1
|
Zhang Z, Chu J, Hu H, Sun H, Zhao X, Du H, Yang M. Enhanced Room-Temperature NO 2 Sensing through Deep Functional Group Hybridization in Nitrogen-Doped Monolayer Ti 3C 2T x. ACS Sens 2024; 9:4134-4142. [PMID: 39096509 DOI: 10.1021/acssensors.4c01081] [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] [Indexed: 08/05/2024]
Abstract
Nitrogen dioxide (NO2) is a significant environmental and human health hazard. Current NO2 sensors often lack sensitivity and selectivity under ambient conditions. This study investigates ammonia pyrolysis modification of monolayer Ti3C2Tx MXene to enhance NO2 detection at room temperature. Nitrogen-doped Ti3C2Tx demonstrates a substantial improvement in sensitivity, with a response of 8.87% to 50 ppm of NO2 compared to 0.65% for the original sensor, representing a 13.8-fold increase. The nitrogen-doped sensor also exhibits superior selectivity and linearity for NO2 under ambient conditions. Theoretical analysis shows that nitrogen incorporation promotes enhanced interaction between Ti3C2Tx and its surface oxygen-containing functional groups through electronic hybridization, resulting in improved adsorption energy (1.80 |eV|) and electron transfer efficiency (0.67 |e|) for NO2, thereby enhancing its gas-sensing performance. This study highlights the potential of ammonia pyrolysis-treated Ti3C2Tx MXene for advancing NO2 sensor technologies with heightened performance at room temperature.
Collapse
Affiliation(s)
- Zhaorui Zhang
- State Key Laboratory of High-performance Precision Manufacturing, Dalian University of Technology, Dalian 116024, China
- Ningbo Research Institute of Dalian University of Technology, Ningbo 315032, China
- School of Environmental Science and Technology, Dalian University of Technology, Dalian 116024, China
| | - Jinkui Chu
- State Key Laboratory of High-performance Precision Manufacturing, Dalian University of Technology, Dalian 116024, China
- Ningbo Research Institute of Dalian University of Technology, Ningbo 315032, China
| | - Huashuai Hu
- School of Environmental Science and Technology, Dalian University of Technology, Dalian 116024, China
| | - Haoming Sun
- School of Environmental Science and Technology, Dalian University of Technology, Dalian 116024, China
| | - Xuefei Zhao
- School of Environmental Science and Technology, Dalian University of Technology, Dalian 116024, China
| | - Haiying Du
- College of Mechanical and Electronic Engineering, Dalian Minzu University, Dalian 116600, China
| | - Minghui Yang
- School of Environmental Science and Technology, Dalian University of Technology, Dalian 116024, China
| |
Collapse
|
2
|
Li CC, Tsai BCK, Annseles Rajula S, Hsu CH, Chen MC, Kuo CH, Yeh CM, Hsieh DJY, Kuo WW, Huang CY. Tannic Acid Impedes the Proliferation of Bladder Cancer Cells by Elevating Mitochondrial Pathways of Apoptosis. Cell Biochem Biophys 2024; 82:1325-1333. [PMID: 38809348 DOI: 10.1007/s12013-024-01286-w] [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] [Accepted: 04/17/2024] [Indexed: 05/30/2024]
Abstract
Bladder cancer stands as a prevailing neoplasm among men globally, distinguished for its pronounced malignancy attributed to invasiveness and metastatic proclivity. Tannic acid (TA), an organic compound in many plants, has garnered recent attention for its discernible anti-mutagenic attributes. This investigation endeavored to scrutinize the repercussions of TA on grade II bladder cancer, with a concerted focus on unraveling its anti-cancer mechanisms. The cytotoxic effects of TA on grade II bladder cancer cells were investigated using multiple techniques, including MTT assay, flow cytometry, TUNEL assay, and western blot. Our findings revealed that elevated concentrations of TA induced cytotoxic effects in grade II bladder cancer cells. Both flow cytometry and the TUNEL assay substantiated the dose-dependent capacity of TA to prompt apoptosis. Western blot analysis corroborated that TA treatment in bladder cancer cells resulted in the upregulation of cleaved caspase-3 expression and PARP. Furthermore, heightened TA dosage elicited an augmentation in the expression of pro-apoptotic proteins, namely Bax and Bak, alongside a reduction in the expression of the anti-apoptotic protein Bcl-2 within bladder cancer cells. This study confirms TA as a potential anticancer agent, demonstrably diminishing the viability of bladder cancer cells. TA exerts cytotoxicity through the activation of mitochondrial apoptotic pathways. Specifically, TA initiates the cleavage of PARP and caspase-3, concurrently augmenting the expression of pro-apoptotic proteins to facilitate apoptosis. Collectively, the present study indicates that TA effectively impedes the proliferation of bladder cancer cells by instigating apoptosis through the intrinsic mitochondrial pathway.
Collapse
Affiliation(s)
- Chi-Cheng Li
- Center of Stem Cell and Precision Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Hematology and Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Bruce Chi-Kang Tsai
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Selvaraj Annseles Rajula
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Chiung-Hung Hsu
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Ming-Cheng Chen
- Division of Colorectal Surgery, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
- Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chia-Hua Kuo
- Department of Sports Sciences, University of Taipei, Taipei, Taiwan
- Laboratory of Exercise Biochemistry, University of Taipei, Tianmu Campus, Taipei, Taiwan
- Department of Kinesiology and Health Science, College of William and Mary, Williamsburg, VA, USA
- School of Physical Education and Sports Science, Soochow University, Suzhou, China
| | - Chung-Min Yeh
- Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan
- Department of Medical Technology, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli, Taiwan
- Department of Surgical Pathology, Changhua Christian Hospital, Changhua, Taiwan
| | - Dennis Jine-Yuan Hsieh
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan.
- Ph.D. Program for Biotechnology Industry, China Medical University, Taichung, Taiwan.
- School of Pharmacy, China Medical University, Taichung, Taiwan.
| | - Chih-Yang Huang
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan.
- Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan.
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.
| |
Collapse
|
3
|
Luo L, Zhang W, You S, Cui X, Tu H, Yi Q, Wu J, Liu O. The role of epithelial cells in fibrosis: Mechanisms and treatment. Pharmacol Res 2024; 202:107144. [PMID: 38484858 DOI: 10.1016/j.phrs.2024.107144] [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: 11/02/2023] [Revised: 02/19/2024] [Accepted: 03/12/2024] [Indexed: 03/19/2024]
Abstract
Fibrosis is a pathological process that affects multiple organs and is considered one of the major causes of morbidity and mortality in multiple diseases, resulting in an enormous disease burden. Current studies have focused on fibroblasts and myofibroblasts, which directly lead to imbalance in generation and degradation of extracellular matrix (ECM). In recent years, an increasing number of studies have focused on the role of epithelial cells in fibrosis. In some cases, epithelial cells are first exposed to external physicochemical stimuli that may directly drive collagen accumulation in the mesenchyme. In other cases, the source of stimulation is mainly immune cells and some cytokines, and epithelial cells are similarly altered in the process. In this review, we will focus on the multiple dynamic alterations involved in epithelial cells after injury and during fibrogenesis, discuss the association among them, and summarize some therapies targeting changed epithelial cells. Especially, epithelial mesenchymal transition (EMT) is the key central step, which is closely linked to other biological behaviors. Meanwhile, we think studies on disruption of epithelial barrier, epithelial cell death and altered basal stem cell populations and stemness in fibrosis are not appreciated. We believe that therapies targeted epithelial cells can prevent the progress of fibrosis, but not reverse it. The epithelial cell targeting therapies will provide a wonderful preventive and delaying action.
Collapse
Affiliation(s)
- Liuyi Luo
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Wei Zhang
- Department of Oral Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Siyao You
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Xinyan Cui
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Hua Tu
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Qiao Yi
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Jianjun Wu
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China.
| | - Ousheng Liu
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China.
| |
Collapse
|
4
|
Wu W, Wang Z, Zhang H, Zhang X, Tian H. circGRHPR inhibits aberrant epithelial-mesenchymal transformation progression of lung epithelial cells associated with idiopathic pulmonary fibrosis. Cell Biol Toxicol 2024; 40:7. [PMID: 38267743 PMCID: PMC10808371 DOI: 10.1007/s10565-024-09839-8] [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: 09/09/2023] [Accepted: 12/05/2023] [Indexed: 01/26/2024]
Abstract
Air pollution has greatly increased the risk of idiopathic pulmonary fibrosis (IPF). Circular RNAs (circRNAs) have been found to play a significant role in the advancement of IPF, but there is limited evidence of correlation between circRNAs and lung epithelial cells (LECs) in IPF. This research aimed to explore the influence of circRNAs on the regulation of EMT progression in LECs, with the objective of elucidating its mechanism and establishing its association with IPF. Our results suggested that the downregulation of circGRHPR in peripheral blood of clinical cases was associated with the diagnosis of IPF. Meanwhile, we found that circGRHPR was downregulated in transforming growth factor-beta1 (TGF-β1)-induced A549 and Beas-2b cells. It is a valid model to study the abnormal EMT progression of IPF-associated LECs in vitro. The overexpression of circGRHPR inhibited the abnormal EMT progression of TGF-β1-induced LECs. Furthermore, as the sponge of miR-665, circGRHPR released the expression of E3 ubiquitin-protein ligase NEDD4-like (NEDD4L), thus promoting its downstream transforming growth factor beta receptor 2 (TGFBR2) ubiquitination. It is helpful to reduce the response of LECs to TGF-β1 signaling. In summary, circGRHPR/miR-665/NEDD4L axis inhibited the abnormal EMT progression of TGF-β1-induced LECs by promoting TGFBR2 ubiquitination, which provides new ideas and potential targets for the treatment of IPF.
Collapse
Affiliation(s)
- Wensi Wu
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, No. 107, Wenhua West Road, Lixia District, Jinan, 250012, People's Republic of China
| | - Zhi Wang
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, No. 107, Yanjiang West Road, Yuexiu District, Guangzhou, 510120, People's Republic of China
| | - Huiying Zhang
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, No. 107, Wenhua West Road, Lixia District, Jinan, 250012, People's Republic of China
| | - Xiaojun Zhang
- Department of Anesthesiology, Qilu Hospital of Shandong University, No. 107, Wenhua West Road, Lixia District, Jinan, 250012, People's Republic of China.
| | - Hui Tian
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, No. 107, Wenhua West Road, Lixia District, Jinan, 250012, People's Republic of China.
| |
Collapse
|
5
|
Wang M, Cao L. Hydrolysable tannins as a potential therapeutic drug for the human fibrosis-associated disease. Drug Dev Res 2023; 84:1096-1113. [PMID: 37386756 DOI: 10.1002/ddr.22089] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 05/22/2023] [Accepted: 06/06/2023] [Indexed: 07/01/2023]
Abstract
Fibrosis is a pathological change with abnormal tissue regeneration due to a response to persistent injury, which is extensively related to organ damage and failure, leading to high morbidity and mortality worldwide. Although the pathogenesis of fibrosis has been comprehensively elucidated, there are few effective therapies for treating fibrotic diseases. Natural products are increasingly regarded as an effective strategy for fibrosis with numerous favorable functions. Hydrolysable tannins (HT) are a type of natural products that have the potential to treat the fibrotic disease. In this review, we describe some biological activities and the therapeutic prospects of HT in organ fibrosis. Furthermore, the underlying mechanisms of inhibition of HT on fibrotic organs in relation to inflammation, oxidative stress, epithelial-mesenchymal transition, fibroblast activation and proliferation, and extracellular matrix accumulation are discussed. Understanding the mechanism of HT against fibrotic diseases will provide a new strategy for the prevention and attenuation of fibrosis progression.
Collapse
Affiliation(s)
- Meiwei Wang
- The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| | - Linghui Cao
- The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| |
Collapse
|
6
|
Kleszcz R, Majchrzak-Celińska A, Baer-Dubowska W. Tannins in cancer prevention and therapy. Br J Pharmacol 2023. [PMID: 37614022 DOI: 10.1111/bph.16224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/31/2023] [Accepted: 08/17/2023] [Indexed: 08/25/2023] Open
Abstract
Tannins are a heterogenous class of polyphenolic natural products with promising cancer chemopreventive and therapeutic potential. Studies undertaken over the last 30 years have demonstrated their capacity to target many cellular pathways and molecules important in the development of cancer. Recently, new mechanisms that might be important in anti-carcinogenic activity, such as inhibition of epithelial-to-mesenchymal transition, reduction of cancer stem cell creation, and modulation of cancer cells metabolism have been described. Along with the mechanisms underlying the anti-cancer activity of tannins, this review focuses on their possible application as chemosensitizers in adjuvant therapy and countering multidrug resistance. Furthermore, characteristic physicochemical properties of some tannins, particularly tannic acid, are useful in the formation of nanovehicles for anticancer drugs or the isolation of circulating cancer cells. These new potential applications of tannins deserve further studies. Well-designed clinical trials, which are scarce, are needed to assess the therapeutic effects of tannins themselves or as adjuvants in cancer treatment.
Collapse
Affiliation(s)
- Robert Kleszcz
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Poznań, Poland
| | | | - Wanda Baer-Dubowska
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Poznań, Poland
| |
Collapse
|
7
|
Ramundo V, Palazzo ML, Aldieri E. TGF-β as Predictive Marker and Pharmacological Target in Lung Cancer Approach. Cancers (Basel) 2023; 15:cancers15082295. [PMID: 37190223 DOI: 10.3390/cancers15082295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/03/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
Lung cancer (LC) represents the leading cause of cancer incidence and mortality worldwide. LC onset is strongly related to genetic mutations and environmental interactions, such as tobacco smoking, or pathological conditions, such as chronic inflammation. Despite advancement in knowledge of the molecular mechanisms involved in LC, this tumor is still characterized by an unfavorable prognosis, and the current therapeutic options are unsatisfactory. TGF-β is a cytokine that regulates different biological processes, particularly at the pulmonary level, and its alteration has been demonstrated to be associated with LC progression. Moreover, TGF-β is involved in promoting invasiveness and metastasis, via epithelial to mesenchymal transition (EMT) induction, where TGF-β is the major driver. Thus, a TGF-β-EMT signature may be considered a potential predictive marker in LC prognosis, and TGF-β-EMT inhibition has been demonstrated to prevent metastasis in various animal models. Concerning a LC therapeutic approach, some TGF-β and TGF-β-EMT inhibitors could be used in combination with chemo- and immunotherapy without major side effects, thereby improving cancer therapy. Overall, targeting TGF-β may be a valid possibility to fight LC, both in improving LC prognosis and cancer therapy, via a novel approach that could open up new effective strategies against this aggressive cancer.
Collapse
Affiliation(s)
- Valeria Ramundo
- Department of Oncology, University of Torino, 10126 Torino, Italy
| | | | | |
Collapse
|
8
|
Repositioning itraconazole for amelioration of bleomycin-induced pulmonary fibrosis: Targeting HMGB1/TLR4 Axis, NLRP3 inflammasome/NF-κB signaling, and autophagy. Life Sci 2023; 313:121288. [PMID: 36528079 DOI: 10.1016/j.lfs.2022.121288] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/07/2022] [Accepted: 12/11/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIMS Bleomycin (BLM) is one of the antitumor medications that had proven efficacy in the treatment of a wide range of malignant conditions. Pulmonary fibrosis which is frequently encountered during the course of bleomycin therapy may significantly reduce the potential efficacy of bleomycin in cancer therapy. This study tested the hypothesis that itraconazole may have mitigating effects on BLM-induced pulmonary fibrosis and tried to delineate the potential mechanisms of these effects. MATERIALS AND METHODS In a rat model of pulmonary fibrosis elicited by BLM, the effect of different doses of itraconazole was explored at the biochemical, histopathological, and electron microscopic levels. KEY FINDINGS Itraconazole, in a dose-dependent manner, exhibited significant effects on the pro-oxidant/antioxidant balance, the inflammatory consequences, high-mobility group box 1/toll-like receptor-4 Axis, autophagy and nuclear factor kappa B/Nod-like receptor protein 3 inflammasome signaling and alleviated the histopathological, immunohistochemical, and electron microscopic perturbations induced by BLM in the pulmonary tissues. SIGNIFICANCE In view of the afore-mentioned data, itraconazole may be a promising drug that efficiently mitigates the deleterious effects of BLM on the pulmonary tissues.
Collapse
|
9
|
Systematic Pharmacology-Based Strategy to Explore the Mechanism of Bufei Huoxue Capsule in the Treatment of Chronic Obstructive Pulmonary Disease. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:1129567. [DOI: 10.1155/2022/1129567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/30/2022] [Accepted: 10/20/2022] [Indexed: 12/12/2022]
Abstract
Objective. To explore the effects and mechanisms of Bufei Huoxue Capsule (BHC) on chronic obstructive pulmonary disease (COPD) based on network pharmacology. Methods. The effective components and related targets of BHC were collected by searching TCMSP, HERB, and ETCM databases, after which the related targets of COPD were obtained on GeneCards and OMIM databases. The common targets were imported into the STRING database and Cytoscape database to construct a target interaction network and screen core targets. Next, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed on the Metascape platform. According to the prediction results of network pharmacology, the action mechanism was further examined in an animal model of COPD. The pathological changes of lung tissue were observed by HE staining; goblet cells and mucus secretion in lung tissue were observed by AB-PAS staining, airway collagen deposition was observed by Masson staining, and the expression of NE, TGF-β1, P-EGFR/EGFR, P-ERK1/2/ERK1/2, P-JNK/JNK, and P-P38/P38MAPK protein was detected by Western blot analysis. Results. A total of 379 targets related to BHC and 7391 targets related to COPD were obtained, including 313 potential targets of BHC in treating chronic obstructive pulmonary disease, with JUN, AKT1, TNF, IL6, EGFR, MAPK1, and MAPK14 as the core targets. Through enrichment analysis, BHC may interfere with COPD by regulating the MAPK signal pathway, HIF-1 signal pathway, NF-κB signal pathway, cAMP signal pathway, cGMP-PKG signal pathway, and so on. Animal experiments showed that the BHC could reduce airway inflammatory cell infiltration, inhibit airway epithelial goblet cell proliferation, reduce mucus secretion, and improve small airway collagen fiber deposition in COPD model rats. Besides, BHC could downregulate the protein expression of NE, TGF-β1, P-EGFR, P-ERK1/2, and P-P38MAPK. Conclusion. BHC can reduce airway inflammation, inhibit mucus hypersecretion, and improve airway remodeling by regulating the MAPK signal transduction pathway.
Collapse
|
10
|
Barboura M, Cornebise C, Hermetet F, Guerrache A, Selmi M, Salek A, Chekir-Ghedira L, Aires V, Delmas D. Tannic Acid, A Hydrolysable Tannin, Prevents Transforming Growth Factor-β-Induced Epithelial-Mesenchymal Transition to Counteract Colorectal Tumor Growth. Cells 2022; 11:cells11223645. [PMID: 36429073 PMCID: PMC9688195 DOI: 10.3390/cells11223645] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/10/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022] Open
Abstract
Despite the medico-surgical progress that has been made in the management of patients with colorectal cancer (CRC), the prognosis at five years remains poor. This resistance of cancer cells partly results from their phenotypic characteristics in connection with the epithelial-mesenchymal transition (EMT). In the present study, we have explored the ability of a polyphenol, tannic acid (TA), to counteract CRC cell proliferation and invasion through an action on the EMT. We highlight that TA decreases human SW480 and SW620 CRC cell and murine CT26 CRC cell viability, and TA inhibits their adhesion in the presence of important factors comprising the extracellular matrix, particularly in the presence of collagen type I and IV, and fibronectin. Moreover, these properties were associated with TA's ability to disrupt CRC cell migration and invasion, which are induced by transforming growth factor-β (TGF-β), as evidence in the video microscopy experiments showing that TA blocks the TGF-β1-induced migration of SW480 and CT26 cells. At the molecular level, TA promotes a reversal of the epithelial-mesenchymal transition by repressing the mesenchymal markers (i.e., Slug, Snail, ZEB1, and N-cadherin) and re-expressing the epithelial markers (i.e., E-cadherin and β-catenin). These effects could result from a disruption of the non-canonical signaling pathway that is induced by TGF-β1, where TA strongly decreases the phosphorylation of extracellular-signal regulated kinase ERK1/2, P38 and the AKT proteins that are well known to contribute to the EMT, the cell motility, and the acquisition of invasive properties by tumor cells. Very interestingly, a preclinical study of mice with subcutaneous murine tumor colon CT26 cells has shown that TA was able to significantly delay the growth of tumors without hepato- and nephrotoxicities.
Collapse
Affiliation(s)
- Mahassen Barboura
- UFR des Sciences de Santé, Université de Bourgogne, 21000 Dijon, France
- INSERM Research Center U1231—Cancer and Adaptive Immune Response Team, Bioactive Molecules and Health Research Group, 21000 Dijon, France
- Research Unit Bioactive Natural Products and Biotechnology UR17ES49, Faculty of Dental Medicine of Monastir, University of Monastir, Avicenne street, Monastir 5000, Tunisia
| | - Clarisse Cornebise
- UFR des Sciences de Santé, Université de Bourgogne, 21000 Dijon, France
- INSERM Research Center U1231—Cancer and Adaptive Immune Response Team, Bioactive Molecules and Health Research Group, 21000 Dijon, France
| | - François Hermetet
- UFR des Sciences de Santé, Université de Bourgogne, 21000 Dijon, France
- INSERM Research Center U1231—Cancer and Adaptive Immune Response Team, Bioactive Molecules and Health Research Group, 21000 Dijon, France
| | - Abderrahmane Guerrache
- UFR des Sciences de Santé, Université de Bourgogne, 21000 Dijon, France
- INSERM Research Center U1231—DesCartes Team, 21000 Dijon, France
| | - Mouna Selmi
- Research Unit Bioactive Natural Products and Biotechnology UR17ES49, Faculty of Dental Medicine of Monastir, University of Monastir, Avicenne street, Monastir 5000, Tunisia
| | - Abir Salek
- Research Unit Bioactive Natural Products and Biotechnology UR17ES49, Faculty of Dental Medicine of Monastir, University of Monastir, Avicenne street, Monastir 5000, Tunisia
| | - Leila Chekir-Ghedira
- Research Unit Bioactive Natural Products and Biotechnology UR17ES49, Faculty of Dental Medicine of Monastir, University of Monastir, Avicenne street, Monastir 5000, Tunisia
| | - Virginie Aires
- UFR des Sciences de Santé, Université de Bourgogne, 21000 Dijon, France
- INSERM Research Center U1231—Cancer and Adaptive Immune Response Team, Bioactive Molecules and Health Research Group, 21000 Dijon, France
| | - Dominique Delmas
- UFR des Sciences de Santé, Université de Bourgogne, 21000 Dijon, France
- INSERM Research Center U1231—Cancer and Adaptive Immune Response Team, Bioactive Molecules and Health Research Group, 21000 Dijon, France
- Centre Anticancéreux Georges François Leclerc Center, 21000 Dijon, France
- Correspondence: ; Tel.: +33-380-39-32-26
| |
Collapse
|
11
|
Sarker P, Nalband DM, Freytes DO, Rojas OJ, Khan SA. High-Axial-Aspect Tannic Acid Microparticles Facilitate Gelation and Injectability of Collagen-Based Hydrogels. Biomacromolecules 2022; 23:4696-4708. [PMID: 36198084 DOI: 10.1021/acs.biomac.2c00916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Injectable collagen-based hydrogels offer great promise for tissue engineering and regeneration, but their use is limited by poor mechanical strength. Herein, we incorporate tannic acid (TA) to tailor the rheology of the corresponding hydrogels while simultaneously adding the therapeutic benefits inherent to this polyphenolic component. TA in the solution form and needle-shaped TA microparticles are combined with collagen and the respective systems studied for their time-dependent sol-gel transitions (from storage to body temperatures, 4-37 °C) as a function of TA concentration. Compared to systems incorporating TA microparticles, those with dissolved TA, applied at a similar concentration, generate a less significant enhancement of the elastic modulus. Premature gelation at a low temperature and associated colloidal arrest of the system are proposed as a main factor explaining this limited performance. A higher yield stress (elastic stress method) is determined for systems loaded with TA microparticles compared to the system with dissolved TA. These results are interpreted in terms of the underlying interactions of TA with collagen, as probed by spectroscopy and isothermal titration calorimetry. Importantly, hydrogels containing TA microparticles show high cell viability (human dermal fibroblasts) and comparative cellular activity relative to the collagen-only hydrogel. Overall, composite hydrogels incorporating TA microparticles demonstrate a new, simple, and better-performance alternative to cell culturing and difficult implantation scenarios.
Collapse
Affiliation(s)
- Prottasha Sarker
- Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Danielle M Nalband
- Joint Department of Biomedical Engineering, North Carolina State University/ University of North Carolina-Chapel Hill, Raleigh, North Carolina 27695, United States.,Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Donald O Freytes
- Joint Department of Biomedical Engineering, North Carolina State University/ University of North Carolina-Chapel Hill, Raleigh, North Carolina 27695, United States.,Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Orlando J Rojas
- Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27695, United States.,Bioproducts Institute, Department of Chemical & Biological Engineering, Department of Chemistry and Department of Wood Science, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Saad A Khan
- Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27695, United States
| |
Collapse
|
12
|
Das SS, Tambe S, Prasad Verma PR, Amin P, Singh N, Singh SK, Gupta PK. Molecular insights and therapeutic implications of nanoengineered dietary polyphenols for targeting lung carcinoma: part I. Nanomedicine (Lond) 2022; 17:1779-1798. [PMID: 36636930 DOI: 10.2217/nnm-2022-0133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Lung cancer is the second leading cause of cancer-related mortality globally, and non-small-cell lung cancer accounts for most lung cancer cases. Nanotechnology-based drug-delivery systems have exhibited immense potential in lung cancer therapy due to their fascinating physicochemical characteristics, in vivo stability, bioavailability, prolonged and targeted delivery, gastrointestinal absorption and therapeutic efficiency of their numerous chemotherapeutic agents. However, traditional chemotherapeutics have systemic toxicity issues; therefore, dietary polyphenols might potentially replace them in lung cancer treatment. Polyphenol-based targeted nanotherapeutics have demonstrated interaction with a multitude of protein targets and cellular signaling pathways that affect major cellular processes. This review summarizes the various molecular mechanisms and targeted therapeutic potentials of nanoengineered dietary polyphenols in the effective management of lung cancer.
Collapse
Affiliation(s)
- Sabya Sachi Das
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India.,School of Pharmaceutical and Population Health Informatics, DIT University, Dehradun, Uttarakhand, 248009, India
| | - Srushti Tambe
- Department of Pharmaceutical Science and Technology, Institute of Chemical Technology, Mumbai, Maharashtra, 400019, India
| | - Priya Ranjan Prasad Verma
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Purnima Amin
- Department of Pharmaceutical Science and Technology, Institute of Chemical Technology, Mumbai, Maharashtra, 400019, India
| | - Neeru Singh
- Department of Biomedical Laboratory Technology, University Polytechnic, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Sandeep Kumar Singh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Piyush Kumar Gupta
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, 201310, India.,Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand, 248002, India.,Faculty of Health and Life Sciences, INTI International University, Nilai 71800, Malaysia
| |
Collapse
|
13
|
Jing W, Xiaolan C, Yu C, Feng Q, Haifeng Y. Pharmacological effects and mechanisms of tannic acid. Biomed Pharmacother 2022; 154:113561. [PMID: 36029537 DOI: 10.1016/j.biopha.2022.113561] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/04/2022] [Accepted: 08/14/2022] [Indexed: 12/18/2022] Open
Abstract
In recent years, increasing attention has been paid to the pharmacological efficacy of tannins. Tannic acid (TA), the simplest hydrolysable tannin that has been approved by the FDA as a safe food additive, is one of the most important components of these traditional medicines. Studies have shown that TA displays a wide range of pharmacological activities, such as anti-inflammatory, neuroprotective, antitumor, cardioprotective, and anti-pathogenic effects. Here, we summarize the known pharmacological effects and associated mechanisms of TA. We focus on the effect and mechanism of TA in various animal models of inflammatory disease and organ, brain, and cardiovascular injury. Moreover, we discuss the possible molecular targets and signaling pathways of TA, in addition to the pharmacological effects of TA-based nanoparticles and TA in combination with chemotherapeutic drugs.
Collapse
Affiliation(s)
- Wang Jing
- Jiangsu Agri-animal Husbandry Vocational College, Taizhou, Jiangsu 225300, PR China.
| | - Chen Xiaolan
- Jiangsu Agri-animal Husbandry Vocational College, Taizhou, Jiangsu 225300, PR China
| | - Chen Yu
- Jiangsu Agri-animal Husbandry Vocational College, Taizhou, Jiangsu 225300, PR China
| | - Qin Feng
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-animal Husbandry Vocational College, Taizhou 225300, PR China
| | - Yang Haifeng
- Jiangsu Agri-animal Husbandry Vocational College, Taizhou, Jiangsu 225300, PR China
| |
Collapse
|
14
|
Gandhi D, Bhandari S, Mishra S, Tiwari RR, Rajasekaran S. Non-malignant respiratory illness associated with exposure to arsenic compounds in the environment. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2022; 94:103922. [PMID: 35779705 DOI: 10.1016/j.etap.2022.103922] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 06/22/2022] [Accepted: 06/25/2022] [Indexed: 06/15/2023]
Abstract
Arsenic (As), a toxic metalloid, primarily originates from both natural and anthropogenic activities. Reports suggested that millions of people globally exposed to high levels of naturally occurring As compounds via inhalation and ingestion. There is evidence that As is a well-known lung carcinogen. However, there has been relatively little evidence suggesting its non-malignant lung effects. This review comprehensively summarises current experimental and clinical studies implicating the association of As exposure and the development of several non-malignant lung diseases. Experimental studies provided evidence that As exposure induces redox imbalance, apoptosis, inflammatory response, epithelial-to-mesenchymal transition (EMT), and affected normal lung development through alteration of the components of intracellular signaling cascades. In addition, we also discuss the sources and possible mechanisms of As influx and efflux in the lung. Finally, current experimental studies on treatment strategies using phytochemicals and our perspective on future research with As are also discussed.
Collapse
Affiliation(s)
- Deepa Gandhi
- Department of Biochemistry, ICMR-National Institute for Research in Environmental, Health, Bhopal, Madhya Pradesh, India
| | - Sneha Bhandari
- Department of Biochemistry, ICMR-National Institute for Research in Environmental, Health, Bhopal, Madhya Pradesh, India
| | - Sehal Mishra
- Department of Biochemistry, ICMR-National Institute for Research in Environmental, Health, Bhopal, Madhya Pradesh, India
| | - Rajnarayan R Tiwari
- Department of Biochemistry, ICMR-National Institute for Research in Environmental, Health, Bhopal, Madhya Pradesh, India
| | - Subbiah Rajasekaran
- Department of Biochemistry, ICMR-National Institute for Research in Environmental, Health, Bhopal, Madhya Pradesh, India.
| |
Collapse
|
15
|
Tsai KF, Shen CJ, Cheung CW, Wang TL, Chow LWC, Leung YM, Wong KL. Lipotoxicity in human lung alveolar type 2 A549 cells: Mechanisms and protection by tannic acid. CHINESE J PHYSIOL 2021; 64:289-297. [PMID: 34975122 DOI: 10.4103/cjp.cjp_68_21] [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] [Indexed: 01/06/2023] Open
Abstract
Palmitic acid (PA) is a saturated free fatty acid which, when being excessive, accounts for lipotoxicity. Using human lung A549 cells as a model for lung alveolar type 2 epithelial cells, we found that challenge of A549 cells with PA resulted in apoptotic cell death, as reflected by positive annexin V and PI staining, and also appearance of cleaved caspase-3. PA treatment also caused depletion of intracellular Ca2+ store, endoplasmic reticulum (ER) stress, and oxidative stress. Tannic acid (TA), a polyphenol present in wines and many beverages, alleviated PA-induced ER stress, oxidative stress and apoptotic death. Thus, our results suggest PA lipotoxicity in lung alveolar type 2 epithelial cells could be protected by TA.
Collapse
Affiliation(s)
- Kun-Feng Tsai
- Gastroenterology and Hepatology Section, Department of Internal Medicine, An Nan Hospital, China Medical University; Department of Medical Sciences Industry, Chang Jung Christian University, Tainan, Taiwan
| | - Chen-Jung Shen
- Endocrinology and Metabolism Section, Department of Internal Medicine, An Nan Hospital, China Medical University, Tainan, Taiwan
| | - Chi-Wai Cheung
- Department of Anesthesiology, University of Hong Kong, China
| | - Tzong-Luen Wang
- School of Medicine, Fu-Jen Catholic University; Department of Emergency Medicine, Fu-Jen Catholic University Hospital, Taipei, Taiwan
| | - Louis W C Chow
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macau; UNIMED Medical Institute, Hong Kong; Organisation for Oncology and Translational Research, Hong Kong, China
| | - Yuk-Man Leung
- Department of Physiology, China Medical University, Taichung, Taiwan
| | - Kar-Lok Wong
- Department of Anesthesiology, University of Hong Kong, China; Department of Anesthesiology, Kuang Tien General Hospital, Shalu, Taichung, Taiwan
| |
Collapse
|
16
|
Carver W, Fix E, Fix C, Fan D, Chakrabarti M, Azhar M. Effects of emodin, a plant-derived anthraquinone, on TGF-β1-induced cardiac fibroblast activation and function. J Cell Physiol 2021; 236:7440-7449. [PMID: 34041746 PMCID: PMC8530838 DOI: 10.1002/jcp.30416] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 04/21/2021] [Accepted: 04/23/2021] [Indexed: 12/15/2022]
Abstract
Cardiac fibrosis accompanies a number of pathological conditions and results in altered myocardial structure, biomechanical properties and function. The signaling networks leading to fibrosis are complex, contributing to the general lack of progress in identifying effective therapeutic approaches to prevent or reverse this condition. Several studies have shown protective effects of emodin, a plant-derived anthraquinone, in animal models of fibrosis. A number of questions remain regarding the mechanisms whereby emodin impacts fibrosis. Transforming growth factor beta 1 (TGF-β1) is a potent stimulus of fibrosis and fibroblast activation. In the present study, experiments were performed to evaluate the effects of emodin on activation and function of cardiac fibroblasts following treatment with TGF-β1. We demonstrate that emodin attenuates TGF-β1-induced fibroblast activation and collagen accumulation in vitro. Emodin also inhibits activation of several canonical (SMAD2/3) and noncanonical (Erk1/2) TGF-β signaling pathways, while activating the p38 pathway. These results suggest that emodin may provide an effective therapeutic agent for fibrosis that functions via specific TGF-β signaling pathways.
Collapse
Affiliation(s)
- Wayne Carver
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209
| | - Ethan Fix
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209
| | - Charity Fix
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209
| | - Daping Fan
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209
| | - Mrinmay Chakrabarti
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209
| | - Mohamad Azhar
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209
| |
Collapse
|
17
|
Li Q, Peng W, Zhang Z, Pei X, Sun Z, Ou Y. A phycocyanin derived eicosapeptide attenuates lung fibrosis development. Eur J Pharmacol 2021; 908:174356. [PMID: 34280398 DOI: 10.1016/j.ejphar.2021.174356] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/23/2021] [Accepted: 07/15/2021] [Indexed: 01/06/2023]
Abstract
Pulmonary fibrosis (PF) is a progressive respiratory disease. Phycocyanin derived eicosapeptide (PP20) is a novel peptide derived from active protein C-phycocyanin in Cyanobacteria. The aim of our study was to explore the anti-fibrotic activity of the PP20 and its underlying mechanism. Characteristic features of pulmonary fibrosis in oleic acid (OA)-induced mice and epithelial-mesenchymal transition (EMT) in TGF-β1-exposed A549 and HFL-1 cells with or without PP20 and the change of TGF-β/Smad and MAPK signaling pathways were examined. Smad and MAPK agonists were used to explore the role of TGF-β/Smad and MAPK signaling in TGF-β1- induced collagen I expression in A549 cells and α-SMA expression in HFL-1 cells when treated with PP20. Our results showed that PP20 significantly alleviated the inflammatory response and tissue destruction, inhibited EMT, restored the imbalance of TIMP-1/MMP-9 and reduced collagen fiber deposition. Moreover, PP20 inhibited TGF-β1-induced EMT and collagen I expression in A549 cells. PP20 could also inhibit the proliferation, and decrease TGF-β1-induced the expression of collagen I and transformation of fibroblasts into myofibroblasts in HFL-1 cells. Additionally, animal experiments and cell experiments combined with pathway agonists have shown that PP20 can negatively regulate TGF-β/Smad and MAPK pathways and show anti-fibrotic properties. PP20 may be a promising drug candidate for protection against pulmonary fibrosis.
Collapse
Affiliation(s)
- Qihao Li
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China
| | - Wen Peng
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China
| | - Zhaoyu Zhang
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China
| | - Xin Pei
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China
| | - Zhongkan Sun
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China
| | - Yu Ou
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China.
| |
Collapse
|
18
|
Chemical Investigation of Diketopiperazines and N-Phenethylacetamide Isolated from Aquimarina sp. MC085 and Their Effect on TGF-β-Induced Epithelial–Mesenchymal Transition. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11198866] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Chemical investigations of Aquimarina sp. MC085, which suppressed TGF-β-induced epithelial–mesenchymal transition (EMT) in A549 human lung cancer cells, led to the isolation of compounds 1–3. Structural characterization using spectroscopic data analyses in combination with Marfey’s analysis revealed that they were two diketopiperazines [cyclo(l-Pro-l-Leu) (1) and cyclo(l-Pro-l-Ile) (2)] and one N-phenethylacetamide (3). Cyclo(l-Pro-l-Leu) (1) and N-phenethylactamide (3) inhibited the TGF-β/Smad pathway and suppressed the metastasis of A549 cells by affecting TGF-β-induced EMT. However, cyclo(l-Pro-l-Ile) (2) downregulated mesenchymal factors via a non-Smad-mediated signaling pathway.
Collapse
|
19
|
Rajasekar N, Sivanantham A, Ravikumar V, Rajasekaran S. An overview on the role of plant-derived tannins for the treatment of lung cancer. PHYTOCHEMISTRY 2021; 188:112799. [PMID: 33975161 DOI: 10.1016/j.phytochem.2021.112799] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 06/12/2023]
Abstract
Lung cancer is the leading cause of cancer-related death globally. Despite many advanced approaches to treat cancer, they are often ineffective due to resistance to classical anti-cancer drugs and distant metastases. Currently, alternative medicinal agents derived from plants are the major interest due to high bioavailability and fewer adverse effects. Tannins are polyphenolic compounds existing as specialized products in a wide variety of vegetables, fruits, and nuts. Many tannins have been found to possess protective properties, such as anti-inflammatory, anti-fibrotic, anti-microbial, anti-diabetic, and so on. This review aims to summarize the current knowledge addressing the anti-cancer effects of dietary tannins and their underlying molecular mechanisms. In vivo and in vitro studies provide evidences that anti-cancer effects of various tannins are predominantly mediated through negative regulation of transcription factors, growth factors, receptor kinases, and many oncogenic molecules. In addition, we also discussed the absorption, distribution, metabolism, excretion, and toxicity (ADMET) properties of tannins, clinical trial results as well as our perspective on future research with tannins.
Collapse
Affiliation(s)
- Nandhine Rajasekar
- Department of Biotechnology, BIT-Campus, Anna University, Tiruchirappalli, Tamil Nadu, India
| | - Ayyanar Sivanantham
- Department of Biotechnology, BIT-Campus, Anna University, Tiruchirappalli, Tamil Nadu, India
| | - Vilwanathan Ravikumar
- Department of Biochemistry, School of Life Science, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Subbiah Rajasekaran
- Department of Biochemistry, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh, India.
| |
Collapse
|
20
|
Zhu J, Wang F, Feng X, Li B, Ma L, Zhang J. Family with sequence similarity 13 member A mediates TGF-β1-induced EMT in small airway epithelium of patients with chronic obstructive pulmonary disease. Respir Res 2021; 22:192. [PMID: 34210319 PMCID: PMC8247231 DOI: 10.1186/s12931-021-01783-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 06/24/2021] [Indexed: 12/28/2022] Open
Abstract
Background To explore the role of family with sequence similarity 13 member A (FAM13A) in TGF-β1-induced EMT in the small airway epithelium of patients with chronic obstructive pulmonary disease (COPD). Methods Small airway wall thickness and protein levels of airway remodeling markers, EMT markers, TGF-β1, and FAM13A were measured in lung tissue samples from COPD and non-COPD patients. The correlations of FAM13A expression with COPD severity and EMT marker expression were evaluated. Gain- and loss-of-function assays were performed to explore the functions of FAM13A in cell proliferation, motility, and TGF-β1-induced EMT marker alterations in human bronchial epithelial cell line BEAS-2B. Results Independent of smoking status, lung tissue samples from COPD patients exhibited significantly increased small airway thickness and collagen fiber deposition, along with enhanced protein levels of remodeling markers (collagen I, fibronectin, and MMP-9), mesenchymal markers (α-SMA, vimentin, and N-cadherin), TGF-β1, and FAM13A, compared with those from non-COPD patients. FAM13A expression negatively correlated with FEV1% and PO2 in COPD patients. In small airway epithelium, FAM13A expression negatively correlated with E-cadherin protein levels and positively correlated with vimentin protein levels. In BEAS-2B cells, TGF-β1 dose-dependently upregulated FAM13A protein levels. FAM13A overexpression significantly promoted cell proliferation and motility in BEAS-2B cells, whereas FAM13A silencing showed contrasting results. Furthermore, FAM13A knockdown partially reversed TGF-β1-induced EMT marker protein alterations in BEAS-2B cells. Conclusions FAM13A upregulation is associated with TGF-β1-induced EMT in the small airway epithelium of COPD patients independent of smoking status, serving as a potential therapeutic target for anti-EMT therapy in COPD. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-021-01783-z.
Collapse
Affiliation(s)
- Jinyuan Zhu
- Department of Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan, 750004, People's Republic of China
| | - Faxuan Wang
- School of Public Health and Management, Ningxia Medical University, Yinchuan, 750004, People's Republic of China
| | - Xueyan Feng
- Ningxia Medical University, Yinchuan, 750004, People's Republic of China
| | - Beibei Li
- Ningxia Medical University, Yinchuan, 750004, People's Republic of China
| | - Liqiong Ma
- Department of Pathology, General Hospital of Ningxia Medical University, Yinchuan, 750004, People's Republic of China
| | - Jin Zhang
- Department of Respiratory and Critical Care Medicine, General Hospital of Ningxia Medical University, 804 Shengli South Street, Xingqing District, Yinchuan, 750004, People's Republic of China.
| |
Collapse
|
21
|
Rajasekar N, Sivanantham A, Kar A, Mukhopadhyay S, Mahapatra SK, Paramasivam SG, Rajasekaran S. Anti-asthmatic effects of tannic acid from Chinese natural gall nuts in a mouse model of allergic asthma. Int Immunopharmacol 2021; 98:107847. [PMID: 34126339 DOI: 10.1016/j.intimp.2021.107847] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/01/2021] [Accepted: 06/01/2021] [Indexed: 12/31/2022]
Abstract
Asthma is a chronic inflammatory disease of the airways, which is characterized by infiltration of inflammatory cells, airway hyperresponsiveness (AHR), and airway remodeling. This study aimed to explore the role and mechanism of tannic acid (TA), a naturally occurring plant-derived polyphenol, in murine asthma model. BALB/c mice were given ovalbumin (OVA) to establish an allergic asthma model. The results revealed that TA treatment significantly decreased OVA-induced AHR, inflammatory cells infiltration, and the expression of various inflammatory mediators (Th2 and Th1 cytokines, eotaxin, and total IgE). Additionally, TA treatment also attenuated increases in mucins (Muc5ac and Muc5b) expression, mucus production in airway goblet cells, mast cells infiltration, and airway remodeling induced by OVA exposure. Furthermore, OVA-induced NF-κB (nuclear factor- kappa B) activation and cell adhesion molecules expression in the lungs was suppressed by TA treatment. In conclusion, TA effectively attenuated AHR, inflammatory response, and airway remodeling in OVA-challenged asthmatic mice. Therefore, TA may be a potential therapeutic option against allergic asthma in clinical settings.
Collapse
Affiliation(s)
- Nandhine Rajasekar
- Department of Biotechnology, BIT-Campus, Anna University, Tiruchirappalli, Tamil Nadu, India
| | - Ayyanar Sivanantham
- Department of Biotechnology, BIT-Campus, Anna University, Tiruchirappalli, Tamil Nadu, India
| | - Amrita Kar
- Department of Paramedical and Allied Health Sciences, Midnapore City College, Midnapore, West Bengal, India
| | - Sramana Mukhopadhyay
- Department of Pathology and Lab Medicine, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, India
| | - Santanu Kar Mahapatra
- Department of Paramedical and Allied Health Sciences, Midnapore City College, Midnapore, West Bengal, India
| | | | - Subbiah Rajasekaran
- Department of Biochemistry, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh, India.
| |
Collapse
|
22
|
Yang YP, Zhao JQ, Gao HB, Li JJ, Li XL, Niu XL, Lei YH, Li X. Tannic acid alleviates lipopolysaccharide‑induced H9C2 cell apoptosis by suppressing reactive oxygen species‑mediated endoplasmic reticulum stress. Mol Med Rep 2021; 24:535. [PMID: 34080663 PMCID: PMC8170226 DOI: 10.3892/mmr.2021.12174] [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: 10/12/2020] [Accepted: 04/30/2021] [Indexed: 01/08/2023] Open
Abstract
Sepsis-induced myocardial dysfunction is one of the features of multiple organ dysfunction in sepsis, which is associated with extremely high mortality and is characterized by impaired myocardial compliance. To date, there are few effective treatment options available to cure sepsis. Tannic acid (TA) is reportedly protective during sepsis; however, the underlying mechanisms by which TA protects against septic heart injury remain elusive. The present study investigated the potential effects and underlying mechanisms of TA in alleviating lipopolysaccharide (LPS)-induced H9C2 cardiomyocyte cell apoptosis. H9C2 cells were treated with LPS (15 µg/ml), TA (10 µM) and TA + LPS; control cells were treated with medium only. Apoptosis was measured using flow cytometry, reverse transcription-quantitative PCR (RT-qPCR) and western blot analysis. Additionally, the levels of cellular reactive oxygen species (ROS), malondialdehyde and nicotinamide adenine dinucleotide phosphate were evaluated. Western blotting and RT-qPCR were also employed to detect the expression levels of endoplasmic reticulum (ER) stress-associated functional proteins. The present findings demonstrated that TA reduced the degree of LPS-induced H9C2 cell injury, including inhibition of ROS production and ER stress (ERS)-associated apoptosis. ERS-associated functional proteins, including activating transcription factor 6, protein kinase-like ER kinase, inositol-requiring enzyme 1, spliced X box-binding protein 1 and C/EBP-homologous protein were suppressed in response to TA treatment. Furthermore, the expression levels of ERS-associated apoptotic proteins, including c-Jun N-terminal kinase, Bax, cytochrome c, caspase-3, caspase-12 and caspase-9 were reduced following treatment with TA. Additionally, the protective effects of TA on LPS-induced H9C2 cells were partially inhibited following treatment with the ROS inhibitor N-acetylcysteine, which demonstrated that ROS mediated ERS-associated apoptosis and TA was able to decrease ROS-mediated ERS-associated apoptosis. Collectively, the present findings demonstrated that the protective effects of TA against LPS-induced H9C2 cell apoptosis may be associated with the amelioration of ROS-mediated ERS. These findings may assist the development of potential novel therapeutic methods to inhibit the progression of myocardial cell injury.
Collapse
Affiliation(s)
- Yan-Ping Yang
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Jie-Qiong Zhao
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Hai-Bo Gao
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Jin-Jing Li
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Xiao-Li Li
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Xiao-Lin Niu
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Yong-Hong Lei
- Department of Plastic Surgery, General Hospital of Chinese PLA, Beijing 100853, P.R. China
| | - Xue Li
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| |
Collapse
|
23
|
Rajasekaran S, Rajasekar N, Sivanantham A. Therapeutic potential of plant-derived tannins in non-malignant respiratory diseases. J Nutr Biochem 2021; 94:108632. [PMID: 33794331 DOI: 10.1016/j.jnutbio.2021.108632] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/06/2021] [Accepted: 03/23/2021] [Indexed: 12/24/2022]
Abstract
Respiratory diseases are the major cause of human illness and death around the world. Despite advances in detection and treatment, very few classes of safe and effective therapy have been introduced to date. At present, phytochemicals are getting more attention because of their diverse beneficial activities and minimal toxicity. Tannins are polyphenolic secondary metabolites with high molecular weights, which are naturally present in a wide variety of fruits, vegetables, cereals, and leguminous seeds. Many tannins are endowed with well-recognized protective properties, such as anti-cancer, anti-microbial, anti-oxidant, anti-hyperglycemic, and many others. This review summarizes a large body of experimental evidence implicating that tannins are helpful in tackling a wide range of non-malignant respiratory diseases including acute lung injury (ALI), pulmonary fibrosis, asthma, pulmonary hypertension, and chronic obstructive pulmonary disease (COPD). Mechanistic pathways by which various classes of tannins execute their beneficial effects are discussed. In addition, clinical trials and our perspective on future research with tannins are also reviewed.
Collapse
Affiliation(s)
- Subbiah Rajasekaran
- Department of Biochemistry, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh, India.
| | - Nandhine Rajasekar
- Department of Biotechnology, BIT-Campus, Anna University, Tiruchirappalli, Tamil Nadu, India
| | - Ayyanar Sivanantham
- Department of Biotechnology, BIT-Campus, Anna University, Tiruchirappalli, Tamil Nadu, India
| |
Collapse
|
24
|
A. Youness R, Kamel R, A. Elkasabgy N, Shao P, A. Farag M. Recent Advances in Tannic Acid (Gallotannin) Anticancer Activities and Drug Delivery Systems for Efficacy Improvement; A Comprehensive Review. Molecules 2021; 26:1486. [PMID: 33803294 PMCID: PMC7967207 DOI: 10.3390/molecules26051486] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/23/2021] [Accepted: 02/26/2021] [Indexed: 12/24/2022] Open
Abstract
Tannic acid is a chief gallo-tannin belonging to the hydrolysable tannins extracted from gall nuts and other plant sources. A myriad of pharmaceutical and biological applications in the medical field has been well recognized to tannic acid. Among these effects, potential anticancer activities against several solid malignancies such as liver, breast, lung, pancreatic, colorectal and ovarian cancers have been reported. Tannic acid was found to play a maestro-role in tuning several oncological signaling pathways including JAK/STAT, RAS/RAF/mTOR, TGF-β1/TGF-β1R axis, VEGF/VEGFR and CXCL12/CXCR4 axes. The combinational beneficial effects of tannic acid with other conventional chemotherapeutic drugs have been clearly demonstrated in literature such as a synergistic anticancer effect and enhancement of the chemo-sensitivity in several resistant cases. Yet, clinical applications of tannic acid have been limited owing to its poor lipid solubility, low bioavailability, off-taste, and short half-life. To overcome such obstacles, novel drug delivery systems have been employed to deliver tannic acid with the aim of improving its applications and/or efficacy against cancer cells. Among these drug delivery systems are several types of organic and metallic nanoparticles. In this review, the authors focus on the molecular mechanisms of tannic acid in tuning several neoplastic diseases as well as novel drug delivery systems that can be used for its clinical applications with an attempt to provide a systemic reference to promote the development of tannic acid as a cheap drug and/or drug delivery system in cancer management.
Collapse
Affiliation(s)
- Rana A. Youness
- The Molecular Genetics Research Team, Department of Pharmaceutical Biology, Faculty of Pharmacy andBiotechnology, German University in Cairo, Cairo 12622, Egypt;
| | - Rabab Kamel
- Pharmaceutical Technology Department, National Research Centre, Cairo 12622, Egypt;
| | - Nermeen A. Elkasabgy
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt;
| | - Ping Shao
- Department of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China;
| | - Mohamed A. Farag
- Pharmacognosy Department, College of Pharmacy, Cairo University, Kasr El Aini St., Cairo 11562, Egypt
- Chemistry Department, School of Sciences & Engineering, The American University in Cairo, New Cairo 11835, Egypt
| |
Collapse
|
25
|
Javed Z, Khan K, Rasheed A, Sadia H, Raza S, Salehi B, Cho WC, Sharifi-Rad J, Koch W, Kukula-Koch W, Głowniak-Lipa A, Helon P. MicroRNAs and Natural Compounds Mediated Regulation of TGF Signaling in Prostate Cancer. Front Pharmacol 2021; 11:613464. [PMID: 33584291 PMCID: PMC7873640 DOI: 10.3389/fphar.2020.613464] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 11/24/2020] [Indexed: 12/25/2022] Open
Abstract
Prostate cancer (PCa) is with rising incidence in male population globally. It is a complex anomaly orchestrated by a plethora of cellular processes. Transforming growth factor-beta (TGF-β) signaling is one of the key signaling pathways involved in the tumorigenesis of PCa. TGF-β signaling has a dual role in the PCa, making it difficult to find a suitable therapeutic option. MicroRNAs (miRNAs) mediated regulation of TGF-β signaling is responsible for the TGF-ß paradox. These are small molecules that modulate the expression of target genes and regulate cancer progression. Thus, miRNAs interaction with different signaling cascades is of great attention for devising new diagnostic and therapeutic options for PCa. Natural compounds have been extensively studied due to their high efficacy and low cytotoxicity. Here, we discuss the involvement of TGF-ß signaling in PCa with the interplay between miRNAs and TGF-β signaling and also review the role of natural compounds for the development of new therapeutics for PCa.
Collapse
Affiliation(s)
- Zeeshan Javed
- Office for Research Innovation and Commercialization, Lahore Garrison University, Lahore, Pakistan
| | - Khushbukhat Khan
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Amna Rasheed
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Haleema Sadia
- Department of Biotechnology, Balochistan University of Information Technology, Engineering and Management Sciences, Quetta, Pakistan
| | - Shahid Raza
- Office for Research Innovation and Commercialization, Lahore Garrison University, Lahore, Pakistan
| | - Bahare Salehi
- Medical Ethics and Law Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, China
| | - Javad Sharifi-Rad
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Facultad de Medicina, Universidad del Azuay, Cuenca, Ecuador
| | - Wojciech Koch
- Chair and Department of Food and Nutrition, Medical University of Lublin, Lublin, Poland
| | | | - Anna Głowniak-Lipa
- Department of Cosmetology, University of Information Technology and Management in Rzeszów, Rzeszów, Poland
| | - Paweł Helon
- Branch in Sandomierz, Jan Kochanowski University in Kielce, Sandomierz, Poland
| |
Collapse
|
26
|
Wang J, Zhao X, Feng W, Li Y, Peng C. Inhibiting TGF-[Formula: see text] 1-Mediated Cellular Processes as an Effective Strategy for the Treatment of Pulmonary Fibrosis with Chinese Herbal Medicines. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2021; 49:1965-1999. [PMID: 34961416 DOI: 10.1142/s0192415x21500932] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Pulmonary fibrosis (PF) is a chronic and irreversible interstitial lung disease that even threatens the lives of some patients infected with COVID-19. PF is a multicellular pathological process, including the initial injuries of epithelial cells, recruitment of inflammatory cells, epithelial-mesenchymal transition, activation and differentiation of fibroblasts, etc. TGF-[Formula: see text]1 acts as a key effect factor that participates in these cellular processes of PF. Recently, much attention was paid to inhibiting TGF-[Formula: see text]1 mediated cell processes in the treatment of PF with Chinese herbal medicines (CHM), an important part of traditional Chinese medicine. Here, this review first summarized the effects of TGF-[Formula: see text]1 in different cellular processes of PF. Then, this review summarized the recent research on CHM (compounds, multi-components, single medicines and prescriptions) to directly and/or indirectly inhibit TGF-[Formula: see text]1 signaling (TLRs, PPARs, micrRNA, etc.) in PF. Most of the research focused on CHM natural compounds, including but not limited to alkaloids, flavonoids, phenols and terpenes. After review, the research perspectives of CHM on TGF-[Formula: see text]1 inhibition in PF were further discussed. This review hopes that revealing the inhibiting effects of CHM on TGF-[Formula: see text]1-mediated cellular processes of PF can promote CHM to be better understood and utilized, thus transforming the therapeutic activities of CHM into practice.
Collapse
Affiliation(s)
- Jing Wang
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| | - Xingtao Zhao
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| | - Wuwen Feng
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| | - Yunxia Li
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| | - Cheng Peng
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| |
Collapse
|
27
|
Bao C, Liu T, Qian L, Xiao C, Zhou X, Ai H, Wang J, Fan W, Pan J. Shikonin inhibits migration and invasion of triple-negative breast cancer cells by suppressing epithelial-mesenchymal transition via miR-17-5p/PTEN/Akt pathway. J Cancer 2021; 12:76-88. [PMID: 33391404 PMCID: PMC7738816 DOI: 10.7150/jca.47553] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 10/21/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Triple-negative breast cancer (TNBC) is a great threat to global women's health due to its high metastatic potential. Epithelial-to-mesenchymal transition (EMT) is considered as a key event in the process of metastasis. So the pharmacological targeting of EMT might be a promising strategy in improving the therapeutic efficacy of TNBC. Here, we investigated the effect of shikonin exerting on EMT and consequently the metastasis of TNBC cells and its underlying mechanism. Methods: The invasive and migratory capacities of MDA-MB-231 and BT549 cells were tested using transwell invasion and wound healing assay. MiR-17-5p expression was examined by qRT-PCR. MiR-17-5p targeted genes were predicted with different bioinformatic algorithms from four databases (TargetScan, miRanda, PITA and picTar) and further screened by Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. The differential expressions of predicted genes and their correlations with miR-17-5p were identified in breast cancer patients based on The Cancer Genome Atlas (TCGA) database. The interaction between phosphatase and tensin homolog deleted on chromosome ten (PTEN) and miR-17-5p was analyzed by luciferase reporter assay. The overexpression vector and small interfering RNA were constructed to investigate the role PTEN played in metastasis and EMT regulation. The expressions of EMT markers, protein kinase B (Akt) and phospho-Akt (p-Akt) were evaluated by western blot. Results: Shikonin suppressed the migration and invasion of MDA-MB-231 and BT549 cells and meanwhile the corresponding alterations of EMT biomarkers were observed in shikonin treated MDA-MB-231 cells. Shikonin inhibited the expression of miR-17-5p, which was upregulated in breast cancer. The 3'-untranslated region (3'-UTR) of PTEN was found to be direct binding target of miR-17-5p by luciferase reporter assays. PTEN functioned as a suppressor both in the metastasis and EMT of TNBC cells. Moreover, Akt and p-Akt (Ser473) were involved in the process of inhibition in cancer cell migration, invasion and EMT by shikonin. Conclusions: Shikonin inhibits migration and invasion of TNBC cells by suppressing EMT via miR-17-5p/PTEN/Akt pathway. This suggests shikonin as a promising therapeutic agent to counteract metastasis in the TNBC patients.
Collapse
Affiliation(s)
- Chang Bao
- Program of Innovative Cancer Therapeutics, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, People's Republic of China
- Key Laboratory of Organ Transplantation, Hangzhou 310003, People's Republic of China
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Hangzhou310003, People's Republic of China
| | - Tao Liu
- Department of Respiratory Medicine, Hospital of Traditional Chinese Medicine of Pingxiang city, No.10 Pingchuxi Road, Pingxiang 337000, People's Republic of China
| | - Lingbo Qian
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, No.481 Binwen Road, Hangzhou 310053, People's Republic of China
| | - Chi Xiao
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, No.481 Binwen Road, Hangzhou 310053, People's Republic of China
| | - Xinru Zhou
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, No.481 Binwen Road, Hangzhou 310053, People's Republic of China
| | - Heng Ai
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, No.481 Binwen Road, Hangzhou 310053, People's Republic of China
| | - Jue Wang
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, No.481 Binwen Road, Hangzhou 310053, People's Republic of China
| | - Weimin Fan
- Program of Innovative Cancer Therapeutics, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, People's Republic of China
- Key Laboratory of Organ Transplantation, Hangzhou 310003, People's Republic of China
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Hangzhou310003, People's Republic of China
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Jie Pan
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, No.481 Binwen Road, Hangzhou 310053, People's Republic of China
| |
Collapse
|
28
|
Phan THG, Paliogiannis P, Nasrallah GK, Giordo R, Eid AH, Fois AG, Zinellu A, Mangoni AA, Pintus G. Emerging cellular and molecular determinants of idiopathic pulmonary fibrosis. Cell Mol Life Sci 2020; 78:2031-2057. [PMID: 33201251 PMCID: PMC7669490 DOI: 10.1007/s00018-020-03693-7] [Citation(s) in RCA: 162] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 10/08/2020] [Accepted: 10/28/2020] [Indexed: 12/17/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF), the most common form of idiopathic interstitial pneumonia, is a progressive, irreversible, and typically lethal disease characterized by an abnormal fibrotic response involving vast areas of the lungs. Given the poor knowledge of the mechanisms underpinning IPF onset and progression, a better understanding of the cellular processes and molecular pathways involved is essential for the development of effective therapies, currently lacking. Besides a number of established IPF-associated risk factors, such as cigarette smoking, environmental factors, comorbidities, and viral infections, several other processes have been linked with this devastating disease. Apoptosis, senescence, epithelial-mesenchymal transition, endothelial-mesenchymal transition, and epithelial cell migration have been shown to play a key role in IPF-associated tissue remodeling. Moreover, molecules, such as chemokines, cytokines, growth factors, adenosine, glycosaminoglycans, non-coding RNAs, and cellular processes including oxidative stress, mitochondrial dysfunction, endoplasmic reticulum stress, hypoxia, and alternative polyadenylation have been linked with IPF development. Importantly, strategies targeting these processes have been investigated to modulate abnormal cellular phenotypes and maintain tissue homeostasis in the lung. This review provides an update regarding the emerging cellular and molecular mechanisms involved in the onset and progression of IPF.
Collapse
Affiliation(s)
- Thị Hằng Giang Phan
- Department of Immunology and Pathophysiology, University of Medicine and Pharmacy, Hue University, Hue City, Vietnam
| | - Panagiotis Paliogiannis
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100, Sassari, Italy
| | - Gheyath K Nasrallah
- Department of Biomedical Sciences, College of Health Sciences Member of QU Health, Qatar University, P.O. Box 2713, Doha, Qatar. .,Biomedical Research Center Qatar University, P.O Box 2713, Doha, Qatar.
| | - Roberta Giordo
- Department of Medical Laboratory Sciences, College of Health Sciences, and Sharjah Institute for Medical Research, University of Sharjah, University City Rd, Sharjah, 27272, United Arab Emirates
| | - Ali Hussein Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, PO Box 2713, Doha, Qatar.,Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, PO Box 11-0236, Beirut, Lebanon
| | - Alessandro Giuseppe Fois
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100, Sassari, Italy
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, 07100, Sassari, Italy
| | - Arduino Aleksander Mangoni
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, Australia.
| | - Gianfranco Pintus
- Department of Medical Laboratory Sciences, College of Health Sciences, and Sharjah Institute for Medical Research, University of Sharjah, University City Rd, Sharjah, 27272, United Arab Emirates. .,Department of Biomedical Sciences, University of Sassari, 07100, Sassari, Italy.
| |
Collapse
|
29
|
Kato M, Takahashi F, Sato T, Mitsuishi Y, Tajima K, Ihara H, Nurwidya F, Baskoro H, Murakami A, Kobayashi I, Hidayat M, Shimada N, Sasaki S, Mineki R, Fujimura T, Kumasaka T, Niwa SI, Takahashi K. Tranilast Inhibits Pulmonary Fibrosis by Suppressing TGFβ/SMAD2 Pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:4593-4603. [PMID: 33149556 PMCID: PMC7605600 DOI: 10.2147/dddt.s264715] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 09/23/2020] [Indexed: 12/13/2022]
Abstract
Purpose Idiopathic pulmonary fibrosis (IPF) is characterized by the accumulation of extracellular matrix (ECM) protein in the lungs. Transforming growth factor (TGF) β-induced ECM protein synthesis contributes to the development of IPF. Tranilast, an anti-allergy drug, suppresses TGFβ expression and inhibits interstitial renal fibrosis in animal models. However, the beneficial effects of tranilast or its mechanism as a therapy for pulmonary fibrosis have not been clarified. Methods We investigated the in vitro effect of tranilast on ECM production and TGFβ/SMAD2 pathway in TGFβ2-stimulated A549 human alveolar epithelial cells, using quantitative polymerase chain reaction, Western blotting, and immunofluorescence. In vitro observations were validated in the lungs of a murine pulmonary fibrosis model, which we developed by intravenous injection of bleomycin. Results Treatment with tranilast suppressed the expression of ECM proteins, such as fibronectin and type IV collagen, and attenuated SMAD2 phosphorylation in TGFβ2-stimulated A549 cells. In addition, based on a wound healing assay in these cells, tranilast significantly inhibited cell motility, with foci formation that comprised of ECM proteins. Histological analyses revealed that the administration of tranilast significantly attenuated lung fibrosis in mice. Furthermore, tranilast treatment significantly reduced levels of TGFβ, collagen, fibronectin, and phosphorylated SMAD2 in pulmonary fibrotic tissues in mice. Conclusion These findings suggest that tranilast inhibits pulmonary fibrosis by suppressing TGFβ/SMAD2-mediated ECM protein production, presenting tranilast as a promising and novel anti-fibrotic agent for the treatment of IPF.
Collapse
Affiliation(s)
- Motoyasu Kato
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Research Institute for Diseases of Old Ages, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Fumiyuki Takahashi
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Research Institute for Diseases of Old Ages, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tadashi Sato
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Research Institute for Diseases of Old Ages, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoichiro Mitsuishi
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Research Institute for Diseases of Old Ages, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ken Tajima
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Research Institute for Diseases of Old Ages, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hiroaki Ihara
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Research Institute for Diseases of Old Ages, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Fariz Nurwidya
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Research Institute for Diseases of Old Ages, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hario Baskoro
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Research Institute for Diseases of Old Ages, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akiko Murakami
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Research Institute for Diseases of Old Ages, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Isao Kobayashi
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Research Institute for Diseases of Old Ages, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Moulid Hidayat
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Research Institute for Diseases of Old Ages, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Naoko Shimada
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Research Institute for Diseases of Old Ages, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Leading Center for the Development and Research of Cancer Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shinichi Sasaki
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Research Institute for Diseases of Old Ages, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Reiko Mineki
- Laboratory of Proteomics and Biomolecular Science, Research Support Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tsutomu Fujimura
- Laboratory of Bioanalytical Chemistry, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Toshio Kumasaka
- Department of Pathology, Japanese Red Cross Medical Center, Tokyo, Japan
| | | | - Kazuhisa Takahashi
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Research Institute for Diseases of Old Ages, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Leading Center for the Development and Research of Cancer Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
30
|
Orlowski P, Zmigrodzka M, Tomaszewska E, Ranoszek-Soliwoda K, Pajak B, Slonska A, Cymerys J, Celichowski G, Grobelny J, Krzyzowska M. Polyphenol-Conjugated Bimetallic Au@AgNPs for Improved Wound Healing. Int J Nanomedicine 2020; 15:4969-4990. [PMID: 32764930 PMCID: PMC7369312 DOI: 10.2147/ijn.s252027] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 06/10/2020] [Indexed: 12/14/2022] Open
Abstract
Background Polyphenols possess antioxidant, anti-inflammatory and antimicrobial properties and have been used in the treatment of skin wounds and burns. We previously showed that tannic acid-modified AgNPs sized >26 nm promote wound healing, while tannic acid-modified AgNPs sized 13 nm can elicit strong local inflammatory response. In this study, we tested bimetallic Au@AgNPs sized 30 nm modified with selected flavonoid and non-flavonoid compounds for wound healing applications. Methods Bimetallic Au@AgNPs were obtained by growing an Ag layer on AuNPs and further modified with selected polyphenols. After toxicity tests and in vitro scratch assay in HaCaT cells, modified lymph node assay as well as the mouse splint wound model were further used to access the wound healing potential of selected non-toxic modifications. Results Tannic acid, gallic acid, polydatin, resveratrol, catechin, epicatechin, epigallocatechin, epicatechin gallate, epigallocatechin gallate and procyanidin B2 used to modify Au@AgNPs exhibited good toxicological profiles in HaCaT cells. Au@AgNPs modified with 15 μM tannic acid, 200 μM resveratrol, 200 μM epicatechin gallate, 1000 μM gallic acid and 200 μM procyanidin B2 induced wound healing in vivo and did not lead to the local irritation or inflammation. Tannic acid-modified Au@AgNPs induced epithelial-to-mesenchymal transition (EMT) - like re-epithelialization, while other polyphenol modifications of Au@AgNPs acted through proliferation and wound closure. Conclusion Bimetallic Au@AgNPs can be used as a basis for modification with selected polyphenols for topical uses. In addition, we have demonstrated that particular polyphenols used to modify bimetallic nanoparticles may show different effects upon different stages of wound healing.
Collapse
Affiliation(s)
- Piotr Orlowski
- Laboratory of Nanobiology and Biomaterials, Military Institute of Hygiene and Epidemiology, Warsaw, Poland
| | - Magdalena Zmigrodzka
- Department of Pathology and Veterinary Diagnostics, Institute of Veterinary Medicine, Warsaw University of Life Sciences (WULS-SGGW), Warsaw, Poland
| | - Emilia Tomaszewska
- Department of Materials Technology and Chemistry, Faculty of Chemistry, University of Lodz, Lodz, Poland
| | | | - Beata Pajak
- Laboratory of Genetics and Molecular Biology, Military Institute of Hygiene and Epidemiology, Warsaw, Poland
| | - Anna Slonska
- Division of Microbiology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw 02-786, Poland
| | - Joanna Cymerys
- Division of Microbiology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw 02-786, Poland
| | - Grzegorz Celichowski
- Department of Materials Technology and Chemistry, Faculty of Chemistry, University of Lodz, Lodz, Poland
| | - Jaroslaw Grobelny
- Department of Materials Technology and Chemistry, Faculty of Chemistry, University of Lodz, Lodz, Poland
| | - Malgorzata Krzyzowska
- Laboratory of Nanobiology and Biomaterials, Military Institute of Hygiene and Epidemiology, Warsaw, Poland
| |
Collapse
|
31
|
Shahbazi MA, Shrestha N, Pierchala MK, Kadumudi FB, Mehrali M, Hasany M, Préat V, Leeuwenburgh S, Dolatshahi-Pirouz A. A self-healable, moldable and bioactive biomaterial gum for personalised and wearable drug delivery. J Mater Chem B 2020; 8:4340-4356. [PMID: 32363370 DOI: 10.1039/c9tb02156f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
One of the long-standing challenges in materials science involves synthesizing biomaterials that recapitulate important features of native biological tissues. Even though, the number of available biomaterials at the moment are virtually limitless, few of them has unlocked all the secrets of the human body by mimicking the combinatorial-like material properties of our tissues and organs. Inspired by the human body, we have developed a polymeric gum, which combines stretchability, toughness, strength, flexibility, and self-healing. It also exhibits a high bioactivity that can target and eliminate bacterial infections fast and reliably. Notably, this material is moldable into almost any complex shape, and therefore suitable as a building block for wearables designed to conform directly with the curved and personalized anatomy of patients. It also exhibits excellent drug retention and release capacity, which altogether makes it suitable for applications in personalized wearable drug-delivery devices.
Collapse
Affiliation(s)
- Mohammad-Ali Shahbazi
- Department of Micro- and Nanotechnology, Technical University of Denmark, Ørsteds Plads, DK-2800 Kgs, Lyngby, Denmark
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Rajasekar N, Sivanantham A, Kar A, Mahapatra SK, Ahirwar R, Thimmulappa RK, Paramasivam SG, Subbiah R. Tannic acid alleviates experimental pulmonary fibrosis in mice by inhibiting inflammatory response and fibrotic process. Inflammopharmacology 2020; 28:1301-1314. [PMID: 32372165 DOI: 10.1007/s10787-020-00707-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/09/2020] [Indexed: 12/20/2022]
Abstract
Pulmonary fibrosis (PF) is a chronic and irreversible scarring disease in the lung with limited treatment options. Therefore, it is critical to identify new therapeutic options. This study was undertaken to identify the effects of tannic acid (TA), a naturally occurring dietary polyphenol, in a mouse model of PF. Bleomycin (BLM) was intratracheally administered to induce PF. Administration of TA significantly reduced BLM-induced histological alterations, inflammatory cell infiltration and the levels of various inflammatory mediators (nitric oxide, leukotriene B4 and cytokines). Additionally, treatment with TA also impaired BLM-mediated increases in pro-fibrotic (transforming growth factor-β1) and fibrotic markers (alpha-smooth muscle actin, vimentin, collagen 1 alpha and fibronectin) expression. Further investigation indicated that BLM-induced phosphorylation of Erk1/2 (extracellular signal-regulated kinases 1 and 2) in lungs was suppressed by TA treatment. Findings of this study suggest that TA has the potential to mitigate PF through inhibiting the inflammatory response and fibrotic process in lungs and that TA might be useful for the treatment of PF in clinical practice.
Collapse
Affiliation(s)
- Nandhine Rajasekar
- Department of Biotechnology, BIT-Campus, Anna University, Tiruchirappalli, Tamil Nadu, 620024, India
| | - Ayyanar Sivanantham
- Department of Biotechnology, BIT-Campus, Anna University, Tiruchirappalli, Tamil Nadu, 620024, India
| | - Amrita Kar
- Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, Tamil Nadu, 613401, India
| | - Santanu Kar Mahapatra
- Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, Tamil Nadu, 613401, India
| | - Rajesh Ahirwar
- Department of Biochemistry, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh, 462030, India
| | - Rajesh K Thimmulappa
- Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education and Research, Mysore, Karnataka, 570015, India
| | | | - Rajasekaran Subbiah
- Department of Biotechnology, BIT-Campus, Anna University, Tiruchirappalli, Tamil Nadu, 620024, India. .,Department of Biochemistry, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh, 462030, India.
| |
Collapse
|
33
|
Baer-Dubowska W, Szaefer H, Majchrzak-Celińska A, Krajka-Kuźniak V. Tannic Acid: Specific Form of Tannins in Cancer Chemoprevention and Therapy-Old and New Applications. ACTA ACUST UNITED AC 2020. [DOI: 10.1007/s40495-020-00211-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Abstract
Purpose of Review
This short review is aimed at providing an updated and comprehensive report on tannic acid biological activities and molecular mechanisms of action most important for cancer prevention and adjuvant therapy.
Recent Findings
Tannic acid (TA), a mixture of digallic acid esters of glucose, is a common ingredient of many foods. The early studies of its anti-mutagenic and anti-tumorigenic activity were mostly demonstrated in the mouse skin model. This activity has been explained by its ability to inhibit carcinogens activation, as well as antioxidant and anti-inflammatory properties. Recently, the cell cycle arrest, apoptosis induction, reduced rate of proliferation, and cell migration and adhesion of several cancer cell lines as a result of TA treatment were described. The underlining mechanisms include modulation of signaling pathways such as EGFR/Jak2/STATs, or inhibition of PKM2 glycolytic enzyme. Moreover, epithelial-to-mesenchymal transition prevention and decrease of cancer stem cells formation by TA were also reported. Besides, TA was found to be potent chemosensitizer overcoming multidrug resistance. Eventually, its specific physicochemical features were found useful for generation of drug-loaded nanoparticles.
Summary
TA was shown to be a very versatile molecule with possible application not only in cancer prophylaxis, as was initially thought, but also in adjuvant cancer therapy. The latter may refer to chemosensitization and its application as a part of drug delivery systems. More studies are required to better explore this subject. In addition, the effect of TA on normal cells and its bioavailability have to better characterized.
Collapse
|
34
|
Pu Y, Liu YQ, Zhou Y, Qi YF, Liao SP, Miao SK, Zhou LM, Wan LH. Dual role of RACK1 in airway epithelial mesenchymal transition and apoptosis. J Cell Mol Med 2020; 24:3656-3668. [PMID: 32064783 PMCID: PMC7131927 DOI: 10.1111/jcmm.15061] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/04/2020] [Accepted: 01/21/2020] [Indexed: 02/05/2023] Open
Abstract
Airway epithelial apoptosis and epithelial mesenchymal transition (EMT) are two crucial components of asthma pathogenesis, concomitantly mediated by TGF‐β1. RACK1 is the downstream target gene of TGF‐β1 shown to enhancement in asthma mice in our previous study. Balb/c mice were sensitized twice and challenged with OVA every day for 7 days. Transformed human bronchial epithelial cells, BEAS‐2B cells were cultured and exposed to recombinant soluble human TGF‐β1 to induced apoptosis (30 ng/mL, 72 hours) and EMT (10 ng/mL, 48 hours) in vitro, respectively. siRNA and pharmacological inhibitors were used to evaluate the regulation of RACK1 protein in apoptosis and EMT. Western blotting analysis and immunostaining were used to detect the protein expressions in vivo and in vitro. Our data showed that RACK1 protein levels were significantly increased in OVA‐challenged mice, as well as TGF‐β1‐induced apoptosis and EMT of BEAS‐2B cells. Knockdown of RACK1 (siRACK1) significantly inhibited apoptosis and decreased TGF‐β1 up‐regulated EMT related protein levels (N‐cadherin and Snail) in vitro via suppression of JNK and Smad3 activation. Moreover, siSmad3 or siJNK impaired TGF‐β1‐induced N‐cadherin and Snail up‐regulation in vitro. Importantly, JNK gene silencing (siERK) also impaired the regulatory effect of TGF‐β1 on Smad3 activation. Our present data demonstrate that RACK1 is a concomitant regulator of TGF‐β1 induces airway apoptosis and EMT via JNK/Smad/Snail signalling axis. Our findings may provide a new insight into understanding the regulation mechanism of RACK1 in asthma pathogenesis.
Collapse
Affiliation(s)
- Yue Pu
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Yuan-Qi Liu
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Yan Zhou
- Department of Intensive Care Unit, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Yi-Fan Qi
- Grade 2015, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Shi-Ping Liao
- Functional Laboratory, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Shi-Kun Miao
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Li-Ming Zhou
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Li-Hong Wan
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, PR China
| |
Collapse
|
35
|
Han X, Na T, Wu T, Yuan BZ. Human lung epithelial BEAS-2B cells exhibit characteristics of mesenchymal stem cells. PLoS One 2020; 15:e0227174. [PMID: 31900469 PMCID: PMC6941928 DOI: 10.1371/journal.pone.0227174] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 12/06/2019] [Indexed: 12/15/2022] Open
Abstract
BEAS-2B was originally established as an immortalized but non-tumorigenic epithelial cell line from human bronchial epithelium. Because of general recognition for its bronchial epithelial origin, the BEAS-2B cell line has been widely used as an in vitro cell model in a large variety of studies associated with respiratory diseases including lung carcinogenesis. However, very few studies have discussed non-epithelial features of BEAS-2B cells, especially the features associated with mesenchymal stem cells (MSCs), which represent a group of fibroblast-like cells with limited self-renewal and differentiation potential to various cell lineages. In this study, we compared BEAS-2B with a human umbilical cord-derived MSCs (hMSCs) cell line, hMSC1, which served as a representative of hMSCs in terms of expressing common features of hMSCs. It was observed that both BEAS-2B and hMSC1 shared the same expression profile of surface markers of hMSCs and exhibited similar osteogenic and adipogenic differentiation potential. In addition, like hMSC1, the BEAS-2B cell line exhibited suppressive activities on proliferation of mitogen-activated total T lymphocytes as well as Th1 lymphocytes, and IFNγ-induced expression of IDO1, all thus demonstrating that BEAS-2B cells exhibited an almost identical characteristic profile with hMSCs, even though, there was a clear difference between BEAS-2B and hMSCs in the effects on type 2 macrophage polarization. Most importantly, the hMSCs features of BEAS-2B were unlikely a consequence of epithelial-mesenchymal transition. Therefore, this study provided a set of evidence to provoke reconsideration of epithelial origin of BEAS-2B.
Collapse
Affiliation(s)
- Xiaoyan Han
- Cell Collection and Research Center, National Institutes for Food and Drug Control, Beijing, China
| | - Tao Na
- Cell Collection and Research Center, National Institutes for Food and Drug Control, Beijing, China
| | - Tingting Wu
- Cell Collection and Research Center, National Institutes for Food and Drug Control, Beijing, China
| | - Bao-Zhu Yuan
- Cell Collection and Research Center, National Institutes for Food and Drug Control, Beijing, China
- * E-mail:
| |
Collapse
|
36
|
Lee CH. Reversal of Epithelial-Mesenchymal Transition by Natural Anti-Inflammatory and Pro-Resolving Lipids. Cancers (Basel) 2019; 11:E1841. [PMID: 31766574 PMCID: PMC6966475 DOI: 10.3390/cancers11121841] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 11/16/2019] [Accepted: 11/19/2019] [Indexed: 02/08/2023] Open
Abstract
Epithelial mesenchymal transition (EMT) is a key process in the progression of malignant cancer. Therefore, blocking the EMT can be a critical fast track for the development of anticancer drugs. In this paper, we update recent research output of EMT and we explore suppression of EMT by natural anti-inflammatory compounds and pro-resolving lipids.
Collapse
Affiliation(s)
- Chang Hoon Lee
- College of Pharmacy, Dongguk University, Seoul 100-715, Korea
| |
Collapse
|
37
|
Sivanantham A, Pattarayan D, Rajasekar N, Kannan A, Loganathan L, Bethunaickan R, Mahapatra SK, Palanichamy R, Muthusamy K, Rajasekaran S. Tannic acid prevents macrophage-induced pro-fibrotic response in lung epithelial cells via suppressing TLR4-mediated macrophage polarization. Inflamm Res 2019; 68:1011-1024. [DOI: 10.1007/s00011-019-01282-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/27/2019] [Accepted: 08/29/2019] [Indexed: 02/08/2023] Open
|
38
|
Avila-Carrasco L, Majano P, Sánchez-Toméro JA, Selgas R, López-Cabrera M, Aguilera A, González Mateo G. Natural Plants Compounds as Modulators of Epithelial-to-Mesenchymal Transition. Front Pharmacol 2019; 10:715. [PMID: 31417401 PMCID: PMC6682706 DOI: 10.3389/fphar.2019.00715] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 06/05/2019] [Indexed: 12/13/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a self-regulated physiological process required for tissue repair that, in non-controled conditions may lead to fibrosis, angiogenesis, loss of normal organ function or cancer. Although several molecular pathways involved in EMT regulation have been described, this process does not have any specific treatment. This article introduces a systematic review of effective natural plant compounds and their extract that modulates the pathological EMT or its deleterious effects, through acting on different cellular signal transduction pathways both in vivo and in vitro. Thereby, cryptotanshinone, resveratrol, oxymatrine, ligustrazine, osthole, codonolactone, betanin, tannic acid, gentiopicroside, curcumin, genistein, paeoniflorin, gambogic acid and Cinnamomum cassia extracts inhibit EMT acting on transforming growth factor-β (TGF-β)/Smads signaling pathways. Gedunin, carnosol, celastrol, black rice anthocyanins, Duchesnea indica, cordycepin and Celastrus orbiculatus extract downregulate vimectin, fibronectin and N-cadherin. Sulforaphane, luteolin, celastrol, curcumin, arctigenin inhibit β-catenin signaling pathways. Salvianolic acid-A and plumbagin block oxidative stress, while honokiol, gallic acid, piperlongumine, brusatol and paeoniflorin inhibit EMT transcription factors such as SNAIL, TWIST and ZEB. Plectranthoic acid, resveratrol, genistein, baicalin, polyphyllin I, cairicoside E, luteolin, berberine, nimbolide, curcumin, withaferin-A, jatrophone, ginsenoside-Rb1, honokiol, parthenolide, phoyunnanin-E, epicatechin-3-gallate, gigantol, eupatolide, baicalin and baicalein and nitidine chloride inhibit EMT acting on other signaling pathways (SIRT1, p38 MAPK, NFAT1, SMAD, IL-6, STAT3, AQP5, notch 1, PI3K/Akt, Wnt/β-catenin, NF-κB, FAK/AKT, Hh). Despite the huge amount of preclinical data regarding EMT modulation by the natural compounds of plant, clinical translation is poor. Additionally, this review highlights some relevant examples of clinical trials using natural plant compounds to modulate EMT and its deleterious effects. Overall, this opens up new therapeutic alternatives in cancer, inflammatory and fibrosing diseases through the control of EMT process.
Collapse
Affiliation(s)
- Lorena Avila-Carrasco
- Therapeutic and Pharmacology Department, Health and Human Science Research, Academic Unit of Human Medicine and Health Sciences, Autonomous University of Zacatecas, Zacatecas, Mexico
| | - Pedro Majano
- Molecular Biology Unit, Research Institute of University Hospital La Princesa (IP), Madrid, Spain
| | - José Antonio Sánchez-Toméro
- Department and Nephrology, Research Institute of University Hospital La Princesa (IP), Madrid, Spain.,Renal research network REDINREN, Madrid, Spain
| | - Rafael Selgas
- Research Institute of La Paz (IdiPAZ), University Hospital La Paz, Madrid, Spain.,Renal research network REDINREN, Madrid, Spain
| | - Manuel López-Cabrera
- Renal research network REDINREN, Madrid, Spain.,Molecular Biology Research Centre Severo Ochoa, Spanish Council for Scientific Research (CSIC), Madrid, Spain
| | - Abelardo Aguilera
- Molecular Biology Unit, Research Institute of University Hospital La Princesa (IP), Madrid, Spain.,Renal research network REDINREN, Madrid, Spain
| | - Guadalupe González Mateo
- Research Institute of La Paz (IdiPAZ), University Hospital La Paz, Madrid, Spain.,Renal research network REDINREN, Madrid, Spain.,Molecular Biology Research Centre Severo Ochoa, Spanish Council for Scientific Research (CSIC), Madrid, Spain
| |
Collapse
|
39
|
Reed EB, Ard S, La J, Park CY, Culligan L, Fredberg JJ, Smolyaninova LV, Orlov SN, Chen B, Guzy R, Mutlu GM, Dulin NO. Anti-fibrotic effects of tannic acid through regulation of a sustained TGF-beta receptor signaling. Respir Res 2019; 20:168. [PMID: 31358001 PMCID: PMC6664561 DOI: 10.1186/s12931-019-1141-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 07/22/2019] [Indexed: 12/23/2022] Open
Abstract
Background Pulmonary fibrosis is a progressive disease characterized by structural distortion of the lungs. Transforming growth factor-beta (TGF-beta) is a key cytokine implicated in the pathogenesis of pulmonary fibrosis. TGF-beta-induced myofibroblast differentiation characterized by expression of smooth muscle alpha-actin and extracellular matrix proteins is a key process in pathogenesis of fibrotic disease. Tannic acid is a natural polyphenol with diverse applications. In this study, we investigated the effect of tannic acid on myofibroblast differentiation and pulmonary fibrosis in cultured cells and in bleomycin model of the disease. Methods Primary cultured human lung fibroblasts (HLF) were used. The relative levels of proteins were determined by Western blotting. HLF contraction was measured by traction microscopy. Bleomycin-induced pulmonary fibrosis in mice was used as the disease model. Results Tannic acid inhibited TGF-beta-induced expression of collagen-1 and smooth muscle alpha-actin (SMA) as well as force generation by HLF. Tannic acid did not affect initial phosphorylation of Smad2 in response to TGF-beta, but significantly inhibited sustained Smad2 phosphorylation, which we recently described to be critical for TGF-beta-induced myofibroblast differentiation. Accordingly, tannic acid inhibited Smad-dependent gene transcription in response to TGF-beta, as assessed using luciferase reporter for the activity of Smad-binding elements. Finally, in mouse model of bleomycin-induced pulmonary fibrosis, therapeutic application of tannic acid resulted in a significant reduction of lung fibrosis, decrease in collagen-1 content and of Smad2 phosphorylation in the lungs. Conclusions This study demonstrates the anti-fibrotic effect of tannic acid in vitro and in vivo through a regulation of sustained Smad2 phosphorylation. Electronic supplementary material The online version of this article (10.1186/s12931-019-1141-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Eleanor B Reed
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, the University of Chicago, 5841 S. Maryland Ave, MC6076, Chicago, IL, 60637, USA
| | - Shawn Ard
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, the University of Chicago, 5841 S. Maryland Ave, MC6076, Chicago, IL, 60637, USA
| | - Jennifer La
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, the University of Chicago, 5841 S. Maryland Ave, MC6076, Chicago, IL, 60637, USA
| | - Chan Young Park
- Molecular and Integrative Physiological Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Laura Culligan
- Molecular and Integrative Physiological Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jeffrey J Fredberg
- Molecular and Integrative Physiological Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Larisa V Smolyaninova
- Laboratory of Biomembranes, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russian Federation
| | - Sergei N Orlov
- Laboratory of Biomembranes, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russian Federation.,Siberian Medical State University, Tomsk, Russian Federation
| | - Bohao Chen
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, the University of Chicago, 5841 S. Maryland Ave, MC6076, Chicago, IL, 60637, USA
| | - Robert Guzy
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, the University of Chicago, 5841 S. Maryland Ave, MC6076, Chicago, IL, 60637, USA
| | - Gökhan M Mutlu
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, the University of Chicago, 5841 S. Maryland Ave, MC6076, Chicago, IL, 60637, USA
| | - Nickolai O Dulin
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, the University of Chicago, 5841 S. Maryland Ave, MC6076, Chicago, IL, 60637, USA.
| |
Collapse
|
40
|
Bang IJ, Kim HR, Jeon Y, Jeong MH, Park YJ, Kwak JH, Chung KH. β-Peltoboykinolic Acid from Astilbe rubra Attenuates TGF-β1-Induced Epithelial-to-Mesenchymal Transitions in Lung Alveolar Epithelial Cells. Molecules 2019; 24:molecules24142573. [PMID: 31311194 PMCID: PMC6680586 DOI: 10.3390/molecules24142573] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/09/2019] [Accepted: 07/14/2019] [Indexed: 01/04/2023] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is increasingly recognized as contributing to the pathogenesis of idiopathic pulmonary fibrosis. Therefore, novel plant-based natural, active compounds have been sought for the treatment of fibrotic EMT. The aim of the present study was to investigate the inhibitory effects of Astilbe rubra on TGF-β1-induced EMT in lung alveolar epithelial cells (A549). A. rubra was subjected to extraction using 70% ethanol (ARE), and ethanol extracts of the aerial part and that of the rhizome were further partitioned using various solvents. Protein expression and cell motility were investigated to evaluate the inhibitory effects of ARE on EMT. EMT occurred in A549 cells treated with TGF-β1, but was prevented by co-treatment with ARE. The dichloromethane fractions showed the strongest inhibitory effect on TGF-β1-induced EMT. β-Peltoboykinolic acid was isolated from the dichloromethane fractions of A. rubra by activity-oriented isolation. β-Peltoboykinolic acid not only attenuated TGF-β1-induced EMT, but also the overproduction of extracellular matrix components including type I collagen and fibronectin. The Smad pathway activated by TGF-β1 was inhibited by co-treatment with β-peltoboykinolic acid. Taken together, these results indicate that β-peltoboykinolic acid from A. rubra and dichloromethane fractions shows potential as an antifibrotic agent in A549 cells treated with TGF-β1.
Collapse
Affiliation(s)
- In Jae Bang
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Ha Ryong Kim
- College of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Korea
| | - Yukyoung Jeon
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Mi Ho Jeong
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Yong Joo Park
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Jong Hwan Kwak
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Kyu Hyuck Chung
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea.
| |
Collapse
|
41
|
Kawami M, Harada R, Ojima T, Yamagami Y, Yumoto R, Takano M. Association of cell cycle arrest with anticancer drug-induced epithelial-mesenchymal transition in alveolar epithelial cells. Toxicology 2019; 424:152231. [PMID: 31170432 DOI: 10.1016/j.tox.2019.06.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 05/28/2019] [Accepted: 06/03/2019] [Indexed: 02/07/2023]
Abstract
Many drugs exert serious cytotoxic effects on pulmonary tissues. Although several reports have shown an association of epithelial-mesenchymal transition (EMT) with anticancer drug-induced lung injury, mechanisms of these effects are poorly understood. In the present study, we evaluated mechanisms of anticancer drug-induced EMT, with a focus on involvement of cell cycle arrest. We found that methotrexate (MTX) altered mRNA expression levels of many genes as determined by microarray analysis. Gene set enrichment analysis revealed that cell cycle arrest pathways may be associated with MTX-induced EMT. In addition, thymidine (THY) and nocodazole (NOC), which induce cell cycle arrest at S-phase and G2/M-phase, increased mRNA expression levels of α-smooth muscle actin (SMA), an EMT marker. Furthermore, α-SMA protein expression in cells arrested at S- and G2/M-phases by MTX and paclitaxel (PTX) was significantly higher than that in cells at G1. Notably, co-treatment of cells with THY or NOC and EMT-inducing anticancer drugs did not result in additional upregulation of α-SMA mRNA expression. These findings suggested that cell cycle arrest may be closely associated with anticancer drug-induced EMT in alveolar epithelial cells.
Collapse
Affiliation(s)
- Masashi Kawami
- Department of Pharmaceutics and Therapeutics, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Risako Harada
- Department of Pharmaceutics and Therapeutics, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Takamichi Ojima
- Department of Pharmaceutics and Therapeutics, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Yohei Yamagami
- Department of Pharmaceutics and Therapeutics, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Ryoko Yumoto
- Department of Pharmaceutics and Therapeutics, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Mikihisa Takano
- Department of Pharmaceutics and Therapeutics, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan.
| |
Collapse
|
42
|
Combined Activation of Guanylate Cyclase and Cyclic AMP in Lung Fibroblasts as a Novel Therapeutic Concept for Lung Fibrosis. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1345402. [PMID: 30984775 PMCID: PMC6431482 DOI: 10.1155/2019/1345402] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 12/19/2018] [Accepted: 01/28/2019] [Indexed: 12/21/2022]
Abstract
Remodelling of the peripheral lung tissue and fibrotic foci are the main pathologies of idiopathic pulmonary fibrosis (IPF), a disease that is difficult to treat. TGF-β activation of peripheral lung fibroblasts is indicated as the major cause of tissue remodelling in IPF and is resulting in fibroblast hyperplasia and deposition of extracellular matrix. Soluble guanylate cyclase (sGC) stimulators combined with cyclic AMP (cAMP) activators have been reported to reduce proliferation and matrix deposition in other conditions than IPF. Therefore, this drug combination may present a novel therapeutic concept for IPF. This study investigated the effect of BAY 41-2272 and forskolin on remodelling parameters in primary human lung fibroblasts. The study determined TGF-β induced proliferation by direct cell counts after 3 days; and deposition of collagen type-I, type III, and fibronectin. BAY 41-2272 significantly reduced TGF-β induced fibroblast proliferation, but did not reduce viability. This inhibitory effect was further supported by forskolin. Both BAY 41-2272 and forskolin alone reduced TGF-β induced collagen and fibronectin de novo synthesis as well as deposition. This effect was significantly stronger when the two compounds were combined. Furthermore, the TGF-β induced expression of fibrilar α-smooth muscle actin was reduced by BAY 41-2272 and this effect was strengthened by forskolin. In addition, BAY 41-2272 and forskolin reduced TGF-β induced β-catenin. All effects of BAY 41-2272 were concentration dependent. The findings suggest that BAY 41-2272 in combination with cAMP stimulation may present a novel therapeutic strategy to reduce tissue remodelling in IPF.
Collapse
|
43
|
Yang Y, Hu L, Xia H, Chen L, Cui S, Wang Y, Zhou T, Xiong W, Song L, Li S, Pan S, Xu J, Liu M, Xiao H, Qin L, Shang Y, Yao S. Resolvin D1 attenuates mechanical stretch-induced pulmonary fibrosis via epithelial-mesenchymal transition. Am J Physiol Lung Cell Mol Physiol 2019; 316:L1013-L1024. [PMID: 30724098 DOI: 10.1152/ajplung.00415.2018] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Mechanical ventilation-induced pulmonary fibrosis plays an important role in the high mortality rate of acute respiratory distress syndrome (ARDS). Resolvin D1 (RvD1) displays potent proresolving activities. Epithelial-mesenchymal transition (EMT) has been proved to be an important pathological feature of lung fibrosis. This study aimed to investigate whether RvD1 can attenuate mechanical ventilation-induced lung fibrosis. Human lung epithelial (BEAS-2B) cells were pretreated with RvD1 for 30 min and exposed to acid for 10 min before being subjected to mechanical stretch for 48 h. C57BL/6 mice were subjected to intratracheal acid aspiration followed by mechanical ventilation 24 h later (peak inspiratory pressure 22 cmH2O, positive end-expiratory pressure 2 cmH2O, and respiratory rate 120 breaths/min for 2 h). RvD1 was injected into mice for 5 consecutive days after mechanical ventilation. Treatment with RvD1 significantly inhibited mechanical stretch-induced mesenchymal markers (vimentin and α-smooth muscle actin) and stimulated epithelial markers (E-cadherin). Tert-butyloxycarbonyl 2 (BOC-2), a lipoxin A4 receptor/formyl peptide receptor 2 (ALX/FPR2) antagonist, is known to inhibit ALX/FPR2 function. BOC-2 could reverse the beneficial effects of RvD1. The antifibrotic effect of RvD1 was associated with the suppression of Smad2/3 phosphorylation. This study demonstrated that mechanical stretch could induce EMT and pulmonary fibrosis and that treatment with RvD1 could attenuate mechanical ventilation-induced lung fibrosis, thus highlighting RvD1 as an effective therapeutic agent against pulmonary fibrosis associated with mechanical ventilation.
Collapse
Affiliation(s)
- Yiyi Yang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China
| | - Lisha Hu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China
| | - Haifa Xia
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China
| | - Lin Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China
| | - Shunan Cui
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China
| | - Yaxin Wang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China
| | - Ting Zhou
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China
| | - Wei Xiong
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China
| | - Limin Song
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China
| | - Shengnan Li
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China
| | - Shangwen Pan
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China
| | - Jiqian Xu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China
| | - Min Liu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China
| | - Hairong Xiao
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China
| | - Lu Qin
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China
| | - You Shang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China
| | - Shanglong Yao
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei , China
| |
Collapse
|
44
|
Yao Z, Zheng W, Zhang X, Xiong H, Qian Y, Fan C. Hydroxycamptothecin Prevents Fibrotic Pathways in Fibroblasts In Vitro. IUBMB Life 2019; 71:653-662. [PMID: 30690843 DOI: 10.1002/iub.2013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 12/12/2018] [Accepted: 12/29/2018] [Indexed: 01/23/2023]
Abstract
Peritendinous fibrosis, which leads to impaired tendon function, is a clinical problem worldwide, and it is urgent to explore potential ways to reduce the formation of peritendinous adhesion. Several studies have demonstrated the biological roles of hydroxycamptothecin (HCPT) in inhibiting fibrosis in different tissues. In this study, we investigated whether HCPT could inhibit tendon fibrosis in vitro. Our results revealed that HCPT inhibited transforming growth factor (TGF)-β1-induced cell viability of human fibroblasts, decreased excessive cell hyperproliferation and promoted fibroblasts apoptosis. In addition, HCPT treatment also inhibited expression of fibrosis genes COL3A1 and α-smooth muscle actin (α-SMA). In terms of mechanism, we pretreated fibroblasts with the endoplasmic reticulum stress (ER) inhibitor salubrinal and RNA-dependent protein kinase-like ER kinase (PERK) short hairpin RNA, these treatments abolished the inhibitory effects of HCPT on fibrosis, thereby suggesting that HCPT's inhibition of TGF-β1-induced tendon fibrosis might be mediated by the PERK signaling pathway in vitro. In conclusion, our results suggested that HCPT had protective effects on peritendinous tissue fibrosis and might be promising in future clinical applications. © 2019 IUBMB Life, 71(5):653-662, 2019.
Collapse
Affiliation(s)
- Zhixiao Yao
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Wei Zheng
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xiangqi Zhang
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Hao Xiong
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yun Qian
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Cunyi Fan
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
45
|
Fix C, Carver-Molina A, Chakrabarti M, Azhar M, Carver W. Effects of the isothiocyanate sulforaphane on TGF-β1-induced rat cardiac fibroblast activation and extracellular matrix interactions. J Cell Physiol 2019; 234:13931-13941. [PMID: 30609032 DOI: 10.1002/jcp.28075] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 12/11/2018] [Indexed: 12/30/2022]
Abstract
An important step in many pathological conditions, particularly tissue and organ fibrosis, is the conversion of relatively quiescent cells into active myofibroblasts. These are highly specialized cells that participate in normal wound healing but also contribute to pathogenesis. These cells possess characteristics of smooth muscle cells and fibroblasts, have enhanced synthetic activity secreting abundant extracellular matrix components, cytokines, and growth factors, and are capable of generating contractile force. As such, these cells have become potential therapeutic targets in a number of disease settings. Transforming growth factor β (TGF-β) is a potent stimulus of fibrosis and myofibroblast formation and likewise is an important therapeutic target in several disease conditions. The plant-derived isothiocyanate sulforaphane has been shown to have protective effects in several pathological models including diabetic cardiomyopathy, carcinogenesis, and fibrosis. These studies suggest that sulforaphane may be an attractive preventive agent against disease progression, particularly in conditions involving alterations of the extracellular matrix and activation of myofibroblasts. However, few studies have evaluated the effects of sulforaphane on cardiac fibroblast activation and their interactions with the extracellular matrix. The present studies were carried out to determine the potential effects of sulforaphane on the conversion of quiescent cardiac fibroblasts to an activated myofibroblast phenotype and associated alterations in signaling, expression of extracellular matrix receptors, and cellular physiology following stimulation with TGF-β1. These studies demonstrate that sulforaphane attenuates TGF-β1-induced myofibroblast formation and contractile activity. Sulforaphane also reduces expression of collagen-binding integrins and inhibits canonical and noncanonical TGF-β signaling pathways.
Collapse
Affiliation(s)
- Charity Fix
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina
| | - Amanda Carver-Molina
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina
| | - Mrinmay Chakrabarti
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina
| | - Mohamad Azhar
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina
| | - Wayne Carver
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina
| |
Collapse
|
46
|
Sivanantham A, Pattarayan D, Bethunaickan R, Kar A, Mahapatra SK, Thimmulappa RK, Palanichamy R, Rajasekaran S. Tannic acid protects against experimental acute lung injury through downregulation of TLR4 and MAPK. J Cell Physiol 2018; 234:6463-6476. [DOI: 10.1002/jcp.27383] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 08/17/2018] [Indexed: 01/11/2023]
Affiliation(s)
- Ayyanar Sivanantham
- Department of Biotechnology BIT‐Campus, Anna University Tiruchirappalli India
| | | | | | - Amrita Kar
- Centre for Research in Infectious Diseases (CRID) School of Chemical & Biotechnology, SASTRA Deemed To Be University Thanjavur India
| | - Santanu Kar Mahapatra
- Centre for Research in Infectious Diseases (CRID) School of Chemical & Biotechnology, SASTRA Deemed To Be University Thanjavur India
| | - Rajesh K. Thimmulappa
- Department of Biochemistry Center of Excellence in Molecular Biology & Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education and Research Mysuru India
| | | | - Subbiah Rajasekaran
- Department of Biotechnology BIT‐Campus, Anna University Tiruchirappalli India
| |
Collapse
|
47
|
Song D, Zhao J, Deng W, Liao Y, Hong X, Hou J. Tannic acid inhibits NLRP3 inflammasome-mediated IL-1β production via blocking NF-κB signaling in macrophages. Biochem Biophys Res Commun 2018; 503:3078-3085. [PMID: 30126633 DOI: 10.1016/j.bbrc.2018.08.096] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 08/13/2018] [Indexed: 11/19/2022]
Abstract
The NLRP3 inflammasome rapidly responds to many infections and stress signals and is involved in the pathogenesis of numerous inflammatory disease processes. Tannic acid plays a role in antioxidant, antifungal and antitumor activities. Here, we reported that tannic acid inhibited NLRP3 inflammasome activation by blocking NF-κB signaling to suppress IL-1β secretion. We found that the BMDMs (bone marrow-derived macrophages cells) pre-treated with tannic acid blocked caspase-1 cleavage and inhibited IL-1β secretion in a NLRP3-dependent manner, and suppressed NF-κB signaling activation by inhibiting NF-κB/P65 nuclear localization, suggesting that tannic acid inhibited NLRP3 inflammasome activation. These investigations revealed that tannic acid inhibited NLRP3 inflammasome activation via blocking NF-κB signaling in macrophages, providing us with evidence that tannic acid may be a potent inhibitor for NLRP3-driven diseases.
Collapse
Affiliation(s)
- Dan Song
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Jiabao Zhao
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, 361102, China; Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, Xiamen, Fujian, 361004, China
| | - Weixian Deng
- Respiratory Medicine, Second Affiliated Hospital of Xiamen Medical College, Xiamen, Fujian, 361000, China
| | - Yueting Liao
- Blood Transfusion Department, Zhongshan Hospital of Xiamen University, Xiamen, Fujian, 361004, China
| | - Xuehui Hong
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, Xiamen, Fujian, 361004, China; Department of Gastrointestinal Surgery, Institute of Gastrointestinal Oncology, Medical College of Xiamen University, Xiamen, Fujian, 361004, China.
| | - Jingjing Hou
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, Xiamen, Fujian, 361004, China; Department of Gastrointestinal Surgery, Institute of Gastrointestinal Oncology, Medical College of Xiamen University, Xiamen, Fujian, 361004, China; State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, 361102, China.
| |
Collapse
|
48
|
Rout-Pitt N, Farrow N, Parsons D, Donnelley M. Epithelial mesenchymal transition (EMT): a universal process in lung diseases with implications for cystic fibrosis pathophysiology. Respir Res 2018; 19:136. [PMID: 30021582 PMCID: PMC6052671 DOI: 10.1186/s12931-018-0834-8] [Citation(s) in RCA: 188] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 06/25/2018] [Indexed: 12/21/2022] Open
Abstract
Cystic Fibrosis (CF) is a genetic disorder that arises due to mutations in the Cystic Fibrosis Transmembrane Conductance Regulator gene, which encodes for a protein responsible for ion transport out of epithelial cells. This leads to a disruption in transepithelial Cl-, Na + and HCO3− ion transport and the subsequent dehydration of the airway epithelium, resulting in infection, inflammation and development of fibrotic tissue. Unlike in CF, fibrosis in other lung diseases including asthma, chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis has been well characterised. One of the driving forces behind fibrosis is Epithelial Mesenchymal Transition (EMT), a process where epithelial cells lose epithelial proteins including E-Cadherin, which is responsible for tight junctions. The cell moves to a more mesenchymal phenotype as it gains mesenchymal markers such as N-Cadherin (providing the cells with migration potential), Vimentin and Fibronectin (proteins excreted to help form the extracellular matrix), and the fibroblast proliferation transcription factors Snail, Slug and Twist. This review paper explores the EMT process in a range of lung diseases, details the common links that these have to cystic fibrosis, and explores how understanding EMT in cystic fibrosis may open up novel methods of treating patients with cystic fibrosis.
Collapse
Affiliation(s)
- Nathan Rout-Pitt
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia. .,Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia. .,Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, 72 King William Rd, North Adelaide, South Australia, 5006, Australia.
| | - Nigel Farrow
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia.,Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, 72 King William Rd, North Adelaide, South Australia, 5006, Australia.,Australian Respiratory Epithelium Consortium (AusRec), Perth, Western Australia, 6105, Australia
| | - David Parsons
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia.,Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, 72 King William Rd, North Adelaide, South Australia, 5006, Australia.,Australian Respiratory Epithelium Consortium (AusRec), Perth, Western Australia, 6105, Australia
| | - Martin Donnelley
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia.,Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, 72 King William Rd, North Adelaide, South Australia, 5006, Australia
| |
Collapse
|
49
|
Sunil Gowda SN, Rajasowmiya S, Vadivel V, Banu Devi S, Celestin Jerald A, Marimuthu S, Devipriya N. Gallic acid-coated sliver nanoparticle alters the expression of radiation-induced epithelial-mesenchymal transition in non-small lung cancer cells. Toxicol In Vitro 2018; 52:170-177. [PMID: 29928970 DOI: 10.1016/j.tiv.2018.06.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 06/15/2018] [Accepted: 06/16/2018] [Indexed: 01/03/2023]
Abstract
BACKGROUND Radiotherapy is the most widely used treatment method for treating cancer with or without surgery and chemotherapy. In lung cancer, it is one of the important treatment steps in excising the tumor from the lung tissue; unfortunately, radiation can induce epithelial- mesenchymal transition (EMT), a typical physiological process in which cuboidal shaped epithelial cell loses its phenotype and acquires mesenchymal-like phenotype thus, increases the metastasis progression in the body. To prevent EMT mediated metastasis, we aimed to 1) synthesize silver nanoparticles by using Gallic acid, a potential antioxidant which acts as stabilizing and reducing agent in the form of silver nanoparticle (GA-AgNPs) 2) to analyze its effect on EMT markers during radiation-induced EMT in A549 cells. METHODS A549 cells were irradiated with 8Gy (X-ray) and treated with GA-AgNPs at a fixed concentration under in vitro condition. GA-AgNPs were prepared and characterized for absorption, potential stability, size and morphology by UV-Visible spectrophotometer, Zeta potential and Transmission electron microscopy respectively. After irradiation, the morphology changes were observed using an inverted microscope, the gene and protein expression of EMT markers were analyzed by RT-PCR and western blotting. RESULTS/CONCLUSION GA-AgNPs are in nano size with fair stability. The synthesized nanoparticles suppressed the EMT markers including Vimentin, N-cadherin, Snail-1 and increased E-cadherin expression which might inhibit cancer cells to acquire radio resistant metastasis potential.
Collapse
Affiliation(s)
- S N Sunil Gowda
- School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - S Rajasowmiya
- School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Vellingiri Vadivel
- School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - S Banu Devi
- School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - A Celestin Jerald
- School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - S Marimuthu
- Vishnu Cancer Center, Thanjavur, Tamil Nadu, India
| | - N Devipriya
- School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, India.
| |
Collapse
|
50
|
Zheng W, Qian Y, Chen S, Ruan H, Fan C. Rapamycin Protects Against Peritendinous Fibrosis Through Activation of Autophagy. Front Pharmacol 2018; 9:402. [PMID: 29731718 PMCID: PMC5921906 DOI: 10.3389/fphar.2018.00402] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 04/06/2018] [Indexed: 12/31/2022] Open
Abstract
Dysregulation of autophagy plays a pivotal role in fibrosis in multiple organs. However, the role of autophagy in peritendinous fibrosis is not well understood. Here, we hypothesize that autophagy plays a protective role in preventing adhesion formation. In a rat model of tendon injury, we observed dysregulated autophagy during excessive extracellular matrix deposition. Pharmacological induction of autophagy by rapamycin markedly alleviated the severity of peritendinous fibrosis in vivo. In NIH/3T3 fibroblasts and tenocytes, transforming growth factor β1 (TGF-β1) markedly activated myofibroblasts and increased collagen synthesis. Addition of rapamycin activated autophagy, reduced collagen synthesis, and suppressed myofibroblast activation. In vitro experiments also showed that rapamycin decreased cell proliferation and increased the number of cells arrested in G0/G1 phase. However, following pretreatment with the autophagy inhibitor 3-methyladenine (3-MA), rapamycin was unable to repress the fibrotic changes induced by TGF-β1. Autophagy related protein 5 (Atg5) RNA interference in fibroblasts also abolished the protective effects of rapamycin in vitro. In conclusion, our results point to rapamycin as a potential treatment strategy in the prevention of peritendinous fibrosis after tendon injury.
Collapse
Affiliation(s)
- Wei Zheng
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yun Qian
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Shuai Chen
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Hongjiang Ruan
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Cunyi Fan
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|