1
|
Sanati M, Afshari AR, Amini J, Mollazadeh H, Jamialahmadi T, Sahebkar A. Targeting angiogenesis in gliomas: Potential role of phytochemicals. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105192] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
2
|
β-cyclodextrin encapsulation of curcumin elicits an altered mode of angiogenin inhibition: In vitro and in vivo studies. Int J Biol Macromol 2022; 208:654-666. [PMID: 35341883 DOI: 10.1016/j.ijbiomac.2022.03.127] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/13/2022] [Accepted: 03/20/2022] [Indexed: 12/29/2022]
Abstract
The interaction of curcumin (Cur) with human angiogenin (hAng), a potent blood vessel inducer responsible for angiogenesis is found to change following encapsulation within the β-cyclodextrin (βCD) cavity. The enhanced bioavailability and increase in the binding stoichiometry of hAng:Cur-βCD (1:2) leads to increased affinity, hence an increase in the association constant. The altered mode of hAng inhibition of Cur from a non-competitive (KI = 23.7 ± 2.2 μM) to a mixed type (KI = 19.8 ± 1.4 μM), after encapsulation provides an insight into interaction patterns. Isothermal titration calorimetry (ITC) experiments indicate the formation of multiple favorable non-covalent interactions (also confirmed by docking studies), which implies negative enthalpy changes (-ΔHo) and restriction in the dynamic mobility of the free protein molecule resulting in a very less positive entropy change (TΔSo). This leads to a medium magnitude for the spontaneous free energy change associated with the interaction/binding process. The spontaneity of binding indicates a more favorable value for the Cur-βCD (ΔGo = -7.75 kcal/mol) compared to Cur (ΔGo = -7.49 kcal/mol). In vivo studies also demonstrate the anti-angiogenic effect of Cur/Cur-βCD confirmed by the significant decrease in blood vessel density and branching index.
Collapse
|
3
|
Abdalla A, Murali C, Amin A. Safranal Inhibits Angiogenesis via Targeting HIF-1α/VEGF Machinery: In Vitro and Ex Vivo Insights. Front Oncol 2022; 11:789172. [PMID: 35211395 PMCID: PMC8862147 DOI: 10.3389/fonc.2021.789172] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/30/2021] [Indexed: 12/27/2022] Open
Abstract
Nature has a nearly infinite inventory of unexplored phytochemicals and biomolecules that have the potential to treat a variety of diseases. Safranal exhibits anti-cancer property and the present study explores its antiangiogenic property. Hepatocellular carcinoma (HCC) ranks as the sixth deadliest among all cancer types. Targeting the non-tumor vasculature supporting system is very promising as it has less plasticity, unlike malignant cells that are often associated with issues like drug resistance, poor prognosis, and relapse. In this study, we successfully inhibited the proliferation of primary human umbilical vein endothelial cells (HUVEC) with an IC50 of 300μM and blocked VEGF secretion in HepG2 cells. Furthermore, safranal inhibited VEGF-induced angiogenesis in vitro and ex vivo via scratch wound assay, tube formation assay, transmembrane assay, and aortic ring assay. In addition, safranal downregulated the in vitro expression of HIF-1α, VEGF, VEGFR2, p-AKT, p-ERK1/2, MMP9, p-FAK, and p-STAT3. The present study is the first to reveal the antiangiogenic potential of safranal and propose its possible underlying mechanism in HCC.
Collapse
Affiliation(s)
- Ali Abdalla
- Weinberg Institute for Cognitive Science, University of Michigan, Ann Arbor, MI, United States
| | - Chandraprabha Murali
- Biology Department, College of Science, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Amr Amin
- Biology Department, College of Science, United Arab Emirates University, Al-Ain, United Arab Emirates.,The College, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
4
|
Khan A, Ali S, Murad W, Hayat K, Siraj S, Jawad M, Khan RA, Uddin J, Al-Harrasi A, Khan A. Phytochemical and pharmacological uses of medicinal plants to treat cancer: A case study from Khyber Pakhtunkhwa, North Pakistan. JOURNAL OF ETHNOPHARMACOLOGY 2021; 281:114437. [PMID: 34391861 DOI: 10.1016/j.jep.2021.114437] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/16/2021] [Accepted: 07/17/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cancer is the top death causing disease in the world, due to its occurrence through various mechanism and form. Medicinal plants have been extensively used for the purifications and isolations of phytochemicals for the treatment and prevention of cancer. OBJECTIVES Consequently, this research was designed to document the traditional practices of anti-cancer plants and its phytochemical essay across the districts of KP, Pakistan. MATERIALS AND METHODS Semi-structured interviews were conducted in 24 districts from the informants mostly the traditional herbalists (key informants). The information were compared with the publish data using various authentic search engines including, google, researchgate, google scholar and NCBI. RESULTS One hundred and fifty-four (154) anti-cancer plants were recognized belonging to 69 families among all, Lamiaceae (13 sp.), Asteraceae (12 sp.) and Solanaceae (9 sp.) were the preferred families. The local inhabitants in the area typically prepare ethnomedicinal recipes from leaves (33.70%) and whole plants (23.37%) in the form of decoction and powder (24.67%), respectively. Herbs stayed the most preferred life form (61.68%) followed by shrub (21.4%). Similarly, breast (29.22%) and lung cancer (14.83%) was the common disease type. Literature study also authorize that, the medicinal plants of the research area were rich in phytochemical like quercetin, coumarine, kaempferol, apigenin, colchicine, alliin, rutin, lupeol, allicin, berbarine, lutolin, vanilic acid, urocilic acid and solamargine have revealed significant activates concerning the cancer diseases, that replicating the efficacy of these plants as medicines. CONCLUSION The Khyber Pakhtunkhwa is rural area and the local inhabitants have very strong traditional knowledge about the medicinal plants for different diseases like cancer. The medicinal plants for significant ranked disorder might be pharmacologically and phtyochemicaly explored to demonstrate their efficacy. Moreover, the local flora especially medicinal plants facing overgrazing, overexploitation and inappropriate way of collection, however, proper management strategies like reforestation, controlled grazing, proper permission from concerned department and rangeland strategies among others may be assumed to enhance the proper usage of medicinal plants.
Collapse
Affiliation(s)
- Asif Khan
- Department of Botany, Garden Campus Abdul Wali Khan University, Mardan, Pakistan
| | - Sajid Ali
- Department of Botany, Garden Campus Abdul Wali Khan University, Mardan, Pakistan
| | - Waheed Murad
- Department of Botany, Garden Campus Abdul Wali Khan University, Mardan, Pakistan.
| | - Khizar Hayat
- Key Laboratory of Plant Ecology, Northeast Forestry University, Harbin, China
| | - Shumaila Siraj
- Department of Botany, Garden Campus Abdul Wali Khan University, Mardan, Pakistan
| | - Muhammad Jawad
- Center of Geographical Information System, University of Punjab, Pakistan
| | | | - Jalal Uddin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat Al Mauz, Nizwa, Oman.
| | - Ajmal Khan
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat Al Mauz, Nizwa, Oman.
| |
Collapse
|
5
|
Shabestarian H, Tabrizi MH, Es-Haghi A, Khadem F. The Brassica Napus Extract (BNE)-Loaded PLGA Nanoparticles as an Early Necroptosis and Late Apoptosis Inducer in Human MCF-7 Breast Cancer Cells. Nutr Cancer 2021; 74:2540-2549. [PMID: 34844492 DOI: 10.1080/01635581.2021.2008986] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Loading of the Brassica napus extract (BNE) on PLGA nanoparticle (BNE-PNP) and study its necroptotic activity in human MCF7-breast cancer cells. Double emulsion solvent evaporation methods were used for synthesis of BNE-PNP and DLS, SEM, and surface Zeta-potential analysis were applied for defining the physicochemical properties of BNE-PNP. The cytotoxic impact of BNE-PNP nanoparticles was analyzed by MTT assay and expression of apoptotic (P53 and Cas-3) and necrotic (TNF-α) gene markers were measured by qPCR to evaluate the BNE-PNP-induced cell death type. The stable (-36.07 mV) BNE-PNP were synthesized at 71.07 nm dimension. They significantly decrease the count of metabolically active MCF7 cells (IC50: 170.94 µg/ml after 48 h). The BNE-PNP induced an early programmed necrotic (necroptosis) and late apoptotic death on the MCF7 cancer cells by up-regulating all the P53/TNF-α and Cas-3 gene expression, respectively. The BNE-PNP dose-dependently induced an early cell-selective necroptotic death. Since the necroptotic death is known as a biocompatible cellular death induction, the BNE-PNP have the potential to be used as a safe efficient anticancer compound.
Collapse
Affiliation(s)
- Hanieh Shabestarian
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | | | - Ali Es-Haghi
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | | |
Collapse
|
6
|
Esteghlal S, Mokhtari MJ, Beyzaei Z. Quercetin Can Inhibit Angiogenesis via the Down Regulation of MALAT1 and MIAT LncRNAs in Human Umbilical Vein Endothelial Cells. Int J Prev Med 2021; 12:59. [PMID: 34447501 PMCID: PMC8356977 DOI: 10.4103/ijpvm.ijpvm_103_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 07/30/2020] [Indexed: 12/14/2022] Open
Abstract
Background: Angiogenesis is an important step in cancer metastasis since it enables the growing tumor to receive nutrients and oxygen. Quercetin is a generic flavonoid and has been investigated for its ability to inhibit angiogenesis in different types of cancers. MALAT1 and MIAT lncRNAs are associated with the angiogenesis process. MALAT1 induces hypoxia-driven angiogenesis via the overexpression of angiogenic genes. Down regulation of MIAT1 could inhibit the proliferation of endothelial cells, tube formation, and migration. In this study, we assessed the anti-angiogenic activity of quercetin on human umbilical vein endothelial cells (HUVEC) via the expression of MALAT1 and MIAT genes. Methods: In the present study, HUVEC cells were incubated with various concentrations of quercetin for 24, 48, and 72 h. Cell proliferation was then evaluated by MTT assay. RNA was extracted by TRIzol and cDNA synthesis. The expression levels of MALAT1 and MIAT genes relative to the GAPDH gene were quantified using the highly sensitive real-time PCR method. Results: Our results demonstrated that quercetin has an inhibitory impact on the cell viability of HUVEC cells. The IC50 values of quercetin after 24, 48, and 72 h were 282.05 μM, 228.25 μM, and 131.65 μM, respectively. The MALAT1/GAPDH ratio was computed as 0.21 for 24h, 0.18 for 48h, and 0.29 for 72 h. The MIAT/GAPDH ratio was computed as 0.82 for 24h, 0.84 for 48h, and 0.78 for 72 h. Conclusions: In conclusion, quercetin treatment had an anti-angiogenic effect on HUVEC cells, at least partially via the down regulation of MALAT1 and MIAT LncRNAs gene expression.
Collapse
Affiliation(s)
- Somayeh Esteghlal
- Department of Biology, Zarghan Branch, Islamic Azad University, Zarghan, Iran
| | | | - Zahra Beyzaei
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
7
|
Gallemit PEM, Yoodee S, Malaitad T, Thongboonkerd V. Epigallocatechin-3-gallate plays more predominant roles than caffeine for inducing actin-crosslinking, ubiquitin/proteasome activity and glycolysis, and suppressing angiogenesis features of human endothelial cells. Biomed Pharmacother 2021; 141:111837. [PMID: 34175818 DOI: 10.1016/j.biopha.2021.111837] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/13/2021] [Accepted: 06/14/2021] [Indexed: 01/06/2023] Open
Abstract
A recent expression proteomics study has reported changes in cellular proteome (set of proteins) of human endothelial cells (ECs) induced by caffeine and epigallocatechin-3-gallate (EGCG), the most abundant bioactive compounds in coffee and green tea, respectively. Although both common and differential changes were highlighted by bioinformatics prediction, no experimental validation was performed. Herein, we reanalyzed these proteome datasets and performed protein-protein interactions network analysis followed by functional investigations using various assays to address the relevance of such proteome changes in human ECs functions. Protein-protein interactions network analysis revealed actin-crosslink formation, ubiquitin-proteasome activity and glycolysis as the three main networks among those significantly altered proteins induced by caffeine and EGCG. The experimental data showed predominant increases of actin-crosslink formation, ubiquitin-proteasome activity, and glycolysis (as reflected by increased F-actin and β-actin, declined ubiquitinated proteins and increased intracellular ATP, respectively) in the EGCG-treated cells. Investigations on angiogenesis features revealed that EGCG predominantly reduced ECs proliferation, migration/invasion, endothelial tube formation (as determined by numbers of nodes/junctions and meshes), barrier function (as determined by levels of VE-cadherin, zonula occludens-1 (ZO-1) and transendothelial resistance (TER)), and angiopoietin-2 secretion. However, both caffeine and EGCG had no effects on matrix metalloproteinase-2 (MMP-2) secretion. These data indicate that EGCG exhibits more potent effects on human ECs functions to induce actin-crosslink, ubiquitin-proteasome activity and glycolysis, and to suppress angiogenesis processes that commonly occur in various diseases, particularly cancers.
Collapse
Affiliation(s)
| | - Sunisa Yoodee
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Thanyalak Malaitad
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Visith Thongboonkerd
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.
| |
Collapse
|
8
|
Xin Y, Roh K, Cho E, Park D, Whang W, Jung E. Isookanin Inhibits PGE 2-Mediated Angiogenesis by Inducing Cell Arrest through Inhibiting the Phosphorylation of ERK1/2 and CREB in HMEC-1 Cells. Int J Mol Sci 2021; 22:ijms22126466. [PMID: 34208772 PMCID: PMC8234715 DOI: 10.3390/ijms22126466] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/09/2021] [Accepted: 06/14/2021] [Indexed: 11/16/2022] Open
Abstract
Inflammation is increasingly recognized as a critical mediator of angiogenesis, and unregulated angiogenic responses often involve human diseases. The importance of regulating angiogenesis in inflammatory diseases has been demonstrated through some successful cases of anti-angiogenesis therapies in related diseases, including arthritis, but it has been reported that some synthetic types of antiangiogenic drugs have potential side effects. In recent years, the importance of finding alternative strategies for regulating angiogenesis has begun to attract the attention of researchers. Therefore, identification of natural ingredients used to prevent or treat angiogenesis-related diseases will play a greater role. Isookanin is a phenolic flavonoid presented in Bidens extract, and it has been reported that isookanin possesses some biological properties, including antioxidative and anti-inflammatory effects, anti-diabetic properties, and an ability to inhibit α-amylase. However, its antiangiogenic effects and mechanism thereof have not been studied yet. In this study, our results indicate that isookanin has an effective inhibitory effect on the angiogenic properties of microvascular endothelial cells. Isookanin shows inhibitory effects in multiple stages of PGE2-induced angiogenesis, including the growth, proliferation, migration, and tube formation of microvascular endothelial cells. In addition, isookanin induces cell cycle arrest in S phase, which is also the reason for subsequent inhibition of cell proliferation. The mechanism of inhibiting angiogenesis by isookanin is related to the inhibition of PGE2-mediated ERK1/2 and CREB phosphorylation. These findings make isookanin a potential candidate for the treatment of angiogenesis-related diseases.
Collapse
Affiliation(s)
- Yingji Xin
- Biospectrum Life Science Institute, Yongin 16827, Korea; (Y.X.); (K.R.); (E.C.); (D.P.)
- Department of Global Innovative Drug, Graduate School, College of Pharmacy, Chung-Ang University, Heukseok-dong, Dongjak-gu, Seoul 156756, Korea
| | - Kyungbaeg Roh
- Biospectrum Life Science Institute, Yongin 16827, Korea; (Y.X.); (K.R.); (E.C.); (D.P.)
| | - Eunae Cho
- Biospectrum Life Science Institute, Yongin 16827, Korea; (Y.X.); (K.R.); (E.C.); (D.P.)
| | - Deokhoon Park
- Biospectrum Life Science Institute, Yongin 16827, Korea; (Y.X.); (K.R.); (E.C.); (D.P.)
| | - Wankyunn Whang
- Department of Global Innovative Drug, Graduate School, College of Pharmacy, Chung-Ang University, Heukseok-dong, Dongjak-gu, Seoul 156756, Korea
- Correspondence: (W.W.); (E.J.); Tel.: +82-70-5117-0043 (E.J.)
| | - Eunsun Jung
- Biospectrum Life Science Institute, Yongin 16827, Korea; (Y.X.); (K.R.); (E.C.); (D.P.)
- Correspondence: (W.W.); (E.J.); Tel.: +82-70-5117-0043 (E.J.)
| |
Collapse
|
9
|
Kasprzak-Drozd K, Oniszczuk T, Stasiak M, Oniszczuk A. Beneficial Effects of Phenolic Compounds on Gut Microbiota and Metabolic Syndrome. Int J Mol Sci 2021; 22:3715. [PMID: 33918284 PMCID: PMC8038165 DOI: 10.3390/ijms22073715] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/25/2021] [Accepted: 03/30/2021] [Indexed: 12/12/2022] Open
Abstract
The human intestine contains an intricate community of microorganisms, referred to as the gut microbiota (GM), which plays a pivotal role in host homeostasis. Multiple factors could interfere with this delicate balance, including genetics, age, medicines and environmental factors, particularly diet. Growing evidence supports the involvement of GM dysbiosis in gastrointestinal (GI) and extraintestinal metabolic diseases. The beneficial effects of dietary polyphenols in preventing metabolic diseases have been subjected to intense investigation over the last twenty years. As our understanding of the role of the gut microbiota advances and our knowledge of the antioxidant and anti-inflammatory functions of polyphenols accumulates, there emerges a need to examine the prebiotic role of dietary polyphenols. This review firstly overviews the importance of the GM in health and disease and then reviews the role of dietary polyphenols on the modulation of the gut microbiota, their metabolites and how they impact on host health benefits. Inter-dependence between the gut microbiota and polyphenol metabolites and the vital balance between the two in maintaining the host gut homeostasis are also discussed.
Collapse
Affiliation(s)
- Kamila Kasprzak-Drozd
- Department of Inorganic Chemistry, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Tomasz Oniszczuk
- Department of Thermal Technology and Food Process Engineering, University of Life Sciences in Lublin, Głęboka 31, 20-612 Lublin, Poland
| | - Mateusz Stasiak
- Institute of Agrophysics, Polish Academy of Sciences, Doświadczalna 4, 20-290 Lublin, Poland;
| | - Anna Oniszczuk
- Department of Inorganic Chemistry, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| |
Collapse
|
10
|
Vo TH, Lin YC, Liaw CC, Pan WP, Cheng JJ, Lee CK, Kuo YH. Triterpene glycosides and phenylpropane derivatives from Staurogyne concinnula possessing anti-angiogenic activity. PHYTOCHEMISTRY 2021; 184:112666. [PMID: 33524858 DOI: 10.1016/j.phytochem.2021.112666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 01/06/2021] [Accepted: 01/09/2021] [Indexed: 06/12/2023]
Abstract
After anti-angiogenic activity screening, the potential n-butanol layer partitioned from the ethanol extract of Staurogyne concinnula was conducted. Further purification by Diaion HP20 column and preparative HPLC chromatography, four undescribed triterpenoid saponin derivatives, along with the known baptisiasaponin I, and four known phenylpropanoid glycosides were isolated and characterized from n-butanol layer. The structures of isolated compounds were elucidated by ESI-MS, 1D, and 2D MNR data. Biological evaluation revealed that baptisiasaponin I possessed significant anti-angiogenic effects (IC50 4.0 ± 0.2 μM). Further mechanism of action of baptisiasaponin I by inhibition of integrin/FAK/paxillin signaling pathway and its downstream effectors as MMP2 and MMP9 are also presented.
Collapse
Affiliation(s)
- Thanh-Hoa Vo
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan; National Research Institute of Chinese Medicine, Taipei, 11221, Taiwan
| | - Yu-Chi Lin
- National Research Institute of Chinese Medicine, Taipei, 11221, Taiwan; Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan
| | - Chia-Ching Liaw
- National Research Institute of Chinese Medicine, Taipei, 11221, Taiwan; Department of Biochemical Science and Technology, National Chiayi University, Chiayi, 60004, Taiwan
| | - Wen-Pin Pan
- National Research Institute of Chinese Medicine, Taipei, 11221, Taiwan
| | - Jing-Jy Cheng
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan; National Research Institute of Chinese Medicine, Taipei, 11221, Taiwan.
| | - Ching-Kuo Lee
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan; School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan.
| | - Yao-Haur Kuo
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan; National Research Institute of Chinese Medicine, Taipei, 11221, Taiwan; Graduate Institute of Intergrated Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan.
| |
Collapse
|
11
|
Astaxanthin mediated regulation of VEGF through HIF1α and XBP1 signaling pathway: An insight from ARPE-19 cell and streptozotocin mediated diabetic rat model. Exp Eye Res 2021; 206:108555. [PMID: 33789142 DOI: 10.1016/j.exer.2021.108555] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 03/08/2021] [Accepted: 03/23/2021] [Indexed: 11/21/2022]
Abstract
Breakdown of outer blood-retina barrier (BRB) has been associated with the pathogenesis of diabetic retinopathy (DR) and diabetic macular edema (DME). Vascular endothelial growth factor (VEGF) might play a detrimental role in the pathogenesis of DME, a major clinical manifestation of DR. In the present study, we investigated the inhibitory mechanism of astaxanthin on VEGF and its upstream signaling pathways under in vitro and in vivo conditions. Astaxanthin has been observed to downregulate VEGF expression under hyperglycemic (HG) and CoCl2 induced hypoxic conditions in ARPE-19 cells. There were compelling pieces of evidence for the involvement of transcription factors like HIF1α and XBP1 in the upregulation of VEGF under HG and hypoxic conditions. Thus, we investigated the role of astaxanthin in the expression and nuclear translocation of HIF1α and XBP1. The activation and translocation of HIF1α and XBP1 induced by HG or CoCl2 conditions were hindered by astaxanthin. Additionally, treatment with HIF1α siRNA and IRE1 inhibitor STF-083010 also inhibited the expression of VEGF induced by HG and CoCl2 conditions. These results indicated that the anti-VEGF property of astaxanthin might be associated with the downregulation of HIF1α and XBP1. Furthermore, astaxanthin mitigated the enhanced migration of retinal pigment epithelial (RPE) cells under DR conditions. As well, astaxanthin protected disorganization of zona occludin-1 (ZO-1) tight junction protein in RPE and reduced HG or hypoxic induced permeability of RPE cells. In streptozotocin-induced diabetic rat model, astaxanthin reduced the expression of HIF1α, XBP1, and VEGF as well as protected the abnormalities in the retinal layers induced by diabetes condition. Thus, astaxanthin may be used as a potential nutraceutical to prevent or treat retinal dysfunction in diabetic patients.
Collapse
|
12
|
Theasaponin E 1 Inhibits Platinum-Resistant Ovarian Cancer Cells through Activating Apoptosis and Suppressing Angiogenesis. Molecules 2021; 26:molecules26061681. [PMID: 33802884 PMCID: PMC8002815 DOI: 10.3390/molecules26061681] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/14/2021] [Accepted: 03/15/2021] [Indexed: 12/28/2022] Open
Abstract
Novel therapeutic strategies for ovarian cancer treatment are in critical need due to the chemoresistance and adverse side effects of platinum-based chemotherapy. Theasaponin E1 (TSE1) is an oleanane-type saponin from Camellia sinensis seeds. Its apoptosis-inducing, cell cycle arresting and antiangiogenesis activities against platinum-resistant ovarian cancer cells were elucidated in vitro and using the chicken chorioallantoic membrane (CAM) assay. The results showed that TSE1 had more potent cell growth inhibitory effects on ovarian cancer OVCAR-3 and A2780/CP70 cells than cisplatin and was lower in cytotoxicity to normal ovarian IOSE-364 cells. TSE1 significantly induced OVCAR-3 cell apoptosis via the intrinsic and extrinsic apoptotic pathways, slightly arresting cell cycle at the G2/M phase, and obviously inhibited OVCAR-3 cell migration and angiogenesis with reducing the protein secretion and expression of vascular endothelial growth factor (VEGF). Western bolt assay showed that Serine/threonine Kinase (Akt) signaling related proteins including Ataxia telangiectasia mutated kinase (ATM), Phosphatase and tensin homolog (PTEN), Akt, Mammalian target of rapamycin (mTOR), Ribosome S6 protein kinase (p70S6K) and e IF4E-binding protein 1(4E-BP1) were regulated, and Hypoxia inducible factor-1α (HIF-1α) protein expression was decreased by TSE1 in OVCAR-3 cells. Moreover, TSE1 treatment potently downregulated protein expression of the Notch ligands including Delta-like protein 4 (Dll4) and Jagged1, and reduced the protein level of the intracellular domain (NICD) of Notch1. Combination treatment of TSE1 with the Notch1 signaling inhibitor tert-butyl (2S)-2-[[(2S)-2-[[2-(3,5-difluorophenyl)acetyl]amino]propanoyl]amino]-2-phenylacetate (DAPT), or the Akt signaling inhibitor wortmannin, showed a stronger inhibition toward HIF-1α activation compared with single compound treatment. Taken together, TSE1 might be a potential candidate compound for improving platinum-resistant ovarian cancer treatment via Dll4/Jagged1-Notch1-Akt-HIF-1α axis.
Collapse
|
13
|
Almnhawy M, Jebur M, Alhajamee M, Marai K, Tabrizi MH. PLGA-Based Nano-Encapsulation of Trachyspermum Ammi Seed Essential Oil (TSEO-PNP) as a Safe, Natural, Efficient, Anticancer Compound in Human HT-29 Colon Cancer Cell Line. Nutr Cancer 2020; 73:2808-2820. [PMID: 33319599 DOI: 10.1080/01635581.2020.1862256] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Colorectal cancer is a lethal and commonly diagnosed cancer worldwide. To halt its burden more efficient targeted strategies are needed. Trachyspermum ammi seed essential oil (TSEO) contains several anticancer phytochemicals that maybe more effective via PLGA-based nano-encapsulation. TSEO-PNP nanoparticles were synthesized utilizing evaporation and ultra-sonication-based emulsification methods. Their size, morphology, and stability were defined by DLS, SEM, and surface zeta-potential data, respectively. The TSEO-PNP antioxidant apoptotic, cytotoxic, and antiangiogenic impacts on both cell lines (HT-29 and HUVEC) were studied by FRAP/ABTS, Q-PCR, MTT, and CAM assays, respectively. Moreover, further confirmatory measurements such as AO/EB fluorescent staining and flow cytometry analysis were performed to verify apoptosis. Stable (-32.42 mV) 206.21-nm TSEO-PNP induced apoptosis in the HT-29 cells. Apoptosis was confirmed by significant overexpression of apoptotic genes (Cas-9 and BAX), down-regulation of the anti-apoptotic (BCL-2) gene, fluorescent AO/EB staining, and flow cytometry data following increased TSEO-PNP treatment doses. TSEO-PNP exhibited a meaningful dose- and time-dependent cancer-specific cytotoxic impact on HT-29 cells. The TSEO-PNP has three main anticancer activities on HT-29 colon cancer cells including oxidant reduction, apoptosis induction, and angiogenesis suppression.
Collapse
Affiliation(s)
- Mokhalad Almnhawy
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Mohammed Jebur
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Maitham Alhajamee
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Khadeeja Marai
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | | |
Collapse
|
14
|
Ochiai K, Yamaoka M, Swaminathan A, Shima H, Hiura H, Matsumoto M, Kurotaki D, Nakabayashi J, Funayama R, Nakayama K, Arima T, Ikawa T, Tamura T, Sciammas R, Bouvet P, Kundu TK, Igarashi K. Chromatin Protein PC4 Orchestrates B Cell Differentiation by Collaborating with IKAROS and IRF4. Cell Rep 2020; 33:108517. [PMID: 33357426 DOI: 10.1016/j.celrep.2020.108517] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 10/10/2020] [Accepted: 11/22/2020] [Indexed: 12/24/2022] Open
Abstract
The chromatin protein positive coactivator 4 (PC4) has multiple functions, including chromatin compaction. However, its role in immune cells is largely unknown. We show that PC4 orchestrates chromatin structure and gene expression in mature B cells. B-cell-specific PC4-deficient mice show impaired production of antibody upon antigen stimulation. The PC4 complex purified from B cells contains the transcription factors (TFs) IKAROS and IRF4. IKAROS protein is reduced in PC4-deficient mature B cells, resulting in de-repression of their target genes in part by diminished interactions with gene-silencing components. Upon activation, the amount of IRF4 protein is not increased in PC4-deficient B cells, resulting in reduction of plasma cells. Importantly, IRF4 reciprocally induces PC4 expression via a super-enhancer. PC4 knockdown in human B cell lymphoma and myeloma cells reduces IKAROS protein as an anticancer drug, lenalidomide. Our findings establish PC4 as a chromatin regulator of B cells and a possible therapeutic target adjoining IKAROS in B cell malignancies.
Collapse
Affiliation(s)
- Kyoko Ochiai
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Sendai 980-8575, Japan.
| | - Mari Yamaoka
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Sendai 980-8575, Japan
| | - Amrutha Swaminathan
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore 560064, India
| | - Hiroki Shima
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Sendai 980-8575, Japan
| | - Hitoshi Hiura
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Sendai 980-8575, Japan
| | - Mitsuyo Matsumoto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Sendai 980-8575, Japan; Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Sendai 980-8575, Japan
| | - Daisuke Kurotaki
- Department of Immunology, Yokohama City University Graduate School of Medicine, Fukuura 3-9, Yokohama 236-0004, Japan
| | - Jun Nakabayashi
- Advanced Medical Research Center, Yokohama City University, Fukuura 3-9, Yokohama 236-0004, Japan
| | - Ryo Funayama
- Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Sendai 980-8575, Japan; Division of Cell Proliferation, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Sendai 980-8575, Japan
| | - Keiko Nakayama
- Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Sendai 980-8575, Japan; Division of Cell Proliferation, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Sendai 980-8575, Japan
| | - Takahiro Arima
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Sendai 980-8575, Japan
| | - Tomokatsu Ikawa
- Division of Immunobiology, Tokyo University of Science, Yamazaki 2669, Noda 278-0022, Japan
| | - Tomohiko Tamura
- Department of Immunology, Yokohama City University Graduate School of Medicine, Fukuura 3-9, Yokohama 236-0004, Japan; Advanced Medical Research Center, Yokohama City University, Fukuura 3-9, Yokohama 236-0004, Japan
| | - Roger Sciammas
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, CA 95616, USA
| | - Philippe Bouvet
- Université de Lyon, Ecole Normale Supérieure de Lyon, Centre de Recherche en Cancérologie de Lyon, Cancer Cell Plasticity Department, UMR INSERM 1052 CNRS 5286, Centre Léon Bérard, Lyon, France
| | - Tapas K Kundu
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore 560064, India.
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Sendai 980-8575, Japan; Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Sendai 980-8575, Japan.
| |
Collapse
|
15
|
Limanaqi F, Biagioni F, Mastroiacovo F, Polzella M, Lazzeri G, Fornai F. Merging the Multi-Target Effects of Phytochemicals in Neurodegeneration: From Oxidative Stress to Protein Aggregation and Inflammation. Antioxidants (Basel) 2020; 9:antiox9101022. [PMID: 33092300 PMCID: PMC7589770 DOI: 10.3390/antiox9101022] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/14/2020] [Accepted: 10/16/2020] [Indexed: 02/08/2023] Open
Abstract
Wide experimental evidence has been provided in the last decade concerning the neuroprotective effects of phytochemicals in a variety of neurodegenerative disorders. Generally, the neuroprotective effects of bioactive compounds belonging to different phytochemical classes are attributed to antioxidant, anti-aggregation, and anti-inflammatory activity along with the restoration of mitochondrial homeostasis and targeting alterations of cell-clearing systems. Far from being independent, these multi-target effects represent interconnected events that are commonly implicated in the pathogenesis of most neurodegenerative diseases, independently of etiology, nosography, and the specific misfolded proteins being involved. Nonetheless, the increasing amount of data applying to a variety of neurodegenerative disorders joined with the multiple effects exerted by the wide variety of plant-derived neuroprotective agents may rather confound the reader. The present review is an attempt to provide a general guideline about the most relevant mechanisms through which naturally occurring agents may counteract neurodegeneration. With such an aim, we focus on some popular phytochemical classes and bioactive compounds as representative examples to design a sort of main highway aimed at deciphering the most relevant protective mechanisms which make phytochemicals potentially useful in counteracting neurodegeneration. In this frame, we emphasize the potential role of the cell-clearing machinery as a kernel in the antioxidant, anti-aggregation, anti-inflammatory, and mitochondrial protecting effects of phytochemicals.
Collapse
Affiliation(s)
- Fiona Limanaqi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy;
| | - Francesca Biagioni
- Istituto di Ricovero e Cura a Carattere Scientifico Neuromed, Via Atinense 18, 86077 Pozzilli, Italy; (F.B.); (F.M.)
| | - Federica Mastroiacovo
- Istituto di Ricovero e Cura a Carattere Scientifico Neuromed, Via Atinense 18, 86077 Pozzilli, Italy; (F.B.); (F.M.)
| | - Maico Polzella
- Aliveda Laboratories, Viale Karol Wojtyla 19, 56042 Crespina Lorenzana, Italy;
| | - Gloria Lazzeri
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy;
- Correspondence: (G.L.); (F.F.)
| | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy;
- Istituto di Ricovero e Cura a Carattere Scientifico Neuromed, Via Atinense 18, 86077 Pozzilli, Italy; (F.B.); (F.M.)
- Correspondence: (G.L.); (F.F.)
| |
Collapse
|
16
|
Liskova A, Koklesova L, Samec M, Varghese E, Abotaleb M, Samuel SM, Smejkal K, Biringer K, Petras M, Blahutova D, Bugos O, Pec M, Adamkov M, Büsselberg D, Ciccocioppo R, Adamek M, Rodrigo L, Caprnda M, Kruzliak P, Kubatka P. Implications of flavonoids as potential modulators of cancer neovascularity. J Cancer Res Clin Oncol 2020; 146:3079-3096. [PMID: 32902794 DOI: 10.1007/s00432-020-03383-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 09/01/2020] [Indexed: 02/06/2023]
Abstract
PURPOSE The formation of new blood vessels from previous ones, angiogenesis, is critical in tissue repair, expansion or remodeling in physiological processes and in various pathologies including cancer. Despite that, the development of anti-angiogenic drugs has great potential as the treatment of cancer faces many problems such as development of the resistance to treatment or an improperly selected therapy approach. An evaluation of predictive markers in personalized medicine could significantly improve treatment outcomes in many patients. METHODS This comprehensive review emphasizes the anticancer potential of flavonoids mediated by their anti-angiogenic efficacy evaluated in current preclinical and clinical cancer research. RESULTS AND CONCLUSION Flavonoids are important groups of phytochemicals present in common diet. Flavonoids show significant anticancer effects. The anti-angiogenic effects of flavonoids are currently a widely discussed topic of preclinical cancer research. Flavonoids are able to regulate the process of tumor angiogenesis through modulation of signaling molecules such as VEGF, MMPs, ILs, HIF or others. However, the evaluation of the anti-angiogenic potential of flavonoids within the clinical studies is not frequently discussed and is still of significant scientific interest.
Collapse
Affiliation(s)
- Alena Liskova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Lenka Koklesova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Marek Samec
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Elizabeth Varghese
- Department of Physiology and Biophysics, Weill Cornell Medicine in Qatar, Education City, Qatar Foundation, Doha, 24144, Qatar
| | - Mariam Abotaleb
- Department of Physiology and Biophysics, Weill Cornell Medicine in Qatar, Education City, Qatar Foundation, Doha, 24144, Qatar
| | - Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine in Qatar, Education City, Qatar Foundation, Doha, 24144, Qatar
| | - Karel Smejkal
- Department of Natural Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Brno, Czech Republic
| | - Kamil Biringer
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Martin Petras
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - Dana Blahutova
- Department of Biology and Ecology, Faculty of Education, Catholic University in Ruzomberok, Ruzomberok, Slovakia
| | | | - Martin Pec
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01, Martin, Slovakia
| | - Marian Adamkov
- Department of Histology and Embryology, Jessenius Faculty of Medicine, Comenius University, Martin, Slovakia
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine in Qatar, Education City, Qatar Foundation, Doha, 24144, Qatar.
| | - Rachele Ciccocioppo
- Gastroenterology Unit, Department of Medicine, Azienda Ospedaliera Universitaria Integrata Policlinico GB Rossi, University of Verona, Verona, Italy
| | - Mariusz Adamek
- Department of Thoracic Surgery, Faculty of Medicine and Dentistry, Medical University of Silesia, Katowice, Poland
| | - Luis Rodrigo
- Faculty of Medicine, University of Oviedo, Central University Hospital of Asturias (HUCA), Oviedo, Spain
| | - Martin Caprnda
- 1st Department of Internal Medicine, Faculty of Medicine, Comenius University and University Hospital, Bratislava, Slovakia
| | - Peter Kruzliak
- 2nd Department of Surgery, Faculty of Medicine, Masaryk University, Pekarska 53, 656 91, Brno, Czech Republic. .,St. Anne's University Hospital, Brno, Czech Republic.
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01, Martin, Slovakia.
| |
Collapse
|
17
|
Debele TA, Wu HC, Wu SR, Shan YS, Su WP. Combination Delivery of Alpha-Tocopheryl Succinate and Curcumin Using a GSH-Sensitive Micelle (PAH-SS-PLGA) to Treat Pancreatic Cancer. Pharmaceutics 2020; 12:pharmaceutics12080778. [PMID: 32824299 PMCID: PMC7464675 DOI: 10.3390/pharmaceutics12080778] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/13/2020] [Accepted: 08/14/2020] [Indexed: 01/13/2023] Open
Abstract
Pancreatic cancer is one of the highest causes of mortality throughout the world; thus, it requires an effective treatment strategy. Some chemotherapeutic agents used in the clinics or under clinical trials are hydrophobic and have poor aqueous solubility; consequently, they also have minimal systemic bioavailability. Nanoparticle-based drug delivery tactics have the potential for overcoming these limitations and enhancing their therapeutic efficacy. Herein, a glutathione (GSH)-sensitive micelle (PAH-SS-PLGA) was synthesized for the combined delivery of alpha-tocopheryl succinate (TOS) and curcumin to improve its therapeutic efficacy. The chemical structures of PAH-SS-PLGA were analyzed using Proton Nuclear Magnetic Resonance (1H-NMR) and Fourier Transform Infrared (FTIR) spectroscopy, whereas the particle size, zeta potential, and surface morphology were observed using dynamic light scattering (DLS) and transmission electron microscopy (TEM). In vitro drug release results revealed that more TOS and curcumin were released in the presence of GSH (5 mM) than the physiological pH value. Fluorescence microscopy images revealed that nanoformulated curcumin/rhodamine was uptaken by PAN02 pancreatic cancer cells. In vitro cytotoxicity assays showed higher cytotoxicity for nanoformulated TOS and/or curcumin than free TOS and/or curcumin. In addition, higher cytotoxicity was observed for combination drugs than free drugs alone. Most interestingly, at all tested concentrations of nanoformulated drugs (PAH-SS-PLGA, TOS, and curcumin), the calculated combination index (CI) value was less than one, which shows that TOS and curcumin have a synergistic effect on cellular proliferation inhibition. Overall, synthesized co-polymers are the best carriers for combination drugs, TOS, and curcumin, because they enhance the therapeutic efficacy and improve pancreatic cancer treatments.
Collapse
Affiliation(s)
- Tilahun Ayane Debele
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, No.138, Sheng Li Road, Tainan 704, Taiwan; (T.A.D.); (Y.-S.S.)
| | - Hung-Chang Wu
- Department of Internal Medicine, Chi Mei Medical Center, Tainan 710, Taiwan;
| | - Shang-Rung Wu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan;
- Department of Dentistry & Institute of Oral Medicine, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| | - Yan-Shen Shan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, No.138, Sheng Li Road, Tainan 704, Taiwan; (T.A.D.); (Y.-S.S.)
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| | - Wen-Pin Su
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, No.138, Sheng Li Road, Tainan 704, Taiwan; (T.A.D.); (Y.-S.S.)
- Departments of Oncology and Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
- Correspondence: ; Tel.: +886-6-2353535 (ext. 4252)
| |
Collapse
|
18
|
Kargozar S, Baino F, Hamzehlou S, Hamblin MR, Mozafari M. Nanotechnology for angiogenesis: opportunities and challenges. Chem Soc Rev 2020; 49:5008-5057. [PMID: 32538379 PMCID: PMC7418030 DOI: 10.1039/c8cs01021h] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Angiogenesis plays a critical role within the human body, from the early stages of life (i.e., embryonic development) to life-threatening diseases (e.g., cancer, heart attack, stroke, wound healing). Many pharmaceutical companies have expended huge efforts on both stimulation and inhibition of angiogenesis. During the last decade, the nanotechnology revolution has made a great impact in medicine, and regulatory approvals are starting to be achieved for nanomedicines to treat a wide range of diseases. Angiogenesis therapies involve the inhibition of angiogenesis in oncology and ophthalmology, and stimulation of angiogenesis in wound healing and tissue engineering. This review aims to summarize nanotechnology-based strategies that have been explored in the broad area of angiogenesis. Lipid-based, carbon-based and polymeric nanoparticles, and a wide range of inorganic and metallic nanoparticles are covered in detail. Theranostic and imaging approaches can be facilitated by nanoparticles. Many preparations have been reported to have a bimodal effect where they stimulate angiogenesis at low dose and inhibit it at higher doses.
Collapse
Affiliation(s)
- Saeid Kargozar
- Tissue Engineering Research Group (TERG), Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, 917794-8564 Mashhad, Iran
| | - Francesco Baino
- Institute of Materials Physics and Engineering, Applied Science and Technology Department, Politecnico di Torino, Corso Duca degli Abruzzi 24, 101 29 Torino, Italy
| | - Sepideh Hamzehlou
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Michael R. Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| | - Masoud Mozafari
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
19
|
Fang Y, Wang M, Shen Y, Zhang M, Cao Z, Deng Y. Highly sensitive and selective recognition behaviour for fluoride based on a homoditopic curcumin-difluoroboron receptor. Inorganica Chim Acta 2020. [DOI: 10.1016/j.ica.2020.119413] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
20
|
Majnooni MB, Fakhri S, Smeriglio A, Trombetta D, Croley CR, Bhattacharyya P, Sobarzo-Sánchez E, Farzaei MH, Bishayee A. Antiangiogenic Effects of Coumarins against Cancer: From Chemistry to Medicine. Molecules 2019; 24:molecules24234278. [PMID: 31771270 PMCID: PMC6930449 DOI: 10.3390/molecules24234278] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 11/18/2019] [Accepted: 11/19/2019] [Indexed: 12/21/2022] Open
Abstract
Angiogenesis, the process of formation and recruitment of new blood vessels from pre-existing vessels, plays an important role in the development of cancer. Therefore, the use of antiangiogenic agents is one of the most critical strategies for the treatment of cancer. In addition, the complexity of cancer pathogenicity raises the need for multi-targeting agents. Coumarins are multi-targeting natural agents belonging to the class of benzopyrones. Coumarins have several biological and pharmacological effects, including antimicrobial, antioxidant, anti-inflammation, anticoagulant, anxiolytic, analgesic, and anticancer properties. Several reports have shown that the anticancer effect of coumarins and their derivatives are mediated through targeting angiogenesis by modulating the functions of vascular endothelial growth factor as well as vascular endothelial growth factor receptor 2, which are involved in cancer pathogenesis. In the present review, we focus on the antiangiogenic effects of coumarins and related structure-activity relationships with particular emphasis on cancer.
Collapse
Affiliation(s)
- Mohammad Bagher Majnooni
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah 6714415153, Iran;
| | - Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran;
| | - Antonella Smeriglio
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Palatucci, 98168 Messina, Italy; (A.S.); (D.T.)
| | - Domenico Trombetta
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Palatucci, 98168 Messina, Italy; (A.S.); (D.T.)
| | | | - Piyali Bhattacharyya
- Escuela de Ciencias de la Salud, Universidad Ana G. Méndez, Recinto de Gurabo, Gurabo, PR 00778, USA;
| | - Eduardo Sobarzo-Sánchez
- Laboratory of Pharmaceutical Chemistry, Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; or
- Instituto de Investigación e Innovación en Salud, Facultad de Ciencias de la Salud, Universidad Central de Chile, Santiago 8330507, Chile
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran;
- Correspondence: (M.H.F.); or (A.B.)
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA;
- Correspondence: (M.H.F.); or (A.B.)
| |
Collapse
|
21
|
Han Y, Sengupta S, Lee BJ, Cho H, Kim J, Choi HG, Dash U, Kim JH, Kim ND, Kim JH, Sim T. Identification of a Unique Resorcylic Acid Lactone Derivative That Targets Both Lymphangiogenesis and Angiogenesis. J Med Chem 2019; 62:9141-9160. [PMID: 31513411 DOI: 10.1021/acs.jmedchem.9b01025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We synthesized 11 novel L-783277 derivatives, in which a structure rigidifying phenyl ring is incorporated into the 14-membered chiral resorcylic acid lactone system. The SAR study with these substances demonstrated that 17 possesses excellent kinase selectivity against a panel of 335 kinases in contrast to L-783277 and inhibits VEGFR3, VEGFR2, and FLT3 with single-digit nanomolar IC50 values. Also, we found that 21, a stereoisomer of 17, has excellent potency (IC50 = 9 nM) against VEGFR3 and selectivity over VEGFR2 and FLT3. 17, a potent dual VEGFR3 and VEGFR2 inhibitor, effectively suppresses both lymphangiogenesis and angiogenesis in a 3D-microfluidic tumor lymphangiogenesis assay and in vivo corneal assay while SAR131675 blocks only lymphangiogenesis. In addition, 17 blocks the endothelial tube formation and suppresses proliferation of PHE tumor vascular model. 17 will be a valuable templatefor developing therapeutically active and selective substances that target both lymphangiogenesis and angiogenesis.
Collapse
Affiliation(s)
- Youngsun Han
- KU-KIST Graduate School of Converging Science and Technology , Korea University , 145 Anam-ro, Seongbuk-gu , Seoul 02841 , Republic of Korea
| | - Sandip Sengupta
- Chemical Kinomics Research Center , Korea Institute of Science and Technology (KIST) , 5 Hwarangro 14-gil, Seongbuk-gu , Seoul 02792 , Republic of Korea
| | - Byung Joo Lee
- Fight Against Angiogenesis-Related Blindness Laboratory, Clinical Research Institute , Seoul National University Hospital , 101 Daehak-ro, Jongno-gu , Seoul 110-744 , Republic of Korea.,Department of Biomedical Sciences, College of Medicine , Seoul National University , 103 Daehakro, Jongro-gu , Seoul 110-744 , Republic of Korea
| | - Hanna Cho
- KU-KIST Graduate School of Converging Science and Technology , Korea University , 145 Anam-ro, Seongbuk-gu , Seoul 02841 , Republic of Korea
| | - Jiknyeo Kim
- Chemical Kinomics Research Center , Korea Institute of Science and Technology (KIST) , 5 Hwarangro 14-gil, Seongbuk-gu , Seoul 02792 , Republic of Korea
| | - Hwan Geun Choi
- Chemical Kinomics Research Center , Korea Institute of Science and Technology (KIST) , 5 Hwarangro 14-gil, Seongbuk-gu , Seoul 02792 , Republic of Korea
| | - Uttam Dash
- Chemical Kinomics Research Center , Korea Institute of Science and Technology (KIST) , 5 Hwarangro 14-gil, Seongbuk-gu , Seoul 02792 , Republic of Korea
| | - Jin Hyoung Kim
- Fight Against Angiogenesis-Related Blindness Laboratory, Clinical Research Institute , Seoul National University Hospital , 101 Daehak-ro, Jongno-gu , Seoul 110-744 , Republic of Korea
| | - Nam Doo Kim
- NDBio Therapeutics Inc. , 32 Songdogwahak-ro, Yeonsu-gu , Incheon 21984 , Republic of Korea
| | - Jeong Hun Kim
- Fight Against Angiogenesis-Related Blindness Laboratory, Clinical Research Institute , Seoul National University Hospital , 101 Daehak-ro, Jongno-gu , Seoul 110-744 , Republic of Korea
| | - Taebo Sim
- KU-KIST Graduate School of Converging Science and Technology , Korea University , 145 Anam-ro, Seongbuk-gu , Seoul 02841 , Republic of Korea.,Chemical Kinomics Research Center , Korea Institute of Science and Technology (KIST) , 5 Hwarangro 14-gil, Seongbuk-gu , Seoul 02792 , Republic of Korea
| |
Collapse
|
22
|
Kalampaliki AD, Giannouli V, Skaltsounis AL, Kostakis IK. A Three-Step, Gram-Scale Synthesis of Hydroxytyrosol, Hydroxytyrosol Acetate, and 3,4-Dihydroxyphenylglycol. Molecules 2019; 24:E3239. [PMID: 31492013 PMCID: PMC6767028 DOI: 10.3390/molecules24183239] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 09/01/2019] [Accepted: 09/03/2019] [Indexed: 12/11/2022] Open
Abstract
Hydroxytyrosol and two other polyphenols of olive tree, hydroxytyrosol acetate and 3,4-dihydroxyphenylglycol, are known for a wide range of beneficial activities in human health and prevention from diseases. The inability to isolate high, pure amounts of these natural compounds and the difficult and laborious procedures for the synthesis of them led us to describe herein an efficient, easy, cheap, and scaling up synthetic procedure, from catechol, via microwave irradiation.
Collapse
Affiliation(s)
- Amalia D Kalampaliki
- Division of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771 Athens, Greece.
| | - Vassiliki Giannouli
- Division of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771 Athens, Greece.
| | - Alexios-Leandros Skaltsounis
- Division of Pharmacognosy & Natural Product Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771 Athens, Greece.
| | - Ioannis K Kostakis
- Division of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771 Athens, Greece.
| |
Collapse
|
23
|
Limanaqi F, Biagioni F, Busceti CL, Ryskalin L, Polzella M, Frati A, Fornai F. Phytochemicals Bridging Autophagy Induction and Alpha-Synuclein Degradation in Parkinsonism. Int J Mol Sci 2019; 20:ijms20133274. [PMID: 31277285 PMCID: PMC6651086 DOI: 10.3390/ijms20133274] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 06/30/2019] [Accepted: 07/02/2019] [Indexed: 12/11/2022] Open
Abstract
Among nutraceuticals, phytochemical-rich compounds represent a source of naturally-derived bioactive principles, which are extensively studied for potential beneficial effects in a variety of disorders ranging from cardiovascular and metabolic diseases to cancer and neurodegeneration. In the brain, phytochemicals produce a number of biological effects such as modulation of neurotransmitter activity, growth factor induction, antioxidant and anti-inflammatory activity, stem cell modulation/neurogenesis, regulation of mitochondrial homeostasis, and counteracting protein aggregation through modulation of protein-folding chaperones and the cell clearing systems autophagy and proteasome. In particular, the ability of phytochemicals in restoring proteostasis through autophagy induction took center stage in recent research on neurodegenerative disorders such as Parkinson’s disease (PD). Indeed, autophagy dysfunctions and α-syn aggregation represent two interdependent downstream biochemical events, which concur in the parkinsonian brain, and which are targeted by phytochemicals administration. Therefore, in the present review we discuss evidence about the autophagy-based neuroprotective effects of specific phytochemical-rich plants in experimental parkinsonism, with a special focus on their ability to counteract alpha-synuclein aggregation and toxicity. Although further studies are needed to confirm the autophagy-based effects of some phytochemicals in parkinsonism, the evidence discussed here suggests that rescuing autophagy through natural compounds may play a role in preserving dopamine (DA) neuron integrity by counteracting the aggregation, toxicity, and prion-like spreading of α-syn, which remains a hallmark of PD.
Collapse
Affiliation(s)
- Fiona Limanaqi
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa (PI), Italy
| | | | | | - Larisa Ryskalin
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa (PI), Italy
| | - Maico Polzella
- Aliveda Laboratories, Crespina Lorenzana, 56042 Pisa (PI), Italy
| | | | - Francesco Fornai
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa (PI), Italy.
- I.R.C.C.S Neuromed, Via Atinense, 86077 Pozzilli (IS), Italy.
| |
Collapse
|