1
|
Fialková V, Ďúranová H, Borotová P, Klongová L, Grabacka M, Speváková I. Natural Stilbenes: Their Role in Colorectal Cancer Prevention, DNA Methylation, and Therapy. Nutr Cancer 2024; 76:760-788. [PMID: 38950568 DOI: 10.1080/01635581.2024.2364391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 07/03/2024]
Abstract
The resistance of colorectal cancer (CRC) to conventional therapeutic modalities, such as radiation therapy and chemotherapy, along with the associated side effects, significantly limits effective anticancer strategies. Numerous epigenetic investigations have unveiled that naturally occurring stilbenes can modify or reverse abnormal epigenetic alterations, particularly aberrant DNA methylation status, offering potential avenues for preventing or treating CRC. By modulating the activity of the DNA methylation machinery components, phytochemicals may influence the various stages of CRC carcinogenesis through multiple molecular mechanisms. Several epigenetic studies, especially preclinical research, have highlighted the effective DNA methylation modulatory effects of stilbenes with minimal adverse effects on organisms, particularly in combination therapies for CRC. However, the available preclinical and clinical data regarding the effects of commonly encountered stilbenes against CRC are currently limited. Therefore, additional epigenetic research is warranted to explore the preventive potential of these phytochemicals in CRC development and to validate their therapeutic application in the prevention and treatment of CRC. This review aims to provide an overview of selected bioactive stilbenes as potential chemopreventive agents for CRC with a focus on their modulatory mechanisms of action, especially in targeting alterations in DNA methylation machinery in CRC.
Collapse
Affiliation(s)
- Veronika Fialková
- AgroBioTech Research Centre, Slovak University of Agriculture, Nitra, Slovakia
| | - Hana Ďúranová
- AgroBioTech Research Centre, Slovak University of Agriculture, Nitra, Slovakia
| | - Petra Borotová
- AgroBioTech Research Centre, Slovak University of Agriculture, Nitra, Slovakia
| | - Lucia Klongová
- AgroBioTech Research Centre, Slovak University of Agriculture, Nitra, Slovakia
| | - Maja Grabacka
- Department of Biotechnology and General Technology of Foods, Faculty of Food Technology, University of Agriculture, Cracow, Poland
| | - Ivana Speváková
- AgroBioTech Research Centre, Slovak University of Agriculture, Nitra, Slovakia
| |
Collapse
|
2
|
Yin J, Ren Y, Yang K, Wang W, Wang T, Xiao W, Yang H. The role of hypoxia-inducible factor 1-alpha in inflammatory bowel disease. Cell Biol Int 2021; 46:46-51. [PMID: 34658125 DOI: 10.1002/cbin.11712] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/21/2021] [Accepted: 10/08/2021] [Indexed: 02/06/2023]
Abstract
Inflammatory bowel disease (IBD) develops as a result of a combination of genetic predisposition, dysbiosis of the gut microbiota, and environmental influences, which is mainly represented by ulcerative colitis (UC) and Crohn's disease (CD). IBDs can result in inflammatory hypoxia by causing intestinal inflammation and vascular damage. The hypoxia-inducible factor 1-alpha (HIF-1α), as a transcription factor, can regulate the cellular adaptation to low oxygen levels and support the development and function of the gut barrier. HIF-αplays its functions through translocating into the nucleus, dimerizing with HIF-1β, and binding to hypoxia-responsive elements of HIF-1 target genes. So far, most studies have addressed the function of HIF-1α in murine models of IBD. In this review, we aim to outline the major roles of HIF-1α in the IBD.
Collapse
Affiliation(s)
- Jiuheng Yin
- Department of General Surgery, Xinqiao Hospital, Army Military Medical University, Chongqing, China
| | - Yanbei Ren
- Department of General Surgery, Xinqiao Hospital, Army Military Medical University, Chongqing, China
| | - Kunqiu Yang
- Department of General Surgery, Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Wensheng Wang
- Department of General Surgery, Xinqiao Hospital, Army Military Medical University, Chongqing, China
| | - Ting Wang
- Nursing Department, Nursing School of Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Weidong Xiao
- Department of General Surgery, Xinqiao Hospital, Army Military Medical University, Chongqing, China
| | - Hua Yang
- Department of General Surgery, Xinqiao Hospital, Army Military Medical University, Chongqing, China
| |
Collapse
|
3
|
Eriau E, Paillet J, Kroemer G, Pol JG. Metabolic Reprogramming by Reduced Calorie Intake or Pharmacological Caloric Restriction Mimetics for Improved Cancer Immunotherapy. Cancers (Basel) 2021; 13:cancers13061260. [PMID: 33809187 PMCID: PMC7999281 DOI: 10.3390/cancers13061260] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/27/2021] [Accepted: 03/08/2021] [Indexed: 12/11/2022] Open
Abstract
Caloric restriction and fasting have been known for a long time for their health- and life-span promoting effects, with coherent observations in multiple model organisms as well as epidemiological and clinical studies. This holds particularly true for cancer. The health-promoting effects of caloric restriction and fasting are mediated at least partly through their cellular effects-chiefly autophagy induction-rather than reduced calorie intake per se. Interestingly, caloric restriction has a differential impact on cancer and healthy cells, due to the atypical metabolic profile of malignant tumors. Caloric restriction mimetics are non-toxic compounds able to mimic the biochemical and physiological effects of caloric restriction including autophagy induction. Caloric restriction and its mimetics induce autophagy to improve the efficacy of some cancer treatments that induce immunogenic cell death (ICD), a type of cellular demise that eventually elicits adaptive antitumor immunity. Caloric restriction and its mimetics also enhance the therapeutic efficacy of chemo-immunotherapies combining ICD-inducing agents with immune checkpoint inhibitors targeting PD-1. Collectively, preclinical data encourage the application of caloric restriction and its mimetics as an adjuvant to immunotherapies. This recommendation is subject to confirmation in additional experimental settings and in clinical trials. In this work, we review the preclinical and clinical evidence in favor of such therapeutic interventions before listing ongoing clinical trials that will shed some light on this subject.
Collapse
Affiliation(s)
- Erwan Eriau
- Centre de Cancérologie de Lyon, Université de Lyon, UMR Inserm 1052 CNRS 5286, Centre Léon Bérard, 69008 Lyon, France; or
- Ecole Normale Supérieure de Lyon, 69342 Lyon, France
- Centre de Recherche des Cordeliers, Equipe 11 labellisée par la Ligue Nationale contre le Cancer, INSERM, Sorbonne Université, Université de Paris, 75006 Paris, France or (J.P.); (G.K.)
- Gustave Roussy Cancer Campus, Metabolomics and Cell Biology Platforms, 94800 Villejuif, France
| | - Juliette Paillet
- Centre de Recherche des Cordeliers, Equipe 11 labellisée par la Ligue Nationale contre le Cancer, INSERM, Sorbonne Université, Université de Paris, 75006 Paris, France or (J.P.); (G.K.)
- Gustave Roussy Cancer Campus, Metabolomics and Cell Biology Platforms, 94800 Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, 91190 Kremlin-Bicêtre, France
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe 11 labellisée par la Ligue Nationale contre le Cancer, INSERM, Sorbonne Université, Université de Paris, 75006 Paris, France or (J.P.); (G.K.)
- Gustave Roussy Cancer Campus, Metabolomics and Cell Biology Platforms, 94800 Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, 91190 Kremlin-Bicêtre, France
- Institut Universitaire de France, 75005 Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, Assistance Publique–Hôpitaux de Paris (AP-HP), 75015 Paris, France
- Suzhou Institute for Systems Medicine, Chinese Academy of Sciences, Suzhou 215163, China
- Department of Women’s and Children’s Health, Karolinska University Hospital, 17164 Stockholm, Sweden
| | - Jonathan G. Pol
- Centre de Recherche des Cordeliers, Equipe 11 labellisée par la Ligue Nationale contre le Cancer, INSERM, Sorbonne Université, Université de Paris, 75006 Paris, France or (J.P.); (G.K.)
- Gustave Roussy Cancer Campus, Metabolomics and Cell Biology Platforms, 94800 Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, 91190 Kremlin-Bicêtre, France
- Correspondence: or ; Tel.: +33-1-44-27-76-66
| |
Collapse
|
4
|
Biocatalyzed Reactions towards Functional Food Components 4-Alkylcatechols and Their Analogues. Catalysts 2020. [DOI: 10.3390/catal10091077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Catechols are antioxidants and radical scavengers with a broad medical potential. 4-Methylcatechol (1b) and 4-ethylcatechol (2b) (occurring in some traditional fermented and smoked foods) activate the cell defense against oxidative stress. We examined the biocatalyzed reactions towards 4-n-alkylcatechols with different side chains length, which is a factor important for the biological activities of catechols. 4-n-Alkylcatechols with methyl through heptyl side chains (1b–7b) were obtained in one pot by (i) oxidation of phenols 1a–7a with tyrosinase from Agaricus bisporus followed by (ii) reduction of ortho-quinones (intermediates) with L-ascorbic acid sodium salt. The conversions decreased with increasing side chain length. The preparative reactions were carried out with substrates 1a–5a. The isolated yields of the purified products decreased from 59% in 2b to 10% in 5b in correlation with logP of the substrates. Homology modeling indicated that the affinities of two tyrosinase isoforms (PPO3 and PPO4) to the substrates with side chains longer than C2 decreased with increasing side chain length. This was probably due to steric limitations and to missing interactions of the extended side chains in the active sites. We envisage using the model to predict further substrates of tyrosinase and testing the products, catechols, for radical-scavenging and biological activities.
Collapse
|
5
|
Khare T, Palakurthi SS, Shah BM, Palakurthi S, Khare S. Natural Product-Based Nanomedicine in Treatment of Inflammatory Bowel Disease. Int J Mol Sci 2020; 21:E3956. [PMID: 32486445 PMCID: PMC7312938 DOI: 10.3390/ijms21113956] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/26/2020] [Accepted: 05/29/2020] [Indexed: 02/06/2023] Open
Abstract
: Many synthetic drugs and monoclonal antibodies are currently in use to treat Inflammatory Bowel Disease (IBD). However, they all are implicated in causing severe side effects and long-term use results in many complications. Numerous in vitro and in vivo experiments demonstrate that phytochemicals and natural macromolecules from plants and animals reduce IBD-related complications with encouraging results. Additionally, many of them modify enzymatic activity, alleviate oxidative stress, and downregulate pro-inflammatory transcriptional factors and cytokine secretion. Translational significance of natural nanomedicine and strategies to investigate future natural product-based nanomedicine is discussed. Our focus in this review is to summarize the use of phytochemicals and macromolecules encapsulated in nanoparticles for the treatment of IBD and IBD-associated colorectal cancer.
Collapse
Affiliation(s)
- Tripti Khare
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO 65212, USA;
| | - Sushesh Srivatsa Palakurthi
- Department of Pharmaceutical Sciences, Rangel College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA; (S.S.P.); (B.M.S.); (S.P.)
| | - Brijesh M. Shah
- Department of Pharmaceutical Sciences, Rangel College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA; (S.S.P.); (B.M.S.); (S.P.)
| | - Srinath Palakurthi
- Department of Pharmaceutical Sciences, Rangel College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA; (S.S.P.); (B.M.S.); (S.P.)
| | - Sharad Khare
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO 65212, USA;
- Harry S. Truman Veterans Hospital, Columbia, MO 65201, USA
| |
Collapse
|
6
|
Banik K, Ranaware AM, Harsha C, Nitesh T, Girisa S, Deshpande V, Fan L, Nalawade SP, Sethi G, Kunnumakkara AB. Piceatannol: A natural stilbene for the prevention and treatment of cancer. Pharmacol Res 2020; 153:104635. [DOI: 10.1016/j.phrs.2020.104635] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 12/19/2019] [Accepted: 01/06/2020] [Indexed: 12/13/2022]
|
7
|
Zang J, Ma S, Wang C, Guo G, Zhou L, Tian X, Lv M, Zhang J, Han B. Screening for active constituents in Turkish galls against ulcerative colitis by mass spectrometry guided preparative chromatography strategy: in silico, in vitro and in vivo study. Food Funct 2019; 9:5124-5138. [PMID: 30256363 DOI: 10.1039/c8fo01439f] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Turkish galls have been reported to exhibit remedial effects in ulcerative colitis (UC). However, the active constituents of Turkish galls for the treatment of UC remain unclear. The objective of this study was to screen for anti-inflammatory active constituents and clarify their associated molecular mechanisms. Therefore, systems pharmacology was developed to predict the relationship between constituents and the corresponding targets as well as pathways. In addition, mass spectrometry-guided preparative chromatography technique was used for preparing constituents to evaluate the anti-inflammatory activities and the therapeutic efficacy against UC. In silico, active constituents exhibited a remedial effect on UC possibly by regulating multiple pathways and attacking multiple targets, of which those involved mainly in the NF-κB pathway were selected for verification. In vitro, 5 categories of constituents were screened as active constituents by comparing the cytotoxicity and detecting the level of the pro-inflammatory factors of 9 category constituents. In vivo, dextran sulfate sodium (DSS)-induced UC was significantly ameliorated in active constituents-fed mice. The results indicated that the active fraction comprising methyl gallate, digallic acid, di-O-galloyl-β-d-glucose, and tri-O-galloyl-β-d-glucose primarily contributed to the treatment of UC. Moreover, active fraction could also inhibit the phosphorylation level of IKKβ, thus inhibiting the downstream NF-κB signaling pathway. The approach developed in this study not only clarifies the anti-inflammation effect of Turkish galls but also provides a beneficial reference for the discovery of the base material and functional mechanism of this herbal medicine.
Collapse
Affiliation(s)
- Jie Zang
- School of Chemistry and Chemical Engineering/Key Laboratory for Green Process of Chemical Engineering of Xinjiang Bingtuan/School of Pharmacy/Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education/School of Medicine, Shihezi University, Xinjiang Shihezi 832003, P. R. China.
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Jiang Z, You Q, Zhang X. Medicinal chemistry of metal chelating fragments in metalloenzyme active sites: A perspective. Eur J Med Chem 2019; 165:172-197. [PMID: 30684796 DOI: 10.1016/j.ejmech.2019.01.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 12/22/2018] [Accepted: 01/08/2019] [Indexed: 12/25/2022]
Abstract
Numerous metal-containing enzymes (metalloenzymes) have been considered as drug targets related to diseases such as cancers, diabetes, anemia, AIDS, malaria, bacterial infection, fibrosis, and neurodegenerative diseases. Inhibitors of the metalloenzymes have been developed independently, most of which are mimics of substrates of the corresponding enzymes. However, little attention has been paid to the interactions between inhibitors and active site metal ions. This review is focused on different metal binding fragments and their chelating properties in the metal-containing active binding pockets of metalloenzymes. We have enumerated over one hundred of inhibitors targeting various metalloenzymes and identified over ten kinds of fragments with different binding patterns. Furthermore, we have investigated the inhibitors that are undergoing clinical evaluation in order to help looking for more potential scaffolds bearing metal binding fragments. This review will provide deep insights for the rational design of novel inhibitors targeting the metal-containing binding sites of specific proteins.
Collapse
Affiliation(s)
- Zhensheng Jiang
- Sate Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Qidong You
- Sate Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Xiaojin Zhang
- Sate Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Chemistry, School of Science, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
9
|
Gerogianni PS, Chatziathanasiadou MV, Diamantis DA, Tzakos AG, Galaris D. Lipophilic ester and amide derivatives of rosmarinic acid protect cells against H 2O 2-induced DNA damage and apoptosis: The potential role of intracellular accumulation and labile iron chelation. Redox Biol 2018; 15:548-556. [PMID: 29413966 PMCID: PMC5975196 DOI: 10.1016/j.redox.2018.01.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 01/24/2018] [Accepted: 01/31/2018] [Indexed: 12/31/2022] Open
Abstract
Phenolic acids represent abundant components contained in human diet. However, the negative charge in their carboxylic group limits their capacity to diffuse through biological membranes, thus hindering their access to cell interior. In order to promote the diffusion of rosmarinic acid through biological membranes, we synthesized several lipophilic ester- and amide-derivatives of this compound and evaluated their capacity to prevent H2O2-induced DNA damage and apoptosis in cultured human cells. Esterification of the carboxylic moiety with lipophilic groups strongly enhanced the capacity of rosmarinic acid to protect cells. On the other hand, the amide-derivatives were somewhat less effective but exerted less cytotoxicity at high concentrations. Cell uptake experiments, using ultra-high performance liquid chromatography coupled with tandem mass spectrometry (UHPLC-MS/MS), illustrated different levels of intracellular accumulation among the ester- and amide-derivatives, with the first being more effectively accumulated, probably due to their extensive hydrolysis inside the cells. In conclusion, these results highlight the hitherto unrecognized fundamental importance of derivatization of diet-derived phenolic acids to unveil their biological potential.
Collapse
Affiliation(s)
- Paraskevi S Gerogianni
- Laboratory of Biological Chemistry, School of Health Sciences, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece
| | - Maria V Chatziathanasiadou
- Laboratory of Organic Chemistry and Biochemistry, School of Natural Sciences, Department of Chemistry, University of Ioannina, 45110 Ioannina, Greece
| | - Dimitrios A Diamantis
- Laboratory of Organic Chemistry and Biochemistry, School of Natural Sciences, Department of Chemistry, University of Ioannina, 45110 Ioannina, Greece
| | - Andreas G Tzakos
- Laboratory of Organic Chemistry and Biochemistry, School of Natural Sciences, Department of Chemistry, University of Ioannina, 45110 Ioannina, Greece
| | - Dimitrios Galaris
- Laboratory of Biological Chemistry, School of Health Sciences, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece.
| |
Collapse
|
10
|
Minoxidil Induction of VEGF Is Mediated by Inhibition of HIF-Prolyl Hydroxylase. Int J Mol Sci 2017; 19:ijms19010053. [PMID: 29295567 PMCID: PMC5796003 DOI: 10.3390/ijms19010053] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 12/16/2017] [Accepted: 12/20/2017] [Indexed: 12/25/2022] Open
Abstract
The topical application of minoxidil may achieve millimolar concentrations in the skin. We investigated whether millimolar minoxidil could induce vascular endothelial growth factor (VEGF), a possible effector for minoxidil-mediated hair growth, and how it occurred at the molecular level. Cell-based experiments were performed to investigate a molecular mechanism underlying the millimolar minoxidil induction of VEGF. The inhibitory effect of minoxidil on hypoxia-inducible factor (HIF) prolyl hydroxylase-2 (PHD-2) was tested by an in vitro von Hippel–Lindau protein (VHL) binding assay. To examine the angiogenic potential of millimolar minoxidil, a chorioallantoic membrane (CAM) assay was used. In human keratinocytes and dermal papilla cells, millimolar minoxidil increased the secretion of VEGF, which was not attenuated by a specific adenosine receptor antagonist that inhibits the micromolar minoxidil induction of VEGF. Millimolar minoxidil induced hypoxia-inducible factor-1α (HIF-1α), and the induction of VEGF was dependent on HIF-1. Moreover, minoxidil applied to the dorsal area of mice increased HIF-1α and VEGF in the skin. In an in vitro VHL binding assay, minoxidil directly inhibited PHD-2, thus preventing the hydroxylation of cellular HIF-1α and VHL-dependent proteasome degradation and resulting in the stabilization of HIF-1α protein. Minoxidil inhibition of PHD-2 was reversed by ascorbate, a cofactor of PHD-2, and the minoxidil induction of cellular HIF-1α was abrogated by the cofactor. Millimolar minoxidil promoted angiogenesis in the CAM assay, an in vivo angiogenic test, and this was nullified by the specific inhibition of VEGF. Our data demonstrate that PHD may be the molecular target for millimolar minoxidil-mediated VEGF induction via HIF-1.
Collapse
|
11
|
Botta G, Bizzarri BM, Garozzo A, Timpanaro R, Bisignano B, Amatore D, Palamara AT, Nencioni L, Saladino R. Carbon nanotubes supported tyrosinase in the synthesis of lipophilic hydroxytyrosol and dihydrocaffeoyl catechols with antiviral activity against DNA and RNA viruses. Bioorg Med Chem 2015; 23:5345-51. [PMID: 26260341 PMCID: PMC7125559 DOI: 10.1016/j.bmc.2015.07.061] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 07/24/2015] [Accepted: 07/27/2015] [Indexed: 12/16/2022]
Abstract
Hydroxytyrosol and dihydrocaffeoyl catechols with lipophilic properties have been synthesized in high yield using tyrosinase immobilized on multi-walled carbon nanotubes by the Layer-by-Layer technique. All synthesized catechols were evaluated against a large panel of DNA and RNA viruses, including Poliovirus type 1, Echovirus type 9, Herpes simplex virus type 1 (HSV-1), Herpes simplex virus type 2 (HSV-2), Coxsackievirus type B3 (Cox B3), Adenovirus type 2 and type 5 and Cytomegalovirus (CMV). A significant antiviral activity was observed in the inhibition of HSV-1, HSV-2, Cox B3 and CMV. The mechanism of action of the most active dihydrocaffeoyl derivative was investigated against a model of HSV-1 infection.
Collapse
Affiliation(s)
- Giorgia Botta
- Department of Ecology and Biology, University of Tuscia, Largo dell’Università, 01100 Viterbo (VT), Italy
| | - Bruno Mattia Bizzarri
- Department of Ecology and Biology, University of Tuscia, Largo dell’Università, 01100 Viterbo (VT), Italy
| | - Adriana Garozzo
- Department of Biomedical and Biotechnological Sciences, Microbiological Section, University of Catania (CT), Via Androne, 81 95124 Catania, Italy
| | - Rossella Timpanaro
- Department of Biomedical and Biotechnological Sciences, Microbiological Section, University of Catania (CT), Via Androne, 81 95124 Catania, Italy
| | - Benedetta Bisignano
- Department of Biomedical and Biotechnological Sciences, Microbiological Section, University of Catania (CT), Via Androne, 81 95124 Catania, Italy
| | - Donatella Amatore
- Department of Public Health and Infectious Diseases, ‘Sapienza’ University, 00185 Rome, Italy
- IRCCS San Raffaele Pisana, Telematic University, 00166 Rome, Italy
| | - Anna Teresa Palamara
- Department of Public Health and Infectious Diseases, ‘Sapienza’ University, 00185 Rome, Italy
- IRCCS San Raffaele Pisana, Telematic University, 00166 Rome, Italy
| | - Lucia Nencioni
- Department of Public Health and Infectious Diseases, ‘Sapienza’ University, 00185 Rome, Italy
| | - Raffaele Saladino
- Department of Ecology and Biology, University of Tuscia, Largo dell’Università, 01100 Viterbo (VT), Italy
| |
Collapse
|
12
|
Yum S, Jeong S, Lee S, Nam J, Kim W, Yoo JW, Kim MS, Lee BL, Jung Y. Colon-targeted delivery of piceatannol enhances anti-colitic effects of the natural product: potential molecular mechanisms for therapeutic enhancement. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:4247-58. [PMID: 26273188 PMCID: PMC4532174 DOI: 10.2147/dddt.s88670] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Piceatannol (PCT), an anti-colitic natural product, undergoes extensive Phase II hepatic metabolism, resulting in very low bioavailability. We investigated whether colon-targeted delivery of PCT could enhance anti-colitic effects and how therapeutic enhancement occurred at the molecular level. Molecular effects of PCT were examined in human colon carcinoma cells and inflamed colons. The anti-colitic effects of PCT in a colon-targeted capsule (colon-targeted PCT) were compared with PCT in a gelatin capsule (conventional PCT) in a trinitrobenzene sulfonic acid-induced rat colitis model. Colon-targeted PCT elicited greatly enhanced recovery of the colonic inflammation. In HCT116 cells, PCT inhibited nuclear factor kappaB while activating anti-colitic transcription factors, nuclear factor-erythroid 2 (NF-E2) p45-related factor 2, and hypoxia-inducible factor-1. Colon-targeted PCT, but not conventional PCT, modulated production of the target gene products of the transcription factors in the inflamed colonic tissues. Rectal administration of PCT, which simulates the therapeutic action of colon-targeted PCT, also ameliorated rat colitis and reproduced the molecular effects in the inflamed colonic tissues. Colon-targeted delivery increased therapeutic efficacy of PCT against colitis, likely resulting from multitargeted effects exerted by colon-targeted PCT. The drug delivery technique may be useful for therapeutic optimization of anti-colitic lead compounds including natural products.
Collapse
Affiliation(s)
- Soohwan Yum
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Seongkeun Jeong
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Sunyoung Lee
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Joon Nam
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Wooseong Kim
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Jin-Wook Yoo
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Min-Soo Kim
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Bok Luel Lee
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Yunjin Jung
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
13
|
Yum S, Jeong S, Lee S, Kim W, Nam J, Jung Y. HIF-prolyl hydroxylase is a potential molecular target for esculetin-mediated anti-colitic effects. Fitoterapia 2015; 103:55-62. [PMID: 25797536 DOI: 10.1016/j.fitote.2015.03.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Revised: 03/09/2015] [Accepted: 03/14/2015] [Indexed: 01/24/2023]
Abstract
We investigated a potential molecular target for anti-colitic effects of esculetin, 6,7-dihydroxycoumarin. Esculetin administered rectally effectively ameliorated TNBS-induced rat colitis and attenuated the expression of pro-inflammatory mediators in the inflamed colon. In human colon carcinoma HCT116 cells, esculetin induced hypoxia-inducible factor-1α (HIF-1α), leading to secretion of vascular endothelial growth factor, a HIF-1 target gene product involved in ulcer healing of the gastrointestinal mucosa. Esculetin directly inhibited HIF prolyl hydroxylase-2 (HPH-2), an enzyme playing a major role in negatively regulating HIF-1α protein stability. Esculetin inhibition of HPH and consequent induction of HIF-1α were attenuated by escalating dose of either ascorbate or 2-ketoglutarate, the required factors of the enzyme. Structurally, the catechol moiety in esculetin was required for HPH inhibition. Collectively, HPH may be a molecular target for esculetin-mediated anti-colitic effects and the catechol moiety in esculetin is the pharmacophore for HPH inhibition.
Collapse
Affiliation(s)
- Soohwan Yum
- Laboratory of Biomedicinal Chemistry, College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Seongkeun Jeong
- Laboratory of Biomedicinal Chemistry, College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Sunyoung Lee
- Laboratory of Biomedicinal Chemistry, College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Wooseong Kim
- Laboratory of Biomedicinal Chemistry, College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Joon Nam
- Laboratory of Biomedicinal Chemistry, College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Yunjin Jung
- Laboratory of Biomedicinal Chemistry, College of Pharmacy, Pusan National University, Busan, Republic of Korea.
| |
Collapse
|
14
|
Natural compounds regulate glycolysis in hypoxic tumor microenvironment. BIOMED RESEARCH INTERNATIONAL 2015; 2015:354143. [PMID: 25685782 PMCID: PMC4317583 DOI: 10.1155/2015/354143] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 10/01/2014] [Indexed: 01/19/2023]
Abstract
In the early twentieth century, Otto Heinrich Warburg described an elevated rate of glycolysis occurring in cancer cells, even in the presence of atmospheric oxygen (the Warburg effect). Recently it became a therapeutically interesting strategy and is considered as an emerging hallmark of cancer. Hypoxia inducible factor-1 (HIF-1) is one of the key transcription factors that play major roles in tumor glycolysis and could directly trigger Warburg effect. Thus, how to inhibit HIF-1-depended Warburg effect to assist the cancer therapy is becoming a hot issue in cancer research. In fact, HIF-1 upregulates the glucose transporters (GLUT) and induces the expression of glycolytic enzymes, such as hexokinase, pyruvate kinase, and lactate dehydrogenase. So small molecules of natural origin used as GLUT, hexokinase, or pyruvate kinase isoform M2 inhibitors could represent a major challenge in the field of cancer treatment. These compounds aim to suppress tumor hypoxia induced glycolysis process to suppress the cell energy metabolism or enhance the susceptibility of tumor cells to radio- and chemotherapy. In this review, we highlight the role of natural compounds in regulating tumor glycolysis, with a main focus on the glycolysis under hypoxic tumor microenvironment.
Collapse
|
15
|
Jeong S, Park H, Hong S, Yum S, Kim W, Jung Y. Lipophilic modification enhances anti-colitic properties of rosmarinic acid by potentiating its HIF-prolyl hydroxylases inhibitory activity. Eur J Pharmacol 2015; 747:114-22. [PMID: 25483211 DOI: 10.1016/j.ejphar.2014.11.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 11/21/2014] [Accepted: 11/24/2014] [Indexed: 12/22/2022]
Abstract
Inhibition of hypoxia inducible factor-prolyl hydroxylase-2 (HPH), leading to activation of hypoxia inducible factor (HIF)-1 is a potential therapeutic strategy for the treatment of colitis. Rosmarinic acid (RA), an ester of caffeic acid and 3,4-dihydroxyphenyllactic acid is a naturally occurring polyphenolic compound with two catechols, a or inhibition of HPH. To improve accessibility of highly hydrophilic RA to HPH, an intracellular target, RA was chemically modified to decrease hydrophilicity. Of the less-hydrophilic derivatives, rosmarinic acid methyl ester (RAME) most potently inhibited HPH. Accordingly, RAME prevented hydroxylation of HIF-1α and consequently stabilized HIF-1α protein in cells. RAME inhibition of HPH and induction of HIF-1α were diminished by elevated doses of the required factors of HPH, 2-ketoglutarate and ascorbate. RAME induction of HIF-1α led to activation of an ulcer healing pathway, HIF-1-vascular endothelial growth factor (VEGF), in human colon carcinoma cells. RAME administered rectally ameliorated TNBS-induced rat colitis and substantially decreased the levels of pro-inflammatory mediators in the inflamed colonic tissue. In parallel with the cellular effects of RAME, RAME up-regulated HIF-1α and VEGF in the inflamed colonic tissue. Thus, lipophilic modification of RA improves its ability to inhibit HPH, leading to activation of the HIF-1-VEGF pathway. RAME, a lipophilic RA derivative, may exert anti-colitic effects via activation of the ulcer healing pathway.
Collapse
Affiliation(s)
- Seongkeun Jeong
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Huijeong Park
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Sungchae Hong
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Soohwan Yum
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Wooseong Kim
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Yunjin Jung
- College of Pharmacy, Pusan National University, Busan, Republic of Korea.
| |
Collapse
|
16
|
Madeo F, Pietrocola F, Eisenberg T, Kroemer G. Caloric restriction mimetics: towards a molecular definition. Nat Rev Drug Discov 2014; 13:727-40. [PMID: 25212602 DOI: 10.1038/nrd4391] [Citation(s) in RCA: 187] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Caloric restriction, be it constant or intermittent, is reputed to have health-promoting and lifespan-extending effects. Caloric restriction mimetics (CRMs) are compounds that mimic the biochemical and functional effects of caloric restriction. In this Opinion article, we propose a unifying definition of CRMs as compounds that stimulate autophagy by favouring the deacetylation of cellular proteins. This deacetylation process can be achieved by three classes of compounds that deplete acetyl coenzyme A (AcCoA; the sole donor of acetyl groups), that inhibit acetyl transferases (a group of enzymes that acetylate lysine residues in an array of proteins) or that stimulate the activity of deacetylases and hence reverse the action of acetyl transferases. A unifying definition of CRMs will be important for the continued development of this class of therapeutic agents.
Collapse
Affiliation(s)
- Frank Madeo
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Federico Pietrocola
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, INSERM U1138, F-75006 Paris, France and the Université Paris Descartes, Sorbonne Paris Cité, F-75006 Paris, France
| | - Tobias Eisenberg
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Guido Kroemer
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, INSERM U1138, F-75006 Paris, France; the Université Paris Descartes, Sorbonne Paris Cité, F-75006 Paris, France; the Pôle de Biologie, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Labex Immuno-Oncology, F-75015 Paris, France; and the Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, F-94805 Villejuif, France
| |
Collapse
|
17
|
Yum S, Park H, Hong S, Jeong S, Kim W, Jung Y. N-(2-Mercaptopropionyl)-glycine, a diffusible antioxidant, activates HIF-1 by inhibiting HIF prolyl hydroxylase-2: implication in amelioration of rat colitis by the antioxidant. Biochem Biophys Res Commun 2013; 443:1008-13. [PMID: 24361888 DOI: 10.1016/j.bbrc.2013.12.081] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 12/16/2013] [Indexed: 11/19/2022]
Abstract
We investigated anti-colitic effects of N-(2-mercaptopropionyl)-glycine (NMPG), a diffusible antioxidant, in TNBS-induced rat colitis model and a potential molecular mechanism underlying the pharmacologic effect of the antioxidant. NMPG alleviated colonic injury and effectively lowered myeloperoxidase activity. Moreover, NMPG substantially attenuated expression of pro-inflammatory mediators in the inflamed colon. NMPG induced hypoxia-inducible factor-1α (HIF-1α) in human colon carcinoma cells, leading to elevated secretion of vascular endothelial growth factor (VEGF), a target gene product of HIF-1 involved in ulcer healing of gastrointestinal mucosa. NMPG induction of HIF-1α occurred by inhibiting HIF prolyl hydroxylase-2 (HPH-2), an enzyme that plays a major role in negatively regulating HIF-1α protein stability. In in vitro Von Hippel-Lindau protein binding assay, the inhibitory effect of NMPG on HPH-2 was attenuated by escalating dose of ascorbate but not 2-ketoglutarate, cofactors of the enzyme. Consistent with this, cell-permeable ascorbate significantly attenuated NMPG induction of HIF-1α in cells. Our data suggest that NMPG is an anti-colitic antioxidant that exerts its pharmacologic effects at least partly through activation of an ulcer healing pathway, HIF-1-VEGF.
Collapse
Affiliation(s)
- Soohwan Yum
- College of Pharmacy, Pusan National University, Busan 609-735, South Korea
| | - Huijeong Park
- College of Pharmacy, Pusan National University, Busan 609-735, South Korea
| | - Sungchae Hong
- College of Pharmacy, Pusan National University, Busan 609-735, South Korea
| | - Seongkeun Jeong
- College of Pharmacy, Pusan National University, Busan 609-735, South Korea
| | - Wooseong Kim
- College of Pharmacy, Pusan National University, Busan 609-735, South Korea
| | - Yunjin Jung
- College of Pharmacy, Pusan National University, Busan 609-735, South Korea.
| |
Collapse
|