1
|
Al-qaraghuli S, Gache Y, Goncalves-Maia M, Alcor D, Muzotte E, Mahfouf W, Rezvani HR, Magnaldo T. Xeroderma Pigmentosum Type C Primary Skin Fibroblasts Overexpress HGF and Promote Squamous Cell Carcinoma Invasion in the Absence of Genotoxic Stress. Cancers (Basel) 2024; 16:3277. [PMID: 39409898 PMCID: PMC11475422 DOI: 10.3390/cancers16193277] [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: 04/14/2024] [Revised: 05/15/2024] [Accepted: 05/24/2024] [Indexed: 10/20/2024] Open
Abstract
Xeroderma pigmentosum (XP) is a very rare recessive disease caused by the incapacity to resolve ultraviolet-induced DNA lesions through Nucleotide Excision Repair (NER). Most XP patients suffer from aggressive skin carcinoma and melanoma at a very early age (<8). Our previous results showed that primary XP fibroblasts isolated from healthy (non-photo-exposed) skin negatively impact the extracellular matrix and fail to activate the innate immune system. Here, we show for the first time that XP-C fibroblasts also play a major role in cancer cell invasion ex vivo and in vivo through the overexpression of Hepatocyte Growth Factor/Scatter Factor (HGF/SF) in the absence of genotoxic attacks. The use of inhibitors of the activation of the HGF/SF pathway counteracted the effects of XP fibroblasts on the growth of cancer cells, suggesting new perspectives in the care of XP patients.
Collapse
Affiliation(s)
- Sahar Al-qaraghuli
- INSERM U1081–CNRS UMR7284-UNS, CEDEX 02, F-06107 Nice, France; (S.A.-q.)
- Faculté de Médicine, 2ème étage, CNRS UMR 6267—INSERM U998—UNSA, F-06107 Nice Cedex 2, France
| | - Yannick Gache
- INSERM U1081–CNRS UMR7284-UNS, CEDEX 02, F-06107 Nice, France; (S.A.-q.)
- Faculté de Médicine, 2ème étage, CNRS UMR 6267—INSERM U998—UNSA, F-06107 Nice Cedex 2, France
| | - Maria Goncalves-Maia
- INSERM U1081–CNRS UMR7284-UNS, CEDEX 02, F-06107 Nice, France; (S.A.-q.)
- Faculté de Médicine, 2ème étage, CNRS UMR 6267—INSERM U998—UNSA, F-06107 Nice Cedex 2, France
| | - Damien Alcor
- Faculté de Médicine, 2ème étage, CNRS UMR 6267—INSERM U998—UNSA, F-06107 Nice Cedex 2, France
- INSERM U1065, C3M, Microscopy Facility, F-06200 Nice, France
| | - Elodie Muzotte
- BRIC, UMR 1312, Inserm, Université de Bordeaux, F-33076 Bordeaux, France
| | - Walid Mahfouf
- BRIC, UMR 1312, Inserm, Université de Bordeaux, F-33076 Bordeaux, France
| | - Hamid-Reza Rezvani
- BRIC, UMR 1312, Inserm, Université de Bordeaux, F-33076 Bordeaux, France
- Centre de Référence pour les Maladies Rares de la Peau, CHU de Bordeaux, F-33000 Bordeaux, France
| | - Thierry Magnaldo
- INSERM U1081–CNRS UMR7284-UNS, CEDEX 02, F-06107 Nice, France; (S.A.-q.)
- Faculté de Médicine, 2ème étage, CNRS UMR 6267—INSERM U998—UNSA, F-06107 Nice Cedex 2, France
| |
Collapse
|
2
|
Seth A, Yokokura Y, Choi JY, Shyer JA, Vidyarthi A, Craft J. AP-1-independent NFAT signaling maintains follicular T cell function in infection and autoimmunity. J Exp Med 2023; 220:e20211110. [PMID: 36820828 PMCID: PMC9998660 DOI: 10.1084/jem.20211110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/05/2022] [Accepted: 02/01/2023] [Indexed: 02/24/2023] Open
Abstract
Coordinated gene expression programs enable development and function of T cell subsets. Follicular helper T (Tfh) cells coordinate humoral immune responses by providing selective and instructive cues to germinal center B cells. Here, we show that AP-1-independent NFAT gene expression, a program associated with hyporesponsive T cell states like anergy or exhaustion, is also a distinguishing feature of Tfh cells. NFAT signaling in Tfh cells, maintained by NFAT2 autoamplification, is required for their survival. ICOS signaling upregulates Bcl6 and induces an AP-1-independent NFAT program in primary T cells. Using lupus-prone mice, we demonstrate that genetic disruption or pharmacologic inhibition of NFAT signaling specifically impacts Tfh cell maintenance and leads to amelioration of autoantibody production and renal injury. Our data provide important conceptual and therapeutic insights into the signaling mechanisms that regulate Tfh cell development and function.
Collapse
Affiliation(s)
- Abhinav Seth
- Department of Internal Medicine, Section of Rheumatology, Allergy and Immunology, School of Medicine, Yale University, New Haven, CT, USA
| | - Yoshiyuki Yokokura
- Department of Internal Medicine, Section of Rheumatology, Allergy and Immunology, School of Medicine, Yale University, New Haven, CT, USA
| | - Jin-Young Choi
- Department of Internal Medicine, Section of Rheumatology, Allergy and Immunology, School of Medicine, Yale University, New Haven, CT, USA
| | - Justin A. Shyer
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Aurobind Vidyarthi
- Department of Internal Medicine, Section of Rheumatology, Allergy and Immunology, School of Medicine, Yale University, New Haven, CT, USA
| | - Joe Craft
- Department of Internal Medicine, Section of Rheumatology, Allergy and Immunology, School of Medicine, Yale University, New Haven, CT, USA
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| |
Collapse
|
3
|
Zhang X, Guo Y, Xiao T, Li J, Guo A, Lei L, Jin C, Long Q, Su J, Yin M, Liu H, Chen C, Zhou Z, Zhu S, Tao J, Hu S, Chen X, Peng C. CD147 mediates epidermal malignant transformation through the RSK2/AP-1 pathway. J Exp Clin Cancer Res 2022; 41:246. [PMID: 35964097 PMCID: PMC9375950 DOI: 10.1186/s13046-022-02427-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 07/01/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Malignant transformation of the epidermis is an essential process in the pathogenesis of cutaneous squamous-cell carcinoma (cSCC). Although evidence has demonstrated that CD147 plays key roles in various tumors, the role of CD147 in epidermal malignant transformation in vivo remains unclear.
Methods
Epidermal CD147-overexpression or knockout (EpiCD147-OE or EpiCD147-KO) transgenic mouse models were generated for in vivo study. RNA-sequencing and q-PCR were performed to identify the differentially expressed genes. Immunohistochemistry and flow cytometry were performed to investigate the role of CD147 in regulating myeloid-derived suppressor cells (MDSCs). Immunoprecipitation, EMSA and ChIP assays were performed to investigate the mechanism of CD147 in cell transformation.
Results
We found that specific overexpression of CD147 in the epidermis (EpiCD147-OE) induces spontaneous tumor formation; moreover, a set of chemokines and cytokines including CXCL1, which play essential function in MDSC recruitment, were significantly upregulated in EpiCD147-OE transgenic mice. As expected, overexpression of CD147 in the epidermis remarkably facilitated tumorigenesis by increasing the rate of tumor initiation and the number and size of tumors in the DMBA/TPA mouse model. Interestingly, the expression of CXCL1 and the infiltration of MDSCs were dramatically increased in EpiCD147-OE transgenic mice. Our findings also showed that knockdown of CD147 attenuated EGF-induced malignant transformation as well as CXCL1 expression in HaCaT cells. Consistently, CD147 was found overexpressed in cutaneous squamous cell carcinoma (cSCC), and positively related with the expression of CD33, a myeloid-associated marker. We further identified RSK2, a serine/threonine kinase, as an interacting partner of CD147 at the binding site of CD147D207-230. The interaction of CD147 and RSK2 activated RSK2, thus enhancing AP-1 transcriptional activation. Furthermore, EMSAs and ChIP assays showed that AP-1 could associate with the CXCL1 promoter. Importantly, RSK2 inhibitor suppressed the tumor growth in DMBA/TPA mouse model by inhibiting the recruitment of MDSCs.
Conclusion
Our findings demonstrate that CD147 exerts a key function in epidermal malignant transformation in vivo by activating keratinocytes and recruiting MDSCs via the RSK2/AP-1 pathway.
Collapse
|
4
|
Synthesis and study of thioglycoside conjugates of 4-chloro-1,2-dithiol-3-one as potential cancer-preventive substances in vitro and in vivo. Russ Chem Bull 2022. [DOI: 10.1007/s11172-022-3438-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
5
|
Fedorov SN, Kuzmich AS, Sabutskii YE, Guzii AG, Popov RS, Ogurtsov VA, Rakitin OA, Polonik SG. Synthesis and studies of acetylthioglycoside conjugates of 4-chloro-1,2-dithiole-3-thione as potential antitumor agents. Russ Chem Bull 2021. [DOI: 10.1007/s11172-021-3127-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
6
|
Pore-forming toxins from sea anemones: from protein-membrane interaction to its implications for developing biomedical applications. ADVANCES IN BIOMEMBRANES AND LIPID SELF-ASSEMBLY 2020. [DOI: 10.1016/bs.abl.2020.02.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
7
|
Dihydrocapsaicin Inhibits Epithelial Cell Transformation through Targeting Amino Acid Signaling and c-Fos Expression. Nutrients 2019; 11:nu11061269. [PMID: 31167465 PMCID: PMC6627986 DOI: 10.3390/nu11061269] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 05/29/2019] [Accepted: 06/02/2019] [Indexed: 01/09/2023] Open
Abstract
Chili peppers are one of the most widely consumed spices worldwide. However, research on the health benefits of chili peppers and some of its constituents has raised controversy as to whether chili pepper compounds possess cancer-promoting or cancer-preventive effects. While ample studies have been carried out to examine the effect of capsaicin in carcinogenesis, the chemopreventive effect of other major components in chili pepper, including dihydrocapsaicin, capsiate, and capsanthin, is relatively unclear. Herein, we investigated the inhibitory effect of chili pepper components on malignant cell transformation. Among the tested chili pepper compounds, dihydrocapsaicin displayed the strongest inhibitory activity against epidermal growth factor (EGF)-induced neoplastic transformation. Dihydrocapsaicin specifically suppressed EGF-induced phosphorylations of the p70S6K1-S6 pathway and the expression of c-Fos. A reduction in c-Fos levels by dihydrocapsaicin led to a concomitant downregulation of AP-1 activation. Further analysis of the molecular mechanism responsible for the dihydrocapsaicin-mediated decrease in phospho-p70S6K1, revealed that dihydrocapsaicin can block amino acid-dependent mechanistic targets of rapamycin complex 1 (mTORC1)-p70S6K1-S6 signal activation. Additionally, dihydrocapsaicin was able to selectively augment amino acid deprivation-induced cell death in mTORC1-hyperactive cells. Collectively, dihydrocapsaicin exerted chemopreventive effects through inhibiting amino acid signaling and c-Fos pathways and, thus, might be a promising cancer preventive natural agent.
Collapse
|
8
|
Matsuhashi S, Manirujjaman M, Hamajima H, Ozaki I. Control Mechanisms of the Tumor Suppressor PDCD4: Expression and Functions. Int J Mol Sci 2019; 20:ijms20092304. [PMID: 31075975 PMCID: PMC6539695 DOI: 10.3390/ijms20092304] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/05/2019] [Accepted: 05/07/2019] [Indexed: 02/06/2023] Open
Abstract
PDCD4 is a novel tumor suppressor to show multi-functions inhibiting cell growth, tumor invasion, metastasis, and inducing apoptosis. PDCD4 protein binds to the translation initiation factor eIF4A, some transcription factors, and many other factors and modulates the function of the binding partners. PDCD4 downregulation stimulates and PDCD4 upregulation inhibits the TPA-induced transformation of cells. However, PDCD4 gene mutations have not been found in tumor cells but gene expression was post transcriptionally downregulated by micro environmental factors such as growth factors and interleukins. In this review, we focus on the suppression mechanisms of PDCD4 protein that is induced by the tumor promotors EGF and TPA, and in the inflammatory conditions. PDCD4-protein is phosphorylated at 2 serines in the SCFβTRCP ubiquitin ligase binding sequences via EGF and/or TPA induced signaling pathway, ubiquitinated, by the ubiquitin ligase and degraded in the proteasome system. The PDCD4 protein synthesis is inhibited by microRNAs including miR21.
Collapse
Affiliation(s)
- Sachiko Matsuhashi
- Department of Internal Medicine, Saga Medical School, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan.
| | - M Manirujjaman
- Department of Internal Medicine, Saga Medical School, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan.
| | - Hiroshi Hamajima
- Saga Food & Cosmetics Laboratory, Division of Food Manufacturing Industry Promotion, SAGA Regional Industry Support Center, 114 Yaemizo, Nabesima-Machi, Saga 849-0932, Japan.
| | - Iwata Ozaki
- Health Administration Center, Saga Medical School, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan.
| |
Collapse
|
9
|
Lin L, Li G, Zhang W, Wang YL, Yang H. Low-dose aspirin reduces hypoxia-induced sFlt1 release via the JNK/AP-1 pathway in human trophoblast and endothelial cells. J Cell Physiol 2019; 234:18928-18941. [PMID: 31004367 DOI: 10.1002/jcp.28533] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 02/19/2019] [Accepted: 02/20/2019] [Indexed: 11/06/2022]
Abstract
Pre-eclampsia (PE) is a serious hypertensive disorder of pregnancy that remains a leading cause of perinatal and maternal morbidity and mortality worldwide. Placental ischemia/hypoxia and the secretion of soluble fms-like tyrosine kinase 1 (sFlt1) into maternal circulation are involved in the pathogenesis of PE. Although low-dose aspirin (LDA) has beneficial effects on the prevention of PE, the exact mechanisms of action of LDA, particularly on placental dysfunction, and sFlt1 release, have not been well investigated. This study aimed to determine whether LDA exists the protective effects on placental trophoblast and endothelial functions and prevents PE-associated sFlt1 release. First, we observed that LDA mitigated hypoxia-induced trophoblast apoptosis, showed positive effects on trophoblast cells migration and invasion activity, and increased the tube-forming activity of human umbilical vein endothelial cells (HUVECs). In addition, LDA decreased hypoxia-induced sFlt1 production, and the c-Jun NH2 -terminal kinase/activator protein-1 (JNK/AP-1) pathway was shown to mediate the induction of sFlt1. Moreover, the transcription factor AP-1 was confirmed to regulate the Flt1 gene expression by directly binding to the Flt1 promoter in luciferase assays. The result of chromatin immunoprecipitation assays further demonstrated that LDA could directly decrease the expression of the transcription factor AP-1, and thus decrease sFlt1 production. Finally, the effects of LDA on sFlt1 production were proved in human placental explants. Taken together, our data show the protective effects of LDA against trophoblast and endothelial cell dysfunction and reveal that the LDA-mediated inhibition of sFlt1 via the JNK/AP-1 pathway may be a potential cellular/molecular mechanism for the prevention of PE.
Collapse
Affiliation(s)
- Li Lin
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
| | - Guanlin Li
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Wanyi Zhang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
| | - Yan-Ling Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Science, Beijing, China
| | - Huixia Yang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China.,Beijing Key Laboratory of Maternal-Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| |
Collapse
|
10
|
Bouriez D, Giraud J, Gronnier C, Varon C. Efficiency of All-Trans Retinoic Acid on Gastric Cancer: A Narrative Literature Review. Int J Mol Sci 2018; 19:ijms19113388. [PMID: 30380687 PMCID: PMC6275086 DOI: 10.3390/ijms19113388] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 10/26/2018] [Accepted: 10/26/2018] [Indexed: 12/12/2022] Open
Abstract
Gastric cancer (GC) is the third leading cause of cancer-related death worldwide with a five-year survival rate of around 25%, and 4% when diagnosed at a metastatic stage. Cancer stem cells (CSC) have recently been characterized as being responsible for resistance to radio/chemotherapies and metastasis formation, opening up perspectives for new targeted therapies. Those CSCs express biomarkers such as cluster of differentiation 44 (CD44) and display high aldehyde dehydrogenase activity that converts vitamin A-derived retinal into retinoic acids. All-trans retinoic acid (ATRA), which has pro-differentiating properties, has revolutionized the prognosis of acute promyelotic leukemia by increasing its remission rate from 15% to 85%. Recent studies have started to show that ATRA also has an anti-tumoral role on solid cancers such as GC. The purpose of this review is therefore to summarize the work that evaluated the effects of ATRA in GC and to evaluate whether its anti-cancerous action involves gastric CSCs targeting. It has been demonstrated that ATRA can block the cell cycle, enhance apoptosis, and decrease gastric CSCs properties in GC cell lines, tumorspheres, and patient-derived xenograft mice models. Therefore, retinoids and new synthetic retinoids seem to be a promising step forward in targeted therapy of gastric CSC in combination with existing chemotherapies. Future studies should probably focus on these points.
Collapse
Affiliation(s)
- Damien Bouriez
- INSERM, U1053, Bordeaux Research in Translational Oncology, 33000 Bordeaux, France.
- Department of Digestive Surgery, Haut-Lévêque Hospital, 33000 Bordeaux, France.
| | - Julie Giraud
- INSERM, U1053, Bordeaux Research in Translational Oncology, 33000 Bordeaux, France.
- Department of Life and Health Sciences, University of Bordeaux, 33000 Bordeaux, France.
| | - Caroline Gronnier
- INSERM, U1053, Bordeaux Research in Translational Oncology, 33000 Bordeaux, France.
- Department of Digestive Surgery, Haut-Lévêque Hospital, 33000 Bordeaux, France.
- Department of Life and Health Sciences, University of Bordeaux, 33000 Bordeaux, France.
| | - Christine Varon
- INSERM, U1053, Bordeaux Research in Translational Oncology, 33000 Bordeaux, France.
- Department of Life and Health Sciences, University of Bordeaux, 33000 Bordeaux, France.
| |
Collapse
|
11
|
Kim G, Kim JY, Lim SC, Lee KY, Kim O, Choi HS. SUV39H1/DNMT3A-dependent methylation of the RB1 promoter stimulates PIN1 expression and melanoma development. FASEB J 2018; 32:5647-5660. [PMID: 29750576 DOI: 10.1096/fj.201700645rrrrr] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Melanoma is among the most aggressive and treatment-resistant human cancers. Aberrant histone H3 methylation at Lys 9 (H3K9) correlates with carcinogenic gene silencing, but the significance of suppressor of variegation 3-9 homolog 1 (SUV39H1), an H3K9-specific methyltransferase, in melanoma initiation and progression remains unclear. Here, we show that SUV39H1-mediated H3K9 trimethylation facilitates retinoblastoma ( RB) 1 promoter CpG island methylation by interacting with DNA methyltransferase 3A and decreasing RB mRNA and protein in melanoma cells. Reduced RB abundance, in turn, impairs E2F1 transcriptional inhibition, leading to increased peptidyl-prolyl cis-trans isomerase never-in-mitosis A (NIMA)-interacting 1 (PIN1) levels, human keratinocyte neoplastic cell transformation, and melanoma tumorigenesis via enhanced rapidly accelerated fibrosarcoma 1(RAF1)-MEK-ERK signaling pathway activation. In a synergistic model with B16-F1 murine melanoma cells, SUV39H1 and PIN1 overexpression increased melanoma growth, which was abrogated by their inhibition in SUV39H1-overexpressing B16-F1 mice. SUV39H1 also positively correlated with PIN1 expression in human melanoma. Our studies establish SUV39H1 as an oncogene in melanoma and underscore the role of chromatin factors in regulating tumorigenesis.-Kim, G., Kim, J.-Y., Lim, S.-C., Lee, K. Y., Kim, O., Choi, H. S. SUV39H1/DNMT3A-dependent methylation of the RB1 promoter stimulates PIN1 expression and melanoma development.
Collapse
Affiliation(s)
- Garam Kim
- College of Pharmacy, Chosun University, Gwangju, South Korea
| | - Jin-Young Kim
- College of Pharmacy, Chosun University, Gwangju, South Korea
| | - Sung-Chul Lim
- Department of Pathology, School of Medicine, Chosun University, Gwangju, South Korea
| | - Kwang Youl Lee
- College of Pharmacy, Chonnam National University, Gwangju, South Korea
| | - Okyun Kim
- College of Pharmacy, Chosun University, Gwangju, South Korea
| | - Hong Seok Choi
- College of Pharmacy, Chosun University, Gwangju, South Korea
| |
Collapse
|
12
|
Garg P, Pandey S, Hoon S, Jang KJ, Lee MC, Choung YH, Choung PH, Chung JH. JNK2 silencing and caspase-9 activation by hyperosmotic polymer inhibits tumor progression. Int J Biol Macromol 2018; 120:2215-2224. [PMID: 30003914 DOI: 10.1016/j.ijbiomac.2018.07.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 07/04/2018] [Accepted: 07/06/2018] [Indexed: 11/17/2022]
Abstract
c-Jun N-terminal kinase 2 (JNK2) is primarily responsible for the oncogenic transformation of the transcription factor c-Jun. Expression of the proto-oncogene c-Jun progresses the cell cycle from G1 to S phase, but when its expression becomes awry it leads to uncontrolled proliferation and angiogenesis. Delivering a JNK2 siRNA (siJNK2) in tumor tissue was anticipated to reverse the condition with subsequent onset of apoptosis which predominantly requires an efficient delivering system capable of penetrating through the compact tumor mass. In the present study, it was demonstrated that polymannitol-based vector (PMGT) with inherent hyperosmotic properties was able to penetrate through and deliver the siJNK2 in the subcutaneous tumor of xenograft mice. Hyperosmotic activity of polymannitol was shown to account for the enhanced therapeutic delivery both in vitro and in vivo because of the induction of cyclooxygenase-2 (COX-2) which stimulates caveolin-1 for caveolae-mediated endocytosis of the polyplexes. Further suppression of JNK2 and hence c-Jun expression led to the activation of caspase-9 to induce apoptosis and inhibition of tumor growth in xenograft mice model. The study exemplifies PMGT as an efficient vector for delivering therapeutic molecules in compact tumor tissue and suppression of JNK2 introduces a strategy to inhibit tumor progression.
Collapse
Affiliation(s)
- Pankaj Garg
- Department of Biosystems & Biomaterials Science and Engineering, Seoul National University, Seoul 151-921, Republic of Korea
| | - Shambhavi Pandey
- Department of Biosystems & Biomaterials Science and Engineering, Seoul National University, Seoul 151-921, Republic of Korea
| | - Seonwoo Hoon
- Department of Biosystems & Biomaterials Science and Engineering, Seoul National University, Seoul 151-921, Republic of Korea
| | - Kyoung-Je Jang
- Department of Biosystems & Biomaterials Science and Engineering, Seoul National University, Seoul 151-921, Republic of Korea
| | - Myung Chul Lee
- Department of Biosystems & Biomaterials Science and Engineering, Seoul National University, Seoul 151-921, Republic of Korea
| | - Yun-Hoon Choung
- Department of Otalaryngology, Ajou University School of Medicine, Suwon 443-749, Republic of Korea
| | - Pill-Hoon Choung
- Department of Oral and Maxillofacial Surgery and Dental Research Institute, School of Dentistry, Seoul National University, Seoul 110-774, Republic of Korea.
| | - Jong Hoon Chung
- Department of Biosystems & Biomaterials Science and Engineering, Seoul National University, Seoul 151-921, Republic of Korea; Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul 151-921, Republic of Korea.
| |
Collapse
|
13
|
Wang Q, Yang HS. The role of Pdcd4 in tumour suppression and protein translation. Biol Cell 2018; 110:10.1111/boc.201800014. [PMID: 29806708 PMCID: PMC6261700 DOI: 10.1111/boc.201800014] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 05/03/2018] [Accepted: 05/13/2018] [Indexed: 01/07/2023]
Abstract
Programmed cell death 4 (Pdcd4), a tumour suppressor, is frequently down-regulated in various types of cancer. Pdcd4 has been demonstrated to efficiently suppress tumour promotion, progression and proliferation. The biochemical function of Pdcd4 is a protein translation inhibitor. Although the fact that Pdcd4 inhibits protein translation has been known for more than a decade, the mechanism by which Pdcd4 controls tumorigenesis through translational regulation of its target genes is still not fully understood. Recent studies show that Pdcd4 inhibits translation of stress-activated-protein kinase interacting protein 1 to suppress tumour invasion, depicting a picture of how Pdcd4 inhibits tumorigenesis through translational inhibition. Thus, understanding the mechanism of how Pdcd4 attenuates tumorigenesis by translational control should provide a new strategy for combating cancer.
Collapse
Affiliation(s)
- Qing Wang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky
| | - Hsin-Sheng Yang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky
- Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
14
|
Cleveland KH, Yeung S, Huang KM, Liang S, Andresen BT, Huang Y. Phosphoproteome profiling provides insight into the mechanism of action for carvedilol-mediated cancer prevention. Mol Carcinog 2018; 57:997-1007. [PMID: 29626349 DOI: 10.1002/mc.22820] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 03/16/2018] [Accepted: 04/04/2018] [Indexed: 12/18/2022]
Abstract
Recent studies suggest that the β-blocker drug carvedilol prevents skin carcinogenesis but the mechanism is unknown. Carvedilol is one of a few β-blockers identified as biased agonist based on an ability to promote β-arrestin-mediated processes such as ERK phosphorylation. To understand the role of phosphoproteomic signaling in carvedilol's anticancer activity, the mouse epidermal JB6 P+ cells treated with EGF, carvedilol, or their combination were analyzed using the Phospho Explorer Antibody Array containing 1318 site-specific and phospho-specific antibodies of over 30 signaling pathways. The array data indicated that both EGF and carvedilol increased phosphorylation of ERK's cytosolic target P70S6 K while its nuclear target ELK-1 were activated only by EGF; Furthermore, EGF-induced phosphorylation of ELK-1 and c-Jun was attenuated by carvedilol. Subcellular fractionation analysis indicated that ERK nuclear translocation induced by EGF was blocked by co-treatment with carvedilol. Western blot and luciferase reporter assays confirmed that the biased β-blockers carvedilol and alprenolol blocked EGF-induced phosphorylation and activation of c-Jun/AP-1 and ELK-1. Consistently, both carvedilol and alprenolol strongly prevented EGF-induced neoplastic transformation of JB6 P+ cells. Remarkably, oral carvedilol treatment significantly inhibited the growth of A375 melanoma xenograft in SCID mice. As nuclear translocation of ERK is a key step in carcinogenesis, inhibition of this event is proposed as a novel anticancer mechanism for biased β-blockers such as carvedilol.
Collapse
Affiliation(s)
- Kristan H Cleveland
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California
| | - Steven Yeung
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California
| | - Kevin M Huang
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California
| | - Sherry Liang
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California
| | - Bradley T Andresen
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California
| | - Ying Huang
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California
| |
Collapse
|
15
|
XIAP RING domain mediates miR-4295 expression and subsequently inhibiting p63α protein translation and promoting transformation of bladder epithelial cells. Oncotarget 2018; 7:56540-56557. [PMID: 27447744 PMCID: PMC5302933 DOI: 10.18632/oncotarget.10645] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 06/13/2016] [Indexed: 12/12/2022] Open
Abstract
The X-linked inhibitor of apoptosis protein (XIAP) contains three N-terminal BIR domains that mediate anti-apoptosis and one C-terminal RING finger domain whose function(s) are not fully defined. Here we show that the RING domain of XIAP strongly inhibits the expression of p63α, a known tumor suppressor. XIAP knockdown in urothelial cells or RING deletion in knockin mice markedly upregulates p63α expression. This RING-mediated p63α downregulation is critical for the malignant transformation of normal urothelial cells following EGF treatment. We further show that the RING domain promotes Sp1-mediated transcription of miR-4295 which targets the 3′UTR of p63α mRNA and consequently inhibits p63α translation. Our results reveal a previously unknown function of the RING of XIAP in promoting miR-4295 transcription, thereby reducing p63α translation and enhancing urothelial transformation. Our data offer novel insights into the multifunctional effects of the XIAP RING domain on urothelial tumorigenesis and the potential for targeting this frequently overexpressed protein as a therapeutic alternative.
Collapse
|
16
|
Bai F, Liu K, Li H, Wang J, Zhu J, Hao P, Zhu L, Zhang S, Shan L, Ma W, Bode AM, Zhang W, Li H, Dong Z. Veratramine modulates AP-1-dependent gene transcription by directly binding to programmable DNA. Nucleic Acids Res 2018; 46:546-557. [PMID: 29237043 PMCID: PMC5778533 DOI: 10.1093/nar/gkx1241] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 11/27/2017] [Accepted: 11/30/2017] [Indexed: 12/20/2022] Open
Abstract
Because the transcription factor activator protein-1 (AP-1) regulates a variety of protein-encoding genes, it is a participant in many cellular functions, including proliferation, transformation, epithelial mesenchymal transition (EMT), and apoptosis. Inhibitors targeting AP-1 have potential use in the treatment of cancer and other inflammatory diseases. Here, we identify veratramine as a potent natural modulator of AP-1, which selectively binds to a specific site (TRE 5'-TGACTCA-3') of the AP-1 target DNA sequence and regulates AP-1-dependent gene transcription without interfering with cystosolic signaling cascades that might lead to AP-1 activation. Moreover, RNA-seq experiments demonstrate that veratramine does not act on the Hedgehog signaling pathway in contrast to its analogue, cyclopamine, and likely does not harbor the same teratogenicity and toxicity. Additionally, veratramine effectively suppresses EGF-induced AP-1 transactivation and transformation of JB6 P+ cells. Finally, we demonstrate that veratramine inhibits solar-ultraviolet-induced AP-1 activation in mice. The identification of veratramine and new findings in its specific regulation of AP-1 down stream genes pave ways to discovering and designing regulators to regulate transcription factor.
Collapse
Affiliation(s)
- Fang Bai
- Faculty of Chemical, Environmental, and Biological Science and Technology, Dalian University of Technology, Dalian 116023, China
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005, USA
| | - Kangdong Liu
- Department of Pathophysiology, Basic Medical College, Zhengzhou University, No. 100 Science Road, Zhengzhou, Henan 450001, China
- Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan 450001, China
- The Hormel Institute, University of Minnesota, Austin, MN, USA
- China-US (Henan) Hormel Cancer Institute, No.127 Dongmin Road, Zhengzhou 450008, China
| | - Huiliang Li
- Department of Natural Product Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Jiawei Wang
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Junsheng Zhu
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
- China-US (Henan) Hormel Cancer Institute, No.127 Dongmin Road, Zhengzhou 450008, China
| | - Pei Hao
- Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Lili Zhu
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Shoude Zhang
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Lei Shan
- Department of Natural Product Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Weiya Ma
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Weidong Zhang
- Department of Natural Product Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Honglin Li
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Zigang Dong
- The Hormel Institute, University of Minnesota, Austin, MN, USA
- China-US (Henan) Hormel Cancer Institute, No.127 Dongmin Road, Zhengzhou 450008, China
| |
Collapse
|
17
|
Truong VL, Kong AN, Jeong WS. Red Ginseng Oil Inhibits TPA-Induced Transformation of Skin Epidermal JB6 Cells. J Med Food 2017; 21:380-389. [PMID: 29271701 DOI: 10.1089/jmf.2017.4082] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Red ginseng oil (RGO) has been shown to possess anti-inflammatory and hepatoprotective activity. In this study, we evaluated the inhibitory effect of RGO on 12-O-tetradecanoylphorbol-13-acetate (TPA)-stimulated neoplastic transformation of JB6 P+ cells. RGO pretreatment abolished the transformation of JB6 P+ cells challenged by TPA. RGO suppressed the transactivation of activator protein-1 (AP-1) and nuclear factor kappa B (NF-κB) transcription factors as well as protein levels of cyclooxygenase-2, cyclin D1, cyclin E, and Bcl-2 in the TPA-treated cells. Additionally, TPA-induced phosphorylations of extracellular signal-regulated kinases, 90 kDa ribosomal S6 kinase 2, c-Jun N-terminal kinases, and glycogen synthase kinase 3β were downregulated in the presence of RGO. Furthermore, RGO induced the nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated antioxidant enzyme heme oxygenase-1 (HO-1) expression, and effectively blocked the overproduction of TPA-induced reactive oxygen species. These results suggest that RGO exerts a potent chemopreventive activity in skin cell model.
Collapse
Affiliation(s)
- Van-Long Truong
- 1 Department of Food and Life Sciences, College of BNIT, Inje University , Gimhae, Korea
| | - Ah Ng Kong
- 2 Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey , Piscataway, New Jersey, USA
| | - Woo-Sik Jeong
- 1 Department of Food and Life Sciences, College of BNIT, Inje University , Gimhae, Korea
| |
Collapse
|
18
|
Chakraborty D, Benham V, Jdanov V, Bullard B, Leal AS, Liby KT, Bernard JJ. A BET Bromodomain Inhibitor Suppresses Adiposity-Associated Malignant Transformation. Cancer Prev Res (Phila) 2017; 11:129-142. [PMID: 29246955 DOI: 10.1158/1940-6207.capr-17-0262] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 10/24/2017] [Accepted: 12/07/2017] [Indexed: 11/16/2022]
Abstract
Almost half a million of all new cancers have been attributed to obesity and epidemiologic evidence implicates visceral adipose tissue (VAT) and high-fat diets (HFD) in increasing cancer risk. We demonstrated that VAT-derived fibroblast growth factor 2 (FGF2) from mice fed an HFD or obese individuals stimulates the malignant transformation of epithelial cells. Mechanism-based strategies to prevent this VAT-enhanced tumorigenesis have not been explored. Clinical studies have indicated that bromodomain inhibitors have considerable potential as therapeutic agents for cancer by inhibiting the activity of several oncogenes, including c-Myc; however, their chemopreventive activity is unknown. We show herein that mice with visceral adiposity have elevated nuclear c-Myc expression in their epidermis. We hypothesized that the bromodomain inhibitor I-BET-762 (I-BET) would have efficacy in the prevention of malignant transformation by VAT and FGF2. We tested this hypothesis using our novel models of VAT-stimulated transformation in vitro and FGF2- stimulated tumor formation in vivo We found that I-BET significantly attenuates VAT and FGF2-stimulated transformation and inhibits VAT-induced c-Myc protein expression in several skin and breast epithelial cell lines. Moreover, I-BET attenuated tumor growth significantly in FGF2-treated nude mice. Work is ongoing to determine the role of visceral adiposity in c-Myc activity in several tissues and determine the inhibitory effect of I-BET on VAT-promoted tumors in vivoCancer Prev Res; 11(3); 129-42. ©2017 AACRSee related editorial by Berger and Scacheri, p. 125.
Collapse
Affiliation(s)
- Debrup Chakraborty
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Vanessa Benham
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Vladislav Jdanov
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Blair Bullard
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Ana S Leal
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Karen T Liby
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Jamie J Bernard
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan.
| |
Collapse
|
19
|
Chakraborty D, Benham V, Bullard B, Kearney T, Hsia HC, Gibbon D, Demireva EY, Lunt SY, Bernard JJ. Fibroblast growth factor receptor is a mechanistic link between visceral adiposity and cancer. Oncogene 2017; 36:6668-6679. [PMID: 28783178 PMCID: PMC5709202 DOI: 10.1038/onc.2017.278] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 05/19/2017] [Accepted: 07/03/2017] [Indexed: 02/07/2023]
Abstract
Epidemiological evidence implicates excess adipose tissue in increasing cancer risk. Despite a steeply rising global prevalence of obesity, how adiposity contributes to transformation (stage a non-tumorigenic cell undergoes to become malignant) is unknown. To determine the factors in adipose tissue that stimulate transformation, we used a novel ex vivo system of visceral adipose tissue (VAT)-condition medium-stimulated epithelial cell growth in soft agar. To extend this system in vivo, we used a murine lipectomy model of ultraviolet light B-induced, VAT-promoted skin tumor formation. We found that VAT from mice and obese human donors stimulated growth in soft agar of non-tumorigenic epithelial cells. The difference in VAT activity was associated with fibroblast growth factor-2 (FGF2) levels. Moreover, human and mouse VAT failed to stimulate growth in soft of agar in cells deficient in FGFR-1 (FGF2 receptor). We also demonstrated that circulating levels of FGF2 were associated with non-melanoma tumor formation in vivo. These data implicate FGF2 as a major factor VAT releases to transform epithelial cells-a novel, potential pathway of VAT-enhanced tumorigenesis. Strategies designed to deplete VAT stores of FGF2 or inhibit FGFR-1 in abdominally obese individuals may be important cancer prevention strategies as well as adjuvant therapies for improving outcomes.
Collapse
Affiliation(s)
- D Chakraborty
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - V Benham
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - B Bullard
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - T Kearney
- Division of Surgical Oncology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - H C Hsia
- Section of Plastic Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - D Gibbon
- Summit Medical4 Group, Livingston, NJ, USA
| | - E Y Demireva
- Office for the Vice President for Research and Graduate Studies, Michigan State University, East Lansing, MI, USA
| | - S Y Lunt
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - J J Bernard
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
20
|
Mishra DK, Kim MP. SR 11302, an AP-1 Inhibitor, Reduces Metastatic Lesion Formation in Ex Vivo 4D Lung Cancer Model. CANCER MICROENVIRONMENT 2017; 10:95-103. [PMID: 29177791 PMCID: PMC5750205 DOI: 10.1007/s12307-017-0202-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 10/31/2017] [Indexed: 01/11/2023]
Abstract
Activator protein (AP) -1 is a transcription factor, plays important role in cell differentiation, proliferation and apoptosis. Analysis of tumor cells grown on ex vivo 4D lung cancer model shows increase in components of AP-1, c-Fos and c-Jun in circulating tumor cells (CTC) compared to primary tumor. Our aim was to determine whether the AP-1 inhibitor SR11302 reduces metastatic lesion formation in the 4D model. Human lung cancer cell lines A549, H1299, and H460 were grown in the 4D model and treated with SR11302 (1 μM). We compared the number of cells in the metastatic site upon SR11302 treatment and number of viable CTCs isolated from the 4D model with parental cells treated/untreated with SR11302 on a petri dish. There were significantly fewer tumor cells per high-power field on metastatic site in 4D model seeded with H460 (p = 0.009), A549 (p = 0.01), or H1299 (p = 0.02) cells treated with SR11302. Furthermore, the CTCs from SR11302 treated 4D models, seeded with H460 (p = 0.04), A549 (p = 0.008), or H1299 (p = 0.01) cells had significantly fewer viable tumor cells after 4 days in culture than the respective untreated control. However, the SR11302 had no impact on the viability of parental H460 (p = 0.87), A549 (p = 0.93), or H1299 (p = 0.25) cells grown on a petri dish (2D). SR11302 reduces metastatic lesion formation in the ex vivo 4D lung cancer model due to the presence of an independent yet common pathway among three cell lines. The ex vivo 4D model may provide a tool to better understand the complex process of metastasis.
Collapse
Affiliation(s)
- Dhruva Kumar Mishra
- Department of Surgery, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA.
| | - Min P Kim
- Department of Surgery, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA
- Department of Surgery, Weill Cornell Medical College, Houston Methodist Hospital, Houston, TX, 77030, USA
| |
Collapse
|
21
|
Kuzmich AS, Khomenko TM, Fedorov SN, Makarieva TN, Shubina LK, Komarova NI, Korchagina DV, Rybalova TV, Volcho KP, Salakhutdinov NF. Cytotoxic and cancer preventive activity of benzotrithioles and benzotrithiole oxides, synthetic analogues of varacins. Med Chem Res 2016. [DOI: 10.1007/s00044-016-1759-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
22
|
Guanidine Alkaloids from the Marine Sponge Monanchora pulchra Show Cytotoxic Properties and Prevent EGF-Induced Neoplastic Transformation in Vitro. Mar Drugs 2016; 14:md14070133. [PMID: 27428983 PMCID: PMC4962023 DOI: 10.3390/md14070133] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 07/05/2016] [Accepted: 07/08/2016] [Indexed: 12/13/2022] Open
Abstract
Guanidine alkaloids from sponges Monanchora spp. represent diverse bioactive compounds, however, the mechanisms underlying bioactivity are very poorly understood. Here, we report results of studies on cytotoxic action, the ability to inhibit EGF-induced neoplastic transformation, and the effects on MAPK/AP-1 signaling of eight rare guanidine alkaloids, recently isolated from the marine sponge Monanchora pulchra, namely: monanchocidin A (1), monanchocidin B (2), monanchomycalin C (3), ptilomycalin A (4), monanchomycalin B (5), normonanchocidin D (6), urupocidin A (7), and pulchranin A (8). All of the compounds induced cell cycle arrest (apart from 8) and programmed death of cancer cells. Ptilomycalin A-like compounds 1–6 activated JNK1/2 and ERK1/2, following AP-1 activation and caused p53-independent programmed cell death. Compound 7 induced p53-independent cell death without activation of AP-1 or caspase-3/7, and the observed JNK1/2 activation did not contribute to the cytotoxic effect of the compound. Alkaloid 8 induced JNK1/2 (but not ERK1/2) activation leading to p53-independent cell death and strong suppression of AP-1 activity. Alkaloids 1–4, 7, and 8 were able to inhibit the EGF-induced neoplastic transformation of JB6 P+ Cl41 cells. Our results suggest that investigated guanidine marine alkaloids hold potential to eliminate human cancer cells and prevent cancer cell formation and spreading.
Collapse
|
23
|
Mohd Yusof YA. Gingerol and Its Role in Chronic Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 929:177-207. [PMID: 27771925 DOI: 10.1007/978-3-319-41342-6_8] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Since antiquity, ginger or Zingiber officinale, has been used by humans for medicinal purposes and as spice condiments to enhance flavor in cooking. Ginger contains many phenolic compounds such as gingerol, shogaol and paradol that exhibit antioxidant, anti-tumor and anti-inflammatory properties. The role of ginger and its constituents in ameliorating diseases has been the focus of study in the past two decades by many researchers who provide strong scientific evidence of its health benefit. This review discusses research findings and works devoted to gingerols, the major pungent constituent of ginger, in modulating and targeting signaling pathways with subsequent changes that ameliorate, reverse or prevent chronic diseases in human studies and animal models. The physical, chemical and biological properties of gingerols are also described. The use of ginger and especially gingerols as medicinal food derivative appears to be safe in treating or preventing chronic diseases which will benefit the common population, clinicians, patients, researchers, students and industrialists.
Collapse
Affiliation(s)
- Yasmin Anum Mohd Yusof
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Level 17, Pre-Clinical Building, Jalan Yaacob Latif, Bandar Tun Razak, 56000, Cheras, Kuala Lumpur, Malaysia.
| |
Collapse
|
24
|
Lei M, Lai X, Bai X, Qiu W, Yang T, Liao X, Chuong CM, Yang L, Lian X, Zhong JL. Prolonged overexpression of Wnt10b induces epidermal keratinocyte transformation through activating EGF pathway. Histochem Cell Biol 2015; 144:209-21. [PMID: 25995040 DOI: 10.1007/s00418-015-1330-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2015] [Indexed: 01/25/2023]
Abstract
Wnt10b is a signaling protein regulating skin development and homeostasis, and the expression of Wnt10b is restricted to epidermal keratinocytes in embryonic and postnatal skin. Recent studies indicate an elevated expression of Wnt10b in skin tumors. However, how Wnt10b regulates skin tumorigenesis remains largely unknown. Here we report that continuous expression of Wnt10b mediates transformation of epidermal keratinocytes through activating genes involved in EGF/MAPK signaling pathways. We first established a prolonged Wnt10b overexpression system in JB6P- cells to represent the elevated Wnt10b expression level in skin keratinocytes. Through expression assays and observations under phase-contrast microscopy, prolonged expression of Wnt10b activated Wnt/β-catenin pathway and induced morphological changes of cells showing longer protrusions and multilayer growth, indicating early-stage cell transformation. Wnt10b also increased cellular proliferation and migration according to BrdU incorporation and cell mobility assays. Furthermore, multi-doses of AdWnt10b treatment to JB6P- cells induced colony formation, stronger invasive ability in transwell system, and anchorage-independent growth in agar gel. In molecular level, AdWnt10b treatment induced increased transcriptional expressions of Egf, downstream Mapk pathway factors, and MMPs. Administration of Wnt antagonist DKK1 blocked the tumor promotion process induced by Wnt10b. Taken together, these findings clearly demonstrate that Wnt10b promotes epidermal keratinocyte transformation through induced Egf pathway.
Collapse
Affiliation(s)
- Mingxing Lei
- Department of Cell Biology, the Third Military Medical University, Chongqing, 400038, People's Republic of China,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Kim HG, Shi C, Bode AM, Dong Z. p38α MAPK is required for arsenic-induced cell transformation. Mol Carcinog 2015; 55:910-7. [PMID: 25969347 DOI: 10.1002/mc.22331] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 03/18/2015] [Accepted: 04/02/2015] [Indexed: 12/19/2022]
Abstract
Arsenic exposure has been reported to cause neoplastic transformation through the activation of PcG proteins. In the present study, we show that activation of p38α mitogen-activated protein kinase (MAPK) is required for arsenic-induced neoplastic transformation. Exposure of cells to 0.5 μM arsenic increased CRE and c-Fos promoter activities that were accompanied by increases in p38α MAPK and CREB phosphorylation and expression levels concurrently with AP-1 activation. Introduction of short hairpin (sh) RNA-p38α into BALB/c 3T3 cells markedly suppressed arsenic-induced colony formation compared with wildtype cells. CREB phosphorylation and AP-1 activation were decreased in p38α knockdown cells after arsenic treatment. Arsenic-induced AP-1 activation, measured as c-Fos and CRE promoter activities, and CREB phosphorylation were attenuated by p38 inhibition in BALB/c 3T3 cells. Thus, p38α MAPK activation is required for arsenic-induced neoplastic transformation mediated through CREB phosphorylation and AP-1 activation.
Collapse
Affiliation(s)
- Hong-Gyum Kim
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Chengcheng Shi
- The Hormel Institute, University of Minnesota, Austin, Minnesota.,The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Zigang Dong
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| |
Collapse
|
26
|
Li H, Zhu F, Boardman LA, Wang L, Oi N, Liu K, Li X, Fu Y, Limburg PJ, Bode AM, Dong Z. Aspirin Prevents Colorectal Cancer by Normalizing EGFR Expression. EBioMedicine 2015; 2:447-455. [PMID: 26097892 PMCID: PMC4469241 DOI: 10.1016/j.ebiom.2015.03.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Aspirin intake reduces the risk of colorectal cancer (CRC), but the molecular underpinnings remain elusive. Epidermal growth factor receptor (EGFR), which is overexpressed in about 80% of CRC cases, is implicated in the etiology of CRC. Here, we investigated whether aspirin can prevent CRC by normalizing EGFR expression. METHODS Immunohistochemistry staining was performed on paraffin-embedded tissue sections from normal colon mucosa, adenomatous polyps from FAP patients who were classified as regular aspirin users or nonusers. The interplay between cyclooxygenase-2 (COX-2) and EGFR was studied in primary intestinal epithelial cells isolated from ApcMin mice, immortalized normal human colon epithelial cells (HCEC) as well as murine embryonic fibroblasts (MEFs). RESULTS Immunohistochemistry staining results established that EGFR overexpression is an early event in colorectal tumorigenesis, which can be greatly attenuated by regular use of aspirin. Importantly, EGFR and COX-2 were co-overexpressed and co-localized with each other in FAP patients. Further mechanistic studies revealed that COX-2 overexpression triggers the activation of the c-Jun-dependent transcription factor, activator protein-1 (AP-1), which binds to the Egfr promoter. Binding facilitates the cellular accumulation of EGFR and lowers the threshold required for pre-neoplastic cells to undergo transformation. CONCLUSION Aspirin might exert its chemopreventive activity against CRC, at least partially, by normalizing EGFR expression in gastrointestinal precancerous lesions.
Collapse
Affiliation(s)
- Haitao Li
- The Hormel Institute, University of Minnesota, Austin, MN, USA ; The China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, P.R. China
| | - Feng Zhu
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Lisa A Boardman
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Lei Wang
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Naomi Oi
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Kangdong Liu
- The Hormel Institute, University of Minnesota, Austin, MN, USA ; The China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, P.R. China
| | - Xiang Li
- The Hormel Institute, University of Minnesota, Austin, MN, USA ; The China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, P.R. China
| | - Yang Fu
- The Hormel Institute, University of Minnesota, Austin, MN, USA ; The China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, P.R. China
| | | | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Zigang Dong
- The Hormel Institute, University of Minnesota, Austin, MN, USA ; The China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, P.R. China
| |
Collapse
|
27
|
Lee JH, Kim JE, Jang YJ, Lee CC, Lim TG, Jung SK, Lee E, Lim SS, Heo YS, Seo SG, Son JE, Kim JR, Lee CY, Lee HJ, Lee KW. Dehydroglyasperin C suppresses TPA-induced cell transformation through direct inhibition of MKK4 and PI3K. Mol Carcinog 2015; 55:552-62. [DOI: 10.1002/mc.22302] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Revised: 12/26/2014] [Accepted: 01/21/2015] [Indexed: 01/08/2023]
Affiliation(s)
- Ji Hoon Lee
- WCU Biomodulation Major, Department of Agricultural Biotechnology and Center for Food and Bioconvergence; Seoul National University; Seoul Republic of Korea
- Advanced Institutes of Convergence Technology; Seoul National University; Suwon Republic of Korea
| | - Jong-Eun Kim
- WCU Biomodulation Major, Department of Agricultural Biotechnology and Center for Food and Bioconvergence; Seoul National University; Seoul Republic of Korea
- Advanced Institutes of Convergence Technology; Seoul National University; Suwon Republic of Korea
- Research Institute of Bio Food Industry, Institute of Green Bio Science and Technology; Seoul National University; Pyeongchang Republic of Korea
| | - Young Jin Jang
- WCU Biomodulation Major, Department of Agricultural Biotechnology and Center for Food and Bioconvergence; Seoul National University; Seoul Republic of Korea
- Division of Creative Food Science for Health; Korea Food Research Institute; Seongnam Republic of Korea
| | - Charles C. Lee
- Department of Food Science and Technology; Cornell University; Ithaca NY 14456 USA
| | - Tae-Gyu Lim
- WCU Biomodulation Major, Department of Agricultural Biotechnology and Center for Food and Bioconvergence; Seoul National University; Seoul Republic of Korea
- Advanced Institutes of Convergence Technology; Seoul National University; Suwon Republic of Korea
| | - Sung Keun Jung
- WCU Biomodulation Major, Department of Agricultural Biotechnology and Center for Food and Bioconvergence; Seoul National University; Seoul Republic of Korea
- Division of Creative Food Science for Health; Korea Food Research Institute; Seongnam Republic of Korea
| | - Eunjung Lee
- WCU Biomodulation Major, Department of Agricultural Biotechnology and Center for Food and Bioconvergence; Seoul National University; Seoul Republic of Korea
- Traditional Alcoholic Beverage Research Team; Korea Food Research Institute; Seongnam Republic of Korea
| | - Soon Sung Lim
- Department of Food Science and Nutrition; Hallym University; Chuncheon Republic of Korea
| | - Yong Seok Heo
- Department of Chemistry; Konkuk University; Seoul Republic of Korea
| | - Sang Gwon Seo
- WCU Biomodulation Major, Department of Agricultural Biotechnology and Center for Food and Bioconvergence; Seoul National University; Seoul Republic of Korea
- Advanced Institutes of Convergence Technology; Seoul National University; Suwon Republic of Korea
| | - Joe Eun Son
- WCU Biomodulation Major, Department of Agricultural Biotechnology and Center for Food and Bioconvergence; Seoul National University; Seoul Republic of Korea
- Advanced Institutes of Convergence Technology; Seoul National University; Suwon Republic of Korea
| | - Jong Rhan Kim
- WCU Biomodulation Major, Department of Agricultural Biotechnology and Center for Food and Bioconvergence; Seoul National University; Seoul Republic of Korea
- Advanced Institutes of Convergence Technology; Seoul National University; Suwon Republic of Korea
| | - Chang Yong Lee
- Department of Food Science and Technology; Cornell University; Ithaca NY 14456 USA
- Department of Biochemistry; King Abdulaziz University; Jeddah SA
| | - Hyong Joo Lee
- Research Institute of Bio Food Industry, Institute of Green Bio Science and Technology; Seoul National University; Pyeongchang Republic of Korea
| | - Ki Won Lee
- WCU Biomodulation Major, Department of Agricultural Biotechnology and Center for Food and Bioconvergence; Seoul National University; Seoul Republic of Korea
- Advanced Institutes of Convergence Technology; Seoul National University; Suwon Republic of Korea
- Research Institute of Bio Food Industry, Institute of Green Bio Science and Technology; Seoul National University; Pyeongchang Republic of Korea
- Institute on Aging; Seoul National University; Seoul Republic of Korea
| |
Collapse
|
28
|
Zhuravleva OI, Sobolevskaya MP, Afiyatullov SS, Kirichuk NN, Denisenko VA, Dmitrenok PS, Yurchenko EA, Dyshlovoy SA. Sargassopenillines A-G, 6,6-spiroketals from the alga-derived fungi Penicillium thomii and Penicillium lividum. Mar Drugs 2014; 12:5930-43. [PMID: 25501795 PMCID: PMC4278210 DOI: 10.3390/md12125930] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 11/21/2014] [Accepted: 12/02/2014] [Indexed: 12/22/2022] Open
Abstract
Seven new 6,6-spiroketals, sargassopenillines A-G (1-7) were isolated from the alga-derived fungi Penicillium thomii KMM 4645 and Penicillium lividum KMM 4663. The structures of these metabolites were determined by HR-MS and 1D and 2D NMR. The absolute configurations of compounds 1, 5 and 6 were assigned by the modified Mosher's method and by CD data. Sargassopenilline C (3) inhibited the transcriptional activity of the oncogenic nuclear factor AP-1 with an IC50 value of 15 µM.
Collapse
Affiliation(s)
- Olesya I Zhuravleva
- Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Sciences, Prospect 100-let Vladivostoku 159, Vladivostok 690022, Russia.
| | - Maria P Sobolevskaya
- Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Sciences, Prospect 100-let Vladivostoku 159, Vladivostok 690022, Russia.
| | - Shamil Sh Afiyatullov
- Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Sciences, Prospect 100-let Vladivostoku 159, Vladivostok 690022, Russia.
| | - Natalya N Kirichuk
- Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Sciences, Prospect 100-let Vladivostoku 159, Vladivostok 690022, Russia.
| | - Vladimir A Denisenko
- Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Sciences, Prospect 100-let Vladivostoku 159, Vladivostok 690022, Russia.
| | - Pavel S Dmitrenok
- Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Sciences, Prospect 100-let Vladivostoku 159, Vladivostok 690022, Russia.
| | - Ekaterina A Yurchenko
- Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Sciences, Prospect 100-let Vladivostoku 159, Vladivostok 690022, Russia.
| | - Sergey A Dyshlovoy
- Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Sciences, Prospect 100-let Vladivostoku 159, Vladivostok 690022, Russia.
| |
Collapse
|
29
|
Zhuravleva OI, Sobolevskaya MP, Leshchenko EV, Kirichuk NN, Denisenko VA, Dmitrenok PS, Dyshlovoy SA, Zakharenko AM, Kim NY, Afiyatullov SS. Meroterpenoids from the alga-derived fungi Penicillium thomii Maire and Penicillium lividum Westling. JOURNAL OF NATURAL PRODUCTS 2014; 77:1390-5. [PMID: 24852445 DOI: 10.1021/np500151b] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Ten new austalide meroterpenoids (1-10) were isolated from the alga-derived fungi Penicillium thomii KMM 4645 and Penicillium lividum KMM 4663. Their structures were elucidated by extensive spectroscopic analysis and by comparison with related known compounds. The absolute configurations of some of the metabolites were assigned by the modified Mosher's method and CD data. Compounds 1, 2, 8, and 9 were able to inhibit AP-1-dependent transcriptional activity in JB6 Cl41 cell lines at noncytotoxic concentrations. Austalides 1-5, 8, and 9 exhibited significant inhibitory activity against endo-1,3-β-D-glucanase from a crystalline stalk of the marine mollusk Pseudocardium sachalinensis.
Collapse
Affiliation(s)
- Olesya I Zhuravleva
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Sciences , Prospect 100-let Vladivostoku 159, Vladivostok 690022, Russian Federation
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Li W, Liu J, Zhao Y. PKM2 inhibitor shikonin suppresses TPA-induced mitochondrial malfunction and proliferation of skin epidermal JB6 cells. Mol Carcinog 2014; 53:403-12. [PMID: 23255458 PMCID: PMC4827433 DOI: 10.1002/mc.21988] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 11/09/2012] [Accepted: 11/09/2012] [Indexed: 11/10/2022]
Abstract
Chemoprevention has been a pivotal and effective strategy during the skin cancer treatment. Using human skin normal and tumor samples, we demonstrated that both the expression and activity levels of pyruvate kinase M2 (PKM2) were higher in skin tumor tissues than normal tissues, suggesting that PKM2, one of important metabolic enzyme, might serve as a target for skin cancer prevention and/or therapy. Shikonin, a small-molecule active chemical, has been studied as an anti-cancer drug candidate in human cancer models. However, the mechanism of action and the chemopreventive potential of shikonin are unclear. Herein, we used the skin epidermal JB6 P+ cells and demonstrated that shikonin suppressed the tumor promoter 12-O-tetradecanoylphorbol 13-acetate (TPA) induced neoplastic cell transformation and PKM2 activation in the early stage of carcinogenesis. Mitochondrial functions were inhibited by TPA treatment, as indicated by reduced mitochondrial membrane potential and mitochondrial respiration, which were restored by shikonin. We also examined the levels of lactate as a glycolysis marker, and shikonin suppressed its increase caused by tumor promoter treatment. Modulation of cell metabolism by shikonin was associated with G2-M phase accumulation, and Fra-1 (a major subunit of activator protein 1 in skin tumorigenesis) downregulation. In addition, we demonstrated that AMP-activated protein kinase (AMPK), an energy sensor, which is inactivated by TPA, shikonin could reverse AMPK activity. These results suggest that shikonin bears chemopreventive potential for human skin cancers in which PKM2 is upregulated, which might be mediated by inhibiting oncogenic activation, PKM2 activation, and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Wenjuan Li
- Department of Pharmacology, Toxicology & Neuroscience, LSU Health Sciences Center in Shreveport, Shreveport, Louisiana
| | | | | |
Collapse
|
31
|
Li W, Liu J, Jackson K, Shi R, Zhao Y. Sensitizing the therapeutic efficacy of taxol with shikonin in human breast cancer cells. PLoS One 2014; 9:e94079. [PMID: 24710512 PMCID: PMC3977981 DOI: 10.1371/journal.pone.0094079] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 03/11/2014] [Indexed: 12/03/2022] Open
Abstract
Shikonin, a small-molecule natural product which inhibits the activity of pyruvate kinase M2 (PKM2), has been studied as an anti-cancer drug candidate in human cancer models. Here, our results demonstrate that shikonin is able to sensitize human breast cancer cells to chemotherapy by paclitaxel (taxol). Human breast adenocarcinoma MBA-MD-231 cells, which have higher levels of PKM2 expression and activity compared with MCF-7 cells, were selected to study further. The concentrations of shikonin and taxol were first selected at which they did not significantly induce cytotoxicity when treated alone, whereas the combination induced apoptosis. Surprisingly, PKM2 activity was decreased by shikonin, but not by the combination treatment. To identify the potential targets of this combination, human phospho-kinase antibody array analysis was performed and results indicated that the combination treatment inhibited the activation of ERK, Akt, and p70S6 kinases, which are known to contribute to breast cancer progression. Finally, how the combination affects breast cancer cell growth in vivo was tested using a xenograft tumor model. The results indicated that shikonin plus taxol prolonged animal survival and reduced tumor size than the vehicle treatment group. In summary, our results suggest that shikonin has a potential as an adjuvant for breast cancer therapy.
Collapse
Affiliation(s)
- Wenjuan Li
- Department of Pharmacology, Toxicology & Neuroscience, LSU Health Sciences Center in Shreveport, Shreveport, Los Angeles, United States of America
- School of Basic Medicine, Hebei University, Baoding, Hebei, China
| | - Joan Liu
- Department of Pharmacology, Toxicology & Neuroscience, LSU Health Sciences Center in Shreveport, Shreveport, Los Angeles, United States of America
| | - Kasey Jackson
- Department of Pharmacology, Toxicology & Neuroscience, LSU Health Sciences Center in Shreveport, Shreveport, Los Angeles, United States of America
| | - Runhua Shi
- Feist-Weiller Cancer Center, LSU Health Sciences Center in Shreveport, Shreveport, Los Angeles, United States of America
| | - Yunfeng Zhao
- Department of Pharmacology, Toxicology & Neuroscience, LSU Health Sciences Center in Shreveport, Shreveport, Los Angeles, United States of America
- * E-mail:
| |
Collapse
|
32
|
Magaye R, Zhou Q, Bowman L, Zou B, Mao G, Xu J, Castranova V, Zhao J, Ding M. Metallic nickel nanoparticles may exhibit higher carcinogenic potential than fine particles in JB6 cells. PLoS One 2014; 9:e92418. [PMID: 24691273 PMCID: PMC3972196 DOI: 10.1371/journal.pone.0092418] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 02/21/2014] [Indexed: 01/20/2023] Open
Abstract
While numerous studies have described the pathogenic and carcinogenic effects of nickel compounds, little has been done on the biological effects of metallic nickel. Moreover, the carcinogenetic potential of metallic nickel nanoparticles is unknown. Activator protein-1 (AP-1) and nuclear factor-κB (NF-κB) have been shown to play pivotal roles in tumor initiation, promotion, and progression. Mutation of the p53 tumor suppressor gene is considered to be one of the steps leading to the neoplastic state. The present study examines effects of metallic nickel fine and nanoparticles on tumor promoter or suppressor gene expressions as well as on cell transformation in JB6 cells. Our results demonstrate that metallic nickel nanoparticles caused higher activation of AP-1 and NF-κB, and a greater decrease of p53 transcription activity than fine particles. Western blot indicates that metallic nickel nanoparticles induced a higher level of protein expressions for R-Ras, c-myc, C-Jun, p65, and p50 in a time-dependent manner. In addition, both metallic nickel nano- and fine particles increased anchorage-independent colony formation in JB6 P+ cells in the soft agar assay. These results imply that metallic nickel fine and nanoparticles are both carcinogenetic in vitro in JB6 cells. Moreover, metallic nickel nanoparticles may exhibit higher carcinogenic potential, which suggests that precautionary measures should be taken in the use of nickel nanoparticles or its compounds in nanomedicine.
Collapse
Affiliation(s)
- Ruth Magaye
- Public Health Department of Medical School, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Ningbo University, Ningbo, Zhejiang Province, People's Republic of China
| | - Qi Zhou
- Public Health Department of Medical School, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Ningbo University, Ningbo, Zhejiang Province, People's Republic of China
| | - Linda Bowman
- Pathology and Physiology Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia, United States of America
| | - Baobo Zou
- Public Health Department of Medical School, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Ningbo University, Ningbo, Zhejiang Province, People's Republic of China
| | - Guochuan Mao
- Public Health Department of Medical School, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Ningbo University, Ningbo, Zhejiang Province, People's Republic of China
| | - Jin Xu
- Public Health Department of Medical School, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Ningbo University, Ningbo, Zhejiang Province, People's Republic of China
| | - Vincent Castranova
- Pathology and Physiology Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia, United States of America
| | - Jinshun Zhao
- Public Health Department of Medical School, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Ningbo University, Ningbo, Zhejiang Province, People's Republic of China; Pathology and Physiology Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia, United States of America
| | - Min Ding
- Pathology and Physiology Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia, United States of America
| |
Collapse
|
33
|
Li H, Zhu F, Chen H, Cheng KW, Zykova T, Oi N, Lubet RA, Bode AM, Wang M, Dong Z. 6-C-(E-phenylethenyl)-naringenin suppresses colorectal cancer growth by inhibiting cyclooxygenase-1. Cancer Res 2014; 74:243-52. [PMID: 24220240 PMCID: PMC3947334 DOI: 10.1158/0008-5472.can-13-2245] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recent clinical trials raised concerns regarding the cardiovascular toxicity of selective cyclooxygenase-2 (COX-2) inhibitors and cyclooxygenase-1 (COX-1) is now being reconsidered as a target for chemoprevention. Our aims were to determine whether selective COX-1 inhibition could delay or prevent cancer development and also clarify the underlying mechanisms. Data clearly showed that COX-1 was required for maintenance of malignant characteristics of colon cancer cells or tumor promoter-induced transformation of preneoplastic cells. We also successfully applied a ligand-docking computational method to identify a novel selective COX-1 inhibitor, 6-C-(E-phenylethenyl)-naringenin (designated herein as 6CEPN). 6CEPN could bind to COX-1 and specifically inhibited its activity both in vitro and ex vivo. In colorectal cancer cells, it potently suppressed anchorage-independent growth by inhibiting COX-1 activity. 6CEPN also effectively suppressed tumor growth in a 28-day colon cancer xenograft model without any obvious systemic toxicity. Taken together, COX-1 plays a critical role in human colorectal carcinogenesis, and this specific COX-1 inhibitor merits further investigation as a potential preventive agent against colorectal cancer.
Collapse
Affiliation(s)
- Haitao Li
- The Hormel Institute, University of Minnesota, Austin, Minnesota, United States
| | - Feng Zhu
- The Hormel Institute, University of Minnesota, Austin, Minnesota, United States
| | - Hanyong Chen
- The Hormel Institute, University of Minnesota, Austin, Minnesota, United States
| | - Ka Wing Cheng
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | - Tatyana Zykova
- The Hormel Institute, University of Minnesota, Austin, Minnesota, United States
| | - Naomi Oi
- The Hormel Institute, University of Minnesota, Austin, Minnesota, United States
| | - Ronald A. Lubet
- The National Institutes of Health, National Cancer Institute, Bethesda, MD
| | - Ann M. Bode
- The Hormel Institute, University of Minnesota, Austin, Minnesota, United States
| | - Mingfu Wang
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | - Zigang Dong
- The Hormel Institute, University of Minnesota, Austin, Minnesota, United States
| |
Collapse
|
34
|
Hsieh YH, van der Heyde H, Oh ES, Guan JL, Chang PL. Osteopontin mediates tumorigenic transformation of a preneoplastic murine cell line by suppressing anoikis: An Arg-Gly-Asp-dependent-focal adhesion kinase-caspase-8 axis. Mol Carcinog 2013; 54:379-92. [DOI: 10.1002/mc.22108] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 10/16/2013] [Accepted: 10/23/2013] [Indexed: 11/07/2022]
Affiliation(s)
- Yu-Hua Hsieh
- Department of Nutrition Sciences, 1720 2nd Avenue South; University of Alabama at Birmingham; Birmingham Alabama
| | | | - Eok-Soo Oh
- Division of Molecular Life Sciences and Center for Cell Signaling Research, Department of Life Sciences; Ewha Woman's University; Seoul Korea
| | - Jun-Lin Guan
- Division of Molecular Medicine and Genetics, Department of Internal Medicine; University of Michigan Medical School; Ann Arbor Michigan
| | - Pi-Ling Chang
- Department of Nutrition Sciences, 1720 2nd Avenue South; University of Alabama at Birmingham; Birmingham Alabama
- Department of Dermatology, 1720 2nd Avenue South; University of Alabama at Birmingham; Birmingham Alabama
- Department of Comprehensive Cancer Center, 1720 2nd Avenue South; University of Alabama at Birmingham; Birmingham Alabama
| |
Collapse
|
35
|
Arul N, Cho YY. A Rising Cancer Prevention Target of RSK2 in Human Skin Cancer. Front Oncol 2013; 3:201. [PMID: 23936765 PMCID: PMC3733026 DOI: 10.3389/fonc.2013.00201] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 07/23/2013] [Indexed: 02/06/2023] Open
Abstract
RSK2 is a p90 ribosomal S6 kinase family (p90(RSK)) member regulating cell proliferation and transformation induced by tumor promoters such as epithelial growth factor (EGF) and 12-O-tetradecanoylphorbol-13-acetate. This family of p90(RSK) has classified as a serine/threonine kinase that respond to many growth factors, peptide hormones, neurotransmitters, and environmental stresses such as ultraviolet (UV) light. Our recent study demonstrates that RSK2 plays a key role in human skin cancer development. Activation of RSK2 by EGF and UV through extracellular-activated protein kinases signaling pathway induces cell cycle progression, cell proliferation, and anchorage-independent cell transformation. Moreover, knockdown of RSK2 by si-RNA or sh-RNA abrogates cell proliferation and cell transformation of non-malignant human skin keratinocyte, and colony growth of malignant melanoma (MM) cells in soft agar. Importantly, activated and total RSK2 protein levels are highly detected in human skin cancer tissues including squamous cell carcinoma, basal-cell carcinoma, and MM. Kaempferol and eriodictyol are natural substances to inhibit kinase activity of the RSK2 N-terminal kinase domain, which is a critical kinase domain to transduce their activation signals to the substrates by phosphorylation. In this review, we discuss the role of RSK2 in skin cancer, particularly in activation of signaling pathways and potent natural substances to target RSK2 as chemopreventive and therapeutic agents.
Collapse
Affiliation(s)
- Narayanasamy Arul
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Republic of Korea
| | - Yong-Yeon Cho
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Republic of Korea
| |
Collapse
|
36
|
Bengtsson AM, Jönsson G, Magnusson C, Salim T, Axelsson C, Sjölander A. The cysteinyl leukotriene 2 receptor contributes to all-trans retinoic acid-induced differentiation of colon cancer cells. BMC Cancer 2013; 13:336. [PMID: 23829413 PMCID: PMC3710469 DOI: 10.1186/1471-2407-13-336] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 06/24/2013] [Indexed: 12/20/2022] Open
Abstract
Background Cysteinyl leukotrienes (CysLTs) are potent pro-inflammatory mediators that are increased in samples from patients with inflammatory bowel diseases (IBDs). Individuals with IBDs have enhanced susceptibility to colon carcinogenesis. In colorectal cancer, the balance between the pro-mitogenic cysteinyl leukotriene 1 receptor (CysLT1R) and the differentiation-promoting cysteinyl leukotriene 2 receptor (CysLT2R) is lost. Further, our previous data indicate that patients with high CysLT1R and low CysLT2R expression have a poor prognosis. In this study, we examined whether the balance between CysLT1R and CysLT2R could be restored by treatment with the cancer chemopreventive agent all-trans retinoic acid (ATRA). Methods To determine the effect of ATRA on CysLT2R promoter activation, mRNA level, and protein level, we performed luciferase gene reporter assays, real-time polymerase chain reactions, and Western blots in colon cancer cell lines under various conditions. Results ATRA treatment induces CysLT2R mRNA and protein expression without affecting CysLT1R levels. Experiments using siRNA and mutant cell lines indicate that the up-regulation is retinoic acid receptor (RAR) dependent. Interestingly, ATRA also up-regulates mRNA expression of leukotriene C4 synthase, the enzyme responsible for the production of the ligand for CysLT2R. Importantly, ATRA-induced differentiation of colorectal cancer cells as shown by increased expression of MUC-2 and production of alkaline phosphatase, both of which could be reduced by a CysLT2R-specific inhibitor. Conclusions This study identifies a novel mechanism of action for ATRA in colorectal cancer cell differentiation and demonstrates that retinoids can have anti-tumorigenic effects through their action on the cysteinyl leukotriene pathway.
Collapse
|
37
|
Yun HJ, Kim G, Khanal P, Kim K, Oh CH, Choi HK, Sohn H, Choi HS. Inhibitory effects of a new 1H-pyrrolo[3,2-c]pyridine derivative, KIST101029, on activator protein-1 activity and neoplastic cell transformation induced by insulin-like growth factor-1. Biol Pharm Bull 2013; 36:1466-73. [PMID: 23748899 DOI: 10.1248/bpb.b13-00244] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Diarylureas and diarylamides derivatives are reported to have antitumor activity. Encouraged by the interesting antiproliferative activity of diarylurea and diarylamide derivatives, we synthesized a new series of diarylureas and diarylamides containing pyrrolo[3,2-c]pyridine scaffold. In this study, we demonstrate that a N-(3-(4-benzamido-1H-pyrrolo[3,2-c]pyridin-1-yl)phenyl)-4-morpholino-3-(trifluoromethyl)benzamide, KIST101029, inhibits neoplastic cell transformation induced by insulin-like growth factor 1 (IGF-1) in mouse epidermal JB6 Cl41 cells. The KIST101029 compound inhibited mitogen-activated protein kinase/extracellular signal-regulated kinase kinases (MEK), c-jun N-terminal kinases (JNK), and mechanistic target of rapamycin (mTOR) signaling pathways induced by IGF-1 in JB6 Cl41 cells, resulting in the inhibition of c-fos and c-jun transcriptional activity. In addition, the KIST101029 inhibited the associated activator protein-1 (AP-1) transactivation activity and cell transformation induced by IGF-1 in JB6 Cl41 cells. Consistent with these observations, in vivo chorioallantoic membrane assay also showed that the KIST101029 inhibited IGF-1-induced tumorigenicity of JB6 Cl41 cells. Importantly, KIST101029 suppressed the colony formation of A375 cells in soft agar. Taken together, these results indicate that a KIST101029 might exert chemopreventive effects through the inhibition of phosphorylation of MAPK and mTOR signaling pathway.
Collapse
|
38
|
Morpho-mechanical intestinal remodeling in type 2 diabetic GK rats--is it related to advanced glycation end product formation? J Biomech 2013. [PMID: 23403079 DOI: 10.1016/j.] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Little is known about the mechanisms for the biomechanical remodeling in diabetes. The histomorphology, passive biomechanical properties and expression of advanced glycation end product (N epsilon-(carboxymethyl) lysine, AGE) and its receptor (RAGE) were studied in jejunal segments from 8 GK diabetic rats (GK group) and 10 age-matched normal rats (Normal group). The mechanical test was done by using a ramp distension of fluid into the jejunal segments in vitro. Circumferential stress and strain were computed from the length, diameter and pressure data and from the zero-stress state geometry. AGE and RAGE were detected by immunohistochemistry staining. Linear regression analysis was done to study association between the glucose level and AGE/RAGE expression with the histomorphometric and biomechanical parameters. The blood glucose level, the jejunal weight per length, wall thickness, wall area and layer thickness significantly increased in the GK group compared with the Normal group (P<0.05, P<0.01 and P<0.001). The opening angle and absolute values of residual strain decreased whereas the circumferential stiffness of the jejunal wall increased in the GK group (P<0.05 and P<0.01). Furthermore, stronger AGE expression in the villi and crypt and RAGE expression in the villi were found in the GK group (P<0.05 and P<0.01). Most histomorphometric and biomechanical changes were associated with blood glucose level and AGE/RAGE expression. In conclusion, histomorphometric and biomechanical remodeling occurred in type 2 diabetic GK rats. The increasing blood glucose level and the increased AGE/RAGE expression were associated with the remodeling, indicating a causal relationship.
Collapse
|
39
|
Healy S, Khan P, Davie JR. Immediate early response genes and cell transformation. Pharmacol Ther 2013; 137:64-77. [DOI: 10.1016/j.pharmthera.2012.09.001] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 09/06/2012] [Indexed: 01/20/2023]
|
40
|
The Extracts of Some Marine Invertebrates and Algae Collected off the Coast Waters of Vietnam Induce the Inhibitory Effects on the Activator Protein-1 Transcriptional Activity in JB6 Cl41 Cells. J CHEM-NY 2013. [DOI: 10.1155/2013/896709] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
It has previously been shown that inhibition of the transcriptional activity of the oncogenic nuclear factor AP-1 can result in cancer prevention. Marine invertebrates and alga are a rich source of natural compounds that possess various biological activities. The inhibitory effects of the extracts of Vietnamese marine organisms in relation to the AP-1 transcriptional activity were examined by the luciferase method using JB6 Cl41 cells stably expressing a luciferase reporter gene controlled by AP-1 DNA binding sequence. As was found, 71 species of marine sponges out of 148 species studied contain inhibitors of the AP-1 transcriptional activity. Therefore, marine organisms as a source of biologically active compounds have a great potential for isolation of the new cancer preventive compounds that inhibit the oncogenic AP-1 nuclear factor.
Collapse
|
41
|
Baek S, Kang NJ, Popowicz GM, Arciniega M, Jung SK, Byun S, Song NR, Heo YS, Kim BY, Lee HJ, Holak TA, Augustin M, Bode AM, Huber R, Dong Z, Lee KW. Structural and functional analysis of the natural JNK1 inhibitor quercetagetin. J Mol Biol 2012; 425:411-23. [PMID: 23142567 DOI: 10.1016/j.jmb.2012.10.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 10/26/2012] [Accepted: 10/29/2012] [Indexed: 01/22/2023]
Abstract
c-Jun NH2-terminal kinases (JNKs) and phosphatidylinositol 3-kinase (PI3-K) play critical roles in chronic diseases such as cancer, type II diabetes, and obesity. We describe here the binding of quercetagetin (3,3',4',5,6,7-hydroxyflavone), related flavonoids, and SP600125 to JNK1 and PI3-K by ATP-competitive and immobilized metal ion affinity-based fluorescence polarization assays and measure the effect of quercetagetin on JNK1 and PI3-K activities. Quercetagetin attenuated the phosphorylation of c-Jun and AKT, suppressed AP-1 and NF-κB promoter activities, and also reduced cell transformation. It attenuated tumor incidence and reduced tumor volumes in a two-stage skin carcinogenesis mouse model. Our crystallographic structure determination data show that quercetagetin binds to the ATP-binding site of JNK1. Notably, the interaction between Lys55, Asp169, and Glu73 of JNK1 and the catechol moiety of quercetagetin reorients the N-terminal lobe of JNK1, thereby improving compatibility of the ligand with its binding site. The results of a theoretical docking study suggest a binding mode of PI3-K with the hydroxyl groups of the catechol moiety forming hydrogen bonds with the side chains of Asp964 and Asp841 in the p110γ catalytic subunit. These interactions could contribute to the high inhibitory activity of quercetagetin against PI3-K. Our study suggests the potential use of quercetagetin in the prevention or therapy of cancer and other chronic diseases.
Collapse
Affiliation(s)
- Sohee Baek
- Max Planck Institute for Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Chao DC, Lin LJ, Hsiang CY, Li CC, Lo HY, Liang JA, Kao ST, Wu SL, Ho TY. Evodiamine inhibits 12-O-tetradecanoylphorbol-13-acetate-induced activator protein 1 transactivation and cell transformation in human hepatocytes. Phytother Res 2012; 25:1018-23. [PMID: 21246637 DOI: 10.1002/ptr.3392] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Evodia rutaecarpa has been used to treat inflammatory digestive disorders in Asian countries. However, little is known about the antitumor activities of E. rutaecarpa and its bioactive constituent evodiamine (EVO). The aim of this study was to characterize the antitumor mechanisms of E. rutaecarpa and EVO in human hepatocytes. Human Chang liver cells were transfected with activator protein 1 (AP-1)-luciferase reporter gene and designated as Chang/AP-1 cells. The Chang/AP-1 cells were treated with E. rutaecarpa and its bioactive constituents, and challenged with the AP-1 stimulator 12-O-tetradecanoylphorbol-13- acetate (TPA). The present study showed that the methanol extract of E. rutaecarpa decreased the TPA-induced AP-1 transactivation in Chang/AP-1 cells, with an EC₅₀ value of 24.72 μg/mL. EVO inhibited the TPA-induced AP-1 transactivation and colony formation, with EC₅₀ values of 82 μM and 8.2 μM, respectively. Moreover, EVO significantly diminished the TPA-induced phosphorylation of extracellular signal-regulated kinases (ERKs). These results suggested that EVO treatment suppressed the TPA-induced AP-1 activity via the ERKs pathway. In conclusion, EVO inhibited the AP-1 activity and cellular transformation in human hepatocytes, suggesting that EVO was a potential agent for antitumor therapy.
Collapse
Affiliation(s)
- De-Cheng Chao
- Graduate Institute of Chinese Medicine, China Medical University, Taichung 40402, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Yang Y, Wang H, Wang S, Xu M, Liu M, Liao M, Frank JA, Adhikari S, Bower KA, Shi X, Ma C, Luo J. GSK3β signaling is involved in ultraviolet B-induced activation of autophagy in epidermal cells. Int J Oncol 2012; 41:1782-8. [PMID: 22961228 PMCID: PMC3583618 DOI: 10.3892/ijo.2012.1620] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Accepted: 07/27/2012] [Indexed: 12/25/2022] Open
Abstract
Ultraviolet B (UVB) exposure causes damage to skin and represents the primary etiological agent for skin cancer formation. UVB induces DNA damage and apoptosis in epidermal cells. In this study, we demonstrated that UVB activated autophagy in JB6 epidermal cells, which was evident by the formation of LC3 puncta, the induction of LC3 lipidation, the increase in beclin 1 expression, and the decrease in the levels of p62. Autophagy appeared to be a protective response to UVB-induced damage because inhibition of autophagy exacerbated UVB-induced cell death, and stimulation of autophagy offered protection. Furthermore, we demonstrated that glycogen synthase kinase 3β (GSK3β) was involved in UVB-induced autophagy. UVB inhibited GSK3β activation by simultaneously enhancing phosphorylation at Ser9 and suppressing Tyr216 phosphorylation. GSK3β negatively regulated autophagy; overexpression of wild-type or S9A (constitutive-active) GSK3β mutant inhibited UVB-mediated autophagy, while overexpression of a dominant-negative K85R mutant enhanced UVB-mediated autophagy. Inhibition of GSK3β also offered protection against UVB-mediated damage. UVB activated AMP-activated protein kinase (AMPK), an important regulator of autophagy through the inhibition of GSK3β. Taken together, our results suggest that UVB-stimulated autophagy is a protective response for epidermal cells and is mediated by the GSK3β/AMPK pathway.
Collapse
Affiliation(s)
- Yang Yang
- Department of Internal Medicine, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Kang MI, Baker AR, Dextras CR, Cabarcas SM, Young MR, Colburn NH. Targeting of Noncanonical Wnt5a Signaling by AP-1 Blocker Dominant-Negative Jun When It Inhibits Skin Carcinogenesis. Genes Cancer 2012; 3:37-50. [PMID: 22893789 DOI: 10.1177/1947601912448820] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 04/25/2012] [Indexed: 11/17/2022] Open
Abstract
The transcription factor AP-1 (activator protein-1) regulates a number of genes that drive tumor promotion and progression. While basal levels of AP-1 activity are important for normal cell proliferation and cell survival, overactivated AP-1-dependent gene expression stimulates inflammation, angiogenesis, invasion, and other events that propel carcinogenesis. We seek to discover genes targeted by carcinogenesis inhibitors that do not also inhibit cell proliferation or survival. Transgenic TAM67 (dominant-negative c-Jun) inhibits mouse skin tumorigenesis and tumor progression without inhibiting cell proliferation or induced hyperproliferation. Expression profiling of wild-type and K14-TAM67 mouse epidermis has revealed a number of functionally significant genes that are induced by tumor promoters in wild-type mice but not in those expressing the AP-1 blocker. The current study now identifies Wnt5a signaling as a new target of TAM67 when it inhibits DMBA/TPA-induced carcinogenesis. Wnt5a is required to maintain the tumor phenotype in tumorigenic mouse JB6 cells and Ras-transformed human squamous carcinoma HaCaT-II4 cells, as Wnt5a knockdown suppresses anchorage-independent and tumor xenograft growth. The oncogenic Wnt5a-mediated pathway signals through activation of the protein kinase PKCα and oncogenic transcription factor STAT3 phosphorylation and not through the canonical Wnt/β-catenin pathway. Similar to Wnt5a knockdown, inhibitors of PKCα blocked STAT3 activation in both mouse JB6 and human HaCaT-II4 tumor cells. Moreover, expression of STAT3-regulated genes FAS, MMP3, IRF1, and cyclin D1 was suppressed with Wnt5a knockdown. Treatment of mouse Wnt5a knockdown cells with a PKCα-specific activator rescued phosphorylation of STAT3. Thus, Wnt5a signaling is required for maintaining the tumor phenotype in squamous carcinoma cells, Wnt5a targeting by the AP-1 blockade contributes to inhibition of skin carcinogenesis, and the signaling pathway traverses PKCα and STAT3 activation. Coordinate overactivation of Wnt5a expression and STAT3 signaling is observed in human skin and colon cancers as well as glioblastoma.
Collapse
Affiliation(s)
- Moon-Il Kang
- Laboratory of Cancer Prevention, National Cancer Institute, Frederick, MD, USA
| | | | | | | | | | | |
Collapse
|
45
|
Saikali M, Ghantous A, Halawi R, Talhouk SN, Saliba NA, Darwiche N. Sesquiterpene lactones isolated from indigenous Middle Eastern plants inhibit tumor promoter-induced transformation of JB6 cells. Altern Ther Health Med 2012; 12:89. [PMID: 22776414 PMCID: PMC3439278 DOI: 10.1186/1472-6882-12-89] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 06/19/2012] [Indexed: 12/30/2022]
Abstract
Background Sesquiterpene lactones (SL) are plant secondary metabolites that are known for their anti-fungal, anti-bacterial, anti-inflammatory, and anti-tumor properties. Considering that several SL-derived drugs are currently in cancer clinical trials, we have tested two SL molecules, 3-β-methoxy-iso-seco-tanapartholide (β-tan) isolated from Achillea falcata and salograviolide A (Sal A) isolated from Centaurea ainetensis, for their anti-tumor properties. We used the mouse epidermal JB6P + cells as a model for tumor promotion and cellular transformation. Key players that are involved in cellular transformation and tumorigenesis are the AP-1 and NF-κB transcription factors; therefore, we assessed how β-tan and Sal A modulate their signaling pathways in JB6P + cells. Methods The effects of β-tan and Sal A on the growth of normal and neoplastic keratinocytes and on the tumor promotion-responsive JB6P + cells were determined using the MTT assay. Anchorage-independent cell growth transformation assays were used to evaluate the anti-tumor promoting properties of these SL molecules in JB6P + cells and dual luciferase reporter assays and western blot analysis were used to investigate their effects on tumor promoter-induced AP-1 and NF-κB activities and protein levels of key AP-1 and NF-кB target genes. Results β-tan and Sal A selectively inhibited tumor promoter-induced cell growth and transformation of JB6P + cells at concentrations that do not affect JB6P + and primary keratinocytes basal cell growth. In addition, both molecules reduced basal and tumor promoter-induced NF-κB transcriptional activities, differentially regulated basal and tumor promoter-induced AP-1 transcriptional activities, and modulated key players of the AP-1 and NF-κB signaling pathways. Conclusions These results highlight the anti-tumor promoting properties of β-tan and Sal A. These SL molecules isolated from two plant species native to the Middle East may provide opportunities for complementary medicine practices.
Collapse
|
46
|
Peng C, Zhu F, Wen W, Yao K, Li S, Zykova T, Liu K, Li X, Ma WY, Bode AM, Dong Z. Tumor necrosis factor receptor-associated factor family protein 2 is a key mediator of the epidermal growth factor-induced ribosomal S6 kinase 2/cAMP-responsive element-binding protein/Fos protein signaling pathway. J Biol Chem 2012; 287:25881-92. [PMID: 22685297 DOI: 10.1074/jbc.m112.359521] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
TRAF2 has an important function in mediating the TNF-R signaling pathway toward activation of NF-κB and JNKs. Here we reveal a novel function of TRAF2 in the epidermal growth factor (EGF) signaling pathway. Knockdown of TRAF2 blocked EGF-induced AP-1 activity and anchorage- independent cell transformation. Notably, we showed that EGF induces ribosomal S6 kinase 2 (RSK2) ubiquitination, and knocking down TRAF2 suppresses ubiquitination of RSK2 induced by EGF. We also found that TRAF2 affects RSK2 activity through RSK2 ubiquitination. RSK2 plays a critical role in AP-1 activity mediated through CREB and c-Fos, which regulates anchorage-independent cell transformation. In addition, TRAF2 is overexpressed in colon cancer and required for colon cancer development, suggesting that TRAF2 might be a potential molecular target for cancer prevention and treatment.
Collapse
Affiliation(s)
- Cong Peng
- The Hormel Institute, University of Minnesota, Austin, Minnesota 55912, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Dyshlovoy SA, Fedorov SN, Kalinovsky AI, Shubina LK, Bokemeyer C, Stonik VA, Honecker F. Mycalamide A shows cytotoxic properties and prevents EGF-induced neoplastic transformation through inhibition of nuclear factors. Mar Drugs 2012; 10:1212-1224. [PMID: 22822368 PMCID: PMC3397435 DOI: 10.3390/md10061212] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2012] [Revised: 05/15/2012] [Accepted: 05/21/2012] [Indexed: 02/02/2023] Open
Abstract
Mycalamide A, a marine natural compound previously isolated from sponges, is known as a protein synthesis inhibitor with potent antitumor activity. However, the ability of this compound to prevent malignant transformation of cells has never been examined before. Here, for the first time, we report the isolation of mycalamide A from ascidian Polysincraton sp. as well as investigation of its cancer preventive properties. In murine JB6 Cl41 P+ cells, mycalamide A inhibited epidermal growth factor (EGF)-induced neoplastic transformation, and induced apoptosis at subnanomolar or nanomolar concentrations. The compound inhibited transcriptional activity of the oncogenic nuclear factors AP-1 and NF-κB, a potential mechanism of its cancer preventive properties. Induction of phosphorylation of the kinases MAPK p38, JNK, and ERK was also observed at high concentrations of mycalamide A. The drug shows promising potential for both cancer-prevention and cytotoxic therapy and should be further developed.
Collapse
Affiliation(s)
- Sergey A. Dyshlovoy
- Laboratory of Marine Natural Products Chemistry, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch of the Russian Academy of Sciences, Prospect 100-let Vladivostoku 159, Vladivostok 690022, Russia; (S.N.F.); (A.I.K.); (L.K.S.); (V.A.S.)
- Department of Oncology, Haematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Martinistr. 52, Hamburg 20246, Germany; (C.B.); (F.H.)
- Author to whom correspondence should be addressed; ; Tel.: +7-423-231-11-68; Fax: +7-423-231-40-50
| | - Sergey N. Fedorov
- Laboratory of Marine Natural Products Chemistry, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch of the Russian Academy of Sciences, Prospect 100-let Vladivostoku 159, Vladivostok 690022, Russia; (S.N.F.); (A.I.K.); (L.K.S.); (V.A.S.)
| | - Anatoly I. Kalinovsky
- Laboratory of Marine Natural Products Chemistry, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch of the Russian Academy of Sciences, Prospect 100-let Vladivostoku 159, Vladivostok 690022, Russia; (S.N.F.); (A.I.K.); (L.K.S.); (V.A.S.)
| | - Larisa K. Shubina
- Laboratory of Marine Natural Products Chemistry, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch of the Russian Academy of Sciences, Prospect 100-let Vladivostoku 159, Vladivostok 690022, Russia; (S.N.F.); (A.I.K.); (L.K.S.); (V.A.S.)
| | - Carsten Bokemeyer
- Department of Oncology, Haematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Martinistr. 52, Hamburg 20246, Germany; (C.B.); (F.H.)
| | - Valentin A. Stonik
- Laboratory of Marine Natural Products Chemistry, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-East Branch of the Russian Academy of Sciences, Prospect 100-let Vladivostoku 159, Vladivostok 690022, Russia; (S.N.F.); (A.I.K.); (L.K.S.); (V.A.S.)
- School of Natural Sciences, Far Eastern Federal University, Sukhanova Street, 8, Vladivostok 690091, Russia
| | - Friedemann Honecker
- Department of Oncology, Haematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Martinistr. 52, Hamburg 20246, Germany; (C.B.); (F.H.)
| |
Collapse
|
48
|
Kowalczyk P, Kowalczyk MC, Junco JJ, Tolstykh O, Kinjo T, Truong H, Walaszek Z, Hanausek M, Slaga TJ. The possible separation of 12-O-tetradecanoylphorbol-13-acetate-induced skin inflammation and hyperplasia by compound A. Mol Carcinog 2012; 52:488-96. [PMID: 22351517 DOI: 10.1002/mc.21883] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 01/13/2012] [Accepted: 01/18/2012] [Indexed: 01/08/2023]
Abstract
Activated glucocorticoid receptor (GR) acts via two different mechanisms: transcriptional regulation that requires DNA-binding, and protein-protein interaction between GR and other transcription factors, such as nuclear factor kappa B (NF-κB) or activator protein 1 (AP-1). It has been postulated that many important effects of glucocorticoids, including their anti-inflammatory properties, depend on GR's transrepressive effects on NF-κB and AP-1. In the present study, we have employed a TPA-induced model of skin inflammation and epidermal hyperplasia to determine whether partial activation of the glucocorticoid receptor by compound A (CpdA) is sufficient to reverse the effect of TPA treatment. CpdA is a nonsteroidal GR modulator with high binding affinity, is capable of partial activation of GR. Topical application of TPA twice per week for 2 wk results in inflammation and epidermal hyperplasia. TPA treatment also elevates levels of c-jun (AP-1 component), cyclooxygenase-2 (COX-2), p50 (NF-κB component), interleukin-6 (IL-6), and tumor necrosis factor (TNF) in the skin. Fluocinolone acetonide (FA) (a full GR agonist) was able to completely reverse the above effects of TPA. When applied alone, CpdA increased the epidermal thickness and keratinocyte proliferation as well as levels of c-jun, COX-2, IL-6, and IFN-γ. However, CpdA treatment resulted in a decrease in the number of p50 positive cells induced by TPA, suggesting its role in inhibition of NF-κB. The level of metallothionein-1 mRNA, regulated by GR was also significantly decreased in skin samples treated with CpdA. Our results suggest that CpdA is able to inhibit GR transactivation and activate only some transrepression properties of GR.
Collapse
Affiliation(s)
- Piotr Kowalczyk
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Knockdown of RON inhibits AP-1 activity and induces apoptosis and cell cycle arrest through the modulation of Akt/FoxO signaling in human colorectal cancer cells. Dig Dis Sci 2012; 57:371-80. [PMID: 21901254 DOI: 10.1007/s10620-011-1892-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Accepted: 08/24/2011] [Indexed: 12/30/2022]
Abstract
BACKGROUND/AIMS Altered Recepteur d'Origine nantais (RON) expression transduces signals inducting invasive growth phenotype that includes cell proliferation, migration, matrix invasion, and protection of apoptosis in human cancer cells. The aims of the current study were to evaluate whether RON affects tumor cell behavior and cellular signaling pathways including activator protein-1 (AP-1) and Akt/forkhead box O (FoxO) in human colorectal cancer cells. METHODS To study the biological role of RON on tumor cell behavior and cellular signaling pathways in human colorectal cancer, we used small interfering RNA (siRNA) to knockdown RON gene expression in human colorectal cancer cell line, DKO-1. RESULTS Knockdown of RON diminished migration, invasion, and proliferation of human colorectal cancer cells. Knockdown of RON decreased AP-1 transcriptional activity and expression of AP-1 target genes. Knockdown of RON activated cleaved caspase-3, -7, -9, and PARP, and down-regulated the expression of Mcl-1, survivin and XIAP, leading to induction of apoptosis. Knockdown of RON induced cell cycle arrest in the G2/M phase of cancer cells by an increase of p27 and a decrease of cyclin D3. Knockdown of RON inhibited the phosphorylation of Akt/FoxO signaling proteins such as Ser473 and Thr308 of Akt and FoxO1/3a. CONCLUSIONS These results indicate that knockdown of RON inhibits AP-1 activity and induces apoptosis and cell cycle arrest through the modulation of Akt/FoxO signaling in human colorectal cancer cells.
Collapse
|
50
|
Sung B, Prasad S, Yadav VR, Aggarwal BB. Cancer cell signaling pathways targeted by spice-derived nutraceuticals. Nutr Cancer 2011; 64:173-97. [PMID: 22149093 DOI: 10.1080/01635581.2012.630551] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Extensive research within the last half a century has revealed that cancer is caused by dysregulation of as many as 500 different gene products. Most natural products target multiple gene products and thus are ideally suited for prevention and treatment of various chronic diseases, including cancer. Dietary agents such as spices have been used extensively in the Eastern world for a variety of ailments for millennia, and five centuries ago they took a golden journey to the Western world. Various spice-derived nutraceuticals, including 1'-acetoxychavicol acetate, anethole, capsaicin, cardamonin, curcumin, dibenzoylmethane, diosgenin, eugenol, gambogic acid, gingerol, thymoquinone, ursolic acid, xanthohumol, and zerumbone derived from galangal, anise, red chili, black cardamom, turmeric, licorice, fenugreek, clove, kokum, ginger, black cumin, rosemary, hop, and pinecone ginger, respectively, are the focus of this review. The modulation of various transcription factors, growth factors, protein kinases, and inflammatory mediators by these spice-derived nutraceuticals are described. The anticancer potential through the modulation of various targets is also the subject of this review. Although they have always been used to improve taste and color and as a preservative, they are now also used for prevention and treatment of a wide variety of chronic inflammatory diseases, including cancer.
Collapse
Affiliation(s)
- Bokyung Sung
- Cytokine Research Laboratory, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | |
Collapse
|