1
|
Pukkanasut P, Jaskula-Sztul R, Gomora JC, Velu SE. Therapeutic targeting of voltage-gated sodium channel Na V1.7 for cancer metastasis. Front Pharmacol 2024; 15:1416705. [PMID: 39045054 PMCID: PMC11263763 DOI: 10.3389/fphar.2024.1416705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 06/12/2024] [Indexed: 07/25/2024] Open
Abstract
This review focuses on the expression and function of voltage-gated sodium channel subtype NaV1.7 in various cancers and explores its impact on the metastasis driving cell functions such as proliferation, migration, and invasiveness. An overview of its structural characteristics, drug binding sites, inhibitors and their likely mechanisms of action are presented. Despite the lack of clarity on the precise mechanism by which NaV1.7 contributes to cancer progression and metastasis; many studies have suggested a connection between NaV1.7 and proteins involved in multiple signaling pathways such as PKA and EGF/EGFR-ERK1/2. Moreover, the functional activity of NaV1.7 appears to elevate the expression levels of MACC1 and NHE-1, which are controlled by p38 MAPK activity, HGF/c-MET signaling and c-Jun activity. This cascade potentially enhances the secretion of extracellular matrix proteases, such as MMPs which play critical roles in cell migration and invasion activities. Furthermore, the NaV1.7 activity may indirectly upregulate Rho GTPases Rac activity, which is critical for cytoskeleton reorganization, cell adhesion, and actin polymerization. The relationship between NaV1.7 and cancer progression has prompted researchers to investigate the therapeutic potential of targeting NaV1.7 using inhibitors. The positive outcome of such studies resulted in the discovery of several inhibitors with the ability to reduce cancer cell migration, invasion, and tumor growth underscoring the significance of NaV1.7 as a promising pharmacological target for attenuating cancer cell proliferation and metastasis. The research findings summarized in this review suggest that the regulation of NaV1.7 expression and function by small molecules and/or by genetic engineering is a viable approach to discover novel therapeutics for the prevention and treatment of metastasis of cancers with elevated NaV1.7 expression.
Collapse
Affiliation(s)
- Piyasuda Pukkanasut
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Renata Jaskula-Sztul
- Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL, United States
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Juan Carlos Gomora
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Sadanandan E. Velu
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL, United States
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
2
|
Kim B, Yu JE, Yeo IJ, Son DJ, Lee HP, Roh YS, Lim KH, Yun J, Park H, Han SB, Hong JT. (E)-2-methoxy-4-(3-(4-methoxyphenyl)prop-1-en-1-yl)phenol alleviates inflammatory responses in LPS-induced mice liver sepsis through inhibition of STAT3 phosphorylation. Int Immunopharmacol 2023; 125:111124. [PMID: 37977740 DOI: 10.1016/j.intimp.2023.111124] [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: 07/07/2023] [Revised: 10/13/2023] [Accepted: 10/20/2023] [Indexed: 11/19/2023]
Abstract
Sepsis is a life-threatening disease with limited treatment options, and the inflammatory process represents an important factor affecting its progression. Many studies have demonstrated the critical roles of signal transducer and activator of transcription 3 (STAT3) in sepsis pathophysiology and pro-inflammatory responses. Inhibition of STAT3 activity may therefore represent a promising treatment option for sepsis. We here used a mouse model to demonstrate that (E)-2-methoxy-4-(3-(4-methoxyphenyl)prop-1-en-1-yl)phenol (MMPP) treatment prevented the liver sepsis-related mortality induced by 30 mg/kg lipopolysaccharide (LPS) treatment and reduced LPS-induced increase in alanine transaminase, aspartate transaminase, and lactate dehydrogenase levels, all of which are markers of liver sepsis progression. These recovery effects were associated with decreased LPS-induced STAT3, p65, and JAK1 phosphorylation and proinflammatory cytokine (interleukin 1 beta, interleukin 6, and tumor necrosis factor alpha) level; expression of cyclooxygenase-2 and induced nitric oxide synthase were also reduced by MMPP. In an in vitro study using the normal liver cell line THLE-2, MMPP treatment prevented the LPS-induced increase of STAT3, p65, and JAK1 phosphorylation and inflammatory protein expression in a dose-dependent manner, and this effect was enhanced by combination treatment with MMPP and STAT3 inhibitor. The results clearly indicate that MMPP treatment prevents LPS-induced mortality by inhibiting the inflammatory response via STAT3 activity inhibition. Thus, MMPP represents a novel agent for alleviating LPS-induced liver sepsis.
Collapse
Affiliation(s)
- Boyoung Kim
- College of Pharmacy & Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-21, Osong-eup, Heungduk-gu, Cheongju, Chungbuk 28160, Republic of Korea.
| | - Ji Eun Yu
- College of Pharmacy & Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-21, Osong-eup, Heungduk-gu, Cheongju, Chungbuk 28160, Republic of Korea.
| | - In Jun Yeo
- College of Pharmacy & Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-21, Osong-eup, Heungduk-gu, Cheongju, Chungbuk 28160, Republic of Korea.
| | - Dong Ju Son
- College of Pharmacy & Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-21, Osong-eup, Heungduk-gu, Cheongju, Chungbuk 28160, Republic of Korea.
| | - Hee Pom Lee
- College of Pharmacy & Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-21, Osong-eup, Heungduk-gu, Cheongju, Chungbuk 28160, Republic of Korea.
| | - Yoon Seok Roh
- College of Pharmacy & Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-21, Osong-eup, Heungduk-gu, Cheongju, Chungbuk 28160, Republic of Korea.
| | - Key-Hwan Lim
- College of Pharmacy & Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-21, Osong-eup, Heungduk-gu, Cheongju, Chungbuk 28160, Republic of Korea.
| | - Jaesuk Yun
- College of Pharmacy & Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-21, Osong-eup, Heungduk-gu, Cheongju, Chungbuk 28160, Republic of Korea.
| | - Hanseul Park
- College of Pharmacy & Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-21, Osong-eup, Heungduk-gu, Cheongju, Chungbuk 28160, Republic of Korea.
| | - Sang Bae Han
- College of Pharmacy & Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-21, Osong-eup, Heungduk-gu, Cheongju, Chungbuk 28160, Republic of Korea.
| | - Jin Tae Hong
- College of Pharmacy & Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-21, Osong-eup, Heungduk-gu, Cheongju, Chungbuk 28160, Republic of Korea.
| |
Collapse
|
3
|
Li X, Zhou Y, Wen P, Yuan Y, Xiao Z, Shi H, Zhou H. Tumor suppressor LHX6 upregulation contributes to the inhibitory effect of miR-346 knockdown on colorectal cancer cell growth. ENVIRONMENTAL TOXICOLOGY 2022; 37:435-445. [PMID: 34773443 DOI: 10.1002/tox.23410] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 10/04/2021] [Accepted: 11/02/2021] [Indexed: 06/13/2023]
Abstract
Colorectal cancer (CRC) is one of the prevalent types of human malignancies and ranks as the second leading cause of cancer-associated death worldwide. Dysregulated miRNAs have been promulgated as oncogenes or tumor-suppressive genes participating in the initiation and progression of CRC. A recent study reported that miR-346 was highly expressed in CRC patients. However, the biological role and underlying mechanism of miR-346 in CRC remain elusive. qRT-PCR and western blot assays were employed to detect miR-346 and LIM homeobox domain 6 (LHX6) expression in CRC cells. Cell proliferation was evaluated by CCK-8 and BrdU assays. Apoptosis was evaluated by TUNEL assay. The interaction between miR-346 and LHX6 was assessed by luciferase reporter assay. Results showed that miR-346 expression was increased and LHX6 expression was reduced in CRC cells. miR-346 knockdown and LHX6 overexpression inhibited proliferation and promoted apoptosis of CRC cells. Additionally, we found that miR-346 negatively regulated LHX6 expression in CRC cells by directly targeting LHX6. LHX6 knockdown partially attenuated anti-miR-346-induced proliferation reduction and apoptosis promotion in CRC cells. Furthermore, miR-346 knockdown inhibited the protein kinase B (Akt)/mechanistic target of rapamycin (mTOR) pathway in CRC cells by targeting LHX6. The present study indicated that miR-346 knockdown repressed cell growth in CRC cells by upregulating LHX6, and this was associated with inactivation of the Akt/mTOR pathway.
Collapse
Affiliation(s)
- Xianzhe Li
- Department of General Surgery, Nanshi Hospital, Nanyang, China
| | - Yeqi Zhou
- Department of Radiotherapy, The Second People's Hospital of Huai'an, Huai'an Hospital Affiliated to Xuzhou Medical University, Huai'an, China
| | - Penghao Wen
- Department of Medical Oncology, Nanshi Hospital, Nanyang, China
| | - Yan Yuan
- Department of Radiotherapy, Nanshi Hospital, Nanyang, China
| | - Zhenghong Xiao
- Department of Medical Oncology, Nanshi Hospital, Nanyang, China
| | - Hengwei Shi
- Department of General Surgery, Nanshi Hospital, Nanyang, China
| | - Hailang Zhou
- Department of Gastroenterology, Lianshui County People's Hospital Affiliated to Kangda College of Nanjing Medical University, Huai'an, China
| |
Collapse
|
4
|
Zheng J, Park MH, Lee HP, Hyun BK, Chun HO, Jung SH, Seo HO, Ham YW, Han SB, Hong JT. A small molecule, (E)-2-methoxy-4-(3-(4-methoxyphenyl) prop-1-en-1-yl) phenol suppresses tumor growth via inhibition of IkappaB kinase β in colorectal cancer in vivo and in vitro. Oncotarget 2017; 8:91258-91269. [PMID: 29207641 PMCID: PMC5710921 DOI: 10.18632/oncotarget.20440] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 07/26/2017] [Indexed: 11/25/2022] Open
Abstract
Here we report that a novel synthesized compound (E)-2-methoxy-4-(3-(4-methoxyphenyl)prop-1-en-1-yl)phenol (MMPP) which exhibits better stability, drug-likeness and anti-cancer effect than (E)-2,4-bis(p-hydroxyphenyl)-2-butenal (BHPB) that we previously reported. Of all newly synthesized BHPB analogues, MMPP showed the most significant inhibitory effect on colon cancer cell growth. Thus, we evaluated the anti-cancer effects and possible mechanisms of MMPP in vitro and in vivo. MMPP treatment (0-15 μg/mL) induced apoptotic cell death and enhanced the expression of cleaved caspase-3 and cleaved caspase-8 in a concentration dependent manner. Notably, the expression of death receptor (DR)5 and DR6 was significantly increased by MMPP treatment. Moreover, DR5 siRNA or DR6 siRNA transfection partially abolished MMPP-induced cell growth inhibition. Pull down assay and docking experiment showed that MMPP bound directly to IkappaB kinase β (IKKβ). It was noteworthy that IKKβ mutant (C99S) partially abolished MMPP-induced cell growth inhibition and enhanced expression of DR5 and DR6. In addition, MMPP enhanced TRAIL-induced apoptosis, cell growth inhibition and expression of DRs. In xenograft mice model, MMPP (2.5-5 mg/kg) suppressed tumor growth in a dose dependent manner. Immunohistochemistry analysis showed that the expression levels of DR5 and DR6 and active caspase-3 were increased while the expression levels of PCNA and p-IKKβ were decreased in a dose dependent manner. Thus, MMPP may be a promising anti-cancer agent in colon cancer treatment.
Collapse
Affiliation(s)
- Jie Zheng
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk 28160, Republic of Korea.,Current address: Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Mi Hee Park
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk 28160, Republic of Korea
| | - Hee Pom Lee
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk 28160, Republic of Korea
| | - Byung Kook Hyun
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk 28160, Republic of Korea
| | - Hyung Ok Chun
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk 28160, Republic of Korea
| | - Sung Hee Jung
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk 28160, Republic of Korea
| | - Hyun Ok Seo
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk 28160, Republic of Korea
| | - Young Wan Ham
- Department of Chemistry, Utah Valley University 800 W, University Pkwy, Orem, UT 84058, USA
| | - Sang-Bae Han
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk 28160, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk 28160, Republic of Korea
| |
Collapse
|
5
|
Son DJ, Zheng J, Jung YY, Hwang CJ, Lee HP, Woo JR, Baek SY, Ham YW, Kang MW, Shong M, Kweon GR, Song MJ, Jung JK, Han SB, Kim BY, Yoon DY, Choi BY, Hong JT. MMPP Attenuates Non-Small Cell Lung Cancer Growth by Inhibiting the STAT3 DNA-Binding Activity via Direct Binding to the STAT3 DNA-Binding Domain. Am J Cancer Res 2017; 7:4632-4642. [PMID: 29158850 PMCID: PMC5695154 DOI: 10.7150/thno.18630] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 02/13/2017] [Indexed: 12/12/2022] Open
Abstract
Rationale: Signal transducer and activator of transcription-3 (STAT3) plays a pivotal role in cancer biology. Many small-molecule inhibitors that target STAT3 have been developed as potential anticancer drugs. While designing small-molecule inhibitors that target the SH2 domain of STAT3 remains the leading focus for drug discovery, there has been a growing interest in targeting the DNA-binding domain (DBD) of the protein. Methods: We demonstrated the potential antitumor activity of a novel, small-molecule (E)-2-methoxy-4-(3-(4-methoxyphenyl)prop-1-en-1-yl)phenol (MMPP) that directly binds to the DBD of STAT3, in patient-derived non-small cell lung cancer (NSCLC) xenograft model as well as in NCI-H460 cell xenograft model in nude mice. Results: MMPP effectively inhibited the phosphorylation of STAT3 and its DNA binding activity in vitro and in vivo. It induced G1-phase cell cycle arrest and apoptosis through the regulation of cell cycle- and apoptosis-regulating genes by directly binding to the hydroxyl residue of threonine 456 in the DBD of STAT3. Furthermore, MMPP showed a similar or better antitumor activity than that of docetaxel or cisplatin. Conclusion: MMPP is suggested to be a potential candidate for further development as an anticancer drug that targets the DBD of STAT3.
Collapse
|
6
|
Zheng J, Son DJ, Lee HL, Lee HP, Kim TH, Joo JH, Ham YW, Kim WJ, Jung JK, Han SB, Hong JT. (E)-2-methoxy-4-(3-(4-methoxyphenyl)prop-1-en-1-yl)phenol suppresses ovarian cancer cell growth via inhibition of ERK and STAT3. Mol Carcinog 2017; 56:2003-2013. [PMID: 28277616 DOI: 10.1002/mc.22648] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 02/01/2017] [Accepted: 03/03/2017] [Indexed: 12/14/2022]
Abstract
In the present study, we synthesized several non-aldehyde analogues of (E)-2,4-bis(p-hydroxyphenyl)-2-butenal which showed anti-cancer effect. Interestingly, among the 16 compounds, we found that (E)-2-methoxy-4-(3-(4-methoxyphenyl)prop-1-en-1-yl)phenol (MMPP) showed the most significant anti-proliferative effect on PA-1 and SK-OV-3 ovarian epithelial cancer cells. MMPP treatment (0-15 µg/mL) induced apoptotic cell death, enhanced the expression of cleaved caspase-3, and cleaved caspase-9 in a concentration dependent manner. Notably, DNA binding activity of STAT3, phosphorylation of extracellular signal-regulated kinase (ERK) and p38 was significantly decreased by MMPP treatment. However, ERK siRNA augmented MMPP-induced inhibitory effect on cell growth rather than p38 siRNA or JNK siRNA. Moreover, combination treatment of MMPP with ERK inhibitor U0126 (10 µM) augmented MMPP-induced inhibitory effect on cell growth and DNA binding activity of STAT3, and enhanced expression of cleaved caspase-3 and cleaved caspase-9. In addition, STAT3 siRNA transfection augmented MMPP-induced cell growth inhibition. In PA-1 bearing xenograft mice model, MMPP (5 mg/kg) suppressed tumor growth significantly. Immunohistochemistry staining showed that the expression levels of p-ERK, PCNA, p-STAT3 were decreased while the expression level of caspase-3 was increased by MMPP treatment. Thus, MMPP may be a promising anti-cancer agent in ovarian epithelial cancer treatment.
Collapse
Affiliation(s)
- Jie Zheng
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Dong Ju Son
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Hye Lim Lee
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Hee Pom Lee
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Tae Hoon Kim
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Jung Heun Joo
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Young Wan Ham
- Department of Chemistry, Utah Valley University 800 W, University Pkwy, Orem, Utah
| | - Wun Jae Kim
- College of Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Jae Kyung Jung
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| |
Collapse
|
7
|
Li M, Zhang M, Zhang ZL, Liu N, Han XY, Liu QC, Deng WJ, Liao CX. Induction of Apoptosis by Berberine in Hepatocellular Carcinoma HepG2 Cells via Downregulation of NF-κB. Oncol Res 2017; 25:233-239. [PMID: 28277195 PMCID: PMC7840840 DOI: 10.3727/096504016x14742891049073] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is highly resistant to traditional chemotherapeutic approaches, which causes difficulty in the development of effective drugs for the treatment of HCC. Berberine, a major ingredient of Rhizoma coptidis, is a natural alkaloid used in traditional Chinese medicine. Berberine exhibits potent antitumor activity against HCC due to its high efficiency and low toxicity. In the present study, we found that berberine sensitized HepG cells to NF-κB-mediated apoptosis. Berberine exhibited a significant antiproliferation effect on the HepG2 cells and promoted apoptosis. Both qRT-PCR and immunofluorescence staining revealed that berberine reduced the NF-κB p65 levels in HepG2 cells. Moreover, p65 overexpression rescued berberine-induced cell proliferation and prevented HepG2 cells from undergoing apoptosis. These results suggest that berberine inhibits the growth of HepG2 cells by promoting apoptosis through the NF-κB p65 pathway.
Collapse
Affiliation(s)
- Min Li
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Son DJ, Kim DH, Nah SS, Park MH, Lee HP, Han SB, Venkatareddy U, Gann B, Rodriguez K, Burt SR, Ham YW, Jung YY, Hong JT. Novel synthetic (E)-2-methoxy-4-(3-(4-methoxyphenyl) prop-1-en-1-yl) phenol inhibits arthritis by targeting signal transducer and activator of transcription 3. Sci Rep 2016; 6:36852. [PMID: 27845373 PMCID: PMC5109275 DOI: 10.1038/srep36852] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 10/21/2016] [Indexed: 01/23/2023] Open
Abstract
Rheumatoid arthritis (RA) is a severely debilitating chronic autoimmune disease that leads to long-term joint damage. Signal transducer and activator of transcription 3 (STAT3)-targeted small molecules have shown promise as therapeutic drugs for treating RA. We previously identified (E)-2,4-bis(p-hydroxyphenyl)-2-butenal (BHPB), a tyrosine-fructose Maillard reaction product, as a small molecule with potent anti-inflammatory and anti-arthritic properties, mediated through the inhibition of STAT3 activation. The aim of this study was to develop a novel BHPH derivative with improved anti-arthritic properties and drug-likeness. We designed and synthesised (E)-2-methoxy-4-(3-(4-methoxyphenyl) prop-1-en-1-yl) phenol (MMPP), a novel synthetic BHPB analogue, and investigated its anti-inflammatory and anti-arthritic activities in experimentally-induced RA. We showed that MMPP strongly inhibited pro-inflammatory responses by inhibiting in vitro STAT3 activation and its downstream signalling in murine macrophages and human synoviocytes from patients with RA. Furthermore, we demonstrated that MMPP exhibited potent anti-arthritic activity in a collagen antibody-induced arthritis (CAIA) mouse model in vivo. Collectively, our results suggest that MMPP has great potential for use in the treatment of RA.
Collapse
Affiliation(s)
- Dong Ju Son
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk 28160, Korea
| | - Dae Hwan Kim
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk 28160, Korea
| | - Seong-Su Nah
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, Soonchunhyang University, Asan, Chungnam 31538, Korea
| | - Mi Hee Park
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk 28160, Korea
| | - Hee Pom Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk 28160, Korea
| | - Sang Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk 28160, Korea
| | - Udumula Venkatareddy
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84604, USA
| | - Benjamin Gann
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84604, USA
| | - Kevin Rodriguez
- Department of Chemistry, Utah Valley University, 800 W University Pkwy, Orem, UT 84058, USA
| | - Scott R. Burt
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84604, USA
| | - Young Wan Ham
- Department of Chemistry, Utah Valley University, 800 W University Pkwy, Orem, UT 84058, USA
| | - Yu Yeon Jung
- Department of Dental Hygiene, Gwangyang Health Sciences University, Gwnagyang, Jeonnam 57764, Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk 28160, Korea
| |
Collapse
|
9
|
Xia J, Huang N, Huang H, Sun L, Dong S, Su J, Zhang J, Wang L, Lin L, Shi M, Bin J, Liao Y, Li N, Liao W. Voltage-gated sodium channel Nav 1.7 promotes gastric cancer progression through MACC1-mediated upregulation of NHE1. Int J Cancer 2016; 139:2553-69. [PMID: 27529686 DOI: 10.1002/ijc.30381] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 07/19/2016] [Accepted: 07/26/2016] [Indexed: 12/20/2022]
Abstract
Voltage-gated sodium channels (VGSCs), which are aberrantly expressed in several human cancers, affect cancer cell behavior; however, their role in gastric cancer (GC) and the link between these channels and tumorigenic signaling remain unclear. The aims of this study were to determine the clinicopathological significance and role of the VGSC Nav 1.7 in GC progression and to investigate the associated mechanisms. Here, we report that the SCN9A gene encoding Nav 1.7 was the most abundantly expressed VGSC subtype in GC tissue samples and two GC cell lines (BGC-823 and MKN-28 cells). SCN9A expression levels were also frequently found to be elevated in GC samples compared to nonmalignant tissues by real-time PCR. In the 319 GC specimens evaluated by immunohistochemistry, Nav 1.7 expression was correlated with prognosis, and transporter Na(+) /H(+) exchanger-1 (NHE1) and oncoprotein metastasis-associated in colon cancer-1 (MACC1) expression. Nav 1.7 suppression resulted in reduced voltage-gated sodium currents, decreased NHE1 expression, increased extracellular pH and decreased intracellular pH, and ultimately, reduced invasion and proliferation rates of GC cells and growth of GC xenografts in nude mice. Nav 1.7 inhibition led to reduced MACC1 expression, while MACC1 inhibition resulted in reduced NHE1 expression in vitro and in vivo. Mechanistically, the suppression of Nav 1.7 decreased NF-κB p65 nuclear translocation via p38 activation, thus reducing MACC1 expression. Downregulation of MACC1 decreased c-Jun phosphorylation and subsequently reduced NHE1 expression, whereas the addition of hepatocyte growth factor (HGF), a c-Met physiological ligand, reversed the effect. These results indicate that Nav 1.7 promotes GC progression through MACC1-mediated upregulation of NHE1. Therefore, Nav 1.7 is a potential prognostic marker and/or therapeutic target for GC.
Collapse
Affiliation(s)
- Jianling Xia
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Na Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hongxiang Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Li Sun
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Shaoting Dong
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jinyu Su
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jingwen Zhang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Lin Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Li Lin
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Min Shi
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jianping Bin
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yulin Liao
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Nailin Li
- Karolinska Institute, Department of Medicine-Solna, Clinical Pharmacology Group, Karolinska University Hospital-Solna, Stockholm, 17176, Sweden
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
10
|
Zheng J, Son DJ, Gu SM, Woo JR, Ham YW, Lee HP, Kim WJ, Jung JK, Hong JT. Piperlongumine inhibits lung tumor growth via inhibition of nuclear factor kappa B signaling pathway. Sci Rep 2016; 6:26357. [PMID: 27198178 PMCID: PMC4873819 DOI: 10.1038/srep26357] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 04/29/2016] [Indexed: 12/11/2022] Open
Abstract
Piperlongumine has anti-cancer activity in numerous cancer cell lines via various signaling pathways. But there has been no study regarding the mechanisms of PL on the lung cancer yet. Thus, we evaluated the anti-cancer effects and possible mechanisms of PL on non-small cell lung cancer (NSCLC) cells in vivo and in vitro. Our findings showed that PL induced apoptotic cell death and suppressed the DNA binding activity of NF-κB in a concentration dependent manner (0-15 μM) in NSCLC cells. Docking model and pull down assay showed that PL directly binds to the DNA binding site of nuclear factor-κB (NF-κB) p50 subunit, and surface plasmon resonance (SPR) analysis showed that PL binds to p50 concentration-dependently. Moreover, co-treatment of PL with NF-κB inhibitor phenylarsine oxide (0.1 μM) or p50 siRNA (100 nM) augmented PL-induced inhibitory effect on cell growth and activation of Fas and DR4. Notably, co-treatment of PL with p50 mutant plasmid (C62S) partially abolished PL-induced cell growth inhibition and decreased the enhanced expression of Fas and DR4. In xenograft mice model, PL (2.5-5 mg/kg) suppressed tumor growth of NSCLC dose-dependently. Therefore, these results indicated that PL could inhibit lung cancer cell growth via inhibition of NF-κB signaling pathway in vitro and in vivo.
Collapse
Affiliation(s)
- Jie Zheng
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osong-eup, Heungduk-gu, Cheongju, Chungbuk 28160, Republic of Korea
| | - Dong Ju Son
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osong-eup, Heungduk-gu, Cheongju, Chungbuk 28160, Republic of Korea
| | - Sun Mi Gu
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osong-eup, Heungduk-gu, Cheongju, Chungbuk 28160, Republic of Korea
| | - Ju Rang Woo
- New Drug Development Center, KBio, Osong-eup, Heungduk-gu, Cheongju, Chungbuk 28160, Republic of Korea
| | - Young Wan Ham
- Department of Chemistry, Utah Valley University, 800 West University Parkway, Orem, UT 84508, USA
| | - Hee Pom Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osong-eup, Heungduk-gu, Cheongju, Chungbuk 28160, Republic of Korea
| | - Wun Jae Kim
- College of Medicine, Chungbuk National University, Chungdae-ro 1, Seowon-gu, Cheongju, Chungbuk 28644, Republic of Korea
| | - Jae Kyung Jung
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osong-eup, Heungduk-gu, Cheongju, Chungbuk 28160, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osong-eup, Heungduk-gu, Cheongju, Chungbuk 28160, Republic of Korea
| |
Collapse
|
11
|
Xia ZX, Li ZX, Zhang M, Sun LM, Zhang QF, Qiu XS. CARMA3 regulates the invasion, migration, and apoptosis of non-small cell lung cancer cells by activating NF-кB and suppressing the P38 MAPK signaling pathway. Exp Mol Pathol 2015; 100:353-60. [PMID: 26526492 DOI: 10.1016/j.yexmp.2015.10.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 10/20/2015] [Accepted: 10/21/2015] [Indexed: 11/19/2022]
Abstract
In our previous study, CARMA3 overexpression in lung cancer cells promoted cell proliferation and invasion; however, the mechanism underlying the role of CARMA3 in cancer cell invasion remained unclear. In the present study, knockdown of CARMA3 in A549 and H1299 cells suppressed cell invasion and migration, and downregulated matrix metalloprotease 9 expression at the protein and mRNA levels, as shown by Western blotting and real-time PCR. CARMA3 knockdown increased cell apoptosis, as shown by flow cytometry, increased the mRNA and protein expression levels of Bax and Caspase3, and downregulated Bcl-2 in A549 and H1299 cells. Phosphorylated P38 levels increased and NF-кB activation decreased following knockdown of CARMA3. SB203580, a P38 MAPK inhibitor, activated NF-кB, increased cell migration, and inhibited cell apoptosis after knockdown of CARMA3 compared to knockdown of CARMA3 without SB203580. These findings indicate that CARMA3 may suppress the activation of the P38 MAPK signaling pathway to regulate invasion, migration and apoptosis of lung cancer cells by activating NF-кB (P65) in the nucleus.
Collapse
Affiliation(s)
- Z X Xia
- Department of Pathology, the First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China; Department of Pathology, Center Hospital of Wuhan City, Wuhan, Hubei, China
| | - Z X Li
- Department of Radiology, the First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - M Zhang
- Department of Pathology, College of Basic Medical Sciences, Shenyang Medical College, Shenyang, Liaoning, China
| | - L M Sun
- Department of Pathology, the First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Q F Zhang
- Department of Pathology, the First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - X S Qiu
- Department of Pathology, the First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
12
|
Park MH, Hong JE, Park ES, Yoon HS, Seo DW, Hyun BK, Han SB, Ham YW, Hwang BY, Hong JT. Anticancer effect of tectochrysin in colon cancer cell via suppression of NF-kappaB activity and enhancement of death receptor expression. Mol Cancer 2015; 14:124. [PMID: 26123287 PMCID: PMC4487202 DOI: 10.1186/s12943-015-0377-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 05/01/2015] [Indexed: 12/21/2022] Open
Abstract
Background Flavonoids are a diverse family of natural phenolic compounds commonly found in fruits and vegetables. Epidemiologic studies showed that flavonoids also reduce the risk of colon cancer. Tectochrysin is one of the major flavonoids of Alpinia oxyphylla Miquel. However, the anti-cancer effects and the molecular mechanisms of tectochrysin in colon cancer cells have not yet been reported. We investigated whether tectochrysin could inhibit colon cancer cell growth at 1, 5, 10 μg/ml. In in vivo study, we injected a tectochrysin treatment dose of 5 mg/kg to each mouse. Results Tectochrysin suppressed the growth of SW480 and HCT116 human colon cancer cells. The expression of DR3, DR4 and Fas were significantly increased, and pro-apoptotic proteins were also increased. Tectochrysin treatment also inhibited activity of NF-κB. A docking model indicated that tectochrysin binds directly to the p50 unit. In in vivo, tumor weights and volumes in mice were reduced when treated with tectochrysin. Tectochrysin leads to apoptotic cell death in colon cancer cells through activation of death receptors expression via the inhibition of NF-κB. Conclusions Tectochrysin can be a useful agent for the treatment of colon cancer cell growth as well as an adjuvant agent for chemo-resistant cancer cells growth.
Collapse
Affiliation(s)
- Mi Hee Park
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk, 361-951, Republic of Korea.
| | - Ji Eun Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk, 361-951, Republic of Korea.
| | - Eun Sook Park
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk, 361-951, Republic of Korea.
| | - Hee Sung Yoon
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk, 361-951, Republic of Korea.
| | - Doo Won Seo
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk, 361-951, Republic of Korea.
| | - Byung Kook Hyun
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk, 361-951, Republic of Korea.
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk, 361-951, Republic of Korea.
| | - Young Won Ham
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA.
| | - Bang Yeon Hwang
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk, 361-951, Republic of Korea.
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk, 361-951, Republic of Korea.
| |
Collapse
|
13
|
Ma L, Peng H, Li K, Zhao R, Li L, Yu Y, Wang X, Han Z. Luteolin exerts an anticancer effect on NCI-H460 human non-small cell lung cancer cells through the induction of Sirt1-mediated apoptosis. Mol Med Rep 2015; 12:4196-4202. [PMID: 26096576 PMCID: PMC4526059 DOI: 10.3892/mmr.2015.3956] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Accepted: 04/14/2015] [Indexed: 12/23/2022] Open
Abstract
Luteolin is a falconoid compound, which exhibits anticancer properties, however, its contribution to Sirt1-mediated apoptosis in human non-small cell lung cancer remains to be elucidated. The present study confirmed that the anticancer effect of luteolin on NCI-H460 cells was through Sirt1-mediated apoptosis. The NCI-H460 cells were treated with different concentrations of luteolin, and a 3-(4,5-dimeth yl-2-thiazolyl)-2,5-diphnyl-2H-tetrazolium bromide assay, cell cycle analysis and annexin-V/fluorescein isothiocyanate and propidium double staining were performed to assess the apoptotic effect of luteolin. Wound healing and Transwell assays were performed to confirm the inhibition of NCI-H460 cell migration. The protein levels of Sirt1 were knocked down in the NCI-H460 cells using a lentivirus to further investigate the role of this protein, and the expression levels of the apoptotic associated proteins, Bad, Bcl-2, Bax, caspase-3 and Sirt1, were measured using western blotting. The results of the present study demonstrated that luteolin exerted an anticancer effect against NCI-H460 cells through Sirt1-mediated apoptosis and the inhibition of cell migration.
Collapse
Affiliation(s)
- Liping Ma
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210003, P.R. China
| | - Hongjun Peng
- Department of Pediatrics, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu 210002, P.R. China
| | - Kunsheng Li
- Department of Thoracic Surgery, BenQ Medical Center, Nanjing Medical University, Nanjing, Jiangsu 210019, P.R. China
| | - Runrun Zhao
- Department of Thoracic Surgery, BenQ Medical Center, Nanjing Medical University, Nanjing, Jiangsu 210019, P.R. China
| | - Li Li
- Department of Thoracic Surgery, BenQ Medical Center, Nanjing Medical University, Nanjing, Jiangsu 210019, P.R. China
| | - Yilong Yu
- Department of Thoracic Surgery, BenQ Medical Center, Nanjing Medical University, Nanjing, Jiangsu 210019, P.R. China
| | - Xiaoming Wang
- Department of Thoracic Surgery, BenQ Medical Center, Nanjing Medical University, Nanjing, Jiangsu 210019, P.R. China
| | - Zhifeng Han
- Department of Thoracic Surgery, BenQ Medical Center, Nanjing Medical University, Nanjing, Jiangsu 210019, P.R. China
| |
Collapse
|
14
|
Kwon SM, Jung YY, Hwang CJ, Park MH, Yoon NY, Kim TM, Yu JM, Kim DH, Seo DW, Youn HS, Seo HO, Chung IS, Han SB, Hwang BY, Yoo HS, Jung JK, Lee H, Hong JT. Anti-cancer effect of N-(3,5-bis(trifluoromethyl)phenyl)-5-chloro-2,3-dihydronaphtho[1,2-b]furan-2-carboxamide, a novel synthetic compound. Mol Carcinog 2015; 55:659-70. [DOI: 10.1002/mc.22311] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 12/27/2014] [Accepted: 02/18/2015] [Indexed: 01/05/2023]
Affiliation(s)
- Sun Mi Kwon
- College of Pharmacy and Medical Research Center; Chungbuk National University; Chungbuk Korea
| | - Yu Yeon Jung
- College of Pharmacy and Medical Research Center; Chungbuk National University; Chungbuk Korea
| | - Chul Ju Hwang
- College of Pharmacy and Medical Research Center; Chungbuk National University; Chungbuk Korea
| | - Mi Hee Park
- College of Pharmacy and Medical Research Center; Chungbuk National University; Chungbuk Korea
| | - Na Young Yoon
- College of Pharmacy and Medical Research Center; Chungbuk National University; Chungbuk Korea
| | - Tae Myung Kim
- College of Veterinary Medicine; Chungbuk National University; Chungbuk Korea
| | - Ji Myung Yu
- College of Veterinary Medicine; Chungbuk National University; Chungbuk Korea
| | - Dae Hwan Kim
- College of Pharmacy and Medical Research Center; Chungbuk National University; Chungbuk Korea
| | - Doo Won Seo
- College of Pharmacy and Medical Research Center; Chungbuk National University; Chungbuk Korea
| | - Hyu Seok Youn
- College of Pharmacy and Medical Research Center; Chungbuk National University; Chungbuk Korea
| | - Hyun Ok Seo
- College of Pharmacy and Medical Research Center; Chungbuk National University; Chungbuk Korea
| | - In Sung Chung
- College of Pharmacy and Medical Research Center; Chungbuk National University; Chungbuk Korea
| | - Sang Bae Han
- College of Pharmacy and Medical Research Center; Chungbuk National University; Chungbuk Korea
| | - Bang Yeon Hwang
- College of Pharmacy and Medical Research Center; Chungbuk National University; Chungbuk Korea
| | - Hwan-Soo Yoo
- College of Pharmacy and Medical Research Center; Chungbuk National University; Chungbuk Korea
| | - Jae-Kyung Jung
- College of Pharmacy and Medical Research Center; Chungbuk National University; Chungbuk Korea
| | - Heesoon Lee
- College of Pharmacy and Medical Research Center; Chungbuk National University; Chungbuk Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center; Chungbuk National University; Chungbuk Korea
| |
Collapse
|
15
|
Lee HL, Park MH, Hong JE, Kim DH, Kim JY, Seo HO, Han SB, Yoon JH, Lee WH, Song HS, Lee JI, Lee US, Song MJ, Hong JT. Inhibitory effect of snake venom toxin on NF-κB activity prevents human cervical cancer cell growth via increase of death receptor 3 and 5 expression. Arch Toxicol 2014; 90:463-77. [DOI: 10.1007/s00204-014-1393-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 10/16/2014] [Indexed: 12/12/2022]
|
16
|
Oh SB, Hwang CJ, Song SY, Jung YY, Yun HM, Sok CH, Sung HC, Yi JM, Park DH, Ham YW, Han SB, Hwang BY, Hong JT. Anti-cancer effect of tectochrysin in NSCLC cells through overexpression of death receptor and inactivation of STAT3. Cancer Lett 2014; 353:95-103. [DOI: 10.1016/j.canlet.2014.07.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 06/01/2014] [Accepted: 07/07/2014] [Indexed: 01/12/2023]
|
17
|
Ban JO, Jung YS, Kim DH, Park KR, Yun HM, Lee NJ, Lee HP, Shim JH, Jeong HS, Lee YH, Ham YW, Han SB, Hong JT. (E)-2,4-Bis(p-hydroxyphenyl)-2-butenal inhibits tumor growth via suppression of NF-κB and induction of death receptor 6. Apoptosis 2014; 19:165-78. [PMID: 24052407 DOI: 10.1007/s10495-013-0903-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The Maillard reaction products are known to be effective in chemoprevention. Here, we focused on the anti-cancer effects of (E)-2,4-bis(p-hydroxyphenyl)-2-butenal on in vitro and in vivo colon cancer. We analysed the anti-cancer activity of (E)-2,4-bis(p-hydroxyphenyl)-2-butenal on colon cancer cells by using cell cycle and apoptosis analysis. To elucidate it's mechanism, NF-κB DNA binding activity, docking model as well as pull-down assay. Further, a xenograft model of colon cancer was studied to test the in vivo effects of (E)-2,4-bis(p-hydroxyphenyl)-2-butenal. (E)-2,4-Bis(p-hydroxyphenyl)-2-butenal inhibited colon cancer cells (SW620 and HCT116) growth followed by induction of apoptosis in a concentration-dependent manner via down-regulation of NF-κB activity. In docking model as well as pull-down assay, (E)-2,4-bis(p-hydroxyphenyl)-2-butenal directly binds to three amino acid residues of IKKβ, thereby inhibited IKKβ activity in addition to induction of death receptor 6 (DR6) as well as their target apoptotic genes. Finally, (E)-2,4-bis(p-hydroxyphenyl)-2-butenal suppressed anchorage-independent cancer cell growth, and tumor growth in xenograft model accompanied with apoptosis through inhibition of IKKβ/NF-κB activity, and overexpression of DR6. These results suggest that (E)-2,4-bis(p-hydroxyphenyl)-2-butenal inhibits colon cancer cell growth through inhibition of IKKβ/NF-κB activity and induction of DR6 expression.
Collapse
Affiliation(s)
- Jung Ok Ban
- College of Pharmacy and Medical Research Center, Chungbuk National University, 12, Gaeshin-dong, Heungduk-gu, Cheongju, Chungbuk, 361-763, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Cancer cell growth inhibitory effect of bee venom via increase of death receptor 3 expression and inactivation of NF-kappa B in NSCLC cells. Toxins (Basel) 2014; 6:2210-28. [PMID: 25068924 PMCID: PMC4147578 DOI: 10.3390/toxins6082210] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 06/16/2014] [Accepted: 07/18/2014] [Indexed: 01/24/2023] Open
Abstract
Our previous findings have demonstrated that bee venom (BV) has anti-cancer activity in several cancer cells. However, the effects of BV on lung cancer cell growth have not been reported. Cell viability was determined with trypan blue uptake, soft agar formation as well as DAPI and TUNEL assay. Cell death related protein expression was determined with Western blotting. An EMSA was used for nuclear factor kappaB (NF-κB) activity assay. BV (1–5 μg/mL) inhibited growth of lung cancer cells by induction of apoptosis in a dose dependent manner in lung cancer cell lines A549 and NCI-H460. Consistent with apoptotic cell death, expression of DR3 and DR6 was significantly increased. However, deletion of DRs by small interfering RNA significantly reversed BV induced cell growth inhibitory effects. Expression of pro-apoptotic proteins (caspase-3 and Bax) was concomitantly increased, but the NF-κB activity and expression of Bcl-2 were inhibited. A combination treatment of tumor necrosis factor (TNF)-like weak inducer of apoptosis, TNF-related apoptosis-inducing ligand, docetaxel and cisplatin, with BV synergistically inhibited both A549 and NCI-H460 lung cancer cell growth with further down regulation of NF-κB activity. These results show that BV induces apoptotic cell death in lung cancer cells through the enhancement of DR3 expression and inhibition of NF-κB pathway.
Collapse
|
19
|
Cho SH, Park MH, Lee HP, Back MK, Sung HC, Chang HW, Kim JH, Jeong HS, Han SB, Hong JT. (E)-2,4-Bis(p-hydroxyphenyl)-2-butenal enhanced TRAIL-induced apoptosis in ovarian cancer cells through downregulation of NF-κB/STAT3 pathway. Arch Pharm Res 2014; 37:652-61. [PMID: 24390815 DOI: 10.1007/s12272-013-0326-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 12/24/2013] [Indexed: 12/13/2022]
Abstract
Ovarian cancer is a cancerous growth arising from the ovary and with poor prognosis that usually have resistant to all currently available treatments. Whether (E)-2,4-bis(p-hydroxyphenyl)-2-butenal (butenal) synthesized by Maillard reaction from fructose-tyrosine, has potential therapeutic activity against human ovarian cancer was investigated using two ovarian cancer cell lines (PA-1, SK-OV-3). We found that butenal could inhibit NF-κB/STAT3 activity, thereby inducing apoptotic cell death of ovarian cancer cells. We treated with several concentration of butenal each cell line differently (PA-1; 5, 10 and 15 μg/ml, SK-OV-3; 10, 20 and 30 μg/ml). First, ovarian cancer cell lines exhibited constitutively active NF-κB, and treatment with butenal abolished this activation as indicated by DNA binding activity. Second, butenal suppressed activation of signal transducer and activator of transcription-3 as indicated by decreased phosphorylation and inhibition of Janus kinase-2 phosphorylation. Third, butenal induced expression of pro-apoptotic proteins such as proteolytic cleavage of PARP, Bax and activation of caspase-3, -8 and -9. Lastly, combination of butenal and TRAIL causes enhanced induction of apoptosis. Overall, our results indicate that butenal mediates its anti-proliferative and apoptotic effects through activation of multiple cell signaling pathways and enhances the TRAIL-induced apoptosis. These data suggested that butenal may be a potential anti-cancer agent in ovarian cancer.
Collapse
Affiliation(s)
- Seung Hee Cho
- College of Pharmacy, Medical Research Center, Chungbuk National University, 12, Gaeshin-dong, Heungduk-gu, Ch'ongju, 361-763, Chungbuk, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Wang PY, Sun YX, Zhang S, Pang M, Zhang HH, Gao SY, Zhang C, Lv CJ, Xie SY. Let-7c inhibits A549 cell proliferation through oncogenic TRIB2 related factors. FEBS Lett 2013; 587:2675-81. [PMID: 23850892 DOI: 10.1016/j.febslet.2013.07.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 07/01/2013] [Accepted: 07/01/2013] [Indexed: 01/15/2023]
Abstract
MicroRNAs have tumor suppressive or oncogenic roles in carcinogenesis. This study aimed to investigate the mechanism of let-7c in suppressing lung cancer cell proliferation. First, let-7c was revealed to be able to inhibit lung adenocarcinoma cell proliferation significantly. TRIB2 was further demonstrated to be a novel target and negatively regulated by let-7c. As downstream signals of TRIB2, the activities of C/EBP-α and phosphorylated p38MAPK were increased obviously in let-7c-treated cells compared with controls. Our results demonstrate that, through regulating the expression of TRIB2 and its downstream factors, let-7c can effectively inhibit A549 cell proliferation in vitro and in vivo.
Collapse
Affiliation(s)
- Ping-Yu Wang
- Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology, Binzhou Medical University, YanTai, ShanDong 264003, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|