780 and HCT116 cancer cell viability. However, only complexes bearing iodo- substituted in ligands exhibited significantly low cytotoxicity in normal fibroblasts when compared with cancer cell lines. The antiproliferative effect exhibited by I2-H2L2-Cu complex in A2780 cell line was due to induction of cell death mechanisms, namely by apoptosis and autophagy. I2-H2L2-Cu complex does not cause DNA cleavage but a slight delay in cell cycle was observed for the first 24 h of exposition. High cytotoxicity was related with the induction of intracellular ROS. This increase in intracellular ROS was not accompanied by destabilization of the mitochondrial membrane which is an indication that ROS are being triggered externally by I2-H2L2-Cu complex and in agreement with an extrinsic apoptosis activation. I2-H2L2-Cu complex has a pro-angiogenic effect, increasing the vascularization of the CAM in chicken embryos. This is also a very important characteristic in cancer treatment since the increased vascularization in tumors might facilitate the delivery of therapeutic drugs. Taken together, these results support the potential therapeutic of the I2-H2L2-Cu complex.
Collapse
Affiliation(s)
- Hadi Amiri Rudbari
- Department of Chemistry, University of Isfahan, Isfahan 81746-73441, Iran.
| | - Arezoo Saadati
- Department of Chemistry, University of Isfahan, Isfahan 81746-73441, Iran
| | - Mahnaz Aryaeifar
- Department of Chemistry, University of Isfahan, Isfahan 81746-73441, Iran
| | - Olivier Blacque
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Jose V Cuevas-Vicario
- Department of Chemistry, Universidad de Burgos, Pza. Misael Bañuelos s/n, E-09001 Burgos, Spain
| | - Rui Cabral
- UCIBIO, Departamento Ciências da Vida, NOVA School of Science and Technology, Campus Caparica, 2829-516 Caparica, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Luis R Raposo
- UCIBIO, Departamento Ciências da Vida, NOVA School of Science and Technology, Campus Caparica, 2829-516 Caparica, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Alexandra R Fernandes
- UCIBIO, Departamento Ciências da Vida, NOVA School of Science and Technology, Campus Caparica, 2829-516 Caparica, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal.
| |
Collapse
|
38
|
Liu H, Wu L, Cui J, Wang D. Anticancer Activity of Zn(II) Coordination Polymer Against Cervical Cancer Cells via miR-5571/MDM2. J CLUST SCI 2021. [DOI: 10.1007/s10876-021-02201-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
39
|
Wu A, Zhu Y, Han B, Peng J, Deng X, Chen W, Du J, Ou Y, Peng X, Yu X. Delphinidin induces cell cycle arrest and apoptosis in HER-2 positive breast cancer cell lines by regulating the NF-κB and MAPK signaling pathways. Oncol Lett 2021; 22:832. [PMID: 34712357 PMCID: PMC8548810 DOI: 10.3892/ol.2021.13093] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 06/25/2021] [Indexed: 12/23/2022] Open
Abstract
Delphinidin is an anthocyanidin monomer, commonly found in vegetables and fruits, and has demonstrated antitumor effects in the HER-2-positive MDA-MB-453 breast cancer cell line, with low cytotoxicity on normal breast cells. However, the direct functional mechanisms underlying the effect of delphinidin on HER-2-positive breast cancer cells has not been fully characterized. In the present study, it was found that delphinidin could induce G2/M phase cell cycle arrest by inhibiting the protein expression level of cyclin B1 and Cdk1 in HER-2-positive breast cancer cell lines. In addition, delphinidin promoted the mitochondrial apoptosis pathway by inhibiting the ERK and NF-κB signaling pathway and activating the JNK signaling pathway. Therefore, delphinidin markedly suppressed the viability of the HER-2-positive breast cancer cell lines by modulating the cell cycle and inducing apoptosis. Overall, the findings from the present study demonstrated that delphinidin treatment could induce the mitochondrial apoptosis pathway in human HER-2-positive breast cancer cell lines, providing an experimental basis for the prevention and treatment of HER-2-positive breast cancer by flavonoids.
Collapse
Affiliation(s)
- Ailin Wu
- Basic Medical School, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China.,Ministry of Science and Technology, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan 610051, P.R. China
| | - Yanfeng Zhu
- Graduate School, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Bin Han
- School of Public Health, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Jiayuan Peng
- Basic Medical School, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Xiaomin Deng
- Basic Medical School, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Wei Chen
- Basic Medical School, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Jingchang Du
- School of Public Health, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Yu Ou
- Division of Planning and Finance, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Xiaoli Peng
- School of Public Health, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Xiaoping Yu
- Graduate School, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| |
Collapse
|
40
|
Manganese(I) tricarbonyl complexes as potential anticancer agents. J Biol Inorg Chem 2021; 27:49-64. [PMID: 34713347 DOI: 10.1007/s00775-021-01910-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 09/15/2021] [Indexed: 02/06/2023]
Abstract
The antiproliferative activity of [Mn(CO)3(N^N)Br] (N^N = phendione 1, bipy 3) and of the two newly synthesized Mn complexes [Mn(CO)3(acridine)(phendione)]OTf (2) and [Mn(CO)3(di-triazole)Br] (4) has been evaluated by MTS against three tumor cell lines A2780 (ovarian carcinoma), HCT116 (colorectal carcinoma), HCT116doxR (colorectal carcinoma resistant to doxorubicin), and in human dermal fibroblasts. The antiproliferative assay showed a dose-dependent effect higher in complex 1 and 2 with a selectivity toward ovarian carcinoma cell line 21 times higher than in human fibroblasts. Exposure of A2780 cells to IC50 concentrations of complex 1 and 2 led to an increase of reactive oxygen species that led to the activation of cell death mechanisms, namely via intrinsic apoptosis for 2 and autophagy and extrinsic apoptosis for 1. Both complexes do not target DNA or interfere with cell cycle progression but are able to potentiate cell migration and neovascularization (for 2) an indicative that their application might be directed for initial tumor stages to avoid tumor invasion and metastization and opening a new avenue for complex 2 application in regenerative medicine. Interestingly, both complexes do not show toxicity in both in vivo models (CAM and zebrafish).
Collapse
|
41
|
Wei G, Chen J, Jing Z, Li Y, Li Z, Zheng W, Sun X, Zhao W, Zhang Z, Wang X, Han H, Li C, Zhang Y, Ma P. Glucose transporter 1 (GLUT1)-targeting and hypoxia-activated mitochondria-specific chemo-thermal therapy via a glycosylated poly(amido amine)/celastrol (PAMAM/Cel) complex. J Colloid Interface Sci 2021; 608:1355-1365. [PMID: 34742058 DOI: 10.1016/j.jcis.2021.10.129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/21/2021] [Accepted: 10/22/2021] [Indexed: 12/16/2022]
Abstract
Mitochondria are appealing targets in cancer therapy for providing a suitable microenvironment and energy supply. Herein, we constructed a glycosylated poly(amido amine)/celastrol (PAMAM/Cel) complex for hypoxia-activated mitochondria-specific drug delivery and chemothermal therapy to inhibit tumor growth and metastasis. The complex was characterized by high photothermal conversion efficiency, hypoxia-sensitive polyethylene glycol (PEG) outer layer detachment, and alkaline-sensitive drug release. The complex showed specific cellular uptake in glucose transporter 1 (GLUT1)-overexpressing tumor cells and mitochondrial accumulation in a hypoxic environment. Combined with near-infrared (NIR) laser irradiation, the complex exhibited higher cytotoxicity, apoptosis induction, and metastasis inhibition rates due to the synergistic chemothermal effect. Similarly, the complex also targeted tumors and accumulated in mitochondria in tumor-bearing nude mice, resulting in superior inhibitory effects on tumor growth and metastasis as well as low systematic toxicity. Further mechanistic studies discovered that the complex impaired the mitochondrial membrane, reduced adenosine triphosphate (ATP) content, and regulated metastasis-related protein expression. Thus, the present study provides a promising nanomedicine for tumor therapy.
Collapse
Affiliation(s)
- Guijie Wei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jianhua Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Ziqi Jing
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yanyi Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhihui Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Zheng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiurui Sun
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Wenwen Zhao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhe Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xue Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Hongcui Han
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Chu Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yujie Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.
| | - Pengkai Ma
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
42
|
Lee HS, Park BS, Kang HM, Kim JH, Shin SH, Kim IR. Role of Luteolin-Induced Apoptosis and Autophagy in Human Glioblastoma Cell Lines. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:879. [PMID: 34577802 PMCID: PMC8470580 DOI: 10.3390/medicina57090879] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/25/2021] [Accepted: 08/25/2021] [Indexed: 01/07/2023]
Abstract
Background and Objectives: Malignant glioblastoma (GBM) is caused by abnormal proliferation of glial cells, which are found in the brain. The therapeutic effects of surgical treatment, radiation therapy, and chemo-therapy against GBM are relatively poor compared with their effects against other tumors. Luteolin is abundant in peanut shells and is also found in herbs and other plants, such as thyme, green pepper, and celery. Luteolin is known to be effective against obesity and metabolic syndrome. The anti-inflammatory, and anti-cancer activities of luteolin have been investigated. Most studies have focused on the antioxidant and anti-inflammatory effects of luteolin, which is a natural flavonoid. However, the association between the induction of apoptosis by luteolin in GBM and autophagy has not yet been investigated. This study thus aimed to confirm the occurrence of luteolin-induced apoptosis and autophagy in GBM cells and to assess their relationship. Materials and Methods: A172 and U-373MG glioblastoma cell lines were used for this experiment. We confirmed the apoptosis effect of Luteolin on GBM cells using methods such as 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, immunofluorescence, Flow cytometry (FACS) western blot, and real-time quantitative PCR (qPCR). Results: In the luteolin-treated A172 and U-373MG cells, cell viability decreased in a concentration- and time-dependent manner. In addition, in A172 and U-373MG cells treated with luteolin at concentrations greater than 100 μM, nuclear fragmentation, which is a typical morphological change characterizing apoptosis, as well as fragmentation of caspase-3 and Poly (ADP-ribose) polymerase (PARP), which are apoptosis-related factors, were observed. Autophagy was induced after treatment with at least 50 μM luteolin. Inhibition of autophagy using 3MA allowed for a low concentration of luteolin to more effectively induce apoptosis in A172 and U-373MG cells. Conclusions: Results showed that luteolin induces apoptosis and autophagy and that the luteolin-induced autophagy promotes cell survival. Therefore, an appropriate combination therapy involving luteolin and an autophagy inhibitor is expected to improve the prognosis of GBM treatment.
Collapse
Affiliation(s)
- Hye-Sung Lee
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Pusan National University, Yangsan-si 50612, Korea; (H.-S.L.); (S.-H.S.)
| | - Bong-Soo Park
- Department of Oral Anatomy, School of Dentistry, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si 50612, Korea; (B.-S.P.); (H.-M.K.)
- Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan-si 50612, Korea
| | - Hae-Mi Kang
- Department of Oral Anatomy, School of Dentistry, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si 50612, Korea; (B.-S.P.); (H.-M.K.)
| | - Jung-Han Kim
- Medical Center, Department of Oral and Maxillofacial Surgery, Dong-A University, 26, Daesingongwon-ro, Seo-gu, Busan 49201, Korea;
| | - Sang-Hun Shin
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Pusan National University, Yangsan-si 50612, Korea; (H.-S.L.); (S.-H.S.)
| | - In-Ryoung Kim
- Department of Oral Anatomy, School of Dentistry, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si 50612, Korea; (B.-S.P.); (H.-M.K.)
- Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan-si 50612, Korea
| |
Collapse
|
43
|
Li Y, Wen C, Zhong J, Ling J, Jiang Q. Enterococcus faecalis OG1RF induces apoptosis in MG63 cells via caspase-3/-8/-9 without activation of caspase-1/GSDMD. Oral Dis 2021; 28:2026-2035. [PMID: 34370363 DOI: 10.1111/odi.13996] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 05/05/2021] [Accepted: 08/01/2021] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Regulated cell death is key in the pathogenesis of persistent apical periodontitis. Here, we investigated the mechanisms of regulated cell death in osteoblast-like MG63 cells infected with Enterococcus faecalis OG1RF. MATERIALS AND METHODS MG63 cells were infected with live E. faecalis OG1RF at the indicated multiplicity of infection for the indicated infection time. We evaluated the cells by flow cytometry, terminal deoxynucleotidyl transferase dUTP nick end labelling assay and lactate dehydrogenase release analysis; measured the activity of caspase-1/-3/-8/-9 and the release of interleukin-1β; and determined the expression of apoptosis-associated proteins and gasdermin D by apoptosis antibody array and Western blotting. RESULTS Enterococcus faecalis OG1RF reduced the mitochondrial membrane potential of the infected cells, increased the percentage of apoptotic and terminal deoxynucleotidyl transferase dUTP nick end labelling-positive cells, and enhanced lactate dehydrogenase release. The expression of caspase-3 and survivin and the activity of caspase-3/-8/-9 were upregulated, while the expression of death receptor 6 was downregulated. The activity of caspase-1/gasdermin D and the release of interleukin-1β remained unaltered. CONCLUSION Enterococcus faecalis OG1RF induced both intrinsic and extrinsic MG63 cell apoptosis via caspase-3/-8/-9 activation but did not activate the pyroptotic pathway regulated by caspase-1/gasdermin D.
Collapse
Affiliation(s)
- Yang Li
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Cheng Wen
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Jialin Zhong
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Junqi Ling
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China.,Guangdong Province Key Laboratory of Stomatology, Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Qianzhou Jiang
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| |
Collapse
|
44
|
Das A, Haque I, Ray P, Ghosh A, Dutta D, Quadir M, De A, Gunewardena S, Chatterjee I, Banerjee S, Weir S, Banerjee SK. CCN5 activation by free or encapsulated EGCG is required to render triple-negative breast cancer cell viability and tumor progression. Pharmacol Res Perspect 2021; 9:e00753. [PMID: 33745223 PMCID: PMC7981588 DOI: 10.1002/prp2.753] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/13/2021] [Accepted: 02/15/2021] [Indexed: 12/11/2022] Open
Abstract
Epigallocatechin-3-gallate (EGCG) has been considered an anticancer agent despite conflicting and discrepant bioavailability views. EGCG impairs the viability and self-renewal capacity of triple-negative breast cancer (TNBC) cells and makes them sensitive to estrogen via activating ER-α. Surprisingly, the mechanism of EGCG's action on TNBC cells remains unclear. CCN5/WISP-2 is a gatekeeper gene that regulates viability, ER-α, and stemness in TNBC and other types of cancers. This study aimed to investigate whether EGCG (free or encapsulated in nanoparticles) interacts with the CCN5 protein by emphasizing its bioavailability and enhancing its anticancer effect. We demonstrate that EGCG activates CCN5 to inhibit in vitro cell viability through apoptosis, the sphere-forming ability via reversing TNBC cells' stemness, and suppressing tumor growth in vivo. Moreover, we found EGCG-loaded nanoparticles to be functionally more active and superior in their tumor-suppressing ability than free-EGCG. Together, these studies identify EGCG (free or encapsulated) as a novel activator of CCN5 in TNBC cells and hold promise as a future therapeutic option for TNBC with upregulated CCN5 expression.
Collapse
Affiliation(s)
- Amlan Das
- Cancer Research UnitVA Medical CenterKansas CityMOUSA
- Present address:
National Institute of Biomedical GenomicsKalyaniWest BengalIndia
| | - Inamul Haque
- Cancer Research UnitVA Medical CenterKansas CityMOUSA
| | - Priyanka Ray
- Department of Chemical Biochemical Environmental Engineering (CBEEUniversity of MarylandBaltimoreMDUSA
| | - Arnab Ghosh
- Cancer Research UnitVA Medical CenterKansas CityMOUSA
- Department of Pathology and Laboratory MedicineUniversity of Kansas Medical CenterKansas CityKSUSA
| | - Debasmita Dutta
- Department of Coatings and Polymeric MaterialsNorth Dakota State UniversityFargoNDUSA
| | - Mohiuddin Quadir
- Department of Coatings and Polymeric MaterialsNorth Dakota State UniversityFargoNDUSA
| | - Archana De
- Cancer Research UnitVA Medical CenterKansas CityMOUSA
| | - Sumedha Gunewardena
- Department of Molecular and Integrative PhysiologyUniversity of Kansas Medical CenterKansas CityKSUSA
| | - Indranil Chatterjee
- Cancer Research UnitVA Medical CenterKansas CityMOUSA
- Present address:
Department of Life SciencesCentral University of Tamil NaduThiruvarurIndia
| | - Snigdha Banerjee
- Cancer Research UnitVA Medical CenterKansas CityMOUSA
- Department of Pathology and Laboratory MedicineUniversity of Kansas Medical CenterKansas CityKSUSA
| | - Scott Weir
- Department of PharmacologyToxicology and TherapeuticsUniversity of Kansas Medical CenterKansas CityKSUSA
| | - Sushanta K. Banerjee
- Cancer Research UnitVA Medical CenterKansas CityMOUSA
- Department of Pathology and Laboratory MedicineUniversity of Kansas Medical CenterKansas CityKSUSA
- Lead contact, SKB, Cancer Research UnitKansas CityMOUSA
| |
Collapse
|
45
|
Yu P, Zhang X, Liu N, Tang L, Peng C, Chen X. Pyroptosis: mechanisms and diseases. Signal Transduct Target Ther 2021; 6:128. [PMID: 33776057 PMCID: PMC8005494 DOI: 10.1038/s41392-021-00507-5] [Citation(s) in RCA: 1281] [Impact Index Per Article: 320.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 01/14/2021] [Accepted: 01/20/2021] [Indexed: 02/08/2023] Open
Abstract
Currently, pyroptosis has received more and more attention because of its association with innate immunity and disease. The research scope of pyroptosis has expanded with the discovery of the gasdermin family. A great deal of evidence shows that pyroptosis can affect the development of tumors. The relationship between pyroptosis and tumors is diverse in different tissues and genetic backgrounds. In this review, we provide basic knowledge of pyroptosis, explain the relationship between pyroptosis and tumors, and focus on the significance of pyroptosis in tumor treatment. In addition, we further summarize the possibility of pyroptosis as a potential tumor treatment strategy and describe the side effects of radiotherapy and chemotherapy caused by pyroptosis. In brief, pyroptosis is a double-edged sword for tumors. The rational use of this dual effect will help us further explore the formation and development of tumors, and provide ideas for patients to develop new drugs based on pyroptosis.
Collapse
Affiliation(s)
- Pian Yu
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, Hunan, China
| | - Xu Zhang
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, Hunan, China
| | - Nian Liu
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, Hunan, China
| | - Ling Tang
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, Hunan, China
| | - Cong Peng
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China.
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China.
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, Hunan, China.
| | - Xiang Chen
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China.
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China.
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, Hunan, China.
| |
Collapse
|
46
|
Chen S, Zhang RF, Guo Q, Nie JJ, Li QL, Cheng S, Ma CL. Four triorganotin(IV) esters based on 3,5-bifluorobenzenetelluronic acid: Syntheses, structures, in vitro cytostatic activity and BSA-binding assessment. INORG CHEM COMMUN 2021. [DOI: 10.1016/j.inoche.2020.108404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
47
|
Sequeira D, Baptista PV, Valente R, Piedade MFM, Garcia MH, Morais TS, Fernandes AR. Cu(I) complexes as new antiproliferative agents against sensitive and doxorubicin resistant colorectal cancer cells: synthesis, characterization, and mechanisms of action. Dalton Trans 2021; 50:1845-1865. [PMID: 33470993 DOI: 10.1039/d0dt03566a] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cancer is one of the worst health issues worldwide, representing the second leading cause of death. Current chemotherapeutic drugs face some challenges like the acquired resistance of the tumoral cells and low specificity leading to unwanted side effects. There is an urgent need to develop new compounds that may target resistant cells. The synthesis and characterization of two Cu(i) complexes of general formula [Cu(PP)(LL)][BF4], where PP is a phosphane ligand (triphenylphosphine or 1,2-bis(diphenylphosphano) ethane) and LL = is a heteroaromatic bidentate ligand (4,4'-dimethyl-2,2'-bipyridine and 6,3-(2-pyridyl)-5,6-diphenyl-1,2,4-triazine). The new compounds were fully characterized by spectroscopic techniques (NMR, FTIR and UV-vis.), elemental analysis (C, H, N and S) and two structures were determined by single X-ray diffraction studies. The antiproliferative potential of the new Cu(i) complexes were studied in tumor (breast adenocarcinoma, ovarian carcinoma and in colorectal carcinoma sensitive and resistant to doxorubicin) and normal (fibroblasts) cell lines. Complexes 1-4 did not show any antiproliferative potential. Amongst the complexes 5-8, complex 8 shows high cytotoxic potential against colorectal cancer sensitive and resistant to doxorubicin and low cytotoxicity towards healthy cells. We show that complexes 5-8 can cleave pDNA and, in particular, the in vitro pDNA cleavage is due to an oxidative mechanism. This oxidative mechanism corroborates the induction of reactive oxygen species (ROS), that triggers HCT116 cell death via apoptosis, as proved by the increased expression of BAX protein relative to BCL-2 protein and the depolarization of mitochondrial membrane potential, and via autophagy. Additionally, complex 8 can block the cell cycle in the G1 phase, also exhibiting a cytostatic potential. Proteomic analysis confirmed the apoptotic, autophagic and cytostatic potential of complex 8, as well as its ability to produce ROS and cause DNA damage. The interference of the complex in folding and protein synthesis and its ability to cause post-translational modifications was also verified. Finally, it was observed that the complex causes a reduction in cellular metabolism. The results herein demonstrated the potential of Cu(i) complexes in targeting doxorubicin sensitive and resistant cells which is positive and must be further explored using in vivo animal models.
Collapse
Affiliation(s)
- Diogo Sequeira
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Portugal.
| | - Pedro V Baptista
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Portugal.
| | - Ruben Valente
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Portugal.
| | - M Fátima M Piedade
- DQB-FCUL, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Portugal. and CQE@IST, Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - M Helena Garcia
- DQB-FCUL, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Portugal. and CQE@FCUL, Centro de Química Estrutural, Faculdade de Ciências, Universidade de Lisboa, Portugal
| | - Tânia S Morais
- DQB-FCUL, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Portugal. and CQE@FCUL, Centro de Química Estrutural, Faculdade de Ciências, Universidade de Lisboa, Portugal
| | - Alexandra R Fernandes
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Portugal.
| |
Collapse
|
48
|
Mohan H, Ramalingam V, Lim JM, Lee SW, Kim J, Lee JH, Park YJ, Seralathan KK, Oh BT. E-waste based graphene oxide/V2O5/Pt ternary composite: Enhanced visible light driven photocatalyst for anti-microbial and anti-cancer activity. Colloids Surf A Physicochem Eng Asp 2020. [DOI: 10.1016/j.colsurfa.2020.125469] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
49
|
Xu Z, Zhang Y, Cao Y. The Roles of Apoptosis in Swine Response to Viral Infection and Pathogenesis of Swine Enteropathogenic Coronaviruses. Front Vet Sci 2020; 7:572425. [PMID: 33324698 PMCID: PMC7725767 DOI: 10.3389/fvets.2020.572425] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 10/28/2020] [Indexed: 01/18/2023] Open
Abstract
Apoptosis is a tightly regulated mechanism of cell death that plays important roles in various biological processes including biological evolution, multiple system development, anticancer, and viral infections. Swine enteropathogenic coronaviruses invade and damage villous epithelial cells of the small intestine causing severe diarrhea with high mortality rate in suckling piglets. Transmissible gastroenteritis virus (TGEV), Porcine epidemic diarrhea virus (PEDV), Porcine deltacoronavirus (PDCoV), and Swine acute diarrhea syndrome coronavirus (SADS-CoV) are on the top list of commonly-seen swine coronaviruses with a feature of diarrhea, resulting in significant economic losses to the swine industry worldwide. Apoptosis has been shown to be involved in the pathogenesis process of animal virus infectious diseases. Understanding the roles of apoptosis in host responses against swine enteropathogenic coronaviruses infection contribute to disease prevention and control. Here we summarize the recent findings that focus on the apoptosis during swine coronaviruses infection, in particular, TGEV, PEDV, PDCoV, and SADS-CoV.
Collapse
Affiliation(s)
- Zhichao Xu
- State Key Laboratory of Biocontrol, School of Life Science, Sun Yat-sen University, Guangzhou, China
| | - Yun Zhang
- State Key Laboratory of Biocontrol, School of Life Science, Sun Yat-sen University, Guangzhou, China
| | - Yongchang Cao
- State Key Laboratory of Biocontrol, School of Life Science, Sun Yat-sen University, Guangzhou, China
- Higher Education Mega Center, School of Life Science, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
50
|
Yuan L, van der Mei HC, Busscher HJ, Peterson BW. Two-Stage Interpretation of Changes in TEER of Intestinal Epithelial Layers Protected by Adhering Bifidobacteria During E. coli Challenges. Front Microbiol 2020; 11:599555. [PMID: 33329490 PMCID: PMC7710611 DOI: 10.3389/fmicb.2020.599555] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 10/26/2020] [Indexed: 12/19/2022] Open
Abstract
Mechanisms of gastrointestinal protection by probiotic bacteria against infection involve amongst others, modulation of intestinal epithelial barrier function. Trans-epithelial electrical resistance (TEER) is widely used to evaluate cellular barrier functions. Here, we developed a two-stage interpretative model of the time-dependence of the TEER of epithelial layers grown in a transwell during Escherichia coli challenges in the absence or presence of adhering bifidobacteria. E. coli adhesion in absence or presence of adhering bifidobacteria was enumerated using selective plating. After 4-8 h, E. coli challenges increased TEER to a maximum due to bacterial adhesion and increased expression of a tight-junction protein [zonula occludens-1 (ZO-1)], concurrent with a less dense layer structure, that is indicative of mild epithelial layer damage. Before the occurrence of a TEER-maximum, decreases in electrical conductance (i.e., the reciprocal TEER) did not relate with para-cellular dextran-permeability, but after occurrence of a TEER-maximum, dextran-permeability and conductance increased linearly, indicative of more severe epithelial layer damage. Within 24 h after the occurrence of a TEER maximum, TEER decreased to below the level of unchallenged epithelial layers demonstrating microscopically observable holes and apoptosis. Under probiotic protection by adhering bifidobacteria, TEER-maxima were delayed or decreased in magnitude due to later transition from mild to severe damage, but similar linear relations between conductance and dextran permeability were observed as in absence of adhering bifidobacteria. Based on the time-dependence of the TEER and the relation between conductance and dextran-permeability, it is proposed that bacterial adhesion to epithelial layers first causes mild damage, followed by more severe damage after the occurrence of a TEER-maximum. The mild damage caused by E. coli prior to the occurrence of TEER maxima was reversible upon antibiotic treatment, but the severe damage after occurrence of TEER maxima could not be reverted by antibiotic treatment. Thus, single-time TEER is interpretable in two ways, depending whether increasing to or decreasing from its maximum. Adhering bifidobacteria elongate the time-window available for antibiotic treatment to repair initial pathogen damage to intestinal epithelial layers.
Collapse
Affiliation(s)
| | | | | | - Brandon W. Peterson
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|