1
|
Wang Y, Bai M, Peng Q, Li L, Tian F, Guo Y, Jing C. Angiogenesis, a key point in the association of gut microbiota and its metabolites with disease. Eur J Med Res 2024; 29:614. [PMID: 39710789 DOI: 10.1186/s40001-024-02224-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/15/2024] [Indexed: 12/24/2024] Open
Abstract
The gut microbiota is a complex and dynamic ecosystem that plays a crucial role in human health and disease, including obesity, diabetes, cardiovascular diseases, neurodegenerative diseases, inflammatory bowel disease, and cancer. Chronic inflammation is a common feature of these diseases and is closely related to angiogenesis (the process of forming new blood vessels), which is often dysregulated in pathological conditions. Inflammation potentially acts as a central mediator. This abstract aims to elucidate the connection between the gut microbiota and angiogenesis in various diseases. The gut microbiota influences angiogenesis through various mechanisms, including the production of metabolites that directly or indirectly affect vascularization. For example, short-chain fatty acids (SCFAs) such as butyrate, propionate, and acetate are known to regulate immune responses and inflammation, thereby affecting angiogenesis. In the context of cardiovascular diseases, the gut microbiota promotes atherosclerosis and vascular dysfunction by producing trimethylamine N-oxide (TMAO) and other metabolites that promote inflammation and endothelial dysfunction. Similarly, in neurodegenerative diseases, the gut microbiota may influence neuroinflammation and the integrity of the blood-brain barrier, thereby affecting angiogenesis. In cases of fractures and wound healing, the gut microbiota promotes angiogenesis by activating inflammatory responses and immune effects, facilitating the healing of tissue damage. In cancer, the gut microbiota can either inhibit or promote tumor growth and angiogenesis, depending on the specific bacterial composition and their metabolites. For instance, some bacteria can activate inflammasomes, leading to the production of inflammatory factors that alter the tumor immune microenvironment and activate angiogenesis-related signaling pathways, affecting tumor angiogenesis and metastasis. Some bacteria can directly interact with tumor cells, activating angiogenesis-related signaling pathways. Diet, as a modifiable factor, significantly influences angiogenesis through diet-derived microbial metabolites. Diet can rapidly alter the composition of the microbiota and its metabolic activity, thereby changing the concentration of microbial-derived metabolites and profoundly affecting the host's immune response and angiogenesis. For example, a high animal protein diet promotes the production of pro-atherogenic metabolites like TMAO, activating inflammatory pathways and interfering with platelet function, which is associated with the severity of coronary artery plaques, peripheral artery disease, and cardiovascular diseases. A diet rich in dietary fiber promotes the production of SCFAs, which act as ligands for cell surface or intracellular receptors, regulating various biological processes, including inflammation, tissue homeostasis, and immune responses, thereby influencing angiogenesis. In summary, the role of the gut microbiota in angiogenesis is multifaceted, playing an important role in disease progression by affecting various biological processes such as inflammation, immune responses, and multiple signaling pathways. Diet-derived microbial metabolites play a crucial role in linking the gut microbiota and angiogenesis. Understanding the complex interactions between diet, the gut microbiota, and angiogenesis has the potential to uncover novel therapeutic targets for managing these conditions. Therefore, interventions targeting the gut microbiota and its metabolites, such as through fecal microbiota transplantation (FMT) and the application of probiotics to alter the composition of the gut microbiota and enhance the production of beneficial metabolites, present a promising therapeutic strategy.
Collapse
Affiliation(s)
- Yan Wang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Mingshuai Bai
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Qifan Peng
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Leping Li
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Feng Tian
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China.
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| | - Ying Guo
- Department of Breast and Thyroid Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| | - Changqing Jing
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China.
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| |
Collapse
|
2
|
Motamedzadeh A, Rahmati-Dehkordi F, Heydari H, Behnam M, Rashidi Noshabad FZ, Tamtaji Z, Taheri AT, Nabavizadeh F, Aschner M, Mirzaei H, Tamtaji OR. Therapeutic potential of Phycocyanin in gastrointestinal cancers and related disorders. Mol Biol Rep 2024; 51:741. [PMID: 38874869 DOI: 10.1007/s11033-024-09675-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 05/23/2024] [Indexed: 06/15/2024]
Abstract
Gastrointestinal cancer is the most fatal cancer worldwide. The etiology of gastrointestinal cancer has yet to be fully characterized. Alcohol consumption, obesity, tobacco, Helicobacter pylori and gastrointestinal disorders, including gastroesophageal reflux disease, gastric ulcer, colon polyps and non-alcoholic fatty liver disease are among the several risks factors for gastrointestinal cancers. Phycocyanin which is abundant in Spirulina. Phycocyanin, a member of phycobiliprotein family with intense blue color, is an anti-diabetic, neuroprotective, anti-oxidative, anti-inflammatory, and anticancer compound. Evidence exists supporting that phycocyanin has antitumor effects, exerting its pharmacological effects by targeting a variety of cellular and molecular processes, i.e., apoptosis, cell-cycle arrest, migration and Wnt/β-catenin signaling. Phycocyanin has also been applied in treatment of several gastrointestinal disorders such as, gastric ulcer, ulcerative colitis and fatty liver that is known as a risk factor for progression to cancer. Herein, we summarize various cellular and molecular pathways that are affected by phycocyanin, its efficacy upon combined drug treatment, and the potential for nanotechnology in its gastrointestinal cancer therapy.
Collapse
Affiliation(s)
- Alireza Motamedzadeh
- Department of Internal Medicine, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Rahmati-Dehkordi
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hoora Heydari
- Student Research Committee, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohammad Behnam
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Zeinab Tamtaji
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Abdolkarim Talebi Taheri
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Nabavizadeh
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran.
- Department of Physiology, School of Medicine, Tehran University of medical sciences, Tehran, Iran.
| | - Omid Reza Tamtaji
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Physiology, School of Medicine, Tehran University of medical sciences, Tehran, Iran.
| |
Collapse
|
3
|
O’Neill CE, Sun K, Sundararaman S, Chang JC, Glynn SA. The impact of nitric oxide on HER family post-translational modification and downstream signaling in cancer. Front Physiol 2024; 15:1358850. [PMID: 38601214 PMCID: PMC11004480 DOI: 10.3389/fphys.2024.1358850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/16/2024] [Indexed: 04/12/2024] Open
Abstract
The human epidermal growth factor receptor (HER) family consists of four members, activated by two families of ligands. They are known for mediating cell-cell interactions in organogenesis, and their deregulation has been associated with various cancers, including breast and esophageal cancers. In particular, aberrant epidermal growth factor receptor (EGFR) and HER2 signaling drive disease progression and result in poorer patient outcomes. Nitric oxide (NO) has been proposed as an alternative activator of the HER family and may play a role in this aberrant activation due to its ability to induce s-nitrosation and phosphorylation of the EGFR. This review discusses the potential impact of NO on HER family activation and downstream signaling, along with its role in the efficacy of therapeutics targeting the family.
Collapse
Affiliation(s)
- Ciara E. O’Neill
- Lambe Institute for Translational Research, Discipline of Pathology, School of Medicine, University of Galway, Galway, Ireland
| | - Kai Sun
- Houston Methodist Research Institute, Houston, TX, United States
- Dr Mary and Ron Neal Cancer Center, Houston Methodist Hospital, Houston, TX, United States
| | | | - Jenny C. Chang
- Houston Methodist Research Institute, Houston, TX, United States
- Dr Mary and Ron Neal Cancer Center, Houston Methodist Hospital, Houston, TX, United States
| | - Sharon A. Glynn
- Lambe Institute for Translational Research, Discipline of Pathology, School of Medicine, University of Galway, Galway, Ireland
| |
Collapse
|
4
|
Zhang Y, Wu X, Zhu J, Lu R, Ouyang Y. Knockdown of SLC39A14 inhibits glioma progression by promoting erastin-induced ferroptosis SLC39A14 knockdown inhibits glioma progression. BMC Cancer 2023; 23:1120. [PMID: 37978473 PMCID: PMC10655456 DOI: 10.1186/s12885-023-11637-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Ferroptosis is a newly classified form of regulated cell death with implications in various tumor progression pathways. However, the roles and mechanisms of ferroptosis-related genes in glioma remain unclear. METHODS Bioinformatics analysis was employed to identify differentially expressed ferroptosis-related genes in glioma. The expression levels of hub genes were assessed using real-time reverse transcriptase-polymerase chain reaction (RT-qPCR). To explore the role of SLC39A14 in glioma, a series of in vitro assays were conducted, including cell counting kit-8 (CCK-8), 5-ethynyl-2'-deoxyuridine (EdU), flow cytometry, wound healing, and Transwell assays. Enzyme-linked immunosorbent assay (ELISA) was utilized to measure the levels of indicators associated with ferroptosis. Hematoxylin-eosin (HE) and immunohistochemistry (IHC) staining were performed to illustrate the clinicopathological features of the mouse transplantation tumor model. Additionally, Western blot analysis was used to assess the expression of the cGMP-PKG pathway-related proteins. RESULTS Seven ferroptosis-related hub genes, namely SLC39A14, WWTR1, STEAP3, NOTCH2, IREB2, HIF1A, and FANCD2, were identified, all of which were highly expressed in glioma. Knockdown of SLC39A14 inhibited glioma cell proliferation, migration, and invasion, while promoting apoptosis. Moreover, SLC39A14 knockdown also facilitated erastin-induced ferroptosis, leading to the suppression of mouse transplantation tumor growth. Mechanistically, SLC39A14 knockdown inhibited the cGMP-PKG signaling pathway activation. CONCLUSION Silencing SLC39A14 inhibits ferroptosis and tumor progression, potentially involving the regulation of the cGMP-PKG signaling pathway.
Collapse
Affiliation(s)
- Yunwen Zhang
- Department of Neurosurgery, First Clinical Medical College of Gannan Medical University, No.1 Xueyuan Road, Zhanggong District, Ganzhou City, 341000, Jiangxi Province, China
| | - Xinghai Wu
- Department of Neurosurgery, Zhangye People's Hospital Affiliated to Hexi University, No. 67 Xihuan Road, Ganzhou District, Zhangye City, 734000, Gansu Province, China
| | - Jiyong Zhu
- Department of Neurosurgery, Guilin Municipal Hospital of Traditional Chinese Medicine, Guangxi Zhuang Autonomous Region, No. 2 Lingui Road, Xiangshan District, Guilin City, 541002, China
| | - Ruibin Lu
- Department of Neurosurgery, First Clinical Medical College of Gannan Medical University, No.1 Xueyuan Road, Zhanggong District, Ganzhou City, 341000, Jiangxi Province, China
| | - Yian Ouyang
- Department of Neurosurgery, First Affiliated Hospital of Gannan Medical University, No.23 Qingnian Road, Zhanggong District, Ganzhou City, 341000, Jiangxi Province, China.
| |
Collapse
|
5
|
Xu D, Liu A, Liu Q, Zhang H, Tian M, Bian Y, Zhang X, Ying M, Shen H. Cucurbitacin C suppresses the progression of pancreatic ductal adenocarcinoma via inhibition of the cGMP-PKG-VASP axis. Biochem Pharmacol 2023; 217:115810. [PMID: 37717690 DOI: 10.1016/j.bcp.2023.115810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/14/2023] [Accepted: 09/14/2023] [Indexed: 09/19/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most devastating diseases; it has a considerably poor prognosis and may become the second most lethal malignancy in the next 10 years. Chemotherapeutic resistance is common in PDAC; thus, it is necessary to exploit effective alternative drugs. In recent years, traditional folk medicines and their extracts have shown great potential in cancer treatment. The seed of Lagenaria siceraria (Molina) Standl. is a traditional medicine in Asia. Because of its analgesic effects and ability to reduce swelling, it is often used as an adjuvant treatment for abdominal tumors. Cucurbitacin compounds are extracts abundant in Lagenaria siceraria (Molina) Standl. Here, we found that cucurbitacin C (CuC), a member of the cucurbitacin family, has apparent anti-PDAC therapeutic properties. CuC decreased the viability and suppressed the proliferation of PDAC cells in a time- and dose-dependent manner. Further studies revealed that CuC inhibited cell migration and invasion by inhibiting epithelial-mesenchymal transition (EMT). In addition, G2/M arrest was induced, and the apoptotic pathway was activated. Transcriptomic and bioinformatic analyses showed that CuC inhibited the cGMP-PKG-VASP axis, increasing the content of cGMP to restore tumor characteristics. The antitumor activity of CuC in vivo was verified through animal experiments, and no obvious side effects were observed. Overall, our study indicates a candidate therapeutic compound for PDAC that is worthy of further development.
Collapse
Affiliation(s)
- Dongchao Xu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China; Hangzhou Institute of Digestive Diseases, Hangzhou 310000, China; Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou 310000, China
| | - Ajuan Liu
- Hangzhou Medical College, Hangzhou 311300, China
| | - Qiang Liu
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China; Hangzhou Institute of Digestive Diseases, Hangzhou 310000, China; Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou 310000, China
| | - Hongchen Zhang
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China; Hangzhou Institute of Digestive Diseases, Hangzhou 310000, China; Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou 310000, China
| | - Mengyao Tian
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Ying Bian
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Xiaofeng Zhang
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China; Hangzhou Institute of Digestive Diseases, Hangzhou 310000, China; Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou 310000, China.
| | - Meidan Ying
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Hongzhang Shen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China; Hangzhou Institute of Digestive Diseases, Hangzhou 310000, China; Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou 310000, China.
| |
Collapse
|
6
|
Panneerselvan P, Vasanthakumar K, Muthuswamy K, Krishnan V, Subramaniam S. Insights on the functional dualism of nitric oxide in the hallmarks of cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:189001. [PMID: 37858621 DOI: 10.1016/j.bbcan.2023.189001] [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: 01/29/2023] [Revised: 10/09/2023] [Accepted: 10/09/2023] [Indexed: 10/21/2023]
Abstract
Nitric oxide (NO), a gaseous radical, governs a variety of physiological and pathological processes, including cancer, pro-inflammatory signalling, and vasodilation. The family of nitric oxide synthases (NOS), which comprises the constitutive forms, nNOS and eNOS, and the inducible form, iNOS, produces NO enzymatically. Additionally, NO can be generated non-enzymatically from the nitrate-nitrite-NO pathway. The anti- and pro-oxidant properties of NO and its functional dualism in cancer is due to its highly reactive nature. Numerous malignancies have NOS expression, which interferes with the tumour microenvironment to modulate the tumour's growth in both favourable and unfavourable ways. NO regulates a number of mechanisms in the tumour microenvironment, including metabolism, cell cycle, DNA repair, angiogenesis, and apoptosis/necrosis, depending on its concentration and spatiotemporal profile. This review focuses on the bi-modal impact of nitric oxide on the alteration of a few cancer hallmarks.
Collapse
Affiliation(s)
- Prabha Panneerselvan
- Molecular Physiology Laboratory, Department of Biochemistry, Bharathiar University, Coimbatore, Tamil Nadu 641046, India
| | - Keerthana Vasanthakumar
- Molecular Physiology Laboratory, Department of Biochemistry, Bharathiar University, Coimbatore, Tamil Nadu 641046, India
| | - Karthi Muthuswamy
- Molecular Physiology Laboratory, Department of Biochemistry, Bharathiar University, Coimbatore, Tamil Nadu 641046, India
| | - Vasanth Krishnan
- Molecular Biology Laboratory, Department of Botany, Bharathiar University, Coimbatore, Tamil Nadu 641046, India
| | - Selvakumar Subramaniam
- Molecular Physiology Laboratory, Department of Biochemistry, Bharathiar University, Coimbatore, Tamil Nadu 641046, India.
| |
Collapse
|
7
|
Audero MM, Carvalho TMA, Ruffinatti FA, Loeck T, Yassine M, Chinigò G, Folcher A, Farfariello V, Amadori S, Vaghi C, Schwab A, Reshkin SJ, Cardone RA, Prevarskaya N, Fiorio Pla A. Acidic Growth Conditions Promote Epithelial-to-Mesenchymal Transition to Select More Aggressive PDAC Cell Phenotypes In Vitro. Cancers (Basel) 2023; 15:cancers15092572. [PMID: 37174038 PMCID: PMC10177299 DOI: 10.3390/cancers15092572] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/28/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is characterized by an acidic microenvironment, which contributes to therapeutic failure. So far there is a lack of knowledge with respect to the role of the acidic microenvironment in the invasive process. This work aimed to study the phenotypic and genetic response of PDAC cells to acidic stress along the different stages of selection. To this end, we subjected the cells to short- and long-term acidic pressure and recovery to pHe 7.4. This treatment aimed at mimicking PDAC edges and consequent cancer cell escape from the tumor. The impact of acidosis was assessed for cell morphology, proliferation, adhesion, migration, invasion, and epithelial-mesenchymal transition (EMT) via functional in vitro assays and RNA sequencing. Our results indicate that short acidic treatment limits growth, adhesion, invasion, and viability of PDAC cells. As the acid treatment progresses, it selects cancer cells with enhanced migration and invasion abilities induced by EMT, potentiating their metastatic potential when re-exposed to pHe 7.4. The RNA-seq analysis of PANC-1 cells exposed to short-term acidosis and pHe-selected recovered to pHe 7.4 revealed distinct transcriptome rewiring. We describe an enrichment of genes relevant to proliferation, migration, EMT, and invasion in acid-selected cells. Our work clearly demonstrates that upon acidosis stress, PDAC cells acquire more invasive cell phenotypes by promoting EMT and thus paving the way for more aggressive cell phenotypes.
Collapse
Affiliation(s)
- Madelaine Magalì Audero
- U1003-PHYCELL-Laboratoire de Physiologie Cellulaire, Inserm, University of Lille, Villeneuve d'Ascq, 59000 Lille, France
- Laboratory of Cellular and Molecular Angiogenesis, Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy
| | | | - Federico Alessandro Ruffinatti
- Laboratory of Cellular and Molecular Angiogenesis, Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy
| | - Thorsten Loeck
- Institute of Physiology II, University of Münster, 48149 Münster, Germany
| | - Maya Yassine
- U1003-PHYCELL-Laboratoire de Physiologie Cellulaire, Inserm, University of Lille, Villeneuve d'Ascq, 59000 Lille, France
| | - Giorgia Chinigò
- Laboratory of Cellular and Molecular Angiogenesis, Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy
| | - Antoine Folcher
- U1003-PHYCELL-Laboratoire de Physiologie Cellulaire, Inserm, University of Lille, Villeneuve d'Ascq, 59000 Lille, France
| | - Valerio Farfariello
- U1003-PHYCELL-Laboratoire de Physiologie Cellulaire, Inserm, University of Lille, Villeneuve d'Ascq, 59000 Lille, France
| | - Samuele Amadori
- Laboratory of Cellular and Molecular Angiogenesis, Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy
| | - Chiara Vaghi
- Laboratory of Cellular and Molecular Angiogenesis, Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy
| | - Albrecht Schwab
- Institute of Physiology II, University of Münster, 48149 Münster, Germany
| | - Stephan J Reshkin
- Department of Biosciences, Biotechnologies and Environment, University of Bari, 70126 Bari, Italy
| | - Rosa Angela Cardone
- Department of Biosciences, Biotechnologies and Environment, University of Bari, 70126 Bari, Italy
| | - Natalia Prevarskaya
- U1003-PHYCELL-Laboratoire de Physiologie Cellulaire, Inserm, University of Lille, Villeneuve d'Ascq, 59000 Lille, France
| | - Alessandra Fiorio Pla
- U1003-PHYCELL-Laboratoire de Physiologie Cellulaire, Inserm, University of Lille, Villeneuve d'Ascq, 59000 Lille, France
- Laboratory of Cellular and Molecular Angiogenesis, Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy
| |
Collapse
|
8
|
Fan B, Zheng C, Wang N, Chang Z, Liu Y, Wang C, Xiang J, Tao Y, Wang G, Zhang Q. CircSTK3 drives the metastasis of colorectal cancer by regulating epithelial-mesenchymal transition. iScience 2023; 26:106170. [PMID: 36922993 PMCID: PMC10009203 DOI: 10.1016/j.isci.2023.106170] [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: 07/12/2022] [Revised: 11/24/2022] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Circular RNAs (circRNAs) play crucial roles in malignancies. We aimed to delineate the functions and clinical importance of dysregulated circRNAs in colorectal cancer (CRC). We determined the circRNA expression profile from five CRC and paired adjacent normal tissues using circRNA microarray. We found that a novel circRNA, hsa_circ_0004592 (named circSTK3), was significantly upregulated in CRC tissues and correlated with decreased survival. Loss- and gain-of-function assays revealed that circSTK3 promoted the migration and invasion but not proliferation of cells. Whole genome expression microarray identified potential downstream targets and the regulatory networks of circSTK3; Gene Ontology analysis confirmed circSTK3 involvement in the CRC metastasis phenotype. Abnormal circSTK3 expression affected a subset of genes associated with CRC metastasis and triggered epithelial-mesenchymal transition programming, maintaining a tumor-promoting signature. Moreover, circSTK3 was transcriptionally regulated by CTCF. These findings reveal the functional and prognostic roles of circSTK3 and expose circRNAs as key players in metastasis.
Collapse
Affiliation(s)
- Boyang Fan
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Chaojing Zheng
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Ning Wang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Zewen Chang
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Yunxiao Liu
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Chunlin Wang
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Jun Xiang
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Yangbao Tao
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Guiyu Wang
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Qian Zhang
- Department of Colorectal Surgery, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou 310000, China
| |
Collapse
|
9
|
The Expression of ARMCX1 in Gastric Cancer Contributes to Prognosis and Influences Chemotherapy. J Immunol Res 2023; 2023:2623317. [PMID: 36726491 PMCID: PMC9886469 DOI: 10.1155/2023/2623317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 10/26/2022] [Indexed: 01/24/2023] Open
Abstract
The altered expression of ARMCX1 in patients with gastric cancer has been reported frequently, yet its correlation to prognosis and chemotherapy needs to be unveiled. In combination of the gene expression data retrieved from TCGA database and bioinformatic analysis, this study discovered 590 differentially expressed genes in the cancerous biopsies isolated from gastric patients, compared with controls. Among which, ARMCX1 exhibited great potential to serve as a prognostic biomarker for gastric patients; furthermore, patients with low expression of ARMCX1 could be more sensitive to these 9 chemotherapeutic agents: A-770041, AMG-706, ATRA, BEZ235, bortezomib, CGP60474, dasatinib, HG-64-1, and pazopanib, rather than the other chemotherapeutic agents. This study helps the improvement of evaluating the prognosis of gastric cancer patients, and would help optimize chemotherapeutic strategies in consideration of the expression of ARMCX1.
Collapse
|
10
|
Xu K, Mittal K, Ewald J, Rulli S, Jakubowski JL, George S, Basu N. Transcriptomic points of departure calculated from human intestinal cells exposed to dietary nanoparticles. Food Chem Toxicol 2022; 170:113501. [DOI: 10.1016/j.fct.2022.113501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
|
11
|
Yu B, Deng Y, Jia F, Wang Y, Jin Q, Ji J. A Supramolecular Nitric Oxide Nanodelivery System for Prevention of Tumor Metastasis by Inhibiting Platelet Activation and Aggregation. ACS APPLIED MATERIALS & INTERFACES 2022; 14:48515-48526. [PMID: 36278897 DOI: 10.1021/acsami.2c15882] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Tumor cell-induced platelet aggregation (TCIPA) is known as a critical step in hematogenous tumor metastasis. The endogenous nitric oxide (NO) plays an important role in anticoagulation, which might have great potential to inhibit TCIPA. Herein, a glutathione-sensitive supramolecular nanocarrier is prepared via host-guest interaction for effective delivery of NO and chemotherapeutic agent gemcitabine (GEM). NO could be effectively released in tumor cells and inhibits platelet activation and aggregation. The inhibition of TCIPA by NO could effectively attenuate the migration and invasion of tumor cells in vitro. Furthermore, the in vivo experiments demonstrate that the NO and GEM co-delivered supramolecular nanocarriers can suppress the growth of primary tumor. More importantly, although NO-containing nanocarriers cannot inhibit the growth of primary tumors effectively, they can significantly inhibit tumor metastasis. This NO-based nano-delivery system not only provides new inspiration for multifunctional applications of NO in cancer therapy but also shows great potential in clinical antimetastatic applications.
Collapse
Affiliation(s)
- Bo Yu
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang310027, P. R. China
| | - Yongyan Deng
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang310027, P. R. China
| | - Fan Jia
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang310027, P. R. China
| | - Youxiang Wang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang310027, P. R. China
| | - Qiao Jin
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang310027, P. R. China
| | - Jian Ji
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang310027, P. R. China
| |
Collapse
|
12
|
Salihi A, Al-Naqshabandi MA, Khudhur ZO, Housein Z, Hama HA, Abdullah RM, Hussen BM, Alkasalias T. Gasotransmitters in the tumor microenvironment: Impacts on cancer chemotherapy (Review). Mol Med Rep 2022; 26:233. [PMID: 35616143 PMCID: PMC9178674 DOI: 10.3892/mmr.2022.12749] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 04/07/2022] [Indexed: 11/23/2022] Open
Abstract
Nitric oxide, carbon monoxide and hydrogen sulfide are three endogenous gasotransmitters that serve a role in regulating normal and pathological cellular activities. They can stimulate or inhibit cancer cell proliferation and invasion, as well as interfere with cancer cell responses to drug treatments. Understanding the molecular pathways governing the interactions between these gases and the tumor microenvironment can be utilized for the identification of a novel technique to disrupt cancer cell interactions and may contribute to the conception of effective and safe cancer therapy strategies. The present review discusses the effects of these gases in modulating the action of chemotherapies, as well as prospective pharmacological and therapeutic interfering approaches. A deeper knowledge of the mechanisms that underpin the cellular and pharmacological effects, as well as interactions, of each of the three gases could pave the way for therapeutic treatments and translational research.
Collapse
Affiliation(s)
- Abbas Salihi
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region 44001, Iraq
- Center of Research and Strategic Studies, Lebanese French University, Erbil, Kurdistan Region 44002, Iraq
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum, Karolinska Institutet, SE-17165 Stockholm, Sweden
| | - Mohammed A. Al-Naqshabandi
- Department of Clinical Biochemistry, College of Health Sciences, Hawler Medical University, Erbil, Kurdistan Region 44001, Iraq
| | - Zhikal Omar Khudhur
- Department of Medical Analysis, Faculty of Applied Science, Tishk International University, Erbil, Kurdistan Region 44001, Iraq
| | - Zjwan Housein
- Department of Medical Laboratory Technology, Technical Health and Medical College, Erbil Polytechnique University, Erbil, Kurdistan Region 44002, Iraq
| | - Harmand A. Hama
- Department of Biology, Faculty of Education, Tishk International University, Erbil, Kurdistan Region 44002, Iraq
| | - Ramyar M. Abdullah
- College of Medicine, Hawler Medical University, Erbil, Kurdistan Region 44002, Iraq
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Erbil, Kurdistan Region 44002, Iraq
| | - Twana Alkasalias
- General Directorate of Scientific Research Center, Salahaddin University-Erbil, Erbil, Kurdistan Region 44002, Iraq
- Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, SE-17176 Stockholm, Sweden
| |
Collapse
|
13
|
Harnessing oxidative stress for anti-glioma therapy. Neurochem Int 2022; 154:105281. [PMID: 35038460 DOI: 10.1016/j.neuint.2022.105281] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 12/22/2021] [Accepted: 01/10/2022] [Indexed: 02/06/2023]
Abstract
Glioma cells use intermediate levels of reactive oxygen species (ROS) and reactive nitrogen species (RNS) for growth and invasion, and suppressing these reactive molecules thus may compromise processes that are vital for glioma survival. Increased oxidative stress has been identified in glioma cells, in particular in glioma stem-like cells. Studies have shown that these cells harbor potent antioxidant defenses, although endogenous protection against nitrosative stress remains understudied. The enhancement of oxidative or nitrosative stress offers a potential target for triggering glioma cell death, but whether oxidative and nitrosative stresses can be combined for therapeutic effects requires further research. The optimal approach of harnessing oxidative stress for anti-glioma therapy should include the induction of free radical-induced oxidative damage and the suppression of antioxidant defense mechanisms selectively in glioma cells. However, selective induction of oxidative/nitrosative stress in glioma cells remains a therapeutic challenge, and research into selective drug delivery systems is ongoing. Because of multifactorial mechanisms of glioma growth, progression, and invasion, prospective oncological therapies may include not only therapeutic oxidative/nitrosative stress but also inhibition of oncogenic kinases, antioxidant molecules, and programmed cell death mediators.
Collapse
|
14
|
Affiliation(s)
- Xianxian Yao
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science Fudan University Shanghai China
| | - Binru Yang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science Fudan University Shanghai China
| | - Jian Xu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science Fudan University Shanghai China
| | - Qianjun He
- Guangdong Provincial Key Laboratory of Biomedical Measurements and Ultrasound Imaging National‐Regional Key Technology Engineering Laboratory for Medical Ultrasound School of Biomedical Engineering Health Science Center Shenzhen University Shenzhen China
| | - Wuli Yang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science Fudan University Shanghai China
| |
Collapse
|
15
|
Yang D, Li J, Liang C, Tian L, Shi C, Hui N, Liu Y, Ling M, Xin L, Wan M, Li H, Zhao Q, Ren X, Liu H, Cao W. Syringa microphylla Diels: A comprehensive review of its phytochemical, pharmacological, pharmacokinetic, and toxicological characteristics and an investigation into its potential health benefits. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 93:153770. [PMID: 34678528 DOI: 10.1016/j.phymed.2021.153770] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 07/31/2021] [Accepted: 09/20/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Syringa microphylla Diels is a plant in the family Syringa Linn. For hundreds of years, its flowers and leaves have been used as a folk medicine for the treatment of cough, inflammation, colds, sore throat, acute hepatitis, chronic hepatitis, early liver cirrhosis, fatty liver, and oesophageal cancer. PURPOSE For the first time, we have comprehensively reviewed information on Syringa microphylla Diels that is not included in the Pharmacopoeia, clarified the pharmacological mechanisms of Syringa microphylla Diels and its active ingredients from a molecular biology perspective, compiled in vivo and in vitro animal experimental data and clinical data, and summarized the toxicology and pharmacokinetics of Syringa microphylla Diels. The progress in toxicology research is expected to provide a theoretical basis for the development of new drugs from Syringa microphylla Diels, a natural source of compounds that are potentially beneficial to human health. METHODS The PubMed, Google Scholar, China National Knowledge Infrastructure, Web of Science, SciFinder Scholar and Thomson Reuters databases were utilized to conduct a comprehensive search of published literature as of July 2021 to find original literature related to Syringa microphylla Diels and its active ingredients. RESULTS To date, 72 compounds have been isolated and identified from Syringa microphylla Diels, and oleuropein, verbascoside, isoacteoside, echinacoside, forsythoside B, and eleutheroside B are the main active components. These compounds have antioxidant, antibacterial, anti-inflammatory, and neuroprotective effects, and their safety and effectiveness have been demonstrated in long-term traditional applications. Molecular pharmacology experiments have indicated that the active ingredients of Syringa microphylla Diels exert their pharmacological effects in various ways, primarily by reducing oxidative stress damage via Nrf2/ARE pathway regulation, regulating inflammatory factors and inducing apoptosis through the MAPK and NF-κB pathways. CONCLUSION This comprehensive review of Syringa microphylla Diels provides new insights into the correlations among molecular mechanisms, the importance of toxicology and pharmacokinetics, and potential ways to address the limitations of current research. As Syringa microphylla Diels is a natural low-toxicity botanical medicine, it is worthy of development and utilization and is an excellent choice for treating various diseases.
Collapse
Affiliation(s)
- Dan Yang
- School of Food and Bioengineering, Shaanxi University of Science & Technology, Xi'an 710021, PR China
| | - Jingyi Li
- School of Food and Bioengineering, Shaanxi University of Science & Technology, Xi'an 710021, PR China
| | - Chengyuan Liang
- School of Food and Bioengineering, Shaanxi University of Science & Technology, Xi'an 710021, PR China.
| | - Lei Tian
- School of Food and Bioengineering, Shaanxi University of Science & Technology, Xi'an 710021, PR China; College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science & Technology, Xi'an 710021, PR China
| | - Chunyang Shi
- School of Food and Bioengineering, Shaanxi University of Science & Technology, Xi'an 710021, PR China
| | - Nan Hui
- School of Food and Bioengineering, Shaanxi University of Science & Technology, Xi'an 710021, PR China
| | - Yuan Liu
- School of Food and Bioengineering, Shaanxi University of Science & Technology, Xi'an 710021, PR China
| | - Mei Ling
- School of Food and Bioengineering, Shaanxi University of Science & Technology, Xi'an 710021, PR China
| | - Liang Xin
- School of Food and Bioengineering, Shaanxi University of Science & Technology, Xi'an 710021, PR China
| | - Minge Wan
- School of Medicine and Pharmacy, Shaanxi University of Business & Commerce, Xi'an 712046, PR China
| | - Han Li
- School of Food and Bioengineering, Shaanxi University of Science & Technology, Xi'an 710021, PR China
| | - Qianqian Zhao
- School of Food and Bioengineering, Shaanxi University of Science & Technology, Xi'an 710021, PR China
| | - Xiaodong Ren
- Medical College, Guizhou University, Guiyang 550025, PR China.
| | - Hong Liu
- Zhuhai Jinan Selenium Source Nanotechnology Co., Ltd., Hengqin New Area, Zhuhai 519030, PR China.
| | - Wenqiang Cao
- Zhuhai Jinan Selenium Source Nanotechnology Co., Ltd., Hengqin New Area, Zhuhai 519030, PR China
| |
Collapse
|
16
|
The role of extracellular matrix in tumour angiogenesis: the throne has NOx servants. Biochem Soc Trans 2021; 48:2539-2555. [PMID: 33150941 PMCID: PMC7752075 DOI: 10.1042/bst20200208] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/28/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023]
Abstract
The extracellular matrix (ECM) dynamics in tumour tissue are deregulated compared to the ECM in healthy tissue along with disorganized architecture and irregular behaviour of the residing cells. Nitric oxide (NO) as a pleiotropic molecule exerts different effects on the components of the ECM driving or inhibiting augmented angiogenesis and tumour progression and tumour cell proliferation and metastasis. These effects rely on the concentration of NO within the tumour tissue, the nature of the surrounding microenvironment and the sensitivity of resident cells to NO. In this review article, we summarize the recent findings on the correlation between the levels of NO and the ECM components towards the modulation of tumour angiogenesis in different types of cancers. These are discussed principally in the context of how NO modulates the expression of ECM proteins resulting in either the promotion or inhibition of tumour growth via tumour angiogenesis. Furthermore, the regulatory effects of individual ECM components on the expression of the NO synthase enzymes and NO production were reviewed. These findings support the current efforts for developing effective therapeutics for cancers.
Collapse
|
17
|
Protein Phosphorylation in Cancer: Role of Nitric Oxide Signaling Pathway. Biomolecules 2021; 11:biom11071009. [PMID: 34356634 PMCID: PMC8301900 DOI: 10.3390/biom11071009] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 12/16/2022] Open
Abstract
Nitric oxide (NO), a free radical, plays a critical role in a wide range of physiological and pathological processes. Due to its pleiotropic function, it has been widely investigated in various types of cancers and is strongly associated with cancer development. Mounting pieces of evidence show that NO regulates various cancer-related events, which mainly depends on phosphorylating the key proteins in several signaling pathways. However, phosphorylation of proteins modulated by NO signaling pathway may lead to different effects in different types of cancer, which is complex and remains unclear. Therefore, in this review, we focus on the effect of protein phosphorylation modulated by NO signaling pathway in different types of cancers including breast cancer, lung cancer, prostate cancer, colon cancer, gastric cancer, pancreatic cancer, ovarian cancer, and neuroblastoma. Phosphorylation of key proteins, including p38 MAPK, ERK, PI3K, STAT3, and p53, modified by NO in various signaling pathways affects different cancer-related processes including cell apoptosis, proliferation, angiogenesis, metastasis, and several cancer therapies. Our review links the NO signaling pathway to protein phosphorylation in cancer development and provides new insight into potential targets and cancer therapy.
Collapse
|
18
|
AboYoussef AM, Khalaf MM, Malak MN, Hamzawy MA. Repurposing of sildenafil as antitumour; induction of cyclic guanosine monophosphate/protein kinase G pathway, caspase-dependent apoptosis and pivotal reduction of Nuclear factor kappa light chain enhancer of activated B cells in lung cancer. J Pharm Pharmacol 2021; 73:1080-1091. [PMID: 33856030 DOI: 10.1093/jpp/rgab049] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 02/23/2021] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Lung cancer is one of the most frequent types of cancers that lead to death. Sildenafil is a potent inhibitor of phosphodiesterase-5 and showed potential anticancer effects, which has not yet been fully evaluated. Thus, this study aims to investigate the potential anticancer effect of sildenafil in urethane-induced lung cancer in BALB/c mice. METHODS Five-week-old male BALB/c mice were treated with either (i) normal saline only, (ii) sildenafil only 50 mg kg-1/ P.O every other day for the last four successive weeks, (iii) urethane 1.5 gm kg-1 i.p (at day 1 and day 60), (iv) carboplatin after urethane induction, or (v) sildenafil after urethane induction. KEY FINDINGS It was shown that sildenafil significantly increased the levels of cGMP and Caspase-3 with a reduction of NF-κB, Bcl-2, Cyclin D1, intercellular adhesion molecule 1, matrix metalloproteinase-2 levels and normalisation of Nrf2 along with pronounced improvement in the histological patterns. CONCLUSIONS These results indicated that sildenafil markedly induces cell cycle arrest, apoptosis and inhibits the metastatic activity through activation of cyclic guanosine monophosphate/protein kinase G pathway and down-regulation of cyclin D1 and nuclear factor kappa light chain enhancer of activated B cells with downstream anti-apoptotic gene Bcl-2, which underscores the critical importance of future using sildenafil in the treatment of lung cancer.
Collapse
Affiliation(s)
- Amira M AboYoussef
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Marwa M Khalaf
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Marina N Malak
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Mohamed A Hamzawy
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Fayoum University, Fayoum, Egypt
| |
Collapse
|
19
|
Tan Y, Johnson M, Zhou J, Zhao Y, Kamal MA, Qu X. Antrodia cinnamomea Inhibits Growth and Migration of Lung Cancer Cells through Regulating p53-Bcl2 and MMPs Pathways. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2020; 48:1941-1953. [PMID: 33300478 DOI: 10.1142/s0192415x20500974] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Antrodia cinnamomea has been shown to possess antitumor activity. This study investigated the effects and mechanisms of Antrodia cinnamomea extract (ACE) on growth and migration of human non-small cell lung cancer A549 cells. The effect of ACE on cell viability was determined by MTT assay and fluorescent live-cell imaging. The apoptotic effect of ACE was determined by cell cycle distribution using flow cytometry. A P53-mediated apoptosis pathway was identified by measuring protein expression of p53 and Bcl-2 with Western blotting. Additionally, mRNA expression of p53 and Bcl-2 and Bax was detected by qRT-PCR. The effect of ACE on cancer cell migration was confirmed by a wound-healing assay. Expression of MMP-2 and MMP-9 at the protein and gene levels was determined by western blot and qRT-PCR analysis. This study demonstrates the inhibitory effect of ACE on A549 cell proliferation in a dose-response manner with an [Formula: see text]. It was determined that ACE concentration at [Formula: see text] induced cell cycle arrest at S phase in A549 cells. The apoptosis-regulating protein p53 expression was enhanced and also associated with the downregulation of Bcl-2 in ACE treatment cells. The mRNA expression of p53 and Bcl-2 associated with Bxa was consistent with protein expression. The inhibition of migration of cancer cells treated with ACE was clearly evident. At the same time, suppression of expression of MMP-2 and MMP-9 at protein and mRNA levels was observed. The findings of this study highlight ACE as a potential agent of adjuvant therapy for lung cancer.
Collapse
Affiliation(s)
- Yi Tan
- School of Life Sciences, University of Technology Sydney, NSW 2007, Australia
| | - Michael Johnson
- School of Life Sciences, University of Technology Sydney, NSW 2007, Australia
| | - Jiong Zhou
- School of Life Sciences, University of Technology Sydney, NSW 2007, Australia
| | - Yi Zhao
- School of Life Sciences, University of Technology Sydney, NSW 2007, Australia
| | - Mohammad Amjad Kamal
- School of Life Sciences, University of Technology Sydney, NSW 2007, Australia.,King Fahd Medical Research Center, King Abdulaziz University, Saudi Arabia
| | - Xianqin Qu
- School of Life Sciences, University of Technology Sydney, NSW 2007, Australia
| |
Collapse
|
20
|
Anand CR, Bhavya B, Jayakumar K, Harikrishnan VS, Gopala S. Inorganic nitrite alters mitochondrial dynamics without overt changes in cell death and mitochondrial respiration in cardiomyoblasts under hyperglycemia. Toxicol In Vitro 2020; 70:105048. [PMID: 33161133 DOI: 10.1016/j.tiv.2020.105048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 11/03/2020] [Accepted: 11/04/2020] [Indexed: 10/23/2022]
Abstract
Inorganic nitrate or nitrite supplementation has been reported to demonstrate positive outcomes in rodent models of obesity and diabetes as well as in type 2 diabetic humans and even included in clinical trials pertaining to cardiovascular diseases in the recent decade. However, there are contrasting data regarding the useful and toxic effects of the anions. The primary scope of this study was to analyze the beneficial/detrimental alterations in redox status, mitochondrial dynamics and function, and cellular fitness in cardiomyoblasts inflicted by nitrite under hyperglycemic conditions compared with normoglycemia. Nitrite supplementation in H9c2 myoblasts under high glucose diminishes the Bcl-xL expression and mitochondrial ROS levels without significant initiation of cell death or decline in total ROS levels. Concomitantly, there are tendencies towards lowering of mitochondrial membrane potential, but without noteworthy changes in mitochondrial biogenesis and respiration. The study also revealed that under high glucose stress, nitrite may alter mitochondrial dynamics by Drp1 activation possibly via Akt1-Pim1 axis. Moreover, the study revealed differential effects of Drp1 silencing and/or nitrite under the above glycemic conditions. Overall, the study warrants more research regarding the effects of nitrite therapy in cardiac cells exposed to hyperglycemia.
Collapse
Affiliation(s)
- C R Anand
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram 695011, Kerala, India
| | - Bharathan Bhavya
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram 695011, Kerala, India
| | - K Jayakumar
- Department of Cardiovascular and Thoracic Surgery, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram 695011, Kerala, India.
| | - V S Harikrishnan
- Division of Laboratory Animal Sciences, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram 695011, Kerala, India.
| | - Srinivas Gopala
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram 695011, Kerala, India.
| |
Collapse
|
21
|
Moghadam ER, Ang HL, Asnaf SE, Zabolian A, Saleki H, Yavari M, Esmaeili H, Zarrabi A, Ashrafizadeh M, Kumar AP. Broad-Spectrum Preclinical Antitumor Activity of Chrysin: Current Trends and Future Perspectives. Biomolecules 2020; 10:E1374. [PMID: 32992587 PMCID: PMC7600196 DOI: 10.3390/biom10101374] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 02/06/2023] Open
Abstract
Pharmacological profile of phytochemicals has attracted much attention to their use in disease therapy. Since cancer is a major problem for public health with high mortality and morbidity worldwide, experiments have focused on revealing the anti-tumor activity of natural products. Flavonoids comprise a large family of natural products with different categories. Chrysin is a hydroxylated flavonoid belonging to the flavone category. Chrysin has demonstrated great potential in treating different disorders, due to possessing biological and therapeutic activities, such as antioxidant, anti-inflammatory, hepatoprotective, neuroprotective, etc. Over recent years, the anti-tumor activity of chrysin has been investigated, and in the present review, we provide a mechanistic discussion of the inhibitory effect of chrysin on proliferation and invasion of different cancer cells. Molecular pathways, such as Notch1, microRNAs, signal transducer and activator of transcription 3 (STAT3), nuclear factor-kappaB (NF-κB), PI3K/Akt, MAPK, etc., as targets of chrysin are discussed. The efficiency of chrysin in promoting anti-tumor activity of chemotherapeutic agents and suppressing drug resistance is described. Moreover, poor bioavailability, as one of the drawbacks of chrysin, is improved using various nanocarriers, such as micelles, polymeric nanoparticles, etc. This updated review will provide a direction for further studies in evaluating the anti-tumor activity of chrysin.
Collapse
Affiliation(s)
- Ebrahim Rahmani Moghadam
- Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz 7134814336, Iran;
| | - Hui Li Ang
- Cancer Science Institute of Singapore and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore;
| | - Sholeh Etehad Asnaf
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, North Tehran Branch, IslamicAzad University, Tehran 165115331, Iran;
| | - Amirhossein Zabolian
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran; (A.Z.); (H.S.); (H.E.)
| | - Hossein Saleki
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran; (A.Z.); (H.S.); (H.E.)
| | - Mohammad Yavari
- Nursing and Midwifery Department, Islamic Azad University, Tehran Medical Sciences Branch, Tehran 1916893813, Iran;
| | - Hossein Esmaeili
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran; (A.Z.); (H.S.); (H.E.)
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul 34956, Turkey
| | - Milad Ashrafizadeh
- Faculty of Veterinary Medicine, University of Tabriz, Tabriz 5166616471, Iran
| | - Alan Prem Kumar
- Cancer Science Institute of Singapore and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore;
| |
Collapse
|
22
|
Wang H, Wang L, Xie Z, Zhou S, Li Y, Zhou Y, Sun M. Nitric Oxide (NO) and NO Synthases (NOS)-Based Targeted Therapy for Colon Cancer. Cancers (Basel) 2020; 12:E1881. [PMID: 32668616 PMCID: PMC7408898 DOI: 10.3390/cancers12071881] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/10/2020] [Accepted: 07/10/2020] [Indexed: 12/13/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most lethal malignancies worldwide and CRC therapy remains unsatisfactory. In recent decades, nitric oxide (NO)-a free-radical gas-plus its endogenous producer NO synthases (NOS), have attracted considerable attention. NO exerts dual effects (pro- and anti-tumor) in cancers. Endogenous levels of NO promote colon neoplasms, whereas exogenously sustained doses lead to cytotoxic functions. Importantly, NO has been implicated as an essential mediator in many signaling pathways in CRC, such as the Wnt/β-catenin and extracellular-signal-regulated kinase (ERK) pathways, which are closely associated with cancer initiation, metastasis, inflammation, and chemo-/radio-resistance. Therefore, NO/NOS have been proposed as promising targets in the regulation of CRC carcinogenesis. Clinically relevant NO-donating agents have been developed for CRC therapy to deliver a high level of NO to tumor sites. Notably, inducible NOS (iNOS) is ubiquitously over-expressed in inflammatory-associated colon cancer. The development of iNOS inhibitors contributes to targeted therapies for CRC with clinical benefits. In this review, we summarize the multifaceted mechanisms of NO-mediated networks in several hallmarks of CRC. We review the clinical manifestation and limitations of NO donors and NOS inhibitors in clinical trials. We also discuss the possible directions of NO/NOS therapies in the immediate future.
Collapse
Affiliation(s)
- Hao Wang
- College of Laboratory Medicine, Jilin Medical University, Jilin 132013, China;
| | - Liye Wang
- Department of Pharmacological and Pharmaceutical Science, College of Pharmacy, University of Houston, Houston, TX, 77204, USA; (L.W.); (Z.X.); (S.Z.); (Y.L.)
| | - Zuoxu Xie
- Department of Pharmacological and Pharmaceutical Science, College of Pharmacy, University of Houston, Houston, TX, 77204, USA; (L.W.); (Z.X.); (S.Z.); (Y.L.)
| | - Shuang Zhou
- Department of Pharmacological and Pharmaceutical Science, College of Pharmacy, University of Houston, Houston, TX, 77204, USA; (L.W.); (Z.X.); (S.Z.); (Y.L.)
| | - Yan Li
- Department of Pharmacological and Pharmaceutical Science, College of Pharmacy, University of Houston, Houston, TX, 77204, USA; (L.W.); (Z.X.); (S.Z.); (Y.L.)
| | - Yue Zhou
- Department of Statistics, North Dakota University, Fargo, ND 58105, USA;
| | - Meiyan Sun
- College of Laboratory Medicine, Jilin Medical University, Jilin 132013, China;
| |
Collapse
|
23
|
Choi JH, Moon CM, Shin TS, Kim EK, McDowell A, Jo MK, Joo YH, Kim SE, Jung HK, Shim KN, Jung SA, Kim YK. Lactobacillus paracasei-derived extracellular vesicles attenuate the intestinal inflammatory response by augmenting the endoplasmic reticulum stress pathway. Exp Mol Med 2020; 52:423-437. [PMID: 32123288 PMCID: PMC7156483 DOI: 10.1038/s12276-019-0359-3] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/05/2019] [Accepted: 10/13/2019] [Indexed: 12/18/2022] Open
Abstract
Lactobacillus paracasei is a major probiotic and is well known for its anti-inflammatory properties. Thus, we investigated the effects of L. paracasei-derived extracellular vesicles (LpEVs) on LPS-induced inflammation in HT29 human colorectal cancer cells and dextran sulfate sodium (DSS)-induced colitis in C57BL/6 mice. ER stress inhibitors (salubrinal or 4-PBA) or CHOP siRNA were utilized to investigate the relationship between LpEV-induced endoplasmic reticulum (ER) stress and the inhibitory effect of LpEVs against LPS-induced inflammation. DSS (2%) was administered to male C57BL/6 mice to induce inflammatory bowel disease, and disease activity was measured by determining colon length, disease activity index, and survival ratio. In in vitro experiments, LpEVs reduced the expression of the LPS-induced pro-inflammatory cytokines IL-1α, IL-1β, IL-2, and TNFα and increased the expression of the anti-inflammatory cytokines IL-10 and TGFβ. LpEVs reduced LPS-induced inflammation in HT29 cells and decreased the activation of inflammation-associated proteins, such as COX-2, iNOS and NFκB, as well as nitric oxide. In in vivo mouse experiments, the oral administration of LpEVs also protected against DSS-induced colitis by reducing weight loss, maintaining colon length, and decreasing the disease activity index (DAI). In addition, LpEVs induced the expression of endoplasmic reticulum (ER) stress-associated proteins, while the inhibition of these proteins blocked the anti-inflammatory effects of LpEVs in LPS-treated HT29 cells, restoring the pro-inflammatory effects of LPS. This study found that LpEVs attenuate LPS-induced inflammation in the intestine through ER stress activation. Our results suggest that LpEVs have a significant effect in maintaining colorectal homeostasis in inflammation-mediated pathogenesis. Tiny vesicles released by a bacterial species found in the human gut can reduce symptoms of inflammatory bowel disease (IBD) and prevent disease progression. People with IBD have a decreased abundance of Lactobacilli bacteria in their gut, creating an imbalance that perpetuates the disease. Replenishment of this bacteria may become a valuable therapy. Chang Mo Moon at Ewha Womans University, Yoon-Keun Kim at MD Healthcare, both in Seoul, South Korea, and co-workers demonstrated how extracellular vesicles (EVs) released by Lactobacilli paracasei can actively prevent bowel inflammation. These EVs contain a mixture of proteins, nucleic acids and other biomolecules. The team administered EV to cultured human colorectal cancer cells and to mice with induced colitis. The EVs decreased pro-inflammatory protein activity and boosted levels of protective cellular membrane proteins via augmenting ER stress pathway.
Collapse
Affiliation(s)
- Ji Hyun Choi
- Department of Internal Medicine, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Chang Mo Moon
- Department of Internal Medicine, College of Medicine, Ewha Womans University, Seoul, Republic of Korea. .,Tissue Injury Defense Research Center, Ewha Womans University, Seoul, Republic of Korea.
| | | | | | | | - Min-Kyung Jo
- Department of Internal Medicine, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Yang Hee Joo
- Department of Internal Medicine, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Seong-Eun Kim
- Department of Internal Medicine, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Hye-Kyung Jung
- Department of Internal Medicine, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Ki-Nam Shim
- Department of Internal Medicine, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Sung-Ae Jung
- Department of Internal Medicine, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | | |
Collapse
|
24
|
Hulin JA, Gubareva EA, Jarzebska N, Rodionov RN, Mangoni AA, Tommasi S. Inhibition of Dimethylarginine Dimethylaminohydrolase (DDAH) Enzymes as an Emerging Therapeutic Strategy to Target Angiogenesis and Vasculogenic Mimicry in Cancer. Front Oncol 2020; 9:1455. [PMID: 31993367 PMCID: PMC6962312 DOI: 10.3389/fonc.2019.01455] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 12/05/2019] [Indexed: 01/01/2023] Open
Abstract
The small free radical gas nitric oxide (NO) plays a key role in various physiological and pathological processes through enhancement of endothelial cell survival and proliferation. In particular, NO has emerged as a molecule of interest in carcinogenesis and tumor progression due to its crucial role in various cancer-related events including cell invasion, metastasis, and angiogenesis. The dimethylarginine dimethylaminohydrolase (DDAH) family of enzymes metabolize the endogenous nitric oxide synthase (NOS) inhibitors, asymmetric dimethylarginine (ADMA) and monomethyl arginine (L-NMMA), and are thus key for maintaining homeostatic control of NO. Dysregulation of the DDAH/ADMA/NO pathway resulting in increased local NO availability often promotes tumor growth, angiogenesis, and vasculogenic mimicry. Recent literature has demonstrated increased DDAH expression in tumors of different origins and has also suggested a potential ADMA-independent role for DDAH enzymes in addition to their well-studied ADMA-mediated influence on NO. Inhibition of DDAH expression and/or activity in cell culture models and in vivo studies has indicated the potential therapeutic benefit of this pathway through inhibition of both angiogenesis and vasculogenic mimicry, and strategies for manipulating DDAH function in cancer are currently being actively pursued by several research groups. This review will thus provide a timely discussion on the expression, regulation, and function of DDAH enzymes in regard to angiogenesis and vasculogenic mimicry, and will offer insight into the therapeutic potential of DDAH inhibition in cancer based on preclinical studies.
Collapse
Affiliation(s)
- Julie-Ann Hulin
- Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Ekaterina A Gubareva
- N.N. Petrov National Medical Research Center of Oncology, Saint Petersburg, Russia
| | - Natalia Jarzebska
- Division of Angiology, Department of Internal Medicine III, University Center for Vascular Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Department of Anesthesiology and Intensive Care Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Roman N Rodionov
- Division of Angiology, Department of Internal Medicine III, University Center for Vascular Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Arduino A Mangoni
- Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Sara Tommasi
- Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
25
|
Zhong S, Wu B, Wang X, Sun D, Liu D, Jiang S, Ge J, Zhang Y, Liu X, Zhou X, Jin R, Chen Y. Identification of driver genes and key pathways of prolactinoma predicts the therapeutic effect of genipin. Mol Med Rep 2019; 20:2712-2724. [PMID: 31322266 PMCID: PMC6691206 DOI: 10.3892/mmr.2019.10505] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 04/11/2019] [Indexed: 11/06/2022] Open
Abstract
The purpose of the present study was to identify the potential targets and markers for diagnosis, therapy and prognosis in patients with prolactinoma at the molecular level and to determine the therapeutic effects of genipin in prolactinoma. The gene expression profiles of GSE2175, GSE26966 and GSE36314 were obtained from the Gene Expression Omnibus database. The differentially expressed genes (DEGs) were identified after comparing between gene expression profiles of the prolactinoma tissues and normal tissues. Then, Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis and protein‑protein interaction (PPI) network analysis were conducted. In addition, in vitro, scratch assay, colony‑forming assay, Cell Counting Kit 8 (CCK8) assay and flow cytometry were performed to verify the functional effects of genipin. An aggregate of 12,695, 3,847 and 5,310 DEGs were identified from GSE2175, GSE26966 and GSE36314, respectively. The results of GO and KEGG analysis showed that the DEGs significant and important for prolactinoma were mostly involved with 'spindle pole' and 'oocyte meiosis'. A total of 20 genes were selected as hub genes with high degrees after PPI network analysis, including mitogen‑activated protein kinase 1 (MAPK1), MYC, early growth response 1 (EGR1), Bcl2 and calmodulin 1 (CALM1). CCK8 assay, colony‑forming assay and scratch assay were performed to verify the anti‑prolactinoma effect of genipin. The results of flow cytometry showed that apoptosis was increased by genipin. MAPK1, MYC, EGR1, Bcl2 and CALM1 were screened as main hub genes. Genipin upregulated the expression level of EGR1 and p21 (downstream mediator of EGR1) and EGR1, inhibited the proliferation and migration of prolactinoma cells. Genipin is a promising drug for treatment of patients with prolactinoma.
Collapse
Affiliation(s)
- Sheng Zhong
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin 130012, P.R. China
| | - Bo Wu
- Clinical College, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Xinhui Wang
- Clinical College, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Dandan Sun
- Clinical College, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Daqun Liu
- Clinical College, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Shanshan Jiang
- College of Pharmacy, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Junliang Ge
- Clinical College, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Yuan Zhang
- Clinical College, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Xinrui Liu
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin 130012, P.R. China
| | - Xiaoli Zhou
- Department of Cell Biology, Basic Medical College, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Rihua Jin
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin 130012, P.R. China
| | - Yong Chen
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin 130012, P.R. China
| |
Collapse
|
26
|
Liang Y, Zhang C, Dai DQ. Identification of differentially expressed genes regulated by methylation in colon cancer based on bioinformatics analysis. World J Gastroenterol 2019; 25:3392-3407. [PMID: 31341364 PMCID: PMC6639549 DOI: 10.3748/wjg.v25.i26.3392] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/09/2019] [Accepted: 06/01/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND DNA methylation, acknowledged as a key modification in the field of epigenetics, regulates gene expression at the transcriptional level. Aberrant methylation in DNA regulatory regions could upregulate oncogenes and downregulate tumor suppressor genes without changing the sequences. However, studies of methylation in the control of gene expression are still inadequate. In the present research, we performed bioinformatics analysis to clarify the function of methylation and supply candidate methylation-related biomarkers and drivers for colon cancer.
AIM To identify and analyze methylation-regulated differentially expressed genes (MeDEGs) in colon cancer by bioinformatics analysis.
METHODS We downloaded RNA expression profiles, Illumina Human Methylation 450K BeadChip data, and clinical data of colon cancer from The Cancer Genome Atlas project. MeDEGs were identified by analyzing the gene expression and methylation levels using the edgeR and limma package in R software. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed in the DAVID database and KEGG Orthology-Based Annotation System 3.0, respectively. We then conducted Kaplan–Meier survival analysis to explore the relationship between methylation and expression and prognosis. Gene set enrichment analysis (GSEA) and investigation of protein-protein interactions (PPI) were performed to clarify the function of prognosis-related genes.
RESULTS A total of 5 up-regulated and 81 down-regulated genes were identified as MeDEGs. GO and KEGG pathway analyses indicated that MeDEGs were enriched in multiple cancer-related terms. Furthermore, Kaplan–Meier survival analysis showed that the prognosis was negatively associated with the methylation status of glial cell-derived neurotrophic factor (GDNF) and reelin (RELN). In PPI networks, GDNF and RELN interact with neural cell adhesion molecule 1. Besides, GDNF can interact with GDNF family receptor alpha (GFRA1), GFRA2, GFRA3, and RET. RELN can interact with RAFAH1B1, disabled homolog 1, very low-density lipoprotein receptor, lipoprotein receptor-related protein 8, and NMDA 2B. Based on GSEA, hypermethylation of GDNF and RELN were both significantly associated with pathways including “RNA degradation,” “ribosome,” “mismatch repair,” “cell cycle” and “base excision repair.”
CONCLUSION Aberrant DNA methylation plays an important role in colon cancer progression. MeDEGs that are associated with the overall survival of patients may be potential targets in tumor diagnosis and treatment.
Collapse
Affiliation(s)
- Yu Liang
- Department of Gastrointestinal Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, Liaoning Province, China
| | - Cheng Zhang
- Department of Gastrointestinal Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, Liaoning Province, China
| | - Dong-Qiu Dai
- Department of Gastrointestinal Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, Liaoning Province, China
| |
Collapse
|
27
|
Tolone A, Belhadj S, Rentsch A, Schwede F, Paquet-Durand F. The cGMP Pathway and Inherited Photoreceptor Degeneration: Targets, Compounds, and Biomarkers. Genes (Basel) 2019; 10:genes10060453. [PMID: 31207907 PMCID: PMC6627777 DOI: 10.3390/genes10060453] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/05/2019] [Accepted: 06/11/2019] [Indexed: 12/17/2022] Open
Abstract
Photoreceptor physiology and pathophysiology is intricately linked to guanosine-3’,5’-cyclic monophosphate (cGMP)-signaling. Here, we discuss the importance of cGMP-signaling for the pathogenesis of hereditary retinal degeneration. Excessive accumulation of cGMP in photoreceptors is a common denominator in cell death caused by a variety of different gene mutations. The cGMP-dependent cell death pathway may be targeted for the treatment of inherited photoreceptor degeneration, using specifically designed and formulated inhibitory cGMP analogues. Moreover, cGMP-signaling and its down-stream targets may be exploited for the development of novel biomarkers that could facilitate monitoring of disease progression and reveal the response to treatment in future clinical trials. We then briefly present the importance of appropriate formulations for delivery to the retina, both for drug and biomarker applications. Finally, the review touches on important aspects of future clinical translation, highlighting the need for interdisciplinary cooperation of researchers from a diverse range of fields.
Collapse
Affiliation(s)
- Arianna Tolone
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Strasse 5-7, 72076 Tübingen, Germany.
| | - Soumaya Belhadj
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Strasse 5-7, 72076 Tübingen, Germany.
| | | | - Frank Schwede
- Biolog Life Science Institute, 28199 Bremen, Germany.
| | - François Paquet-Durand
- Institute for Ophthalmic Research, University of Tübingen, Elfriede-Aulhorn-Strasse 5-7, 72076 Tübingen, Germany.
| |
Collapse
|
28
|
Lin CC, Chen KB, Tsai CH, Tsai FJ, Huang CY, Tang CH, Yang JS, Hsu YM, Peng SF, Chung JG. Casticin inhibits human prostate cancer DU 145 cell migration and invasion via Ras/Akt/NF-κB signaling pathways. J Food Biochem 2019; 43:e12902. [PMID: 31353708 DOI: 10.1111/jfbc.12902] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 02/13/2019] [Accepted: 05/04/2019] [Indexed: 12/29/2022]
Abstract
Casticin, a polymethoxyflavone derived from natural plants, has biological activities including induction of cell apoptosis. In this study, we showed the beneficial effects of casticin on the inhibition of prostate cancer cell metastasis. Casticin reduced total viable cell number, thus, we selected low doses of casticin for following experiments. Casticin decreased cell mobility, suppressed cell migration and invasion, and reduced cell gelatinolytic activities of MMP-2/-9. Furthermore, casticin inhibited the protein levels of AKT, GSK3 αβ, Snail, and MMPs (MMP-2, -9, -13, and -7) at 24 and 48 hr treatment. Casticin diminished the expressions of NF-κB p65, GRB2, SOS-1, MEK, p-ERK1/2, and p-JNK1/2 at 48 hr treatment only. However, casticin reduced the level of E-cadherin at 24 hr treatment but elevated at 48 hr. The novel findings suggest that casticin may represent a new and promising therapeutic agent for the metastatic prostate cancer. PRACTICAL APPLICATIONS: Casticin derived from natural plants had been used for Chinese medicine in Chinese population for thousands of years. In the present study, casticin attenuated metastatic effects, including decreasing viable cell number, inhibiting the migration, invasion, and adhesion, and reducing matrix metalloproteinases activity on human prostate DU 145 cancer cells. In addition, the results also provided possible pathways involved in casticin anti-metastasis mechanism. We conclude that casticin may be an aptitude anticancer agent or adjuvant for the metastatic prostate cancer in the future.
Collapse
Affiliation(s)
- Chia-Chang Lin
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Kuen-Bao Chen
- Department of Anesthesiology, China Medical University Hospital, Taichung, Taiwan.,School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Chang-Hai Tsai
- China Medical University Children's Hospital, China Medical University, Taichung, Taiwan.,Department of Healthcare Administration, Asia University, Taichung, Taiwan
| | - Fuu-Jen Tsai
- China Medical University Children's Hospital, China Medical University, Taichung, Taiwan.,School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,Graduate Institute of Chinese Medical Science, China Medical University, Taichung, Taiwan.,Department of Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan
| | - Chih-Hsin Tang
- Department of Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan.,Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan.,Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| | - Jai-Sing Yang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Yuan-Man Hsu
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Shu-Fen Peng
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan.,Department of Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan
| |
Collapse
|
29
|
Chen HY, Jiang YW, Kuo CL, Way TD, Chou YC, Chang YS, Chung JG. Chrysin inhibit human melanoma A375.S2 cell migration and invasion via affecting MAPK signaling and NF-κB signaling pathway in vitro. ENVIRONMENTAL TOXICOLOGY 2019; 34:434-442. [PMID: 30578657 DOI: 10.1002/tox.22697] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 11/25/2018] [Accepted: 11/30/2018] [Indexed: 06/09/2023]
Abstract
Numerous evidences have shown that chrysin induced cytotoxic effects via induced cell cycle arrest and induction of cell apoptosis in human cancer cell lines, however, no information showed that chrysin inhibited skin cancer cell migration and invasion. In this study, we investigated anti-metastasis mechanisms of chrysin in human melanoma cancer A375.S2 cells in vitro. Under sub-lethal concentrations of chrysin (0, 5, 10, and 15 μM) which inhibits cell mobility, migration and invasion of A375.S2 cells that were assayed by wound healing and Transwell filter. That chrysin inhibited MMP-2 activity in A375.S2 cells was investigated by gelatin zymography assay. Western blotting was used to examine protein expression and results indicated that chrysin inhibited the expression of GRB2, SOS-1, PKC, p-AKT (Thr308), NF-κBp65, and NF-κBp50 at 24 and 48 hours treatment, but only at 10-15 μM of chrysin decreased Ras, PI3K, p-c-Jun, and Snail only at 48 hours treatment and only decrease p-AKT(Ser473) at 24 hours treatment. Furthermore, chrysin (5-15 μM) decreased the expression of uPA, N-cadherin and MMP-1 at 24 and 48 hours treatment but only decreased MMP-2 and VEGF at 48 hours treatment at 10-15 μM and 5-15 μM of chrysin, respectively, however, increased E-cadherin at 5-15 μM treatment. Results of confocal laser microscopy systems indicated that chrysin inhibited expression of NF-κBp65 in A375.S2 cells. Based on these observations, we suggest that chrysin can be used in anti-metastasis of human melanoma cells in the future.
Collapse
Affiliation(s)
- Hsin-Yu Chen
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan
| | - Yi-Wen Jiang
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan
| | - Chao-Lin Kuo
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan
| | - Tzong-Der Way
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Yu-Cheng Chou
- Department of Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yuan-Shiun Chang
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
30
|
Maryam A, Vedithi SC, Khalid RR, Alsulami AF, Torres PHM, Siddiqi AR, Blundell TL. The Molecular Organization of Human cGMP Specific Phosphodiesterase 6 (PDE6): Structural Implications of Somatic Mutations in Cancer and Retinitis Pigmentosa. Comput Struct Biotechnol J 2019; 17:378-389. [PMID: 30962868 PMCID: PMC6434069 DOI: 10.1016/j.csbj.2019.03.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/24/2019] [Accepted: 03/03/2019] [Indexed: 01/12/2023] Open
Abstract
In the cyclic guanosine monophosphate (cGMP) signaling pathway, phosphodiesterase 6 (PDE6) maintains a critical balance of the intracellular concentration of cGMP by catalyzing it to 5′ guanosine monophosphate (5′-GMP). To gain insight into the mechanistic impacts of the PDE6 somatic mutations that are implicated in cancer and retinitis pigmentosa, we first defined the structure and organization of the human PDE6 heterodimer using computational comparative modelling. Each subunit of PDE6αβ possesses three domains connected through long α-helices. The heterodimer model indicates that the two chains are likely related by a pseudo two-fold axis. The N-terminal region of each subunit is comprised of two allosteric cGMP-binding domains (Gaf-A & Gaf-B), oriented in the same way and interacting with the catalytic domain present at the C-terminal in a way that would allow the allosteric cGMP-binding domains to influence catalytic activity. Subsequently, we applied an integrated knowledge-driven in silico mutation analysis approach to understand the structural and functional implications of experimentally identified mutations that cause various cancers and retinitis pigmentosa, as well as computational saturation mutagenesis of the dimer interface and cGMP-binding residues of both Gaf-A, and the catalytic domains. We studied the impact of mutations on the stability of PDE6αβ structure, subunit-interfaces and Gaf-cGMP interactions. Further, we discussed the changes in interatomic interactions of mutations that are destabilizing in Gaf-A (R93L, V141 M, F162 L), catalytic domain (D600N, F742 L, F776 L) and at the dimer interface (F426A, F248G, F424 N). This study establishes a possible link of change in PDE6αβ structural stability to the experimentally observed disease phenotypes.
Collapse
Affiliation(s)
- Arooma Maryam
- Department of Biosciences, COMSATS University Islamabad (CUI), Park Road, Islamabad, Pakistan.,Department of Biochemistry, University of Cambridge, 80 Tennis Court Rd, Cambridge CB2 1GA, UK
| | | | - Rana Rehan Khalid
- Department of Biosciences, COMSATS University Islamabad (CUI), Park Road, Islamabad, Pakistan
| | - Ali F Alsulami
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Rd, Cambridge CB2 1GA, UK
| | | | - Abdul Rauf Siddiqi
- Department of Biosciences, COMSATS University Islamabad (CUI), Park Road, Islamabad, Pakistan
| | - Tom L Blundell
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Rd, Cambridge CB2 1GA, UK
| |
Collapse
|
31
|
Ianni A, Celenza G, Franceschini N. Oxaprozin: A new hope in the modulation of matrix metalloproteinase 9 activity. Chem Biol Drug Des 2019; 93:811-817. [PMID: 30582279 DOI: 10.1111/cbdd.13468] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 12/01/2018] [Accepted: 12/16/2018] [Indexed: 12/13/2022]
Abstract
Oxaprozin (4,5-diphenyl-2-oxazolepropionic acid) is a non-steroidal, analgesic and antipyretic propionic acid derivative, whose activity in treating inflammatory disorders is well known. The aim of this study was to investigate the ability of oxaprozin to modulate the activity of matrix metalloproteinase 9 (MMP-9), a zinc-dependent endopeptidase involved in a wide range of physiological and pathological events associated with extracellular matrix (ECM) remodelling. The interaction between oxaprozin and MMP-9 was firstly investigated in silico by molecular docking and analysis with LIGPLOT software. Subsequently, the potential inhibitory activity of oxaprozin against MMP-9 and the possible mechanism of the ligand-enzyme interaction were investigated in vitro. Taking into account the in silico findings, MMP-9 can be considered a potential target of oxaprozin, which seems to be able to chelate the catalytic zinc ion through the nitrogen of the oxazole ring and the carboxylate moiety. Moreover, one of the phenyl rings interact with the S1' inhibitor-binding pocket through hydrophobic interaction. Gelatin zymography and enzymatic inhibition assay confirmed the potential role of oxaprozin as a competitive inhibitor of MMP-9. These observations sound particularly interesting if we consider the pathological role of MMP-9, especially evident in inflammatory conditions and cancer. This work may represent a starting point to improve the understanding of the role of oxaprozin, as well as its structural analogues, in modulating the MMP-9 function.
Collapse
Affiliation(s)
- Andrea Ianni
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Giuseppe Celenza
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Nicola Franceschini
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
32
|
Hsiao YT, Fan MJ, Huang AC, Lien JC, Lin JJ, Chen JC, Hsia TC, Wu RSC, Chung JG. Deguelin Impairs Cell Adhesion, Migration and Invasion of Human Lung Cancer Cells through the NF-[Formula: see text]B Signaling Pathways. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2018; 46:209-229. [PMID: 29402127 DOI: 10.1142/s0192415x1850012x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Deguelin, a rotenoid, is isolated from a natural plant species, and has biological activities including antitumor function. In the present study, we investigated the effect of deguelin on the cell adhesion, migration and invasion of NCI-H292 human lung cancer cells in vitro. Cell viability was analyzed by using flow cytometer. Cell adhesion was determined by using the cell-matrix adhesion assay. Wound healing assay was used to examine cell migration. Cell migration and invasion were investigated using a Boyden chamber assay. The protein expression was measured by Western blotting and confocal laser microscopy. The electrophoretic mobility shift assay was used to measure NF-[Formula: see text]B p65 binding to DNA.We selected the concentrations of deguelin at 0, 0.5, 1.0, 1.5, 2.0 and 2.5[Formula: see text][Formula: see text]M and we found that those concentrations of deguelin did not induce significant cytotoxic effects on NCI-H292 cells. Thus, we selected those concentrations of deguelin for metastasis assay. We found that deguelin inhibited cell adhesion, migration and invasion in dose-dependent manners that was assayed by wound healing and transwell methods, respectively. Deguelin decreased the expression of MMP-2/-9, SOS 1, Rho A, p-AKT (Thr308), p-ERK1/2, p-p38, p-JNK, NF-[Formula: see text]B (p65) and uPA in NCI-H292 cells. Deguelin suppressed the expression of PI3K, SOS 1, NF-[Formula: see text]B (p65), but did not significantly affect PKC and Ras in the nuclei of NCI-H292 cells that were confirmed by confocal laser microscopy. We suggest that deguelin may be used as a novel anticancer metastasis of lung cancer in the future.
Collapse
Affiliation(s)
- Yung-Ting Hsiao
- * Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Ming-Jen Fan
- ¶ Department of Biotechnology, Asia University, Taichung, Taiwan
| | - An-Cheng Huang
- ∥ Department of Nursing, St. Mary's Junior College of Medicine, Nursing and Management, Yilan County, Taiwan
| | - Jin-Cherng Lien
- † School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Jen-Jyh Lin
- ** Division of Cardiology, China Medical University Hospital, Taichung, Taiwan
| | - Jaw-Chyun Chen
- §§ Department of Medicinal Botany and Health Applications, Da-Yeh University, Changhua, Taiwan
| | - Te-Chun Hsia
- ‡ Graduate Institute of Chinese Medicine, China Medical University, Taichung, Taiwan.,†† Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Rick Sai-Chuen Wu
- § School of Medicine, China Medical University, Taichung, Taiwan.,‡‡ Department of Anesthesiology, China Medical University Hospital, Taichung, Taiwan
| | - Jing-Gung Chung
- * Department of Biological Science and Technology, China Medical University, Taichung, Taiwan.,¶ Department of Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
33
|
Sun EY, Fleck AKM, Abu-Hakmeh AE, Kotsakis A, Leonard GR, Wan LQ. Cartilage Metabolism is Modulated by Synovial Fluid Through Metalloproteinase Activity. Ann Biomed Eng 2018; 46:810-818. [PMID: 29589167 DOI: 10.1007/s10439-018-2010-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 12/04/2017] [Indexed: 01/03/2023]
Abstract
Synovial fluid (SF) contains various cytokines that regulate chondrocyte metabolism and is dynamically associated with joint disease. The objective of this study was to investigate the effects of diluted normal SF on catabolic metabolism of articular cartilage under inflammatory conditions. For this purpose, SF was isolated from healthy bovine joints, diluted, and added to cartilage explant cultures stimulated with interleukin-1 (IL-1) for 12 days. The kinetic release of sulfated glycosaminoglycan (sGAG) and collagen, as well as nitric oxide and gelatinase matrix metalloproteinases were analyzed in the supernatant. Chondrocyte survival and matrix integrity in the explants were evaluated with Live/Dead and histological staining. Diluted synovial fluid treatment suppressed sGAG and collagen release, downregulated the production of nitric oxide and matrix metalloproteinases, reduced IL-1-induced chondrocyte death, and rescued matrix depletion. Our results demonstrate that normal SF can counteract inflammation-driven cartilage catabolism. This study reports on the protective function of healthy SF and the therapeutic potential of recapitulation of SF for cartilage repair.
Collapse
Affiliation(s)
- Eric Y Sun
- Laboratory for Tissue Engineering and Morphogenesis, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th St, Troy, NY, 12180, USA.,Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th St, Troy, NY, 12180, USA
| | - Allison K M Fleck
- Laboratory for Tissue Engineering and Morphogenesis, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th St, Troy, NY, 12180, USA
| | - Ahmad E Abu-Hakmeh
- Laboratory for Tissue Engineering and Morphogenesis, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th St, Troy, NY, 12180, USA
| | - Alexandra Kotsakis
- Laboratory for Tissue Engineering and Morphogenesis, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th St, Troy, NY, 12180, USA
| | - Garrett R Leonard
- Division of Orthopaedic Surgery, Albany Medical Center, 43 New Scotland Avenue, Albany, NY, 12208, USA
| | - Leo Q Wan
- Laboratory for Tissue Engineering and Morphogenesis, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th St, Troy, NY, 12180, USA. .,Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th St, Troy, NY, 12180, USA. .,Center for Modeling, Simulation and Imaging in Medicine, Rensselaer Polytechnic Institute, 110 8th St, Troy, NY, 12180, USA. .,Laboratory for Tissue Engineering and Morphogenesis, Rensselaer Polytechnic Institute, Biotech 2147, 110 8th Street, Troy, NY, 12180, USA.
| |
Collapse
|
34
|
Booth L, Roberts JL, Poklepovic A, Gordon S, Dent P. PDE5 inhibitors enhance the lethality of pemetrexed through inhibition of multiple chaperone proteins and via the actions of cyclic GMP and nitric oxide. Oncotarget 2018; 8:1449-1468. [PMID: 27903966 PMCID: PMC5352068 DOI: 10.18632/oncotarget.13640] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 11/15/2016] [Indexed: 12/19/2022] Open
Abstract
Phosphodiesterase 5 (PDE5) inhibitors prevent the breakdown of cGMP that results in prolonged protein kinase G activation and the generation of nitric oxide. PDE5 inhibitors enhanced the anti-NSCLC cell effects of the NSCLC therapeutic pemetrexed. [Pemetrexed + sildenafil] activated an eIF2α – ATF4 – CHOP – Beclin1 pathway causing formation of toxic autophagosomes; activated a protective IRE1 – XBP-1 – chaperone induction pathway; and activated a toxic eIF2α – CHOP – DR4 / DR5 / CD95 induction pathway. [Pemetrexed + sildenafil] reduced the expression of c-FLIP-s, MCL-1 and BCL-XL that was blocked in a cell-type -dependent fashion by either over-expression of HSP90 / GRP78 / HSP70 / HSP27 or by blockade of eIF2α-CHOP signaling. Knock down of PKGI/II abolished the ability of sildenafil to enhance pemetrexed toxicity whereas pan-inhibition of NOS using L-NAME or knock down of [iNOS + eNOS] only partially reduced the lethal drug interaction. Pemetrexed reduced the ATPase activities of HSP90 and HSP70 in an ATM-AMPK-dependent fashion that was enhanced by sildenafil signaling via PKGI/II. The drug combination activated an ATM-AMPK-TSC2 pathway that was associated with reduced mTOR S2448 and ULK-1 S757 phosphorylation and increased ULK-1 S317 and ATG13 S318 phosphorylation. These effects were prevented by chaperone over-expression or by expression of an activated form of mTOR that prevented autophagosome formation and reduced cell killing. In two models of NSCLC, sildenafil enhanced the ability of pemetrexed to suppress tumor growth. Collectively we argue that the combination of [pemetrexed + PDE5 inhibitor] should be explored in a new NSCLC phase I trial.
Collapse
Affiliation(s)
- Laurence Booth
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298-0035, USA
| | - Jane L Roberts
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298-0035, USA
| | - Andrew Poklepovic
- Department of Medicine, Virginia Commonwealth University, Richmond, VA 23298-0035, USA
| | - Sarah Gordon
- Department of Medicine, Virginia Commonwealth University, Richmond, VA 23298-0035, USA
| | - Paul Dent
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298-0035, USA
| |
Collapse
|
35
|
Juan TK, Liu KC, Kuo CL, Yang MD, Chu YL, Yang JL, Wu PP, Huang YP, Lai KC, Chung JG. Tetrandrine suppresses adhesion, migration and invasion of human colon cancer SW620 cells via inhibition of nuclear factor-κB, matrix metalloproteinase-2 and matrix metalloproteinase-9 signaling pathways. Oncol Lett 2018; 15:7716-7724. [PMID: 29731901 PMCID: PMC5921181 DOI: 10.3892/ol.2018.8286] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 10/20/2017] [Indexed: 01/02/2023] Open
Abstract
Tetrandrine (TET) exhibits biological activities, including anticancer activity. In Chinese medicine, TET has been used to treat hypertensive and arrhythmic conditions and has been demonstrated to induce cytotoxic effects on human cancer cell lines. However, to the best of the author's knowledge, no previous studies have revealed that TET affects cell metastasis in SW620 human colon cancer cells. The present study demonstrated that TET decreased the cell number and inhibited cell adhesion and mobility of SW620 cells. Furthermore, a wound healing assay was performed to demonstrate that TET suppressed cell movement, and Transwell chamber assays were used to reveal that TET suppressed the cell migration and invasion of SW620 cells. Western blotting demonstrated that TET significantly reduced protein expression levels of SOS Ras/Rac guanine nucleotide exchange factor 1, phosphatidylinositol 3-kinase, growth factor receptor bound protein 2, phosphorylated (p)-c Jun N-terminal kinase 1/2, p-p38, p38, 14-3-3, Rho A, β-catenin, nuclear factor-κB p65, signal transducer and activator of transcription-1 and cyclooxygenase-2, in comparison with untreated SW620 cells. Overall, the results of the present study suggested that TET may be used as a novel anti-metastasis agent for the treatment of human colon cancer in the future.
Collapse
Affiliation(s)
- Ta-Kuo Juan
- Department of Biological Science and Technology, China Medical University, Taichung 404, Taiwan, R.O.C
| | - Kuo-Ching Liu
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404, Taiwan, R.O.C
| | - Chao-Lin Kuo
- Chinese Medicine Resources, China Medical University, Taichung 404, Taiwan, R.O.C
| | - Mei-Due Yang
- Department of Surgery, China Medical University Hospital, Taichung 404, Taiwan, R.O.C
| | - Yung-Lin Chu
- International Master's Degree Program in Food Science, International College, National Pingtung University of Science and Technology, Pingtung 912, Taiwan, R.O.C
| | - Jiun-Long Yang
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404, Taiwan, R.O.C
| | - Ping-Ping Wu
- School of Pharmacy, China Medical University, Taichung 404, Taiwan, R.O.C
| | - Yi-Ping Huang
- Department of Physiology, China Medical University, Taichung 404, Taiwan, R.O.C
| | - Kuang-Chi Lai
- School of Medicine, China Medical University, Taichung 404, Taiwan, R.O.C.,Department of Medical Laboratory Science and Biotechnology, College of Medicine and Life Science, Chung Hwa University of Medical Technology, Tainan 717, Taiwan, R.O.C.,Department of Surgery, China Medical University Beigang Hospital, Beigang, Yunlin 651, Taiwan, R.O.C
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung 404, Taiwan, R.O.C.,Department of Biotechnology, Asia University, Wufeng, Taichung 413, Taiwan, R.O.C
| |
Collapse
|
36
|
Zhao Y, Tang H, Zeng X, Ye D, Liu J. Resveratrol inhibits proliferation, migration and invasion via Akt and ERK1/2 signaling pathways in renal cell carcinoma cells. Biomed Pharmacother 2017; 98:36-44. [PMID: 29241073 DOI: 10.1016/j.biopha.2017.12.029] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 11/24/2017] [Accepted: 12/07/2017] [Indexed: 02/04/2023] Open
Abstract
Recent studies have shown that resveratrol (RES) inhibits cancer cell growth, migration and invasion. Here, we evaluated RES in two human renal cell carcinoma (RCC) cell lines, ACHN and A498. We investigated the effects of RES on proliferation, cell morphology, colony formation, migration, and invasion. We used a proliferation assay to demonstrate that RES inhibited cell growth with IC50 values 132.9±1.064μM in ACHN, and 112.8±1.191μM in A498, respectively. Using inverted contrast microscopy, we showed that RES reduced cell-to-cell contact and inhibited formation of filopodia. A wound healing assay showed that RES inhibited migration of RCC cells. A Transwell assay showed that RES inhibited RCC migration and invasion. Western blot analysis showed that RES suppresses expression of N-cadherin, Vimentin, Snail, MMP-2, MMP-9, p-Akt and p-ERK1/2, but increased expression of E-cadherin and TIMP-1. In the presence of PD98059, the inhibitor of ERK1/2 pathway, we repeated all of the above experiments, showed that RES acted via the ERK1/2 pathway. Taken together, our results suggested that RES suppressed RCC cell proliferation, migration, and invasion in a concentration- and time-dependent manner. These effects likely resulted from inactivation of the Akt and ERK1/2 signaling pathways.
Collapse
Affiliation(s)
- Yuwan Zhao
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Huancheng Tang
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Xin Zeng
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Dongcai Ye
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Jianjun Liu
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| |
Collapse
|
37
|
Ricciuti B, Foglietta J, Bianconi V, Sahebkar A, Pirro M. Enzymes involved in tumor-driven angiogenesis: A valuable target for anticancer therapy. Semin Cancer Biol 2017; 56:87-99. [PMID: 29128510 DOI: 10.1016/j.semcancer.2017.11.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 11/01/2017] [Accepted: 11/04/2017] [Indexed: 02/07/2023]
Abstract
Angiogenesis plays a pivotal role in cancer progression and is required for tissue invasion and metastasis. Starting with Folkman's initial observations in 1971, basic research continued to shed new molecular insight into this multifaceted process, leading to the development of several anti-angiogenic drugs. To date, anti-vascular endothelial growth factor monoclonal antibodies, such as bevacizumab and ramucirumab, and receptor tyrosine kinase inhibitors (e.g., sorafenib, sunitinib, regorafenib and axitinib) have had a profound impact on the way we treat patients with advanced cancer, providing in some cases unprecedented clinical benefit. The molecular mechanisms underlying tumor-driven angiogenesis have been explored extensively and have unveiled a number of potential clinically relevant targets, including several novel enzymes. In this review, we summarized the current strategies to target tumor-driven angiogenesis through the inhibition of relevant and selected classes of enzymes involved in this process.
Collapse
Affiliation(s)
- Biagio Ricciuti
- Department of Medical Oncology, Santa Maria della Misericordia Hospital, Piazzale L. Severi n. 1, 06132, Perugia, Italy
| | - Jennifer Foglietta
- Department of Medical Oncology, Santa Maria della Misericordia Hospital, Piazzale L. Severi n. 1, 06132, Perugia, Italy
| | - Vanessa Bianconi
- Department of Medicine, University of Perugia, Piazzale L. Severi n. 1, 06132, Perugia, Italy
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Matteo Pirro
- Department of Medicine, University of Perugia, Piazzale L. Severi n. 1, 06132, Perugia, Italy.
| |
Collapse
|
38
|
Shih YL, Au MK, Liu KL, Yeh MY, Lee CH, Lee MH, Lu HF, Yang JL, Wu RSC, Chung JG. Ouabain impairs cell migration, and invasion and alters gene expression of human osteosarcoma U-2 OS cells. ENVIRONMENTAL TOXICOLOGY 2017; 32:2400-2413. [PMID: 28795476 DOI: 10.1002/tox.22453] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 07/07/2017] [Accepted: 07/13/2017] [Indexed: 06/07/2023]
Abstract
Ouabain, the specific Na+ /K+ -ATPase blocker, has biological activity including anti-proliferative and anti-metastasis effects in cancer cell. There is no study to show ouabain inhibiting cell migration and invasion in human osteosarcoma U-2 OS cells. Thus, we investigated the effect of ouabain on the cell migration and invasion of human osteosarcoma U-2 OS cells. Results indicated that ouabain significantly decreased the percentage of viable cells at 2.5-5.0 μM, thus, we selected 0.25-1.0 μM for inhibiting studies. Ouabain inhibited cell migration, invasion and the enzymatic activities of MMP-2, and also affected the expression of metastasis-associated protein in U-2 OS cells. The cDNA microarray assay indicated that CDH1, TGFBR3, SHC3 and MAP2K6 metastasis-related genes were increased, but CCND1, JUN, CDKN1A, TGFB1, 2 and 3, SMAD4, MMP13, MMP2 and FN1 genes were decreased. These findings provide more information regarding ouabain inhibited cell migration and invasion and associated gene expressions in U-2 OS cells after exposed to ouabain.
Collapse
Affiliation(s)
- Yung-Luen Shih
- Department of Pathology and Laboratory Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
- School of Medical Laboratory Science and Biotechnology, Taipei Medical University, Taipei, Taiwan
- School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Man-Kuan Au
- Department of Orthopedics, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Ko-Lin Liu
- Department of Pathology and Laboratory Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Ming-Yang Yeh
- Office of Director, Cheng-Hsin General Hospital, Taipei, Taiwan
| | - Ching-Hsiao Lee
- Department of Medical Technology, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli County, Taiwan
| | - Mei-Hui Lee
- Department of Genetic Counseling Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Hsu-Feng Lu
- Restaurant, Hotel and Institutional Management, Fu-Jen Catholic University, New Taipei City, Taiwan
- Department of Clinical Pathology, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Jiun-Long Yang
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan
| | | | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Wufeng, Taichung, Taiwan
| |
Collapse
|
39
|
Cai X, Zhu H, Li Y. PKCζ, MMP‑2 and MMP‑9 expression in lung adenocarcinoma and association with a metastatic phenotype. Mol Med Rep 2017; 16:8301-8306. [PMID: 28983601 DOI: 10.3892/mmr.2017.7634] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 08/01/2017] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate protein kinase C ζ type (PKCζ), matrix metalloproteinase (MMP)‑2 and MMP‑9 expression in lung adenocarcinoma and to define their association with in vitro invasion and metastatic capacity. PKCζ, MMP‑2 and MMP‑9 expression was assessed by immunohistochemistry in 110 cases of lung adenocarcinoma. PKCζ small interfering (si)RNA was transfected into A549 cells, and western blotting was used to confirm PKCζ‑knockdown in transfected cells and to measure MMP‑2 and MMP‑9 levels. A Transwell invasion assay was used to detect in vitro invasive capacity. The rates of positive PKCζ, MMP‑2 and MMP‑9 staining in lung adenocarcinoma tissues were 52.73, 55.45 and 61.82%, respectively. PKCζ expression was increased in malignant tissues compared with adjacent normal lung tissues and was associated with lymph node metastasis (P<0.05), although it was not associated with any other clinicopathological parameters, including sex, age, tumor size, smoking status or distant metastases (all P>0.05). PKCζ, MMP‑2 and MMP‑9 expression was markedly decreased in siPKCζ‑treated A549 cells, which exhibited a significantly decreased invasive capacity in the Transwell invasion assay (P<0.05). In conclusion, PKCζ promoted lung adenocarcinoma invasion and metastasis, and its expression was associated with MMP‑2 and MMP‑9 expression. PKCζ may be a potential target for gene therapy in lung adenocarcinoma.
Collapse
Affiliation(s)
- Xiaoshan Cai
- Department of Pathology, Second People's Hospital of Weifang, Weifang, Shandong 261041, P.R. China
| | - Hongguang Zhu
- Department of Dentistry, Weifang People's Hospital, Weifang, Shandong 261000, P.R. China
| | - Ying Li
- Department of Pathology, Second People's Hospital of Weifang, Weifang, Shandong 261041, P.R. China
| |
Collapse
|
40
|
Shih YL, Chou HM, Chou HC, Lu HF, Chu YL, Shang HS, Chung JG. Casticin impairs cell migration and invasion of mouse melanoma B16F10 cells via PI3K/AKT and NF-κB signaling pathways. ENVIRONMENTAL TOXICOLOGY 2017; 32:2097-2112. [PMID: 28444820 DOI: 10.1002/tox.22417] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Revised: 03/01/2017] [Accepted: 03/05/2017] [Indexed: 06/07/2023]
Abstract
Casticin, a polymethoxyflavone, is one of the major active components obtained from Fructus viticis, which have been shown to have anticancer activities including induce cell apoptosis in human cancer cells. The aim of this study was to investigate the molecular mechanisms by which casticin inhibits cell migration and invasion of mouse melanoma B16F10 cells. Cell viability was examined by MTT assay and the results indicated that casticin decreased the total percentages of viable cells in dose-dependent manners. Casticin affected cell migration and invasion in B16F10 cells were examined by wound healing mobility assay and Boyden chamber migration and invasion assay and results indicated that casticin inhibited cell migration and invasion in dose-dependent manners. Western blotting was used to examine the protein expression of B16F10 cells after exposed to casticin and the results showed that casticin decreased the expressions of MMP-9, MMP-2, MMP-1, FAK, 14-3-3, GRB2, Akt, NF-κB p65, SOS-1, p-EGFR, p-JNK 1/2, uPA, and Rho A in B16F10 cells. Furthermore, cDNA microarray assay was used to show that casticin affected associated gene expression of cell migration and invasion and the results indicated that casticin affected some of the gene expression such as increased SCN1B (cell adhesion molecule 1) and TIMP2 (TIMP metallopeptidase inhibitor 2) and decreased NDUFS4 (NADH dehydrogenase (ubiquinone) Fe-S protein4), VEGFA (vascular endothelial growth factor A), and DDIT3 (DNA-damage-inducible transcript 3) which associated cell migration and invasion in B16F10 cells. Based on those observations, we suggest that casticin could be used as a novel anticancer metastasis of melanoma cancer in the future.
Collapse
Affiliation(s)
- Yung-Luen Shih
- Department of Pathology and Laboratory Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
- School of Medical Laboratory Science and Biotechnology, Taipei Medical University, Taipei, Taiwan
- School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei, Taiwan
| | - Hsiao-Min Chou
- Department of Pathology and Laboratory Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Hsiu-Chen Chou
- Department of Pathology and Laboratory Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Hsu-Feng Lu
- Department of Restaurant, Hotel and Institutional Management, Fu-Jen Catholic University, New Taipei, Taiwan
- Department of Clinical Pathology, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Yung-Lin Chu
- International Master's Degree Program in Food Science, International College, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Hung-Sheng Shang
- Department of Pathology, National Defense Medical Center, Division of Clinical Pathology, Tri-Service General Hospital, Taipei, Taiwan
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Wufeng, Taichung, Taiwan
| |
Collapse
|
41
|
Booth L, Roberts JL, Poklepovic A, Dent P. PDE5 inhibitors enhance the lethality of [pemetrexed + sorafenib]. Oncotarget 2017; 8:13464-13475. [PMID: 28088782 PMCID: PMC5355112 DOI: 10.18632/oncotarget.14562] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 01/02/2017] [Indexed: 01/02/2023] Open
Abstract
The combination of pemetrexed and sorafenib has significant clinical activity against a wide variety of tumor types in patients and the present studies were performed to determine whether sildenafil enhances the killing potential of [pemetrexed + sorafenib]. In multiple genetically diverse lung cancer cell lines, sildenafil enhanced the lethality of [pemetrexed + sorafenib]. The three-drug combination reduced the activities of AKT, mTOR and STAT transcription factors; increased the activities of eIF2α and ULK-1; lowered the expression of MCL-1, BCL-XL, thioredoxin and SOD2; and increased the expression of Beclin1. Enhanced cell killing by sildenafil was blocked by inhibition of death receptor signaling and autophagosome formation. Enforced activation of STAT3 and AKT or inhibition of JNK significantly reduced cell killing. The enhanced cell killing caused by sildenafil was more reliant on increased PKG signaling than on the generation of nitric oxide. In vivo sildenafil enhanced the anti-tumor properties of [pemetrexed + sorafenib]. Based on our data we argue that additional clinical studies combining pemetrexed, sorafenib and sildenafil are warranted.
Collapse
Affiliation(s)
- Laurence Booth
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298-0035, USA
| | - Jane L Roberts
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298-0035, USA
| | - Andrew Poklepovic
- Department of Biochemistry and Medicine, Virginia Commonwealth University, Richmond, VA 23298-0035, USA
| | - Paul Dent
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298-0035, USA
| |
Collapse
|
42
|
Type 5 phosphodiesterase regulates glioblastoma multiforme aggressiveness and clinical outcome. Oncotarget 2017; 8:13223-13239. [PMID: 28099939 PMCID: PMC5355091 DOI: 10.18632/oncotarget.14656] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 12/12/2016] [Indexed: 01/27/2023] Open
Abstract
Expression of type 5 phosphodiesterase (PDE5), a cGMP-specific hydrolytic enzyme, is frequently altered in human cancer, but its specific role in tumorigenesis remains controversial. Herein, by analyzing a cohort of 69 patients affected by glioblastoma multiforme (GBM) who underwent chemo- and radiotherapy after surgical resection of the tumor, we found that PDE5 was strongly expressed in cancer cells in about 50% of the patients. Retrospective analysis indicated that high PDE5 expression in GBM cells significantly correlated with longer overall survival of patients. Furthermore, silencing of endogenous PDE5 by short hairpin lentiviral transduction (sh-PDE5) in the T98G GBM cell line induced activation of an invasive phenotype. Similarly, pharmacological inhibition of PDE5 activity strongly enhanced cell motility and invasiveness in T98G cells. This invasive phenotype was accompanied by increased secretion of metallo-proteinase 2 (MMP-2) and activation of protein kinase G (PKG). Moreover, PDE5 silencing markedly enhanced DNA damage repair and cell survival following irradiation. The enhanced radio-resistance of sh-PDE5 GBM cells was mediated by an increase of poly(ADP-ribosyl)ation (PARylation) of cellular proteins and could be counteracted by poly(ADP-ribose) polymerase (PARP) inhibitors. Conversely, PDE5 overexpression in PDE5-negative U87G cells significantly reduced MMP-2 secretion, inhibited their invasive potential and interfered with DNA damage repair and cell survival following irradiation. These studies identify PDE5 as a favorable prognostic marker for GBM, which negatively affects cell invasiveness and survival to ionizing radiation. Moreover, our work highlights the therapeutic potential of targeting PKG and/or PARP activity in this currently incurable subset of brain cancers.
Collapse
|
43
|
Roberts JL, Poklepovic A, Booth L. Curcumin interacts with sildenafil to kill GI tumor cells via endoplasmic reticulum stress and reactive oxygen/ nitrogen species. Oncotarget 2017; 8:99451-99469. [PMID: 29245915 PMCID: PMC5725106 DOI: 10.18632/oncotarget.19807] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 04/15/2017] [Indexed: 12/18/2022] Open
Abstract
The present studies focused on the ability of the phosphodiesterase 5 (PDE5) inhibitor sildenafil to enhance the anti-cancer properties of clinically relevant concentrations of the dietary diarylheptanoid curcumin. In gastrointestinal tumor cells, sildenafil and curcumin interacted in a greater than additive fashion to kill. Inhibition of the extrinsic apoptotic pathway suppressed killing by ∼50%, as did blockade of the intrinsic apoptotic pathway. Sildenafil and curcumin reduced mTORC1 and mTORC2 activity and increased Beclin1 levels and the numbers of autophagosomes and autolysosomes in cells in a PERK-eIF2α-dependent fashion. Knock down of Beclin1 or ATG5 partially suppressed killing. In contrast, stable knock out of ATG16-L1 unexpectedly enhanced killing, an effect not altered by Beclin1/ATG5 knock down. Curcumin and sildenafil exposure reduced the expression of MCL-1, BCL-XL, thioredoxin and superoxide dismutase 2 (SOD2) in an eIF2α-dependent fashion. Curcumin and sildenafil interacted in a greater than additive fashion to increase the levels of reactive oxygen species; knock down of thioredoxin or SOD2 enhanced killing and over-expression of thioredoxin or SOD2 suppressed killing. In vivo, curcumin and sildenafil interacted to suppress the growth of colon cancer tumors. Multiplex analyses of plasma taken after drug exposure at animal nadir indicated that the levels of M-CSF, CXCL-9, PDGF and G-CSF were significantly increased by [curcumin + sildenafil] and that expression of CXCL1 and CCL5 were significantly reduced. Cells isolated from in vivo treated [curcumin + sildenafil] tumors were resistant to in vitro [curcumin + sildenafil] exposure, a phenotype that was blocked by the colon cancer therapeutic regorafenib.
Collapse
Affiliation(s)
- Jane L Roberts
- Departments of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298-0035, USA
| | - Andrew Poklepovic
- Departments of Biochemistry and Medicine, Virginia Commonwealth University, Richmond, VA 23298-0035, USA
| | - Laurence Booth
- Departments of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298-0035, USA
| |
Collapse
|
44
|
Cao JY, Yin HS, Li HS, Yu XQ, Han X. Interleukin-27 augments the inhibitory effects of sorafenib on bladder cancer cells. ACTA ACUST UNITED AC 2017; 50:e6207. [PMID: 28746469 PMCID: PMC5520222 DOI: 10.1590/1414-431x20176207] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 05/22/2017] [Indexed: 01/08/2023]
Abstract
Both sorafenib and interleukin-27 (IL-27) are antineoplastic drugs. This study aimed to investigate the synergistic effect of these two drugs on bladder cancer cells. HTB-9 and T24 cells were stimulated with IL-27 (50 ng/mL), sorafenib (2 μM) or the synergistic action of these two drugs. The cells without treatment acted as control. Cell proliferation, apoptosis and invasion were measured by bromodeoxyuridine assay, flow cytometry and modified Boyden chamber, respectively. Simultaneously, both modified Boyden chamber and scratch assay were used to assess cell migration. Finally, the phosphorylation levels of key kinases in the Akt/mechanistic target of rapamycin (mTOR)/mitogen-activated protein kinase (MAPK) pathway, and expression levels of matrix metalloproteinase (MMP)-2 and MMP-9 were detected by western blot analysis. Stimulation with IL-27 or sorafenib repressed proliferation, migration and invasion but promoted apoptosis, and the effects were all enhanced by the combination of these two drugs in HTB-9 cells. The effect of the combined treatment on bladder cancer cells was verified in T24 cells. Additionally, the phosphorylation levels of AKT, mTOR and MAPK as well as the expression levels of MMP-2 and MMP-9 were all decreased by a single treatment of IL-27 or sorafenib, and further decreased by the combined treatment of these two drugs. The combination of IL-27 and sorafenib inhibited proliferation, migration and invasion and promoted apoptosis of bladder cancer cells compared with mono-drug treatment. Additionally, the AKT/mTOR/MAPK pathway might be implicated in the functional effects by down-regulations of MMP-2 and MMP-9.
Collapse
Affiliation(s)
- J Y Cao
- Department of Urology, Binzhou Medical University Hospital, Binzhou, China
| | - H S Yin
- Department of Urology, Binzhou Medical University Hospital, Binzhou, China
| | - H S Li
- Department of Gastrointestinal Surgery, Binzhou Medical University Hospital, Binzhou, China
| | - X Q Yu
- Department of Gastrointestinal Surgery, Binzhou Medical University Hospital, Binzhou, China
| | - X Han
- Department of Gastrointestinal Surgery, Binzhou Medical University Hospital, Binzhou, China
| |
Collapse
|
45
|
Gu XD, Xu LL, Zhao H, Gu JZ, Xie XH. Cantharidin suppressed breast cancer MDA-MB-231 cell growth and migration by inhibiting MAPK signaling pathway. ACTA ACUST UNITED AC 2017; 50:e5920. [PMID: 28678918 PMCID: PMC5496155 DOI: 10.1590/1414-431x20175920] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Accepted: 04/17/2017] [Indexed: 01/05/2023]
Abstract
As an active constituent of the beetle Mylabris used in traditional Chinese medicine, cantharidin is a potent and selective inhibitor of protein phosphatase 2A (PP2A) that plays a crucial role in cell cycle progression, apoptosis, and cell fate. The role and possible mechanisms exerted by cantharidin in cell growth and metastasis of breast cancer were investigated in this study. Cantharidin was found to inhibit cell viability and clonogenic potential in a time- and dose-dependent manner. Cell cycle analysis revealed that cell percentage in G2/M phase decreased, whereas cells in S and G1 phases progressively accumulated with the increasing doses of cantharidin treatment. In a xenograft model of breast cancer, cantharidin inhibited tumor growth in a dose-dependent manner. Moreover, high doses of cantharidin treatment inhibited cell migration in wound and healing assay and downregulated protein levels of major matrix metalloproteinases (MMP)-2 and MMP-9. MDA-MB-231 cell migration and invasion were dose-dependently inhibited by cantharidin treatment. Interestingly, the members of the mitogen-activated protein kinase (MAPK) signaling family were less phosphorylated as the cantharidin dose increased. Cantharidin was hypothesized to exert its anticancer effect through the MAPK signaling pathway. The data of this study also highlighted the possibility of using PP2A as a therapeutic target for breast cancer treatment.
Collapse
Affiliation(s)
- X-D Gu
- Department of Breast Surgery, First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - L-L Xu
- First Clinical Medical School, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - H Zhao
- Department of Breast Surgery, First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - J-Z Gu
- Oncology Department, First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - X-H Xie
- Department of Breast Surgery, First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
46
|
de Oliveira GA, Cheng RYS, Ridnour LA, Basudhar D, Somasundaram V, McVicar DW, Monteiro HP, Wink DA. Inducible Nitric Oxide Synthase in the Carcinogenesis of Gastrointestinal Cancers. Antioxid Redox Signal 2017; 26:1059-1077. [PMID: 27494631 PMCID: PMC5488308 DOI: 10.1089/ars.2016.6850] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
SIGNIFICANCE Gastrointestinal (GI) cancer taken together constitutes one of the most common cancers worldwide with a broad range of etiological mechanisms. In this review, we have examined the impact of nitric oxide (NO) on the etiology of colon, colorectal, gastric, esophageal, and liver cancers. Recent Advances: Despite differences in etiology, initiation, and progression, chronic inflammation has been shown to be a common element within these cancers showing interactions of numerous pathways. NO generated at the inflammatory site contributes to the initiation and progression of disease. The amount of NO generated, time, and site vary and are an important determinant of the biological effects initiated. Among the nitric oxide synthase enzymes, the inducible isoform has the most diverse range, participating in numerous carcinogenic processes. There is emerging evidence showing that inducible nitric oxide synthase (NOS2) plays a central role in the process of tumor initiation and/or development. CRITICAL ISSUES Redox inflammation through NOS2 and cyclooxygenase-2 participates in driving the mechanisms of initiation and progression in GI cancers. FUTURE DIRECTIONS Understanding the underlying mechanism involved in NOS2 activation can provide new insights into important prevention and treatment strategies. Antioxid. Redox Signal. 26, 1059-1077.
Collapse
Affiliation(s)
- Graciele Almeida de Oliveira
- 1 Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Frederick, Maryland
| | - Robert Y S Cheng
- 1 Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Frederick, Maryland
| | - Lisa A Ridnour
- 1 Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Frederick, Maryland
| | - Debashree Basudhar
- 1 Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Frederick, Maryland
| | - Veena Somasundaram
- 1 Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Frederick, Maryland
| | - Daniel W McVicar
- 1 Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Frederick, Maryland
| | - Hugo Pequeno Monteiro
- 2 Laboratório de Sinalização Celular, Universidade Federal de São Paulo , São Paulo, Brazil
| | - David A Wink
- 1 Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Frederick, Maryland
| |
Collapse
|
47
|
Ikeda M, Ishima Y, Chuang VTG, Ikeda T, Kinoshita R, Watanabe H, Ishida T, Otagiri M, Maruyama T. Apoptosis induction of poly-S-nitrosated human serum albumin in resistant solid tumor under hypoxia can be restored by phosphodiesterase 5 inhibition. Nitric Oxide 2017; 69:28-34. [PMID: 28414103 DOI: 10.1016/j.niox.2017.04.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 03/15/2017] [Accepted: 04/09/2017] [Indexed: 12/14/2022]
Abstract
Poly-S-nitrosated human serum albumin (Poly-SNO-HSA) delivered and accumulated nitric oxide (NO) in tumors and induces apoptosis. Tumor hypoxia is strongly associated with malignant progression and tumor resistance to therapy. In this study, we examined the cytotoxic effect of Poly-SNO-HSA under hypoxia on the murine colon 26 adenocarcinoma (C26) cells in vitro and in vivo. Under hypoxia, at about 4 times LD50 dose of Poly-SNO-HSA in vitro, the reactive oxygen species production was hindered but apoptotic cells were induced via cGMP pathway as the effect was suppressed by a soluble guanylate cyclase inhibitor, NS2028. The apoptosis induction effect of low dose Poly-SNO-HSA on C26 cells in vitro under hypoxia can be restored by a phosphodiesterase 5 (PDE5) inhibitor, vardenafil. In C26-bearing mice, Poly-SNO-HSA/vardenafil combination treatment significantly suppressed the tumor volume compared with Poly-SNO-HSA or vardenafil treatment alone. Furthermore, the core tumor tissues showed increased expression of caspase-3 than the non-core tissue. The expression of caspase-3 appeared to overlap with the hypoxic zone of tumor tissues. Similar results were also obtained when the experiments were repeated using Epimedium extract, a natural herbal supplement with PDE5 inhibition activity. In conclusion, Poly-SNO-HSA/PDE5 inhibitors combination therapy is a promising approach for enhancing the anticancer therapeutic effects of Poly-SNO-HSA against not only anti-cancer drug resistance but also hypoxic stress related solid tumor resistance.
Collapse
Affiliation(s)
- Mayumi Ikeda
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Kumamoto 862-0973, Japan
| | - Yu Ishima
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| | - Victor T G Chuang
- School of Pharmacy, Faculty of Health Sciences, Curtin University, GPO Box U1987, Perth 6845, Western Australia, Australia
| | - Tsuyoshi Ikeda
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan
| | - Ryo Kinoshita
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Kumamoto 862-0973, Japan
| | - Hiroshi Watanabe
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Kumamoto 862-0973, Japan
| | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| | - Masaki Otagiri
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan; DDS Research Institute, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan.
| | - Toru Maruyama
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Kumamoto 862-0973, Japan.
| |
Collapse
|
48
|
Chai Y, Xu J, Yan B. The anti-metastatic effect of baicalein on colorectal cancer. Oncol Rep 2017; 37:2317-2323. [DOI: 10.3892/or.2017.5437] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 12/14/2016] [Indexed: 11/06/2022] Open
|
49
|
Lee K, Lindsey AS, Li N, Gary B, Andrews J, Keeton AB, Piazza GA. β-catenin nuclear translocation in colorectal cancer cells is suppressed by PDE10A inhibition, cGMP elevation, and activation of PKG. Oncotarget 2017; 7:5353-65. [PMID: 26713600 PMCID: PMC4868691 DOI: 10.18632/oncotarget.6705] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 12/07/2015] [Indexed: 12/21/2022] Open
Abstract
Phosphodiesterase 10A (PDE10) is a cGMP and cAMP degrading PDE isozyme that is highly expressed in the brain striatum where it appears to play an important role in cognition and psychomotor activity. PDE10 inhibitors are being developed for the treatment of schizophrenia and Huntington's disease and are generally well tolerated, possibly because of low expression levels in most peripheral tissues. We recently reported high levels of PDE10 in colon tumors and that genetic silencing of PDE10 by siRNA or inhibition with small molecule inhibitors can suppress colon tumor cell growth with a high degree of selectivity over normal colonocytes (Li et al., Oncogene 2015). These observations suggest PDE10 may have an unrecognized role in tumorigenesis. Here we report that the concentration range by which the highly specific PDE10 inhibitor, Pf-2545920 (MP-10), inhibits colon tumor cell growth parallels the concentration range required to increase cGMP and cAMP levels, and activates PKG and PKA, respectively. Moreover, PDE10 knockdown by shRNA reduces the sensitivity of colon tumor cells to the growth inhibitory activity of Pf-2545920. Pf-2545920 also inhibits the translocation of β-catenin to the nucleus, thereby reducing β-catenin mediated transcription of survivin, resulting in caspase activation and apoptosis. PDE10 mRNA was also found to be elevated in colon tumors compared with normal tissues. These findings suggest that PDE10 can be targeted for cancer therapy or prevention whereby inhibition results in cGMP elevation and PKG activation to reduce β-catenin-mediated transcription of survival proteins leading to the selective apoptosis of cancer cells.
Collapse
Affiliation(s)
- Kevin Lee
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Ashley S Lindsey
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Nan Li
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Bernard Gary
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Joel Andrews
- Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Adam B Keeton
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Gary A Piazza
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| |
Collapse
|
50
|
Lee K, A Piazza G. The interaction between the Wnt/β-catenin signaling cascade and PKG activation in cancer. J Biomed Res 2016; 31:189-196. [PMID: 28808213 PMCID: PMC5460607 DOI: 10.7555/jbr.31.20160133] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The activation of the Wnt/β-catenin signaling cascade has been well studied and documented in colorectal cancer (CRC). The long-term use of non-steroidal anti-inflammatory drugs (NSAIDs) has been shown to reduce the incidence and risk of death from CRC in numerous epidemiological studies. The NSAID sulindac has also been reported to cause regression of precancerous adenomas in individuals with familial adenomatous polyposis who are at high risk of developing CRC. The mechanism responsible for cancer chemopreventive activity of NSAIDs is not well understood but may be unrelated to their cyclooxygenase inhibitory activity. Emerging evidence suggests that sulindac inhibits the growth of colon tumor cells by suppressing the activity of certain phosphodiesterase isozymes to activate cGMP-dependent protein kinase, PKG, through the elevation of the second messenger cyclic guanosine monophosphote, cGMP. PKG activation has been shown to inhibit the nuclear translocation of β-catenin, reduce β-catenin mRNA and protein levels, and suppress the transcriptional activity of β-catenin. This review describes the relationship between the Wnt/β-catenin signaling cascade and the activation of PKG through PDE inhibition and elevation of intracellular cGMP levels.
Collapse
Affiliation(s)
- Kevin Lee
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604-1405, USA
| | - Gary A Piazza
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604-1405, USA
| |
Collapse
|