1
|
Kofman K, Levin M. Bioelectric pharmacology of cancer: A systematic review of ion channel drugs affecting the cancer phenotype. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2024; 191:25-39. [PMID: 38971325 DOI: 10.1016/j.pbiomolbio.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/21/2024] [Accepted: 07/04/2024] [Indexed: 07/08/2024]
Abstract
Cancer is a pernicious and pressing medical problem; moreover, it is a failure of multicellular morphogenesis that sheds much light on evolutionary developmental biology. Numerous classes of pharmacological agents have been considered as cancer therapeutics and evaluated as potential carcinogenic agents; however, these are spread throughout the primary literature. Here, we briefly review recent work on ion channel drugs as promising anti-cancer treatments and present a systematic review of the known cancer-relevant effects of 109 drugs targeting ion channels. The roles of ion channels in cancer are consistent with the importance of bioelectrical parameters in cell regulation and with the functions of bioelectric signaling in morphogenetic signals that act as cancer suppressors. We find that compounds that are well-known for having targets in the nervous system, such as voltage-gated ion channels, ligand-gated ion channels, proton pumps, and gap junctions are especially relevant to cancer. Our review suggests further opportunities for the repurposing of numerous promising candidates in the field of cancer electroceuticals.
Collapse
Affiliation(s)
- Karina Kofman
- Faculty of Dentistry, University of Toronto, Toronto, Canada
| | - Michael Levin
- Allen Discovery Center at Tufts University, USA; Wyss Institute for Biologically Inspired Engineering at Harvard University, USA.
| |
Collapse
|
2
|
Transcriptome Analysis of the Anti-TGFβ Effect of Schisandra chinensis Fruit Extract and Schisandrin B in A7r5 Vascular Smooth Muscle Cells. Life (Basel) 2021; 11:life11020163. [PMID: 33672474 PMCID: PMC7926316 DOI: 10.3390/life11020163] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/14/2021] [Accepted: 02/17/2021] [Indexed: 12/19/2022] Open
Abstract
Schisandra chinensis fruit extract (SCE) has been used as a traditional medicine for treating vascular diseases. However, little is known about how SCE and schisandrin B (SchB) affect transcriptional output-a crucial factor for shaping the fibrotic responses of the transforming growth factor β (TGFβ) signaling pathways in in vascular smooth muscle cells (VSMC). In this study, to assess the pharmacological effect of SCE and SchB on TGFβ-induced transcriptional output, we performed DNA microarray experiments in A7r5 VSMCs. We found that TGFβ induced distinctive changes in the gene expression profile and that these changes were considerably reversed by SCE and SchB. Gene Set Enrichment Analysis (GSEA) with Hallmark signature suggested that SCE or SchB inhibits a range of fibrosis-associated biological processes, including inflammation, cell proliferation and migration. With our VSMC-specific transcriptional interactome network, master regulator analysis identified crucial transcription factors that regulate the expression of SCE- and SchB-effective genes (i.e., TGFβ-reactive genes whose expression are reversed by SCE and SchB). Our results provide novel perspective and insight into understanding the pharmacological action of SCE and SchB at the transcriptome level and will support further investigations to develop multitargeted strategies for the treatment of vascular fibrosis.
Collapse
|
3
|
Park S, Lim JM, Chun JN, Lee S, Kim TM, Kim DW, Kim SY, Bae DJ, Bae SM, So I, Kim HG, Choi JY, Jeon JH. Altered expression of fucosylation pathway genes is associated with poor prognosis and tumor metastasis in non‑small cell lung cancer. Int J Oncol 2019; 56:559-567. [PMID: 31894325 PMCID: PMC6959459 DOI: 10.3892/ijo.2019.4953] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 11/25/2019] [Indexed: 12/28/2022] Open
Abstract
Fucosylation is a post‑translational modification that attaches fucose residues to protein‑ or lipid‑bound oligosaccharides. Certain fucosylation pathway genes are aberrantly expressed in several types of cancer, including non‑small cell lung cancer (NSCLC), and this aberrant expression is associated with poor prognosis in patients with cancer. However, the molecular mechanism by which these fucosylation pathway genes promote tumor progression has not been well‑characterized. The present study analyzed public microarray data obtained from NSCLC samples. Multivariate analysis revealed that altered expression of fucosylation pathway genes, including fucosyltransferase 1 (FUT1), FUT2, FUT3, FUT6, FUT8 and GDP‑L‑fucose synthase (TSTA3), correlated with poor survival in patients with NSCLC. Inhibition of FUTs by 2F‑peracetyl‑fucose (2F‑PAF) suppressed transforming growth factor β (TGFβ)‑mediated Smad3 phosphorylation and nuclear translocation in NSCLC cells. In addition, wound‑healing and Transwell migration assays demonstrated that 2F‑PAF inhibited TGFβ‑induced NSCLC cell migration and invasion. Furthermore, in vivo bioluminescence imaging analysis revealed that 2F‑PAF attenuated the metastatic capacity of NSCLC cells. These results may help characterize the oncogenic role of fucosylation in NSCLC biology and highlight its potential for developing cancer therapeutics.
Collapse
Affiliation(s)
- Soonbum Park
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Jin-Muk Lim
- Biomedical Knowledge Engineering Laboratory, Seoul National University, Seoul 08826, Republic of Korea
| | - Jung Nyeo Chun
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Sanghoon Lee
- Department of Biochemistry, University of
Utah School of Medicine, Salt Lake City, UT 84112‑5650, USA
| | - Tae Min Kim
- Seoul National University Cancer Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Dong-Wan Kim
- Seoul National University Cancer Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Sang-Yeob Kim
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05535, Republic of Korea
| | - Dong-Jun Bae
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05535, Republic of Korea
| | - Sang-Mun Bae
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05535, Republic of Korea
| | - Insuk So
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Hong-Gee Kim
- Biomedical Knowledge Engineering Laboratory, Seoul National University, Seoul 08826, Republic of Korea
| | - Ji-Yeob Choi
- Seoul National University Cancer Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Ju-Hong Jeon
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| |
Collapse
|
4
|
Chun JN, Cho M, Park S, So I, Jeon JH. The conflicting role of E2F1 in prostate cancer: A matter of cell context or interpretational flexibility? Biochim Biophys Acta Rev Cancer 2019; 1873:188336. [PMID: 31870703 DOI: 10.1016/j.bbcan.2019.188336] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 12/20/2019] [Indexed: 02/07/2023]
Abstract
The transcription factor E2F1 plays a crucial role in mediating multiple cancer hallmark capabilities that regulate cell cycle, survival, apoptosis, metabolism, and metastasis. Aberrant activation of E2F1 is closely associated with a poor clinical outcome in various human cancers. However, E2F1 has conflictingly been reported to exert tumor suppressive activity, raising a question as to the nature of its substantive role in the control of cell fate. In this review, we summarize deregulated E2F1 activity and its role in prostate cancer. We highlight the recent advances in understanding the molecular mechanism by which E2F1 regulates the development and progression of prostate cancer, providing insight into how cell context or data interpretation shapes the role of E2F1 in prostate cancer. This review will aid in translating biomedical knowledge into therapeutic strategies for prostate cancer.
Collapse
Affiliation(s)
- Jung Nyeo Chun
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Republic of Korea
| | - Minsoo Cho
- Undergraduate Research Program, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Soonbum Park
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Insuk So
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Republic of Korea
| | - Ju-Hong Jeon
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Republic of Korea.
| |
Collapse
|
5
|
Gupta S, Silveira DA, Mombach JCM. ATM/miR‐34a‐5p axis regulates a p21‐dependent senescence‐apoptosis switch in non‐small cell lung cancer: a Boolean model of G1/S checkpoint regulation. FEBS Lett 2019; 594:227-239. [DOI: 10.1002/1873-3468.13615] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 08/16/2019] [Accepted: 09/19/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Shantanu Gupta
- Department of Physics Universidade Federal de Santa Maria Brazil
| | | | | |
Collapse
|
6
|
Park S, Kim YS, Kim DY, So I, Jeon JH. PI3K pathway in prostate cancer: All resistant roads lead to PI3K. Biochim Biophys Acta Rev Cancer 2018; 1870:198-206. [DOI: 10.1016/j.bbcan.2018.09.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 09/13/2018] [Accepted: 09/13/2018] [Indexed: 12/19/2022]
|
7
|
Schisandrol B and schisandrin B inhibit TGFβ1-mediated NF-κB activation via a Smad-independent mechanism. Oncotarget 2017; 9:3121-3130. [PMID: 29423034 PMCID: PMC5790451 DOI: 10.18632/oncotarget.23213] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 11/15/2017] [Indexed: 01/05/2023] Open
Abstract
Aberrant transforming growth factor β1 (TGFβ1) signaling plays a pathogenic role in the development of vascular fibrosis. We have reported that Schisandra chinensis fruit extract (SCE), which has been used as a traditional oriental medicine, suppresses TGFβ1-mediated phenotypes in vascular smooth muscle cells (VSMCs). However, it is still largely unknown about the pharmacologic effects of SCE on various TGFβ1 signaling components. In this study, we found that SCE attenuated TGFβ1-induced NF-κB activation and nuclear translocation in VSMCs. Among the five active ingredients of SCE that were examined, schisandrol B (SolB) and schisandrin B (SchB) most potently suppressed TGFβ1-mediated NF-κB activation. In addition, SolB and SchB effectively inhibited IKKα/β activation and IκBα phosphorylation in TGFβ1-treated VSMCs. The pharmacologic effects of SolB and SchB on NF-κB activation were independent of the Smad-mediated canonical pathway. Therefore, our study demonstrates that SCE and its active constituents SolB and SchB suppress TGFβ1-mediated NF-κB signaling pathway in a Smad-independent mechanism. Our results may help further investigations to develop novel multi-targeted therapeutic strategies that treat or prevent vascular fibrotic diseases.
Collapse
|
8
|
Ewanchuk BW, Allan ERO, Warren AL, Ramachandran R, Yates RM. The cooling compound icilin attenuates autoimmune neuroinflammation through modulation of the T-cell response. FASEB J 2017; 32:1236-1249. [PMID: 29114087 DOI: 10.1096/fj.201700552r] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The synthetic supercooling drug, icilin, and its primary receptor target, the cation channel transient receptor potential (TRP) melastatin-8 (TRPM8), have been described as potent negative regulators of inflammation in the colon. The aim of this study was to determine whether the anti-inflammatory action of icilin could potentially be used to treat autoimmune neuroinflammatory disorders, such as multiple sclerosis (MS). During experimental autoimmune encephalomyelitis (EAE)-a CD4+ T cell-driven murine model of MS-we found that both wild-type (WT) and TRPM8-deficient EAE mice were protected from disease progression during icilin treatment, as evidenced by delays in clinical onset and reductions in neuroinflammation. In vitro, icilin potently inhibited the proliferation of murine and human CD4+ T cells, with the peripheral expansion of autoantigen-restricted T cells similarly diminished by the administration of icilin in mice. Attenuation of both TRPM8-/- and TRP ankyrin-1-/- T-cell proliferation by icilin was consistent with the WT phenotype, which suggests a mechanism that is independent of these channels. In addition, icilin treatment altered the expressional profile of activated CD4+ T cells to one that was indicative of restricted effector function and limited neuroinflammatory potential. These findings identify a potent anti-inflammatory role for icilin in lymphocyte-mediated neuroinflammation and highlight clear pleiotropic effects of the compound beyond classic TRP channel activation.-Ewanchuk, B. W., Allan, E. R. O., Warren, A. L., Ramachandran, R., Yates, R. M. The cooling compound icilin attenuates autoimmune neuroinflammation through modulation of the T-cell response.
Collapse
Affiliation(s)
- Benjamin W Ewanchuk
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Euan R O Allan
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Amy L Warren
- Department of Veterinary Clinical and Diagnostic Sciences, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Rithwik Ramachandran
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Robin M Yates
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
9
|
Park S, Lee S, Park EJ, Kang M, So I, Jeon JH, Chun JN. TGFβ1 induces stress fiber formation through upregulation of TRPC6 in vascular smooth muscle cells. Biochem Biophys Res Commun 2017; 483:129-134. [DOI: 10.1016/j.bbrc.2016.12.179] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 12/27/2016] [Indexed: 01/01/2023]
|
10
|
Lee S, Park YR, Kim SH, Park EJ, Kang MJ, So I, Chun JN, Jeon JH. Geraniol suppresses prostate cancer growth through down-regulation of E2F8. Cancer Med 2016; 5:2899-2908. [PMID: 27683099 PMCID: PMC5083744 DOI: 10.1002/cam4.864] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 07/04/2016] [Accepted: 07/19/2016] [Indexed: 12/23/2022] Open
Abstract
Geraniol, an acyclic dietary monoterpene, has been found to suppress cancer survival and growth. However, the molecular mechanism underlying the antitumor action of geraniol has not been investigated at the genome-wide level. In this study, we analyzed the microarray data obtained from geraniol-treated prostate cancer cells. Geraniol potently altered a gene expression profile and primarily down-regulated cell cycle-related gene signatures, compared to linalool, another structurally similar monoterpene that induces no apparent phenotypic changes. Master regulator analysis using the prostate cancer-specific regulatory interactome identified that the transcription factor E2F8 as a specific target molecule regulates geraniol-specific cell cycle signatures. Subsequent experiments confirmed that geraniol down-regulated E2F8 expression and the knockdown of E2F8 was sufficient to suppress cell growth by inducing G2 /M arrest. Epidemiological analysis showed that E2F8 is up-regulated in metastatic prostate cancer and associated with poor prognosis. These results indicate that E2F8 is a crucial transcription regulator controlling cell cycle and survival in prostate cancer cells. Therefore, our study provides insight into the role of E2F8 in prostate cancer biology and therapeutics.
Collapse
Affiliation(s)
- Sanghoon Lee
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, Utah, 84112 5650
| | - Yu Rang Park
- Office of Clinical Research Information, Asan Medical Center, Seoul, 05535, Korea
| | - Su-Hwa Kim
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Eun-Jung Park
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Min Ji Kang
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, 05535, Korea
| | - Insuk So
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea.,Institute of Human-Environment Interface Biology, Seoul National University, Seoul, 03080, Korea
| | - Jung Nyeo Chun
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea. .,Institute of Human-Environment Interface Biology, Seoul National University, Seoul, 03080, Korea.
| | - Ju-Hong Jeon
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea. .,Institute of Human-Environment Interface Biology, Seoul National University, Seoul, 03080, Korea.
| |
Collapse
|
11
|
Pérez de Vega MJ, Gómez-Monterrey I, Ferrer-Montiel A, González-Muñiz R. Transient Receptor Potential Melastatin 8 Channel (TRPM8) Modulation: Cool Entryway for Treating Pain and Cancer. J Med Chem 2016; 59:10006-10029. [PMID: 27437828 DOI: 10.1021/acs.jmedchem.6b00305] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
TRPM8 ion channels, the primary cold sensors in humans, are activated by innocuous cooling (<28 °C) and cooling compounds (menthol, icilin) and are implicated in sensing unpleasant cold stimuli as well as in mammalian thermoregulation. Overexpression of these thermoregulators in prostate cancer and in other life-threatening tumors, along with their contribution to an increasing number of pathological conditions, opens a plethora of medicinal chemistry opportunities to develop receptor modulators. This Perspective seeks to describe current known modulators for this ion channel because both agonists and antagonists may be useful for the treatment of most TRPM8-mediated pathologies. We primarily focus on SAR data for the different families of compounds and the pharmacological properties of the most promising ligands. Furthermore, we also address the knowledge about the channel structure, although still in its infancy, and the role of the TRPM8 protein signalplex to channel function and dysfunction. We finally outline the potential future prospects of the challenging TRPM8 drug discovery field.
Collapse
Affiliation(s)
| | - Isabel Gómez-Monterrey
- Dipartimento di Farmacia, Università "Federico II" de Napoli , Via D. Montesano 49, 80131, Naples, Italy
| | - Antonio Ferrer-Montiel
- Instituto de Biología Molecular y Celular. Universitas Miguel Hernández . 03202 Alicante, Spain
| | | |
Collapse
|
12
|
Kam SC, Lee SH, Jeon JH, So I, Chae MR, Park JK, Lee SW. Gene expression profile comparison in the penile tissue of diabetes and cavernous nerve injury-induced erectile dysfunction rat model. Investig Clin Urol 2016; 57:286-97. [PMID: 27437539 PMCID: PMC4949699 DOI: 10.4111/icu.2016.57.4.286] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 06/09/2016] [Indexed: 01/02/2023] Open
Abstract
PURPOSE To investigate the effects of cavernous nerve injury (CNI) on gene expression profiles in the cavernosal tissue of a CNI-induced erectile dysfunction (ED) model and to provide a basis for future investigations to discover potential target genes for ED treatment. MATERIALS AND METHODS Young adult rats were divided randomly into 2 groups: sham operation and bilateral CN resection. At 12 weeks after CNI we measured erectile responses and performed microarray experiments and gene set enrichment analysis to reveal gene signatures that were enriched in the CNI-induced ED model. Alterations in gene signatures were compared with those in the diabetes-induced ED model. The diabetic-induced ED data is taken from GSE2457. RESULTS The mean ratio of intracavernosal pressure/blood pressure for the CNI group (0.54±0.4 cmH2O) was significantly lower than that in the sham operation group (0.73±0.8 cmH2O, p<0.05). Supervised and unsupervised clustering analysis showed that the diabetes- and CNI-induced ED cavernous tissues had different gene expression profiles from normal cavernous tissues. We identified 46 genes that were upregulated and 77 genes that were downregulated in both the CNI- and diabetes-induced ED models. CONCLUSIONS Our genome-wide and computational studies provide the groundwork for understanding complex mechanisms and molecular signature changes in ED.
Collapse
Affiliation(s)
- Sung Chul Kam
- Department of Urology, Gyeongsang National University Changwon Hospital, Gyeongsang National University School of Medicine, Changwon, Korea
| | - Sang Hoon Lee
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Ju Hong Jeon
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Institute of Human-Environment Interface Biology, Seoul National University, Seoul, Korea
| | - Insuk So
- Department of Physiology and Biophysics, Seoul National University College of Medicine, Seoul, Korea
| | - Mee Ree Chae
- Department of Urology, Samsung Medical Center, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jong Kwan Park
- Department of Urology, Chonbuk National University Medical School, Institute for Medical Sciences, Chonbuk National University, Research Institute and Clinical Trial Center of Medical Device of Chonbuk National University Hospital, Jeonju, Korea
| | - Sung Won Lee
- Department of Urology, Samsung Medical Center, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
13
|
Inhibitory effect of positively charged triazine antagonists of prokineticin receptors on the transient receptor vanilloid type-1 (TRPV1) channel. Pharmacol Res 2015; 99:362-9. [DOI: 10.1016/j.phrs.2015.07.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 07/10/2015] [Accepted: 07/10/2015] [Indexed: 11/22/2022]
|
14
|
Galle M, Kladniew BR, Castro MA, Villegas SM, Lacunza E, Polo M, de Bravo MG, Crespo R. Modulation by geraniol of gene expression involved in lipid metabolism leading to a reduction of serum-cholesterol and triglyceride levels. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2015; 22:696-704. [PMID: 26141755 DOI: 10.1016/j.phymed.2015.04.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 04/18/2015] [Accepted: 04/24/2015] [Indexed: 06/04/2023]
Abstract
BACKGROUND Geraniol (G) is a natural isoprenoid present in the essential oils of several aromatic plants, with various biochemical and pharmacologic properties. Nevertheless, the mechanisms of action of G on cellular metabolism are largely unknown. HYPOTHESIS/PURPOSE We propose that G could be a potential agent for the treatment of hyperlipidemia that could contribute to the prevention of cardiovascular disease. The aim of the present study was to advance our understanding of its mechanism of action on cholesterol and TG metabolism. STUDY DESIGN/METHODS NIH mice received supplemented diets containing 25, 50, and 75 mmol G/kg chow. After a 3-week treatment, serum total-cholesterol and triglyceride levels were measured by commercial kits and lipid biosynthesis determined by the [(14)C] acetate incorporated into fatty acids plus nonsaponifiable and total hepatic lipids of the mice. The activity of the mRNA encoding HMGCR-the rate-limiting step in cholesterol biosynthesis-along with the enzyme levels and catalysis were assessed by real-time RT-PCR, Western blotting, and HMG-CoA-conversion assays, respectively. In-silico analysis of several genes involved in lipid metabolism and regulated by G in cultured cells was also performed. Finally, the mRNA levels encoded by the genes for the low-density-lipoprotein receptor (LDLR), the sterol-regulatory-element-binding transcription factor (SREBF2), the very-low-density-lipoprotein receptor (VLDLR), and the acetyl-CoA carboxylase (ACACA) were determined by real-time RT-PCR. RESULTS Plasma total-cholesterol and triglyceride levels plus hepatic fatty-acid, total-lipid, and nonsaponifiable-lipid biosynthesis were significantly reduced by feeding with G. Even though an up-regulation of the mRNA encoding HMGCR occurred in the G treated mouse livers, the protein levels and specific activity of the enzyme were both inhibited. G also enhanced the mRNAs encoding the LDL and VLDL receptors and reduced ACACA mRNA, without altering the transcription of the mRNA encoding the SREBF2. CONCLUSIONS The following mechanisms may have mediated the decrease in plasma lipids levels in mice: a down-regulation of hepatocyte-cholesterol synthesis occurred as a result of decreased HMGCR protein levels and catalytic activity; the levels of LDLR mRNA became elevated, thus suggesting an increase in the uptake of serum LDL, especially by the liver; and TG synthesis became reduced very likely because of a decrease in fatty-acid synthesis.
Collapse
Affiliation(s)
- Marianela Galle
- INIBIOLP (UNLP-CONICET CCT La Plata), Facultad de Ciencias Médicas, Calles 60 y 120, La Plata, Argentina
| | - Boris Rodenak Kladniew
- INIBIOLP (UNLP-CONICET CCT La Plata), Facultad de Ciencias Médicas, Calles 60 y 120, La Plata, Argentina
| | - María Agustina Castro
- INIBIOLP (UNLP-CONICET CCT La Plata), Facultad de Ciencias Médicas, Calles 60 y 120, La Plata, Argentina
| | - Sandra Montero Villegas
- INIBIOLP (UNLP-CONICET CCT La Plata), Facultad de Ciencias Médicas, Calles 60 y 120, La Plata, Argentina
| | - Ezequiel Lacunza
- CINIBA (UNLP-CONICET CCT La Plata), Facultad de Ciencias MéG dicas, Calles 60 y 120, La Plata, Argentina
| | - Mónica Polo
- INIBIOLP (UNLP-CONICET CCT La Plata), Facultad de Ciencias Médicas, Calles 60 y 120, La Plata, Argentina
| | - Margarita García de Bravo
- INIBIOLP (UNLP-CONICET CCT La Plata), Facultad de Ciencias Médicas, Calles 60 y 120, La Plata, Argentina
| | - Rosana Crespo
- INIBIOLP (UNLP-CONICET CCT La Plata), Facultad de Ciencias Médicas, Calles 60 y 120, La Plata, Argentina.
| |
Collapse
|
15
|
De Petrocellis L, Ortar G, Schiano Moriello A, Serum EM, Rusterholz DB. Structure-activity relationships of the prototypical TRPM8 agonist icilin. Bioorg Med Chem Lett 2015; 25:2285-90. [PMID: 25935641 DOI: 10.1016/j.bmcl.2015.04.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 04/10/2015] [Accepted: 04/12/2015] [Indexed: 01/28/2023]
Abstract
A series of structural analogues of the TRPM8 agonist icilin was prepared. The compounds were examined for their ability to exert agonist or antagonist effects in HEK-293 cells expressing the TRPM8 receptor. Most structural modifications of the icilin structure largely met with diminished TRPM8 agonist activity. Cinnamamide 'open-chain' analogs of icilin, however, demonstrated significant antagonistic actions at the TRPM8 receptor. Optimal potency (IC50=73 nM) was observed in the 3-iodo derivative 18l.
Collapse
Affiliation(s)
- Luciano De Petrocellis
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, National Research Council, Via Campi Flegrei 34, Comprensorio Olivetti, 80078 Pozzuoli, Naples, Italy.
| | - Giorgio Ortar
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza-Università di Roma, piazzale Aldo Moro 5, 00185 Roma, Italy
| | - Aniello Schiano Moriello
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, National Research Council, Via Campi Flegrei 34, Comprensorio Olivetti, 80078 Pozzuoli, Naples, Italy
| | - Eric M Serum
- Department of Chemistry and Biochemistry, North Dakota State University, Fargo, ND 58108, United States
| | - David B Rusterholz
- Department of Chemistry, University of Wisconsin-River Falls, 410 S. Third St., River Falls, WI 54022, United States
| |
Collapse
|
16
|
Stegeman S, Moya L, Selth LA, Spurdle AB, Clements JA, Batra J. A genetic variant of MDM4 influences regulation by multiple microRNAs in prostate cancer. Endocr Relat Cancer 2015; 22:265-76. [PMID: 25670033 DOI: 10.1530/erc-15-0013] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The oncogene MDM4, also known as MDMX or HDMX, contributes to cancer susceptibility and progression through its capacity to negatively regulate a range of genes with tumour-suppressive functions. As part of a recent genome-wide association study it was determined that the A-allele of the rs4245739 SNP (A>C), located in the 3'-UTR of MDM4, is associated with an increased risk of prostate cancer. Computational predictions revealed that the rs4245739 SNP is located within a predicted binding site for three microRNAs (miRNAs): miR-191-5p, miR-887 and miR-3669. Herein, we show using reporter gene assays and endogenous MDM4 expression analyses that miR-191-5p and miR-887 have a specific affinity for the rs4245739 SNP C-allele in prostate cancer. These miRNAs do not affect MDM4 mRNA levels, rather they inhibit its translation in C-allele-containing PC3 cells but not in LNCaP cells homozygous for the A-allele. By analysing gene expression datasets from patient cohorts, we found that MDM4 is associated with metastasis and prostate cancer progression and that targeting this gene with miR-191-5p or miR-887 decreases in PC3 cell viability. This study is the first, to our knowledge, to demonstrate regulation of the MDM4 rs4245739 SNP C-allele by two miRNAs in prostate cancer, and thereby to identify a mechanism by which the MDM4 rs4245739 SNP A-allele may be associated with an increased risk for prostate cancer.
Collapse
Affiliation(s)
- Shane Stegeman
- School of Biomedical SciencesInstitute of Health and Biomedical Innovation, Translational Research Institute Pty Ltd, Australian Prostate Cancer Research Centre - Queensland, Queensland University of Technology, 37 Kent Street, Woolloongabba, Brisbane, Queensland 4102, AustraliaDame Roma Mitchell Cancer Research LaboratoriesSchool of Medicine, Adelaide Prostate Cancer Research CentreSchool of MedicineFreemasons Foundation Centre for Men's Health, The University of Adelaide, Adelaide, South Australia 5005, AustraliaMolecular Cancer Epidemiology LaboratoryGenetics and Computational Biology Division, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Leire Moya
- School of Biomedical SciencesInstitute of Health and Biomedical Innovation, Translational Research Institute Pty Ltd, Australian Prostate Cancer Research Centre - Queensland, Queensland University of Technology, 37 Kent Street, Woolloongabba, Brisbane, Queensland 4102, AustraliaDame Roma Mitchell Cancer Research LaboratoriesSchool of Medicine, Adelaide Prostate Cancer Research CentreSchool of MedicineFreemasons Foundation Centre for Men's Health, The University of Adelaide, Adelaide, South Australia 5005, AustraliaMolecular Cancer Epidemiology LaboratoryGenetics and Computational Biology Division, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Luke A Selth
- School of Biomedical SciencesInstitute of Health and Biomedical Innovation, Translational Research Institute Pty Ltd, Australian Prostate Cancer Research Centre - Queensland, Queensland University of Technology, 37 Kent Street, Woolloongabba, Brisbane, Queensland 4102, AustraliaDame Roma Mitchell Cancer Research LaboratoriesSchool of Medicine, Adelaide Prostate Cancer Research CentreSchool of MedicineFreemasons Foundation Centre for Men's Health, The University of Adelaide, Adelaide, South Australia 5005, AustraliaMolecular Cancer Epidemiology LaboratoryGenetics and Computational Biology Division, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia School of Biomedical SciencesInstitute of Health and Biomedical Innovation, Translational Research Institute Pty Ltd, Australian Prostate Cancer Research Centre - Queensland, Queensland University of Technology, 37 Kent Street, Woolloongabba, Brisbane, Queensland 4102, AustraliaDame Roma Mitchell Cancer Research LaboratoriesSchool of Medicine, Adelaide Prostate Cancer Research CentreSchool of MedicineFreemasons Foundation Centre for Men's Health, The University of Adelaide, Adelaide, South Australia 5005, AustraliaMolecular Cancer Epidemiology LaboratoryGenetics and Computational Biology Division, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Amanda B Spurdle
- School of Biomedical SciencesInstitute of Health and Biomedical Innovation, Translational Research Institute Pty Ltd, Australian Prostate Cancer Research Centre - Queensland, Queensland University of Technology, 37 Kent Street, Woolloongabba, Brisbane, Queensland 4102, AustraliaDame Roma Mitchell Cancer Research LaboratoriesSchool of Medicine, Adelaide Prostate Cancer Research CentreSchool of MedicineFreemasons Foundation Centre for Men's Health, The University of Adelaide, Adelaide, South Australia 5005, AustraliaMolecular Cancer Epidemiology LaboratoryGenetics and Computational Biology Division, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Judith A Clements
- School of Biomedical SciencesInstitute of Health and Biomedical Innovation, Translational Research Institute Pty Ltd, Australian Prostate Cancer Research Centre - Queensland, Queensland University of Technology, 37 Kent Street, Woolloongabba, Brisbane, Queensland 4102, AustraliaDame Roma Mitchell Cancer Research LaboratoriesSchool of Medicine, Adelaide Prostate Cancer Research CentreSchool of MedicineFreemasons Foundation Centre for Men's Health, The University of Adelaide, Adelaide, South Australia 5005, AustraliaMolecular Cancer Epidemiology LaboratoryGenetics and Computational Biology Division, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Jyotsna Batra
- School of Biomedical SciencesInstitute of Health and Biomedical Innovation, Translational Research Institute Pty Ltd, Australian Prostate Cancer Research Centre - Queensland, Queensland University of Technology, 37 Kent Street, Woolloongabba, Brisbane, Queensland 4102, AustraliaDame Roma Mitchell Cancer Research LaboratoriesSchool of Medicine, Adelaide Prostate Cancer Research CentreSchool of MedicineFreemasons Foundation Centre for Men's Health, The University of Adelaide, Adelaide, South Australia 5005, AustraliaMolecular Cancer Epidemiology LaboratoryGenetics and Computational Biology Division, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| |
Collapse
|
17
|
miR-223 inhibits dengue virus replication by negatively regulating the microtubule-destabilizing protein STMN1 in EAhy926 cells. Microbes Infect 2014; 16:911-22. [PMID: 25181337 PMCID: PMC7110837 DOI: 10.1016/j.micinf.2014.08.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 08/15/2014] [Accepted: 08/25/2014] [Indexed: 12/23/2022]
Abstract
The pathogenesis of dengue virus (DENV) infection is not completely understood. Endothelial cells may act as a target of the virus and be involved in disease pathogenesis. Therefore, the identification of host cell components involved in DENV replication would provide useful information for better understanding DENV infection. In this study, a significantly decreased level of miR-223 was found in DENV2-infected EAhy926 cells, a human endothelial-like cell line, whereas miR-223 overexpression inhibited DENV2 replication. Furthermore, we identified that miR-223 directly targeted the 3' untranslated region (3'UTR) of the messenger RNA (mRNA) for microtubule-destabilizing protein stathmin 1 (STMN1), thereby reducing its mRNA and protein levels. The depletion of miR-223 or overexpression of STMN1 enhanced DENV2 replication, whereas the opposite (increased miR-223 or decreased STMN1) suppressed DENV2 replication, indicating that miR-223 down-regulates STMN1 expression by targeting the 3'UTR of the STMN1 gene to inhibit DENV2 replication. Finally, we demonstrated that two transcription factors, C/EBPα and E2F1, are involved in the regulation of miR-223 levels after DENV2 infection in EAhy926 cells. Collectively, our results suggest that miR-223 may act as a novel antiviral factor, which may open an avenue to limit DENV infection.
Collapse
|