1
|
Chung CL, Chen CL. Fluoroquinolones upregulate insulin-like growth factor-binding protein 3, inhibit cell growth and insulin-like growth factor signaling. Eur J Pharmacol 2024; 969:176421. [PMID: 38423242 DOI: 10.1016/j.ejphar.2024.176421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 01/27/2024] [Accepted: 02/13/2024] [Indexed: 03/02/2024]
Abstract
Fluoroquinolones (FQs), commonly known for their antibiotic properties, exhibit additional pharmacological potential with anti-proliferative effects on various malignant cell types and immunomodulatory responses. Despite these observed effects, the precise mechanisms of action remain elusive. This study elucidates the biological impact of FQs on insulin-like growth factor-binding protein 3 (IGFBP-3) productions in a p53-dependent manner. Cultured cells and mouse models treated with FQs demonstrated increased IGFBP-3 mRNA expression and protein secretion. The FQ-induced IGFBP-3 was identified to impede cell growth by inhibiting IGF-I signaling and exerting effects through an IGF-independent pathway. Notably, FQ-mediated suppression of cell proliferation was reversed in p53-null and p53 knockdown cells, suggesting the pivotal role of p53 in FQ-induced IGFBP-3 production and IGFBP-3-mediated growth inhibition. Additionally, ciprofloxacin, a clinically used FQ, exhibited the induction of tumor cell apoptosis and attenuation of tumor growth in a syngeneic mouse hepatocellular carcinoma (HCC) model. These findings unveil a novel mechanism through which FQs act as anti-proliferative agents, prompting further exploration of their potential utility or derivative compounds in cancer treatment and prevention.
Collapse
Affiliation(s)
- Chih-Ling Chung
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
| | - Chun-Lin Chen
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan; Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| |
Collapse
|
2
|
Tai SB, Huang CY, Chung CL, Sung PJ, Wen ZH, Chen CL. Prodigiosin Inhibits Transforming Growth Factor β Signaling by Interfering Receptor Recycling and Subcellular Translocation in Epithelial Cells. Mol Pharmacol 2024; 105:286-300. [PMID: 38278554 DOI: 10.1124/molpharm.123.000776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/05/2023] [Accepted: 01/02/2024] [Indexed: 01/28/2024] Open
Abstract
Prodigiosin (PG) is a naturally occurring polypyrrole red pigment produced by numerous microorganisms including some Serratia and Streptomyces strains. PG has exhibited promising anticancer activity; however, the molecular mechanisms of action of PG on malignant cells remain ambiguous. Transforming growth factor-β (TGF-β) is a multifunctional cytokine that governs a wide array of cellular processes in development and tissue homeostasis. Malfunctions of TGF-β signaling are associated with numerous human cancers. Emerging evidence underscores the significance of internalized TGF-β receptors and their intracellular trafficking in initiating signaling cascades. In this study, we identified PG as a potent inhibitor of the TGF-β pathway. PG blocked TGF-β signaling by targeting multiple sites of this pathway, including facilitating the sequestering of TGF-β receptors in the cytoplasm by impeding the recycling of type II TGF-β receptors to the cell surface. Additionally, PG prompts a reduction in the abundance of receptors on the cell surface through the disruption of the receptor glycosylation. In human Caucasian lung carcinoma cells and human hepatocellular cancer cell line cells, nanomolar concentrations of PG substantially diminish TGF-β-triggered phosphorylation of Smad2 protein. This attenuation is further reflected in the suppression of downstream target gene expression, including those encoding fibronectin, plasminogen activator inhibitor-1, and N-cadherin. SIGNIFICANCE STATEMENT: Prodigiosin (PG) emerges from this study as a potent TGF-β pathway inhibitor, disrupting receptor trafficking and glycosylation and reducing TGF-β signaling and downstream gene expression. These findings not only shed light on PG's potential therapeutic role but also present a captivating avenue towards future anti-TGF-β strategies.
Collapse
Affiliation(s)
- Shun-Ban Tai
- Departments of Marine Biotechnology and Resources (S.-B.T., Z.-H.W.) and Biological Sciences (C.-L.Chu., C.-L.Che.), National Sun Yat-Sen University, Kaohsiung, Taiwan; Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Zuoying Armed Forces General Hospital, Kaohsiung, Taiwan (S.-B.T.); Department of Orthopaedics, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan (C.-y.H.); National Museum of Marine Biology and Aquarium, Pingtung, Taiwan (P.-J.S.); and Department of Biotechnology (C.-L.Che.) and Graduate Institute of Natural Products, College of Pharmacy (C.-L.Che.), Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Yin Huang
- Departments of Marine Biotechnology and Resources (S.-B.T., Z.-H.W.) and Biological Sciences (C.-L.Chu., C.-L.Che.), National Sun Yat-Sen University, Kaohsiung, Taiwan; Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Zuoying Armed Forces General Hospital, Kaohsiung, Taiwan (S.-B.T.); Department of Orthopaedics, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan (C.-y.H.); National Museum of Marine Biology and Aquarium, Pingtung, Taiwan (P.-J.S.); and Department of Biotechnology (C.-L.Che.) and Graduate Institute of Natural Products, College of Pharmacy (C.-L.Che.), Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Ling Chung
- Departments of Marine Biotechnology and Resources (S.-B.T., Z.-H.W.) and Biological Sciences (C.-L.Chu., C.-L.Che.), National Sun Yat-Sen University, Kaohsiung, Taiwan; Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Zuoying Armed Forces General Hospital, Kaohsiung, Taiwan (S.-B.T.); Department of Orthopaedics, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan (C.-y.H.); National Museum of Marine Biology and Aquarium, Pingtung, Taiwan (P.-J.S.); and Department of Biotechnology (C.-L.Che.) and Graduate Institute of Natural Products, College of Pharmacy (C.-L.Che.), Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ping-Jyun Sung
- Departments of Marine Biotechnology and Resources (S.-B.T., Z.-H.W.) and Biological Sciences (C.-L.Chu., C.-L.Che.), National Sun Yat-Sen University, Kaohsiung, Taiwan; Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Zuoying Armed Forces General Hospital, Kaohsiung, Taiwan (S.-B.T.); Department of Orthopaedics, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan (C.-y.H.); National Museum of Marine Biology and Aquarium, Pingtung, Taiwan (P.-J.S.); and Department of Biotechnology (C.-L.Che.) and Graduate Institute of Natural Products, College of Pharmacy (C.-L.Che.), Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Zhi-Hong Wen
- Departments of Marine Biotechnology and Resources (S.-B.T., Z.-H.W.) and Biological Sciences (C.-L.Chu., C.-L.Che.), National Sun Yat-Sen University, Kaohsiung, Taiwan; Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Zuoying Armed Forces General Hospital, Kaohsiung, Taiwan (S.-B.T.); Department of Orthopaedics, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan (C.-y.H.); National Museum of Marine Biology and Aquarium, Pingtung, Taiwan (P.-J.S.); and Department of Biotechnology (C.-L.Che.) and Graduate Institute of Natural Products, College of Pharmacy (C.-L.Che.), Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chun-Lin Chen
- Departments of Marine Biotechnology and Resources (S.-B.T., Z.-H.W.) and Biological Sciences (C.-L.Chu., C.-L.Che.), National Sun Yat-Sen University, Kaohsiung, Taiwan; Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Zuoying Armed Forces General Hospital, Kaohsiung, Taiwan (S.-B.T.); Department of Orthopaedics, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan (C.-y.H.); National Museum of Marine Biology and Aquarium, Pingtung, Taiwan (P.-J.S.); and Department of Biotechnology (C.-L.Che.) and Graduate Institute of Natural Products, College of Pharmacy (C.-L.Che.), Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
3
|
Mateusz M, Seweryn KM, Janusz S, Piotr K, Panek MG. Assessment of the effectiveness of the peptide inhibitor homologous to the transforming growth factor β cytokine blocking the TGFβRI/TGFβRII receptor complex-pilot study. Clin Transl Allergy 2024; 14:e12320. [PMID: 38282199 PMCID: PMC10758017 DOI: 10.1002/clt2.12320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 10/18/2023] [Accepted: 11/21/2023] [Indexed: 01/30/2024] Open
Abstract
BACKGROUND A key player in the fibrotic process is the transforming growth factor β (TGF-β) which enhances extracellular matrix production by increasing the transcription of matrix proteins. The cytokine TGF-β first binds to the TGFβRII receptor (dimer), resulting in the recruitment of the TGFβRI receptor (dimer). The complex thus formed leads to the phosphorylation of the kinase domain of TGFβRI, which in turn results in activation of the Smad pathway. This is therefore a targeted pathway for research into the application of peptide inhibitors in blocking the TGF-β-Smad signaling pathway. The aim of this study was to design a peptide inhibitor (homologous to the cytokine TGF-β) which, after binding to the TGFβRI/TGFβRII receptor, would block the cytokine binding and thus prevent the formation of an activating complex. METHODS Preliminary work on the design and synthesis of inhibitors for TGFβRI/TGFβRII has allowed us to identify and describe five key regions of the TGF-β-TGFβRI/TGFβRII interface. The following five peptide inhibitors were synthesized for Region 1: 1.1 ALDAAYCFR, 1.2 LDAAYCFRN, 1.3 DAAYCFRNV, 1.4 AAYCFRNVQ, 1.5 AYCFRNVQD. The expression of the SEAP reporter gene, Smad2, Smad3, Smad4, and JNK1 gene was measured using quantitative real-time polymerase chain reaction. RESULTS For Region 1 peptide inhibitors tested for TGFβRI/TGFβRII, reduced SEAP (reporter gene) expression was observed in cells of the MFB-F11 line, which suggests inhibited the formation of cytokine-receptor complexes. CONCLUSIONS For IP1_2, 1_3 and 1_5 Region 1 peptides tested for TGFβRI/TGFβRII, reduced cytokine-receptor signal by adding newly designed inhibitors. The study revealed an impact of these peptide inhibitors on the reduction of mRNA expression of Smad2, Smad3, Smad4 and JNK1 genes.
Collapse
Affiliation(s)
- Marynowski Mateusz
- Department of Internal Medicine, Asthma and AllergyMedical University of LodzLodzLodzkiePoland
| | | | - Szemraj Janusz
- Department of Medical BiochemistryMedical University of LodzLodzPoland
| | - Kuna Piotr
- Department of Internal Medicine, Asthma and AllergyMedical University of LodzLodzLodzkiePoland
| | - Michał Gabriel Panek
- Department of Internal Medicine, Asthma and AllergyMedical University of LodzLodzLodzkiePoland
| |
Collapse
|
4
|
Feng L, Guo L, Tanaka Y, Su L. Tumor-Derived Small Extracellular Vesicles Involved in Breast Cancer Progression and Drug Resistance. Int J Mol Sci 2022; 23:ijms232315236. [PMID: 36499561 PMCID: PMC9736664 DOI: 10.3390/ijms232315236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/28/2022] [Accepted: 12/01/2022] [Indexed: 12/09/2022] Open
Abstract
Breast cancer is one of the most serious and terrifying threats to the health of women. Recent studies have demonstrated that interaction among cancer cells themselves and those with other cells, including immune cells, in a tumor microenvironment potentially and intrinsically regulate and determine cancer progression and metastasis. Small extracellular vesicles (sEVs), a type of lipid-bilayer particles derived from cells, with a size of less than 200 nm, are recognized as one form of important mediators in cell-to-cell communication. sEVs can transport a variety of bioactive substances, including proteins, RNAs, and lipids. Accumulating evidence has revealed that sEVs play a crucial role in cancer development and progression, with a significant impact on proliferation, invasion, and metastasis. In addition, sEVs systematically coordinate physiological and pathological processes, such as coagulation, vascular leakage, and stromal cell reprogramming, to bring about premetastatic niche formation and to determine metastatic organ tropism. There are a variety of oncogenic factors in tumor-derived sEVs that mediate cellular communication between local stromal cells and distal microenvironment, both of which are important in cancer progression and metastasis. Tumor-derived sEVs contain substances that are similar to parental tumor cells, and as such, sEVs could be biomarkers in cancer progression and potential therapeutic targets, particularly for predicting and preventing future metastatic development. Here, we review the mechanisms underlying the regulation by tumor-derived sEVs on cancer development and progression, including proliferation, metastasis, drug resistance, and immunosuppression, which coordinately shape the pro-metastatic microenvironment. In addition, we describe the application of sEVs to the development of cancer biomarkers and potential therapeutic modalities and discuss how they can be engineered and translated into clinical practice.
Collapse
Affiliation(s)
- Lingyun Feng
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lijuan Guo
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yoshimasa Tanaka
- Center for Medical Innovation, Nagasaki University, 1-7-1, Sakamoto, Nagasaki 852-8588, Japan
- Correspondence: (Y.T.); (L.S.); Tel.: +81-95-819-7063 (Y.T.); +86-27-8779-2024 (L.S.); Fax: +81-95-819-2189 (Y.T.); +86-27-8779-2072 (L.S.)
| | - Li Su
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Correspondence: (Y.T.); (L.S.); Tel.: +81-95-819-7063 (Y.T.); +86-27-8779-2024 (L.S.); Fax: +81-95-819-2189 (Y.T.); +86-27-8779-2072 (L.S.)
| |
Collapse
|
5
|
Zhang S, Zhu N, Li HF, Gu J, Zhang CJ, Liao DF, Qin L. The lipid rafts in cancer stem cell: a target to eradicate cancer. Stem Cell Res Ther 2022; 13:432. [PMID: 36042526 PMCID: PMC9429646 DOI: 10.1186/s13287-022-03111-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/03/2022] [Indexed: 11/10/2022] Open
Abstract
Cancer stem cells (CSCs) are a subpopulation of cancer cells with stem cell properties that sustain cancers, which may be responsible for cancer metastasis or recurrence. Lipid rafts are cholesterol- and sphingolipid-enriched microdomains in the plasma membrane that mediate various intracellular signaling. The occurrence and progression of cancer are closely related to lipid rafts. Emerging evidence indicates that lipid raft levels are significantly enriched in CSCs compared to cancer cells and that most CSC markers such as CD24, CD44, and CD133 are located in lipid rafts. Furthermore, lipid rafts play an essential role in CSCs, specifically in CSC self-renewal, epithelial-mesenchymal transition, drug resistance, and CSC niche. Therefore, lipid rafts are critical regulatory platforms for CSCs and promising therapeutic targets for cancer therapy.
Collapse
Affiliation(s)
- Shuo Zhang
- Laboratory of Stem Cell Regulation With Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, 300 Xueshi Road, Hanpu Science and Education District, 410208, Changsha, Hunan, People's Republic of China
| | - Neng Zhu
- Department of Urology, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Hong Fang Li
- Laboratory of Stem Cell Regulation With Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, 300 Xueshi Road, Hanpu Science and Education District, 410208, Changsha, Hunan, People's Republic of China
| | - Jia Gu
- Laboratory of Stem Cell Regulation With Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, 300 Xueshi Road, Hanpu Science and Education District, 410208, Changsha, Hunan, People's Republic of China
| | - Chan Juan Zhang
- Laboratory of Stem Cell Regulation With Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, 300 Xueshi Road, Hanpu Science and Education District, 410208, Changsha, Hunan, People's Republic of China
| | - Duan Fang Liao
- Laboratory of Stem Cell Regulation With Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, 300 Xueshi Road, Hanpu Science and Education District, 410208, Changsha, Hunan, People's Republic of China
| | - Li Qin
- Laboratory of Stem Cell Regulation With Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, 300 Xueshi Road, Hanpu Science and Education District, 410208, Changsha, Hunan, People's Republic of China. .,Institutional Key Laboratory of Vascular Biology and Translational Medicine in Hunan Province, Hunan University of Chinese Medicine, Changsha, China. .,Hunan Province Engineering Research Center of Bioactive Substance Discovery of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China.
| |
Collapse
|
6
|
A novel role of BK potassium channel activity in preventing the development of kidney fibrosis. Kidney Int 2022; 101:945-962. [PMID: 34968553 DOI: 10.1016/j.kint.2021.11.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 11/03/2021] [Accepted: 11/22/2021] [Indexed: 11/21/2022]
Abstract
Kidney fibrosis is a common characteristic of chronic kidney disease and while the large conductance voltage and calcium-activated potassium channel (BK) is widely expressed in kidneys, its role in kidney fibrosis is unknown. To evaluate this, we found that BK protein expression was decreased in the fibrotic kidneys. Accompanying this was increased fibrotic marker protein expression of fibronectin, vimentin and α-smooth muscle actin and increased mRNA expressions of fibronectin, α-smooth muscle actin, collagen III and collagen I. These changes occurred in the unilateral ureteral obstruction and folic acid models of fibrosis and were more pronounced in BK knockout than in wild-type mice. Activation of BK activity by chemical NS1619 or BMS191011 channel openers attenuated kidney fibrosis in these two models while protecting kidney function in wild-type mice. BK deficiency up-regulated transforming growth factor-β (TGF-β)/transcription factor Smad2/3 signaling in the fibrotic kidney, whereas activation of BK activity inhibited this signaling pathway both in vivo and in vitro. BK channel activation increased the degradation of TGF-β receptors induced by TGF-β1 in vivo and in vitro. Furthermore, in cell lines HK-2, NRK49, and NRK-52E, BK channel activation by NS1619 led to increased caveolae formation and facilitated localization of TGF-β receptors in the microdomains of lipid rafts. Thus, our data demonstrated that BK activation has an anti-fibrotic effect on kidney fibrosis by inhibiting the TGF-β signaling pathway through accelerating TGF-β receptor degradation via the caveolae route. Hence, our study provides innovative insight into BK as a potential therapeutic target for the treatment of kidney fibrosis.
Collapse
|
7
|
Modulation of the tumour microenvironment in hepatocellular carcinoma by tyrosine kinase inhibitors: from modulation to combination therapy targeting the microenvironment. Cancer Cell Int 2022; 22:73. [PMID: 35148789 PMCID: PMC8840552 DOI: 10.1186/s12935-021-02435-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/28/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer deaths worldwide. Tyrosine kinase inhibitors (TKIs) remain the backbone of systematic therapy for advanced hepatocellular carcinoma. Sorafenib and lenvatinib are currently approved as first-line therapeutic drugs, and regorafenib and cabozantinib are applied as second-line treatments. With inhibition of angiogenesis as the main target, TKIs exert a profound effect on the tumour microenvironment (TME). The TME is a complex mixture of cellular and noncellular components surrounding the tumour mass, and is associated with tumour progression partially through the epithelial-mesenchymal transition. Specifically, the TME of HCC is characterized by profound extracellular matrix remodelling and an immunosuppressive microenvironment. The purpose of this review is to provide a summary of TME remodelling mediated by four Food and Drug Administration approved TKIs in HCC and thus summarize the rationale and potential targets for combination therapy. The modulatory effect of TKIs on the TME of HCC was reported to enhance the antitumour effect of TKIs through pyroptosis of macrophages and subsequent natural killer cell activation, T cell activation, regulatory T cell reduction in HCC. Meanwhile, TKIs also induce drug resistance via M2 polarization and accumulation, recruitment of tumour-associated neutrophils, and induction of the epithelial-mesenchymal transition. In conclusion, the effect of TKIs on TME can enhance its antitumour effect, but might also partially contribute to the drug resistance that hinders the progression of TKIs as treatment for HCC. Additionally, the effect of TKIs also provides the rationale for combination therapy, including combining TKIs with immune checkpoint inhibitors, to facilitate increased drug efficacy of TKIs.
Collapse
|
8
|
Fluoroquinolones Suppress TGF-β and PMA-Induced MMP-9 Production in Cancer Cells: Implications in Repurposing Quinolone Antibiotics for Cancer Treatment. Int J Mol Sci 2021; 22:ijms222111602. [PMID: 34769032 PMCID: PMC8584204 DOI: 10.3390/ijms222111602] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/22/2021] [Accepted: 10/24/2021] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Fluoroquinolones (FQs) are potent antimicrobials with multiple effects on host cells and tissues. Although FQs can attenuate cancer invasion and metastasis, the underlying molecular mechanisms remain unclear. Matrix metalloproteinase-9 (MMP-9) has functional roles in tumor angiogenesis, invasion, and metastasis, and is associated with cancer progression and poor prognosis, suggesting that inhibitors of MMP-9 activity and transcription are prime candidates for cancer therapy. Despite numerous preclinical data supporting the use of MMP-9 inhibitors as anticancer drugs, the few available examples are not therapeutically useful due to low specificity and off-target effects. We examined the effects of FQs on MMP-9 production in cancer cells following transforming growth factor beta (TGF-β) and phorbol 12-myristate 13-acetate (PMA) stimulation. EXPERIMENTAL APPROACHES Using confluent cultures of HepG2 and A549 cells, the effects of FQs (ciprofloxacin, levofloxacin, clinafloxacin, gatifloxacin, and enrofloxacin) on TGF-β and PMA-induced MMP-9 mRNA expression and production were studied in RNA extracts and culture supernatants, respectively. FQs specifically abrogated TGF-β and PMA-induced MMP-9 levels and activity in a concentration and time-dependent manner, without affecting other MMPs or proteins involved in epithelial-mesenchymal transition. Additionally, FQs inhibited TGF-β and PMA-induced cell migration via p38 and cyclic AMP signaling pathways. CONCLUSIONS AND IMPLICATIONS Overall, we demonstrated that FQs inhibit cancer cell migration and invasion by downregulating MMP-9 expression and revealed the cellular mechanisms underlying their potential value in cancer treatment.
Collapse
|
9
|
Cai J, Cui Y, Yang J, Wang S. Epithelial-mesenchymal transition: When tumor cells meet myeloid-derived suppressor cells. Biochim Biophys Acta Rev Cancer 2021; 1876:188564. [PMID: 33974950 DOI: 10.1016/j.bbcan.2021.188564] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 05/05/2021] [Accepted: 05/05/2021] [Indexed: 12/12/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous myeloid cell population characterized by protumoral functions in the tumor immune network. An increasing number of studies have focused on the biological functions of MDSCs in tumor immunity. Epithelial-mesenchymal transition (EMT) is a cellular plasticity process accompanied by a loss of epithelial phenotypes and an acquisition of mesenchymal phenotypes. In general, tumor cells that undergo EMT are more likely to invade and metastasize. Recently, extensive evidence suggests that EMT is closely related to a highly immunosuppressive environment. This review will summarize the immunosuppressive capacities of MDSC subsets and their distinct role in tumor EMT and further discuss immunotherapy for tumor EMT by targeting MDSCs.
Collapse
Affiliation(s)
- Jingshan Cai
- Department of Laboratory Medicine, the Affiliated People's Hospital, Jiangsu University, Zhenjiang, China; Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yudan Cui
- Department of Laboratory Medicine, the Affiliated People's Hospital, Jiangsu University, Zhenjiang, China; Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Jun Yang
- Department of Laboratory Medicine, the Affiliated People's Hospital, Jiangsu University, Zhenjiang, China.
| | - Shengjun Wang
- Department of Laboratory Medicine, the Affiliated People's Hospital, Jiangsu University, Zhenjiang, China; Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China.
| |
Collapse
|
10
|
Wang YL, Zheng CM, Lee YH, Cheng YY, Lin YF, Chiu HW. Micro- and Nanosized Substances Cause Different Autophagy-Related Responses. Int J Mol Sci 2021; 22:4787. [PMID: 33946416 PMCID: PMC8124422 DOI: 10.3390/ijms22094787] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 04/27/2021] [Accepted: 04/29/2021] [Indexed: 02/07/2023] Open
Abstract
With rapid industrialization, humans produce an increasing number of products. The composition of these products is usually decomposed. However, some substances are not easily broken down and gradually become environmental pollutants. In addition, these substances may cause bioaccumulation, since the substances can be fragmented into micro- and nanoparticles. These particles or their interactions with other toxic matter circulate in humans via the food chain or air. Whether these micro- and nanoparticles interfere with extracellular vesicles (EVs) due to their similar sizes is unclear. Micro- and nanoparticles (MSs and NSs) induce several cell responses and are engulfed by cells depending on their size, for example, particulate matter with a diameter ≤2.5 μm (PM2.5). Autophagy is a mechanism by which pathogens are destroyed in cells. Some artificial materials are not easily decomposed in organisms. How do these cells or tissues respond? In addition, autophagy operates through two pathways (increasing cell death or cell survival) in tumorigenesis. Many MSs and NSs have been found that induce autophagy in various cells and tissues. As a result, this review focuses on how these particles interfere with cells and tissues. Here, we review MSs, NSs, and PM2.5, which result in different autophagy-related responses in various tissues or cells.
Collapse
Affiliation(s)
- Yung-Li Wang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-L.W.); (Y.-F.L.)
| | - Cai-Mei Zheng
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan;
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei 11031, Taiwan
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Hsuan Lee
- Department of Cosmeceutics, China Medical University, Taichung 406040, Taiwan;
| | - Ya-Yun Cheng
- Department of Environmental Health, Harvard University T.H. Chan School of Public Health, Boston, MA 02115, USA;
| | - Yuh-Feng Lin
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-L.W.); (Y.-F.L.)
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan;
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei 11031, Taiwan
| | - Hui-Wen Chiu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-L.W.); (Y.-F.L.)
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei 11031, Taiwan
- Department of Medical Research, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
| |
Collapse
|
11
|
Combined Inhibition of TGF-β1-Induced EMT and PD-L1 Silencing Re-Sensitizes Hepatocellular Carcinoma to Sorafenib Treatment. J Clin Med 2021; 10:jcm10091889. [PMID: 33925488 PMCID: PMC8123871 DOI: 10.3390/jcm10091889] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/06/2021] [Accepted: 04/21/2021] [Indexed: 12/14/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary hepatic malignancy. HCC is one of the leading causes of cancer deaths worldwide. The oral multi-tyrosine kinase inhibitor Sorafenib is the standard first-line therapy in patients with advanced unresectable HCC. Despite the significant survival benefit in HCC patients post treatment with Sorafenib, many patients had progressive disease as a result of acquiring drug resistance. Circumventing resistance to Sorafenib by exploring and targeting possible molecular mechanisms and pathways is an area of active investigation worldwide. Epithelial-to-mesenchymal transition (EMT) is a cellular process allowing epithelial cells to assume mesenchymal traits. HCC tumour cells undergo EMT to become immune evasive and develop resistance to Sorafenib treatment. Immune checkpoint molecules control immune escape in many tumours, including HCC. The aim of this study is to investigate whether combined inhibition of EMT and immune checkpoints can re-sensitise HCC to Sorafenib treatment. Post treatment with Sorafenib, HCC cells PLC/PRF/5 and Hep3B were monitored for induction of EMT and immune checkpoint molecules using quantitative reverse transcriptase (qRT)- PCR, western blot, immunofluorescence, and motility assays. The effect of combination treatment with SB431542, a specific inhibitor of the transforming growth factor (TGF)-β receptor kinase, and siRNA mediated knockdown of programmed cell death protein ligand-1 (PD-L1) on Sorafenib resistance was examined using a cell viability assay. We found that three days of Sorafenib treatment activated EMT with overexpression of TGF-β1 in both HCC cell lines. Following Sorafenib exposure, increase in the expression of PD-L1 and other immune checkpoints was observed. SB431542 blocked the TGF-β1-mediated EMT in HCC cells and also repressed PD-L1 expression. Likewise, knockdown of PD-L1 inhibited EMT. Moreover, the sensitivity of HCC cells to Sorafenib was enhanced by combining a blockade of EMT with SB431542 and knockdown of PD-L1 expression. Sorafenib-induced motility was attenuated with the combined treatment of SB431542 and PD-L1 knockdown. Our findings indicate that treatment with Sorafenib induces EMT and expression of immune checkpoint molecules, which contributes to Sorafenib resistance in HCC cells. Thus, the combination treatment strategy of inhibiting EMT and immune checkpoint molecules can re-sensitise HCC cells to Sorafenib.
Collapse
|
12
|
Huang SF, Wang YL, Chen JJ, Huang YB, Tai SB, Chung CL, Chen CL. Garcimultiflorone K from Garcinia multiflora attenuates hepatocellular carcinoma metastasis by suppressing transforming growth factor-β signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 84:153502. [PMID: 33647775 DOI: 10.1016/j.phymed.2021.153502] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Transforming growth factor‑β (TGF-β) signaling is a crucial inducer of tissue fibrosis and extracellular matrix accumulation and a vital suppressor of epithelial cell proliferation and cancer metastasis. The nature of this multifunctional cytokine has prompted the development of TGF-β signaling inhibitors as therapeutic agents. Our research group has recently isolated the polyprenylated polycyclic acylphloroglucinol garcimultiflorone K (GMK) from the stems of Garcinia multiflora; GMK exhibits antiangiogenic activity in endothelial cells. PURPOSE In the current study, we aimed to explore the antitumor effect and detailed mechanisms of Garcimultiflorone K in hepatocellular carcinoma cells. METHODS Cell proliferation and viability were evaluated using the MTT assay. The migratory ability of HepG2 cells was measured using wound healing assays. The inhibitory effect of GMK against the nuclear translocation of Smad by TGF-β was assessed through immunofluorescence staining and Western blotting. To investigate TGF-β-dependent gene expression profiles upon GMK stimulation, RNA transcript levels were determined using reverse transcription polymerase chain reaction. The effects of GMK in Smad2-driven transcriptomic activities were studied using a reporter gene assay. Protein levels were detected using Western blotting. RESULTS Our data revealed that GMK inhibited TGF-β-induced cellular responses, including Smad protein phosphorylation, cell migration, and extracellular matrix production, during epithelial-mesenchymal transition (EMT). Mechanistic studies further demonstrated that GMK suppressed TGF-β signaling by downregulating TGF-β receptor II (TβRII). CONCLUSION These findings elucidate that TβRII expression in hepatic cells can be specifically suppressed by GMK to attenuate metastasis and the disease-promoting effects of EMT, representing a therapeutic approach.
Collapse
Affiliation(s)
- Shu-Fang Huang
- Department of Medicine Chest, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan ROC
| | - Yu-Lun Wang
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan ROC
| | - Jih-Jung Chen
- Faculty of Pharmacy, School of Pharmaceutical Sciences, National Yang-Ming Chiao-Tung University, Taipei 11221, Taiwan ROC; Department of Medical Research, China Medical University Hospital, Taichung 40402, Taiwan ROC
| | - Yaw-Bin Huang
- Department of Pharmacy, School of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan ROC
| | - Shun-Ban Tai
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan ROC; Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Zuoying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung 81342, Taiwan ROC
| | - Chih-Ling Chung
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan ROC
| | - Chun-Lin Chen
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan ROC; Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan ROC; Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan ROC.
| |
Collapse
|
13
|
Huang CY, Chung CL, Hu TH, Chen JJ, Liu PF, Chen CL. Recent progress in TGF-β inhibitors for cancer therapy. Biomed Pharmacother 2020; 134:111046. [PMID: 33341049 DOI: 10.1016/j.biopha.2020.111046] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/16/2020] [Accepted: 11/19/2020] [Indexed: 01/18/2023] Open
Abstract
Transforming growth factor-β (TGF-β) is a multifunctional cytokine that is involved in proliferation, metastasis, and many other important processes in malignancy. Inhibitors targeting TGF-β have been considered by pharmaceutical companies for cancer therapy, and some of them are in clinical trial now. Unfortunately, several of these programs have recently been relinquished, and most companies that remain in the contest are progressing slowly and cautiously. This review summarizes the TGF-β signal transduction pathway, its roles in oncogenesis and fibrotic diseases, and advancements in antibodies and small-molecule inhibitors of TGF-β.
Collapse
Affiliation(s)
- Cheng-Yi Huang
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan, ROC; Department of Pathology, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan, ROC
| | - Chih-Ling Chung
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan, ROC
| | - Tsung-Hui Hu
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan, ROC
| | - Jih-Jung Chen
- Faculty of Pharmacy, School of Pharmaceutical Sciences, National Yang-Ming University, Taipei 11221, Taiwan, ROC; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan
| | - Pei-Feng Liu
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan ROC
| | - Chun-Lin Chen
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan, ROC; Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan ROC; Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan ROC.
| |
Collapse
|
14
|
Adenina S, Louisa M, Soetikno V, Arozal W, Wanandi SI. The Effect of Alpha Mangostin on Epithelial-Mesenchymal Transition on Human Hepatocellular Carcinoma HepG2 Cells Surviving Sorafenib via TGF-β/Smad Pathways. Adv Pharm Bull 2020; 10:648-655. [PMID: 33062605 PMCID: PMC7539313 DOI: 10.34172/apb.2020.078] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 02/10/2020] [Accepted: 02/10/2020] [Indexed: 12/14/2022] Open
Abstract
Purpose: This study was intended to find out the impact of alpha mangostin administration on the epithelial-mesenchymal transition (EMT) markers and TGF-β/Smad pathways in hepatocellular carcinoma Hep-G2 cells surviving sorafenib. Methods: Hepatocellular carcinoma HepG2 cells were treated with sorafenib 10 μM. Cells surviving sorafenib treatment (HepG2surv) were then treated vehicle, sorafenib, alpha mangostin, or combination of sorafenib and alpha mangostin. Afterward, cells were observed for their morphology with an inverted microscope and counted for cell viability. The concentrations of transforming growth factor (TGF)-β1 in a culture medium were examined using ELISA. The mRNA expressions of TGF-β1, TGF-β1-receptor, Smad3, Smad7, E-cadherin, and vimentin were evaluated using quantitative reverse transcriptase–polymerase chain reaction. The protein level of E-cadherin was also determined using western blot analysis. Results: Treatment of alpha mangostin and sorafenib caused a significant decrease in the viability of sorafenib-surviving HepG2 cells versus control (both groups with P <0.05). Our study found that alpha mangostin treatment increased the expressions of vimentin (P <0.001 versus control). In contrast, alpha mangostin treatment tends to decrease the expressions of Smad7 and E-cadherin (both with P >0.05). In line with our findings, the expressions of TGF-β1 and Smad3 are significantly upregulated after alpha mangostin administration (both with P <0.05) versus control. Conclusion: Alpha mangostin reduced cell viability of sorafenib-surviving HepG2 cells; however, it also enhanced epithelial–mesenchymal transition markers by activating TGF-β/Smad pathways.
Collapse
Affiliation(s)
- Syarinta Adenina
- Master Program in Biomedical Sciences, Faculty of Medicine, Universitas Indonesia
| | - Melva Louisa
- Department of Pharmacology and Therapeutics Faculty of Medicine, Universitas Indonesia
| | - Vivian Soetikno
- Department of Pharmacology and Therapeutics Faculty of Medicine, Universitas Indonesia
| | - Wawaimuli Arozal
- Department of Pharmacology and Therapeutics Faculty of Medicine, Universitas Indonesia
| | | |
Collapse
|
15
|
Zhou BY, Wang WB, Wu XL, Zhang WJ, Zhou GD, Gao Z, Liu W. Nintedanib inhibits keloid fibroblast functions by blocking the phosphorylation of multiple kinases and enhancing receptor internalization. Acta Pharmacol Sin 2020; 41:1234-1245. [PMID: 32327724 PMCID: PMC7608201 DOI: 10.1038/s41401-020-0381-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 02/13/2020] [Indexed: 01/22/2023] Open
Abstract
Keloid is a benign skin tumor characterized by its cell hyperproliferative activity, invasion into normal skin, uncontrolled growth, overproduction and deposition of extracellular matrices and high recurrence rate after various therapies. Nintedanib is a receptor tyrosine kinase inhibitor targeting VEGF, PDGF, FGF, and TGF-β receptors with proved efficacy in anti-angiogenesis and in treating various types of cancers. In this study, we investigated the effects of nintedanib on keloid fibroblasts in both in vitro and ex vivo models. Keloid fibroblasts were prepared from 54 keloid scar samples in active stages collected from 49 patients. We found that nintedanib (1−4 μM) dose-dependently suppressed cell proliferation, induced G0/G1 cell cycle arrest, and inhibited migration and invasion of keloid fibroblasts. The drug also significantly inhibited the gene and protein expression of collagen I (COL-1) and III (COL-3), fibronectin (FN), and connective growth factor (CTGF), as well as the gene expression of other pathological factors, such as alpha smooth muscle actin (α-SMA), plasminogen activator inhibitor-1 (PAI-1), FK506-binding protein 10 (FKBP10), and heat shock protein 47 (HSP47) in keloid fibroblasts. Furthermore, nintedanib treatment significantly suppressed the phosphorylation of p38, JNK, ERK, STAT3, and Smad, enhanced endocytosis of various growth factor receptors. Using an ex vivo tissue explant model, we showed that nintedanib significantly suppressed cell proliferation, migration, and collagen production. The drug also significantly disrupted microvessel structure ex vivo. In summary, our results demonstrate that nintedanib is likely to become a potential targeted drug for keloid systemic therapy.
Collapse
|
16
|
Wu Y, Wang J, Zheng X, Chen Y, Huang M, Huang Q, Xiao W, Wei H, Tian Z, Sun R, Sun C. Establishment and Preclinical Therapy of Patient-derived Hepatocellular Carcinoma Xenograft Model. Immunol Lett 2020; 223:33-43. [PMID: 32335145 DOI: 10.1016/j.imlet.2020.04.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 12/06/2019] [Accepted: 04/18/2020] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is a world-wide health problem. Poor and delayed diagnoses as well as high recurrence rate resulting in high mortality rate. In this study, we established a patient-derived xenograft (PDX) model from HCC patient, and continuously maintained with subcutaneous passage more than 20 times. This HCC PDX tumor exhibited the same histological characteristics with the HCC patient and could be used to verify therapeutic effect of liver cancer. We further evaluated this PDX model by experimental chemotherapy, demonstrating that this HCC PDX model was sensitive to sorafenib treatment. Further, the potential of natural killer cell-based immunotherapy for HCC was tested using this model. We found that NK92 cells effectively suppressed the tumor growth in vivo and prolonged the survival time of HCC-bearing PDX mice. This study indicates that HCC PDX model is a good platform to testify the efficacy of preclinical chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Yuwei Wu
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, China; Institute of Immunology, University of Science and Technology of China, China
| | - Jinyu Wang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, China; Institute of Immunology, University of Science and Technology of China, China
| | - Xiaodong Zheng
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, China; Institute of Immunology, University of Science and Technology of China, China
| | - Yongyan Chen
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, China; Institute of Immunology, University of Science and Technology of China, China
| | - Mei Huang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, China
| | - Qiang Huang
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science & Technology of China, Hefei, China
| | - Weihua Xiao
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, China; Institute of Immunology, University of Science and Technology of China, China
| | - Haiming Wei
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, China; Institute of Immunology, University of Science and Technology of China, China
| | - Zhigang Tian
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, China; Institute of Immunology, University of Science and Technology of China, China
| | - Rui Sun
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, China; Institute of Immunology, University of Science and Technology of China, China.
| | - Cheng Sun
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, China; Institute of Immunology, University of Science and Technology of China, China; Transplant & Immunology Laboratory, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
17
|
Hou FJ, Guo LX, Zheng KY, Song JN, Wang Q, Zheng YG. Chelidonine enhances the antitumor effect of lenvatinib on hepatocellular carcinoma cells. Onco Targets Ther 2019; 12:6685-6697. [PMID: 31695406 PMCID: PMC6707434 DOI: 10.2147/ott.s215103] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 08/08/2019] [Indexed: 12/15/2022] Open
Abstract
Background Lenvatinib is a newly approved molecular targeted drug for the treatment of advanced hepatocellular carcinoma (HCC). However, the high cost associated with this treatment poses a huge financial burden on patients and the entire public health system. Therefore, there is an urgent need to develop novel strategies that enhance the antitumor effect of lenvatinib. Methods The antitumor effects of chelidonine or/and lenvatinib on HCC cell lines MHCC97-H and LM-3 were examined using the 3-[4,5-dimethyl-2-thiazolyl]-2,5-diphenyl-2- H-tetrazolium bromide (MTT) assay. For the in-vivo investigation, the effect on subcutaneous or intrahepatic tumor growth in nude mice was also determined. The mRNA levels of epithelial mesenchymal transition (EMT)-related factors were examined through quantitative polymerase chain reaction or Western blot. Results In the present study, we found that treatment with chelidonine enhanced the apoptotic effect of lenvatinib on HCC cells and the in-vivo growth of HCC tumors in nude mice. Mechanistically, treatment with chelidonine increased the expression of epithelial indicator E-cadherin, whereas it decreased the expression of mesenchymal indicators N-cadherin and Vimentin. These findings suggest that chelidonine restricted the EMT in HCC cells. Conclusion Chelidonine inhibits the process of EMT and enhances the antitumor effect of lenvatinib on HCC cells.
Collapse
Affiliation(s)
- Fang-Jie Hou
- Hebei University of Chinese Medicine, Shijiazhuang City, Hebei Province 050200, People's Republic of China
| | - Li-Xiao Guo
- Hebei University of Chinese Medicine, Shijiazhuang City, Hebei Province 050200, People's Republic of China
| | - Kai-Yan Zheng
- Hebei University of Chinese Medicine, Shijiazhuang City, Hebei Province 050200, People's Republic of China
| | - Jun-Na Song
- Hebei University of Chinese Medicine, Shijiazhuang City, Hebei Province 050200, People's Republic of China
| | - Qian Wang
- Hebei University of Chinese Medicine, Shijiazhuang City, Hebei Province 050200, People's Republic of China
| | - Yu-Guang Zheng
- Hebei University of Chinese Medicine, Shijiazhuang City, Hebei Province 050200, People's Republic of China
| |
Collapse
|
18
|
He Z, Du X, Wu Y, Hua L, Wan L, Yan N. Simvastatin promotes endothelial dysfunction by activating the Wnt/β‑catenin pathway under oxidative stress. Int J Mol Med 2019; 44:1289-1298. [PMID: 31432100 PMCID: PMC6713427 DOI: 10.3892/ijmm.2019.4310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 07/11/2019] [Indexed: 12/15/2022] Open
Abstract
Atherosclerosis is a major pathogenic factor in patients with cardiovascular diseases, and endothelial dysfunction (ED) plays a primary role in its occurrence and development. Simvastatin is a lipid‑lowering drug, which is commonly used to prevent or treat risk factors of cardiovascular diseases with a significant anti‑atherogenic effect. However, its impact on endothelial cells under conditions of oxidative stress and broader mechanisms of action remain unclear. The present study evaluated the effect of simvastatin on human umbilical vein endothelial cells (HUVECs) under oxidative stress with H2O2, and the associated mechanisms. At a high dose (1 µM), simvastatin exacerbated H2O2‑induced endothelial cell dysfunction. Moreover, inhibition of the Wnt/β‑catenin pathway by salinomycin significantly suppressed the simvastatin‑associated HUVEC dysfunction. Western blot analysis further demonstrated that simvastatin promoted the phosphorylation of low‑density lipoprotein receptor‑related protein 6 (LRP6) and activated the Wnt/β‑catenin pathway. Simvastatin also activated endoplasmic reticulum (ER) stress, which was reversed by salinomycin treatment. Based on these results, it was hypothesized that simvastatin may promote ER stress by facilitating LRP6 phosphorylation and the subsequent activation of the Wnt/β‑catenin pathway, thereby enhancing H2O2‑induced ED. Therefore, high‑dose simvastatin treatment could have potential toxic side effects, indicating the need for close clinical management, monitoring and patient selection.
Collapse
Affiliation(s)
- Zhiqiang He
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xinyue Du
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yifan Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Lingyue Hua
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Linxi Wan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Nianlong Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
19
|
He Z, He X, Liu M, Hua L, Wang T, Liu Q, Chen L, Yan N. Simvastatin Attenuates H 2O 2-Induced Endothelial Cell Dysfunction by Reducing Endoplasmic Reticulum Stress. Molecules 2019; 24:molecules24091782. [PMID: 31071981 PMCID: PMC6539125 DOI: 10.3390/molecules24091782] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 05/07/2019] [Accepted: 05/08/2019] [Indexed: 12/18/2022] Open
Abstract
Atherosclerosis is the pathological basis of cardiovascular disease, whilst endothelial dysfunction (ED) plays a primary role in the occurrence and development of atherosclerosis. Simvastatin has been shown to possess significant anti-atherosclerosis activity. In this study, we evaluated the protective effect of simvastatin on endothelial cells under oxidative stress and elucidated its underlying mechanisms. Simvastatin was found to attenuate H2O2-induced human umbilical vein endothelial cells (HUVECs) dysfunction and inhibit the Wnt/β-catenin pathway; however, when this pathway was activated by lithium chloride, endothelial dysfunction was clearly enhanced. Further investigation revealed that simvastatin did not alter the expression or phosphorylation of LRP6, but reduced intracellular cholesterol deposition and inhibited endoplasmic reticulum (ER) stress. Inducing ER stress with tunicamycin activated the Wnt/β-catenin pathway, whereas reducing ER stress with 4-phenylbutyric acid inhibited it. We hypothesize that simvastatin does not affect transmembrane signal transduction in the Wnt/β-catenin pathway, but inhibits ER stress by reducing intracellular cholesterol accumulation, which blocks intracellular signal transduction in the Wnt/β-catenin pathway and ameliorates endothelial dysfunction.
Collapse
Affiliation(s)
- Zhiqiang He
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science; Nanchang University, Nanchang 330006, China.
| | - Xuanhong He
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science; Nanchang University, Nanchang 330006, China.
| | - Menghan Liu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science; Nanchang University, Nanchang 330006, China.
| | - Lingyue Hua
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science; Nanchang University, Nanchang 330006, China.
| | - Tian Wang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science; Nanchang University, Nanchang 330006, China.
| | - Qian Liu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science; Nanchang University, Nanchang 330006, China.
| | - Lai Chen
- Laboratory Animal Research Center for Science and Technology, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China.
| | - Nianlong Yan
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science; Nanchang University, Nanchang 330006, China.
| |
Collapse
|
20
|
Which is the best combination of TACE and Sorafenib for advanced hepatocellular carcinoma treatment? A systematic review and network meta-analysis. Pharmacol Res 2018; 135:89-101. [PMID: 29959032 DOI: 10.1016/j.phrs.2018.06.021] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 06/13/2018] [Accepted: 06/20/2018] [Indexed: 02/06/2023]
Abstract
The aim of this study was to assess the comparative efficacy and safety of combination therapy with transarterial chemoembolization (TACE) and Sorafenib for patients with advanced hepatocellular carcinoma (HCC) through a systematic review and network meta-analysis and identify the best combination of TACE and Sorafenib. We searched databases for publications prior to May 2018. The prespecified efficacy outcomes were the objective response rate, overall survival rate, and time to progression. adverse effects included dermatologic, gastrointestinal, and general disorders. Subgroup analyses, meta-regression, and a network meta-analysis regarding two types of outcomes by different chemotherapy agents in TACE (5-fluorouracil, Adriamycin, Platinum, mitomycin C, hydroxycamptothecin) were included. The study is registered with PROSPERO (CRD42018098541). For efficacy outcomes, subgroups which included 5-fluorouracil and hydroxycamptothecin ranked higher than other chemotherapy agents, while mitomycin C ranked the lowest. For advanced effects, the use of mitomycin C or 5-fluorouracil as the chemotherapy agent ranked higher, while hydroxycamptothecin ranked the lowest. Therefore, we excluded 5-Fu and Mitomycin C in subsequent studies. Additionally, in the evaluation of primary adverse effects by the network meta-analysis, Platinum ranked the highest while hydroxycamptothecin ranked the lowest. Therefore, we excluded Platinum this time. Furthermore, all types of Adriamycin are not same, and some studies included two types of Adriamycin. The network meta-analysis results showed that the TACE (hydroxycamptothecin + pirarubicin) +Sorafenib arm and TACE (hydroxycamptothecin + epirubicin) +Sorafenib arm had significant efficacy differences. In conclusion, for patients with advanced HCC, combination therapy with HCPT plus THP/EPI in TACE and Sorfenib may be used as a first-line treatment.
Collapse
|