1
|
Kadry MO. Resveratrol-based nano-formulations as an emerging therapeutic strategy for ovarian carcinoma: autophagy stimulation and SIRT-1/Beclin/MMP-9/P53/AKT signaling. Cancer Nanotechnol 2024; 15:36. [DOI: 10.1186/s12645-024-00274-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 06/25/2024] [Indexed: 01/05/2025] Open
Abstract
Abstract
Background
Resveratrol (RVS) is a stilbene derivative polyphenolic compound extensively recognized for its anti-inflammatory, antioxidant and anti-aging properties, along with its enormous promise in carcinoma treatment. Unfortunately, the oral supplementation of RVS possesses physicochemical and pharmacokinetic constraints that hinder its effects, necessitating the development of suitable administration strategies to improve its effectiveness. As a result, the current study evaluates the use of resveratrol nano-formulations in ovarian cancer therapy. Ovarian cancer was induced in rats using (35 mg/kg BW) 20-Methyl cholanthrene (20-MC) followed by resveratrol and resveratrol nano-formulations therapy for one month.
Results
20-MC highlighted a noticeable alleviation in autophagy (ATF) biomarkers SIRT-1 and Beclin, inflammatory and apoptotic biomarkers MMP-9, P53 and AKT in addition to oxidative and nitrosative stress biomarkers TAC and NOX and ovarian cancer tumor biomarker CA-125.
Conclusions
Resveratrol and resveratrol nano-formulations modulated autophagy, inflammatory and oxidative stress biomarkers with the upper effect for resveratrol nano-formulations in competing 20-MC-induced ovarian cancer.
Collapse
|
2
|
Najafiyan B, Bokaii Hosseini Z, Esmaelian S, Firuzpour F, Rahimipour Anaraki S, Kalantari L, Hheidari A, Mesgari H, Nabi-Afjadi M. Unveiling the potential effects of resveratrol in lung cancer treatment: Mechanisms and nanoparticle-based drug delivery strategies. Biomed Pharmacother 2024; 172:116207. [PMID: 38295754 DOI: 10.1016/j.biopha.2024.116207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 03/03/2024] Open
Abstract
Lung cancer ranks among the most prevalent forms of cancer and remains a significant factor in cancer-related mortality across the world. It poses significant challenges to healthcare systems and society as a whole due to its high incidence, mortality rates, and late-stage diagnosis. Resveratrol (RV), a natural compound found in various plants, has shown potential as a nanomedicine for lung cancer treatment. RV has varied effects on cancer cells, including promoting apoptosis by increasing pro-apoptotic proteins (Bax and Bak) and decreasing anti-apoptotic proteins (Bcl-2). It also hinders cell proliferation by influencing important signaling pathways (MAPK, mTOR, PI3K/Akt, and Wnt/β-catenin) that govern cancer progression. In addition, RV acts as a potent antioxidant, diminishing oxidative stress and safeguarding cells against DNA damage. However, using RV alone in cancer treatment has drawbacks, such as low bioavailability, lack of targeting ability, and susceptibility to degradation. In contrast, nanoparticle-based delivery systems address these limitations and hold promise for improving treatment outcomes in lung cancer; nanoparticle formulations of RV offer advantages such as improved drug delivery, increased stability, controlled release, and targeted delivery to lung cancer cells. This article will provide an overview of lung cancer, explore the potential of RV as a therapeutic agent, discuss the benefits and challenges of nanoparticle-based drug delivery, and highlight the promise of RV nanoparticles for cancer treatment, including lung cancer. By optimizing these systems for clinical application, future studies aim to enhance overall treatment outcomes and improve the prognosis for lung cancer patients.
Collapse
Affiliation(s)
- Behnam Najafiyan
- Faculty of Pharmacy, Shiraz University of Medical Science, Shiraz, Iran
| | | | - Samar Esmaelian
- Faculty of Dentistry, Islamic Azad University, Tehran Branch, Tehran, Iran
| | - Faezeh Firuzpour
- Student of Research Committee, Babol University of Medical Sciences, Babol, Iran
| | | | - Leila Kalantari
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Ali Hheidari
- Department of Mechanical Engineering, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Hassan Mesgari
- Oral and Maxillofacial Surgery Department, Faculty of Dentistry, Islamic Azad University, Tehran Branch, Tehran, Iran.
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
3
|
Fatehi R, Rashedinia M, Akbarizadeh AR, Zamani M, Firouzabadi N. Metformin enhances anti-cancer properties of resveratrol in MCF-7 breast cancer cells via induction of apoptosis, autophagy and alteration in cell cycle distribution. Biochem Biophys Res Commun 2023; 644:130-139. [PMID: 36641965 DOI: 10.1016/j.bbrc.2022.12.069] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 11/26/2022] [Accepted: 12/22/2022] [Indexed: 12/25/2022]
Abstract
Breast cancer is the fifth leading cause of death, worldwide affecting both genders. Accumulating evidence suggests that metformin, an oral hypoglycemic agent used in the management of type 2 diabetes, exerts anti-tumor effects in many cancers, including the breast cancer. Resveratrol, a natural product found abundantly in many fruits, exhibits marked cytotoxic and pro-oxidant effects. This study was designed to investigate the effect of metformin in combination with resveratrol and cisplatin in MCF-7 cells. Study groups were as follows: untreated control group, single treatment groups (metformin, resveratrol, and cisplatin), double treatment groups (metformin + resveratrol, metformin + cisplatin, and cisplatin + resveratrol) and triple treatment groups (metformin + resveratrol + cisplatin). Our results indicated that metformin inhibits proliferation of MCF-7 cells, an effect that was associated with ROS production and G0/G1 cell cycle arrest, but not apoptosis. Moreover, resveratrol suppressed the proliferation of MCF-7 cells by induction of apoptosis as well as cell cycle arrest. Notably, a significant inhibitory effect in the co-treatment of metformin, resveratrol, and cisplatin was observed which was attributed to induction of autophagy-mediated cell death and apoptosis along cell cycle arrest. In conclusion, our results advocate the anti-cancer properties of metformin and resveratrol on MCF-7 cell s via induction of cell cycle arrest. Additionally, synergistic anti-cancer effects of metformin in a triple combination with cisplatin and resveratrol was attributed to induction of autophagy-mediated cell death and apoptosis along cell cycle arrest. Based on our findings it is proposed that patients may benefit from addition of a drug with a safe profile to conventional anticancer therapies.
Collapse
Affiliation(s)
- Reihaneh Fatehi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marzieh Rashedinia
- Food and Supplements Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Medicinal Plants Processing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amin Reza Akbarizadeh
- Department of Quality Control, Food and Drug, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mozhdeh Zamani
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Firouzabadi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
4
|
Otálora-Otálora BA, González Prieto C, Guerrero L, Bernal-Forigua C, Montecino M, Cañas A, López-Kleine L, Rojas A. Identification of the Transcriptional Regulatory Role of RUNX2 by Network Analysis in Lung Cancer Cells. Biomedicines 2022; 10:3122. [PMID: 36551878 PMCID: PMC9775089 DOI: 10.3390/biomedicines10123122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 12/07/2022] Open
Abstract
The use of a new bioinformatics pipeline allowed the identification of deregulated transcription factors (TFs) coexpressed in lung cancer that could become biomarkers of tumor establishment and progression. A gene regulatory network (GRN) of lung cancer was created with the normalized gene expression levels of differentially expressed genes (DEGs) from the microarray dataset GSE19804. Moreover, coregulatory and transcriptional regulatory network (TRN) analyses were performed for the main regulators identified in the GRN analysis. The gene targets and binding motifs of all potentially implicated regulators were identified in the TRN and with multiple alignments of the TFs' target gene sequences. Six transcription factors (E2F3, FHL2, ETS1, KAT6B, TWIST1, and RUNX2) were identified in the GRN as essential regulators of gene expression in non-small-cell lung cancer (NSCLC) and related to the lung tumoral process. Our findings indicate that RUNX2 could be an important regulator of the lung cancer GRN through the formation of coregulatory complexes with other TFs related to the establishment and progression of lung cancer. Therefore, RUNX2 could become an essential biomarker for developing diagnostic tools and specific treatments against tumoral diseases in the lung after the experimental validation of its regulatory function.
Collapse
Affiliation(s)
- Beatriz Andrea Otálora-Otálora
- Grupo de Investigación INPAC, Unidad de Investigación, Fundación Universitaria Sanitas, Bogotá 110131, Colombia
- Facultad de Medicina, Universidad Nacional de Colombia, Bogotá 11001, Colombia
| | | | - Lucia Guerrero
- Departamento de Estadística, Universidad Nacional de Colombia, Bogotá 11001, Colombia
| | - Camila Bernal-Forigua
- Instituto de Genética Humana, Facultad de Medicina, Pontificia Universidad Javeriana, Bogotá 110211, Colombia
| | - Martin Montecino
- Institute of Biomedical Sciences, Facultad de Medicina y Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago 8370134, Chile
| | - Alejandra Cañas
- Departamento de Medicina Interna, Facultad de Medicina, Pontificia Universidad Javeriana, Bogotá 110211, Colombia
- Unidad de Neumología, Hospital Universitario San Ignacio, Bogotá 110211, Colombia
| | - Liliana López-Kleine
- Departamento de Estadística, Universidad Nacional de Colombia, Bogotá 11001, Colombia
| | - Adriana Rojas
- Instituto de Genética Humana, Facultad de Medicina, Pontificia Universidad Javeriana, Bogotá 110211, Colombia
| |
Collapse
|
5
|
Jiang B, Tian Q, Shu C, Zhao J, Xue M, Zhu S. Resveratrol Enhances the Anti-Cancer Effects of Cis-Platinum on Human Cervical Cancer Cell Lines by Activating the SIRT3 Relative Anti-Oxidative Pathway. Front Pharmacol 2022; 13:916876. [PMID: 35865961 PMCID: PMC9294406 DOI: 10.3389/fphar.2022.916876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/27/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Cervical cancer exerts considerable mortality in the world. The combinations of chemotherapy with cis-platinum were the first-line treatment in late-stage cervical cancer but may cause severe adverse effects. Resveratrol (RES, 3,5,4′-trihydroxy-trans-stilbene) is a phytoalexin, and it showed anti-cancer effects but with low toxicity and side effects. Herein, we examined the anti-cancer effects of cis-platinum combined with RES in human cervical cancer cell lines. Methods: The antiproliferative effect was examined by cell counting and short-term MTT assay. Cell apoptosis was detected. The cell cycle distribution was determined by flow cytometry. Intracellular reactive oxygen species and mitochondrial transmembrane potential change were observed and calculated by confocal microscopy. The Si-RNA interference of SIRT3 in cancer cells was performed. Protein expression was checked by Western blotting. Results: RES inhibited the growth of SiHa cell lines, and it significantly enhanced the cis-platinum-induced cell apoptosis and cell cycle arresting in 48 h. The activation of the SIRT3 relative anti-oxidative pathway was proved to be the reason for the enhanced anti-cancer effects of cis-platinum and RES combination. Si-RNA interference of SIRT3 compromised the anti-cancer effect of cis-platinum and RES combination. Furthermore, the silencing of SIRT3 RNA inhibited the expression of the anti-oxidant enzyme (MnSOD, GPx, SOD-1, and CAT) and decreased the generation of H2O2 in the cis-platinum and RES combination group. Conclusion: RES enhances the anti-cancer effects of cis-platinum on SiHa cells by activating the SIRT3 relative anti-oxidative pathway. RES may act as a potential synergistic agent and be useful in the treatment of cervical cancer.
Collapse
Affiliation(s)
- Bin Jiang
- Department of Obstetrics and Gynecology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Qi Tian
- Department of Obstetrics and Gynecology, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
- National Health Commission Key Laboratory of Birth Defects Research, Prevention and Treatment, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
| | - Chuqiang Shu
- Department of Obstetrics and Gynecology, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
| | - Jing Zhao
- Department of Obstetrics and Gynecology, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
| | - Min Xue
- Department of Obstetrics and Gynecology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Shujuan Zhu
- Department of Obstetrics and Gynecology, The Third Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Shujuan Zhu,
| |
Collapse
|
6
|
Lim C, Lee P, Shim S, Jang SW. HS‑1793 inhibits cell proliferation in lung cancer by interfering with the interaction between p53 and MDM2. Oncol Lett 2022; 24:290. [PMID: 35928802 PMCID: PMC9344265 DOI: 10.3892/ol.2022.13410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/16/2022] [Indexed: 12/05/2022] Open
Abstract
The transcription factor or tumor suppressor protein p53 regulates numerous cellular functions, including cell proliferation, invasion, migration, senescence and apoptosis, in various types of cancer. HS-1793 is an analog of resveratrol, which exhibits anti-cancer effects on various types of cancer, including breast, prostate, colon and renal cancer, and multiple myeloma. However, to the best of our knowledge, the role of HS-1793 in lung cancer remains to be examined. The present study aimed to investigate the anti-cancer effect of HS-1793 on lung cancer and to determine its association with p53. The results revealed that HS-1793 reduced cell proliferation in lung cancer and increased p53 stability, thereby elevating the expression levels of the target genes p21 and mouse double minute 2 homolog (MDM2). When the levels of MDM2, a negative regulator of p53, are increased under normal conditions, MDM2 binds and degrades p53; however, HS-1793 inhibited this binding, confirming that p53 protein stability was increased. In conclusion, the findings of the present study provide new evidence that HS-1793 may inhibit lung cancer proliferation by disrupting the p53-MDM2 interaction.
Collapse
Affiliation(s)
- Chungun Lim
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 138‑736, Republic of Korea
| | - Peter Lee
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 138‑736, Republic of Korea
| | - Sungbo Shim
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju, North Chungcheong 28644, Republic of Korea
| | - Sung-Wuk Jang
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 138‑736, Republic of Korea
| |
Collapse
|
7
|
Dall’Asta M, Barbato M, Rocchetti G, Rossi F, Lucini L, Marsan PA, Colli L. Nutrigenomics: an underestimated contribution to the functional role of polyphenols. Curr Opin Food Sci 2022. [DOI: 10.1016/j.cofs.2022.100880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
8
|
Sánchez-Melgar A, Muñoz-López S, Albasanz JL, Martín M. Antitumoral Action of Resveratrol Through Adenosinergic Signaling in C6 Glioma Cells. Front Neurosci 2021; 15:702817. [PMID: 34539333 PMCID: PMC8440868 DOI: 10.3389/fnins.2021.702817] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/06/2021] [Indexed: 12/15/2022] Open
Abstract
Gliomas are the most common and aggressive primary tumors in the central nervous system. The nucleoside adenosine is considered to be one major constituent within the tumor microenvironment. The adenosine level mainly depends on two enzymatic activities: 5′-nucleotidase (5′NT or CD73) that synthesizes adenosine from AMP, and adenosine deaminase (ADA) that converts adenosine into inosine. Adenosine activates specific G-protein coupled receptors named A1, A2A, A2B, and A3 receptors. Resveratrol, a natural polyphenol present in grapes, peanuts, and berries, shows several healthy effects, including protection against cardiovascular, endocrine, and neurodegenerative diseases and cancer. However, the molecular mechanisms of resveratrol actions are not well known. Recently, we demonstrated that resveratrol acts as an agonist for adenosine receptors in rat C6 glioma cells. The present work aimed to investigate the involvement of adenosine metabolism and adenosine receptors in the molecular mechanisms underlying the antitumoral action of resveratrol. Results presented herein show that resveratrol was able to decrease cell numbers and viability and to reduce CD73 and ADA activities, leading to the increase of extracellular adenosine levels. Some resveratrol effects were reduced by the blockade of A1 or A3 receptors by DPCPX or MRS1220, respectively. These results suggest that reduced CD73 activity located in the plasma membrane in addition to a fine-tuned modulatory role of adenosine receptors could be involved, at least in part, in the antiproliferative action of resveratrol in C6 glioma cells.
Collapse
Affiliation(s)
- Alejandro Sánchez-Melgar
- Department of Inorganic, Organic Chemistry and Biochemistry, Faculty of Chemical and Technological Sciences, School of Medicine of Ciudad Real, Regional Center of Biomedical Research (CRIB), Universidad de Castilla-La Mancha, Ciudad Real, Spain
| | - Sonia Muñoz-López
- Department of Inorganic, Organic Chemistry and Biochemistry, Faculty of Chemical and Technological Sciences, School of Medicine of Ciudad Real, Regional Center of Biomedical Research (CRIB), Universidad de Castilla-La Mancha, Ciudad Real, Spain
| | - José Luis Albasanz
- Department of Inorganic, Organic Chemistry and Biochemistry, Faculty of Chemical and Technological Sciences, School of Medicine of Ciudad Real, Regional Center of Biomedical Research (CRIB), Universidad de Castilla-La Mancha, Ciudad Real, Spain
| | - Mairena Martín
- Department of Inorganic, Organic Chemistry and Biochemistry, Faculty of Chemical and Technological Sciences, School of Medicine of Ciudad Real, Regional Center of Biomedical Research (CRIB), Universidad de Castilla-La Mancha, Ciudad Real, Spain
| |
Collapse
|
9
|
Sharifi-Rad J, Quispe C, Mukazhanova Z, Knut E, Turgumbayeva A, Kipchakbayeva A, Seitimova G, Mahomoodally MF, Lobine D, Koay A, Wang J, Sheridan H, Leyva-Gómez G, Prado-Audelo MLD, Cortes H, Rescigno A, Zucca P, Sytar O, Imran M, Rodrigues CF, Cruz-Martins N, Ekiert H, Kumar M, Abdull Razis AF, Sunusi U, Kamal RM, Szopa A. Resveratrol-Based Nanoformulations as an Emerging Therapeutic Strategy for Cancer. Front Mol Biosci 2021; 8:649395. [PMID: 34540888 PMCID: PMC8440914 DOI: 10.3389/fmolb.2021.649395] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/17/2021] [Indexed: 12/13/2022] Open
Abstract
Resveratrol is a polyphenolic stilbene derivative widely present in grapes and red wine. Broadly known for its antioxidant effects, numerous studies have also indicated that it exerts anti-inflammatory and antiaging abilities and a great potential in cancer therapy. Regrettably, the oral administration of resveratrol has pharmacokinetic and physicochemical limitations such as hampering its effects so that effective administration methods are demanding to ensure its efficiency. Thus, the present review explores the published data on the application of resveratrol nanoformulations in cancer therapy, with the use of different types of nanodelivery systems. Mechanisms of action with a potential use in cancer therapy, negative effects, and the influence of resveratrol nanoformulations in different types of cancer are also highlighted. Finally, the toxicological features of nanoresveratrol are also discussed.
Collapse
Affiliation(s)
- Javad Sharifi-Rad
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Cristina Quispe
- Facultad de Ciencias de la Salud, Universidad Arturo Prat, Iquique, Chile
| | - Zhazira Mukazhanova
- Department of Natural Sciences and Technologies, Sarsen Amanzholov East Kazakhstan State University, Ust-Kamenogorsk, Kazakhstan
| | - Ewa Knut
- Chair and Department of Pharmaceutical Botany, Faculty of Pharmacy, Jagiellonian University, Medical College, Kraków, Poland
| | - Aknur Turgumbayeva
- Asfendiyarov Kazakh National Medical University, School Pharmacy, Almaty, Kazakhstan
- Al-Farabi Kazakh National University, Higher School of Medicine, Almaty, Kazakhstan
| | - Aliya Kipchakbayeva
- Faculty of Chemistry and Chemical Technology, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Gulnaz Seitimova
- Faculty of Chemistry and Chemical Technology, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Mohamad Fawzi Mahomoodally
- Department of Health Sciences, Faculty of Medicine and Health Sciences, University of Mauritius, Réduit, Mauritius
| | - Devina Lobine
- Department of Health Sciences, Faculty of Medicine and Health Sciences, University of Mauritius, Réduit, Mauritius
| | - Aaron Koay
- Trinity College Dublin, NatPro (Natural Products Research Centre), School of Pharmacy and Pharmaceutical Science, Dublin, Ireland
| | - Jinfan Wang
- Trinity College Dublin, NatPro (Natural Products Research Centre), School of Pharmacy and Pharmaceutical Science, Dublin, Ireland
| | - Helen Sheridan
- Trinity College Dublin, NatPro (Natural Products Research Centre), School of Pharmacy and Pharmaceutical Science, Dublin, Ireland
| | - Gerardo Leyva-Gómez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, Mexico City, Mexico
| | - María L. Del Prado-Audelo
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, Mexico City, Mexico
| | - Hernán Cortes
- Laboratorio de Medicina Genómica, Departamento de Genética, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Ciudad de México, Mexico City, Mexico
| | - Antonio Rescigno
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, Cagliari, Italy
| | - Paolo Zucca
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria, Cagliari, Italy
| | - Oksana Sytar
- Department of Plant Biology, Institute of Biology, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
- Department of Plant Physiology, Slovak University of Agriculture, Nitra, Slovakia
| | - Muhammad Imran
- University Institute of Diet and Nutritional Sciences, The University of Lahore, Lahore, Pakistan
| | - Célia F. Rodrigues
- Laboratory for Process Engineering, Environment, Biotechnology and Energy—Department of Chemical Engineering, Faculty of Engineering, University of Porto, Porto, Portugal
| | - Natália Cruz-Martins
- Faculty of Medicine, University of Porto, Porto, Portugal
- Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal
- Laboratory of Neuropsychophysiology, Faculty of Psychology and Education Sciences, University of Porto, Porto, Portugal
| | - Halina Ekiert
- Chair and Department of Pharmaceutical Botany, Faculty of Pharmacy, Jagiellonian University, Medical College, Kraków, Poland
| | - Manoj Kumar
- Chemical and Biochemical Processing Division, ICAR – Central Institute for Research on Cotton Technology, Mumbai, India
| | - Ahmad Faizal Abdull Razis
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Selangor, Malaysia
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Selangor, Malaysia
| | - Usman Sunusi
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Selangor, Malaysia
- Department of Biochemistry, Bayero University Kano, Kano, Nigeria
| | - Ramla Muhammad Kamal
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Selangor, Malaysia
- Department of Pharmacology, Federal University Dutse, Dutse, Nigeria
| | - Agnieszka Szopa
- Chair and Department of Pharmaceutical Botany, Faculty of Pharmacy, Jagiellonian University, Medical College, Kraków, Poland
| |
Collapse
|
10
|
Bhattacharya T, Dutta S, Akter R, Rahman MH, Karthika C, Nagaswarupa HP, Murthy HCA, Fratila O, Brata R, Bungau S. Role of Phytonutrients in Nutrigenetics and Nutrigenomics Perspective in Curing Breast Cancer. Biomolecules 2021; 11:1176. [PMID: 34439842 PMCID: PMC8394348 DOI: 10.3390/biom11081176] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/15/2021] [Accepted: 08/05/2021] [Indexed: 02/07/2023] Open
Abstract
Breast cancer (BC) is one of the most common type of cancer and an important contributor to female mortality. Several genes and epigenetic modifications are involved in the development and progression of BC. Research in phytochemistry, nutrigenomics, and nutrigenetics has provided strong evidence that certain phytonutrients are able to modulate gene expression at transcriptional and post-transcriptional levels. Such phytonutrients may also be beneficial to prevent and treat BC. In this review, we will focus on the nutrigenomic effects of various phytochemicals including polyphenols, phytosterols, terpenoids, alkaloids, and other compounds from different sources. Overall, these phytonutrients are found to inhibit BC cell proliferation, differentiation, invasion, metastasis, angiogenesis, and induce apoptotic cell death by targeting various molecular pathways. They also alter epigenetic mechanisms and enhance the chemosensitivity and radiosensitivity of cancer cells. Such phytochemicals may be used for the effective management of BC patients in the clinical setting in the future. The present article aims to summarize the specific molecular pathways involved in the genetic effects of phytochemicals in BC.
Collapse
Affiliation(s)
- Tanima Bhattacharya
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China;
- Techno India NJR Institute of Technology, Udaipur, Rajasthan 313003, India
| | - Soumam Dutta
- Food and Nutrition Division, University of Calcutta, Calcutta 700027, India;
| | - Rokeya Akter
- Department of Pharmacy, Jagannath University, Sadarghat, Dhaka 1100, Bangladesh;
- Department of Global Medical Science, Yonsei University Wonju College of Medicine, Yonsei University, Wonju 26426, Gangwon-do, Korea
| | - Md. Habibur Rahman
- Department of Global Medical Science, Yonsei University Wonju College of Medicine, Yonsei University, Wonju 26426, Gangwon-do, Korea
- Department of Pharmacy, Southeast University, Banani, Dhaka 1213, Bangladesh
| | - Chenmala Karthika
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty 643001, India;
| | | | - Hanabe Chowdappa Ananda Murthy
- Department of Applied Chemistry, School of Applied Natural Science, Adama Science and Technology University, Adama P.O. Box 1888, Ethiopia;
| | - Ovidiu Fratila
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (O.F.); (R.B.)
| | - Roxana Brata
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (O.F.); (R.B.)
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania
| |
Collapse
|
11
|
Augimeri G, Montalto FI, Giordano C, Barone I, Lanzino M, Catalano S, Andò S, De Amicis F, Bonofiglio D. Nutraceuticals in the Mediterranean Diet: Potential Avenues for Breast Cancer Treatment. Nutrients 2021; 13:2557. [PMID: 34444715 PMCID: PMC8400469 DOI: 10.3390/nu13082557] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 07/22/2021] [Accepted: 07/25/2021] [Indexed: 12/12/2022] Open
Abstract
The traditional Mediterranean Diet constitutes a food model that refers to the dietary patterns of the population living in countries bordering the Mediterranean Sea in the early 1960s. A huge volume of literature data suggests that the Mediterranean-style diet provides several dietary compounds that have been reported to exert beneficial biological effects against a wide spectrum of chronic illnesses, such as cardiovascular and neurodegenerative diseases and cancer including breast carcinoma. Among bioactive nutrients identified as protective factors for breast cancer, natural polyphenols, retinoids, and polyunsaturated fatty acids (PUFAs) have been reported to possess antioxidant, anti-inflammatory, immunomodulatory and antitumoral properties. The multiple anticancer mechanisms involved include the modulation of molecular events and signaling pathways associated with cell survival, proliferation, differentiation, migration, angiogenesis, antioxidant enzymes and immune responses. This review summarizes the anticancer action of some polyphenols, like resveratrol and epigallocatechin 3-gallate, retinoids and omega-3 PUFAs by highlighting the important hallmarks of cancer in terms of (i) cell cycle growth arrest, (ii) apoptosis, (iii) inflammation and (iv) angiogenesis. The data collected from in vitro and in vivo studies strongly indicate that these natural compounds could be the prospective candidates for the future anticancer therapeutics in breast cancer disease.
Collapse
Affiliation(s)
- Giuseppina Augimeri
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy; (G.A.); (F.I.M.); (C.G.); (I.B.); (M.L.); (S.C.); (S.A.); (F.D.A.)
| | - Francesca Ida Montalto
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy; (G.A.); (F.I.M.); (C.G.); (I.B.); (M.L.); (S.C.); (S.A.); (F.D.A.)
| | - Cinzia Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy; (G.A.); (F.I.M.); (C.G.); (I.B.); (M.L.); (S.C.); (S.A.); (F.D.A.)
- Centro Sanitario, University of Calabria, 87036 Arcavacata di Rende, Italy
| | - Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy; (G.A.); (F.I.M.); (C.G.); (I.B.); (M.L.); (S.C.); (S.A.); (F.D.A.)
- Centro Sanitario, University of Calabria, 87036 Arcavacata di Rende, Italy
| | - Marilena Lanzino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy; (G.A.); (F.I.M.); (C.G.); (I.B.); (M.L.); (S.C.); (S.A.); (F.D.A.)
- Centro Sanitario, University of Calabria, 87036 Arcavacata di Rende, Italy
| | - Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy; (G.A.); (F.I.M.); (C.G.); (I.B.); (M.L.); (S.C.); (S.A.); (F.D.A.)
- Centro Sanitario, University of Calabria, 87036 Arcavacata di Rende, Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy; (G.A.); (F.I.M.); (C.G.); (I.B.); (M.L.); (S.C.); (S.A.); (F.D.A.)
- Centro Sanitario, University of Calabria, 87036 Arcavacata di Rende, Italy
| | - Francesca De Amicis
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy; (G.A.); (F.I.M.); (C.G.); (I.B.); (M.L.); (S.C.); (S.A.); (F.D.A.)
- Centro Sanitario, University of Calabria, 87036 Arcavacata di Rende, Italy
| | - Daniela Bonofiglio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy; (G.A.); (F.I.M.); (C.G.); (I.B.); (M.L.); (S.C.); (S.A.); (F.D.A.)
- Centro Sanitario, University of Calabria, 87036 Arcavacata di Rende, Italy
| |
Collapse
|
12
|
Resveratrol Modulation of Apoptosis and Cell Cycle Response to Cisplatin in Head and Neck Cancer Cell Lines. Int J Mol Sci 2021; 22:ijms22126322. [PMID: 34204834 PMCID: PMC8231609 DOI: 10.3390/ijms22126322] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 12/13/2022] Open
Abstract
In head and neck cancers, the effectiveness of cisplatin (CisPt) treatment is limited by its toxicity, especially when higher doses are necessary, and the possible occurrence of cisplatin resistance. This study evaluated the effects of resveratrol (RSV) on the expression of different genes involved in the response of human tumor cells (FaDu, PE/CA-PJ49) to cisplatin therapy. Our results revealed that RSV induced apoptosis amplification in both FaDu and PE/CA-PJ49 cells and modulated the expression of specific genes differently than in normal HaCaT cells. In FaDu cells, combined CisPt + RSV treatment induced an increase in apoptosis, which was associated with an increase in c-MYC and TP53 and a decrease in BCL-2 expression. While CisPt + RSV treatment induced apoptosis in PE/CA-PJ49 cells by inhibition of BCL-2 associated with high levels of MDM-2 and subsequently led to inhibition of TP53 gene expression. Decreased c-MYC expression in PE/CA-PJ49 treated with CisPt + RSV was accompanied by cell cycle blockage in G0/G1 phase. In conclusion, RSV influences tumor cell response to CisPt by inducing apoptosis and modulating gene expression. In addition, in normal HaCaT cells, RSV was able to reduce the harmful effects of CisPt.
Collapse
|
13
|
Network Pharmacological Analysis through a Bioinformatics Approach of Novel NSC765600 and NSC765691 Compounds as Potential Inhibitors of CCND1/ CDK4/ PLK1/ CD44 in Cancer Types. Cancers (Basel) 2021; 13:cancers13112523. [PMID: 34063946 PMCID: PMC8196568 DOI: 10.3390/cancers13112523] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/26/2021] [Accepted: 05/18/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Around 14 million new cancer cases, rate are reported annually, with high mortality worldswide, several mechanisms are associated with complexities in cancer, which leads to resistance to current therapeutic interventions in cancer patients. The aim of this study was to identify molecular genes responsible for cancer development, progression and resistances to therapeutic intervention, and also evaluate the potency of our novel compounds NSC7565600 and NSC765691 as potential target for these oncogenes. Using bioinformatics, we successfully identified CCND1/CDK4/PLK1/CD44 as oncogenic signatures, which drives cancer progression and resistance to treatment, and as potential druggable candidates for both NSC7565600 and NSC765691 small molecules. We also showed the antiproliferative and cytotoxic effects of these compounds against a panel of NCI-60 cancer cell lines. This suggests the potential of NSC765600 and NSC765691 compounds to inhibit CCND1/CDK4/PLK1/CD44 expressions in cancer. Abstract Cyclin D1 (CCND1) and cyclin-dependent kinase 4 (CDK4) both play significant roles in regulating cell cycle progression, while polo-like kinase 1 (PLK1) regulates cell differentiation and tumor progression, and activates cancer stem cells (CSCs), with the cluster of differentiation 44 (CD44) surface marker mostly being expressed. These oncogenes have emerged as promoters of metastasis in a variety of cancer types. In this study, we employed comprehensive computational and bioinformatics analyses to predict drug targets of our novel small molecules, NSC765600 and NSC765691, respectively derived from diflunisal and fostamatinib. The target prediction tools identified CCND1/CDK4/PLK1/CD44 as target genes for NSC765600 and NSC765691 compounds. Additionally, the results of our in silico molecular docking analysis showed unique ligand–protein interactions with putative binding affinities of NSC765600 and NSC765691 with CCND1/CDK4/PLK1/CD44 oncogenic signaling pathways. Moreover, we used drug-likeness precepts as our guidelines for drug design and development, and found that both compounds passed the drug-likeness criteria of molecular weight, polarity, solubility, saturation, flexibility, and lipophilicity, and also exhibited acceptable pharmacokinetic properties. Furthermore, we used development therapeutics program (DTP) algorithms and identified similar fingerprints and mechanisms of NSC765600 and NSC765691 with synthetic compounds and standard anticancer agents in the NCI database. We found that NSC765600 and NSC765691 displayed antiproliferative and cytotoxic effects against a panel of NCI-60 cancer cell lines. Based on these finding, NSC765600 and NSC765691 exhibited satisfactory levels of safety with regard to toxicity, and met all of the required criteria for drug-likeness precepts. Currently, further in vitro and in vivo investigations in tumor-bearing mice are in progress to study the potential treatment efficacies of the novel NSC765600 and NSC765691 small molecules.
Collapse
|
14
|
Vafaee R, Nikzamir A, Razzaghi M, Rezaei Tavirani S, Ahmadzadeh A, Emamhadi M. An Investigation of Post-radiation Gene Expression Profiles: A System Biology Study. J Lasers Med Sci 2020; 11:S101-S106. [PMID: 33995977 PMCID: PMC7956041 DOI: 10.34172/jlms.2020.s16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Introduction: Genomics and bioinformatics are useful methods for exploring unclear aspects of radiation effects on biological systems. Many radiation-induced alterations in irradiated samples are post-radiation time-dependent. This study aims to evaluate the post-irradiation effects of the gamma ray on human Jurkat cells. Methods: Gene expression profiles of the samples harvested 6 and 24 hours after radiation to find the critical differential expressed genes and the related pathways. Samples are provided from Gene Expression Omnibus (GEO) and analyzed by ClueGO. Results: Twnety-nine critical genes were determined as the important affected genes and 7 classes of related pathways were introduced. CCNE2, PSMD11, CDC25C, ANAPC1, PLK1, AURKA, and CCNB1 that were associated with more than 6 pathways were related to one of the determined pathway groups. Conclusion: Cell protecting pathways were associated with the genes (HSPA5, HSPA8, HSP90B1, HMMR, CEBPB, RXRA, and PSMD11) which were related to the minimum numbers of pathways. The finding of this study corresponds to repair processes which depend on post-radiation time. It seems these sets of genes are suitable candidates for further investigation.
Collapse
Affiliation(s)
- Reza Vafaee
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abdolrahim Nikzamir
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohhamadreza Razzaghi
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sina Rezaei Tavirani
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Ahmadzadeh
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - MohammadAli Emamhadi
- Forensic Medicine Specialist, Forensic Medicine Department, Shahid Beheshti Medical University, Tehran, Iran
| |
Collapse
|
15
|
Still Living Better through Chemistry: An Update on Caloric Restriction and Caloric Restriction Mimetics as Tools to Promote Health and Lifespan. Int J Mol Sci 2020; 21:ijms21239220. [PMID: 33287232 PMCID: PMC7729921 DOI: 10.3390/ijms21239220] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 11/30/2020] [Accepted: 11/30/2020] [Indexed: 02/06/2023] Open
Abstract
Caloric restriction (CR), the reduction of caloric intake without inducing malnutrition, is the most reproducible method of extending health and lifespan across numerous organisms, including humans. However, with nearly one-third of the world’s population overweight, it is obvious that caloric restriction approaches are difficult for individuals to achieve. Therefore, identifying compounds that mimic CR is desirable to promote longer, healthier lifespans without the rigors of restricting diet. Many compounds, such as rapamycin (and its derivatives), metformin, or other naturally occurring products in our diets (nutraceuticals), induce CR-like states in laboratory models. An alternative to CR is the removal of specific elements (such as individual amino acids) from the diet. Despite our increasing knowledge of the multitude of CR approaches and CR mimetics, the extent to which these strategies overlap mechanistically remains unclear. Here we provide an update of CR and CR mimetic research, summarizing mechanisms by which these strategies influence genome function required to treat age-related pathologies and identify the molecular fountain of youth.
Collapse
|
16
|
Vargas JE, Puga R, Lenz G, Trindade C, Filippi-Chiela E. Cellular Mechanisms Triggered by the Cotreatment of Resveratrol and Doxorubicin in Breast Cancer: A Translational In Vitro-In Silico Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5432651. [PMID: 33204396 PMCID: PMC7654215 DOI: 10.1155/2020/5432651] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/22/2020] [Accepted: 10/06/2020] [Indexed: 12/14/2022]
Abstract
Doxorubicin (Doxo) is the most effective chemotherapeutic agent for the treatment of breast cancer. However, resistance to Doxo is common. Adjuvant compounds capable of modulating mechanisms involved in Doxo resistance may potentiate the effectiveness of the drug. Resveratrol (Rsv) has been tested as an adjuvant in mammary malignancies. However, the cellular and molecular mechanisms underlying the effects of cotreatment with Doxo and Rsv in breast cancer are poorly understood. Here, we combined in vitro and in silico analysis to characterize these mechanisms. In vitro, we employed a clinically relevant experimental design consisting of acute (24 h) treatment followed by 15 days of analysis. Acute Rsv potentiated the long-lasting effect of Doxo through the induction of apoptosis and senescence. Cells that survived to the cotreatment triggered high levels of autophagy. Autophagy inhibition during its peak of activation but not concomitant with Doxo+Rsv increased the long-term toxicity of the cotreatment. To uncover key proteins potentially associated with in vitro effects, an in silico multistep strategy was implemented. Chemical-protein networks were predicted based on constitutive gene expression of MCF7 cells and interatomic data from breast cancer. Topological analysis, KM survival analysis, and a quantitative model based on the connectivity between apoptosis, senescence, and autophagy were performed. We found seven putative genes predicted to be modulated by Rsv in the context of Doxo treatment: CCND1, CDH1, ESR1, HSP90AA1, MAPK3, PTPN11, and RPS6KB1. Six out of these seven genes have been experimentally proven to be modulated by Rsv in cancer cells, with 4 of the 6 genes in MCF7 cells. In conclusion, acute Rsv potentiated the long-term toxicity of Doxo in breast cancer potentially through the modulation of genes and mechanisms involved in Doxo resistance. Rational autophagy inhibition potentiated the effects of Rsv+Doxo, a strategy that should be further tested in animal models.
Collapse
Affiliation(s)
- José Eduardo Vargas
- Instituto de Ciências Biológicas, Universidade de Passo Fundo, Brazil
- Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Renato Puga
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Guido Lenz
- Centro de Biotecnologia e Departamento de Biofísica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Cristiano Trindade
- Facultad de Ciencias Básicas y Biomédicas, Universidad Simón Bolívar, Barranquilla, Colombia
| | - Eduardo Filippi-Chiela
- Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Departamento de Ciências Morfológicas, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
17
|
Shreberk-Shaked M, Dassa B, Sinha S, Di Agostino S, Azuri I, Mukherjee S, Aylon Y, Blandino G, Ruppin E, Oren M. A Division of Labor between YAP and TAZ in Non-Small Cell Lung Cancer. Cancer Res 2020; 80:4145-4157. [PMID: 32816858 DOI: 10.1158/0008-5472.can-20-0125] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 06/07/2020] [Accepted: 08/04/2020] [Indexed: 11/16/2022]
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide. The paralogous transcriptional cofactors Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ, also called WWTR1), the main downstream effectors of the Hippo signal transduction pathway, are emerging as pivotal determinants of malignancy in lung cancer. Traditionally, studies have tended to consider YAP and TAZ as functionally redundant transcriptional cofactors with similar biological impact. However, there is growing evidence that each of them also possesses distinct attributes. Here we sought to systematically characterize the division of labor between YAP and TAZ in non-small cell lung cancer (NSCLC), the most common histological subtype of lung cancer. Representative NSCLC cell lines as well as patient-derived data showed that the two paralogs orchestrated nonoverlapping transcriptional programs in this cancer type. YAP preferentially regulated gene sets associated with cell division and cell-cycle progression, whereas TAZ preferentially regulated genes associated with extracellular matrix organization. Depletion of YAP resulted in growth arrest, whereas its overexpression promoted cell proliferation. Likewise, depletion of TAZ compromised cell migration, whereas its overexpression enhanced migration. The differential effects of YAP and TAZ on key cellular processes were also associated with differential response to anticancer therapies. Uncovering the different activities and downstream effects of YAP and TAZ may thus facilitate better stratification of patients with lung cancer for anticancer therapies. SIGNIFICANCE: Thease findings show that oncogenic paralogs YAP and TAZ have distinct roles in NSCLC and are associated with differential response to anticancer drugs, knowledge that may assist lung cancer therapy decisions.
Collapse
Affiliation(s)
| | - Bareket Dassa
- Bioinformatics Unit, Department of Life Sciences Core Facilities, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Sanju Sinha
- Cancer Data Science Laboratory, NCI, NIH, Bethesda, Maryland.,Center for Bioinformatics and Computational Biology & Department of Computer Sciences, University of Maryland, College Park, Maryland
| | - Silvia Di Agostino
- Oncogenomic and Epigenetic Lab., IRCCS Regina Elena National Cancer Institute-IFO, Rome, Italy
| | - Ido Azuri
- Bioinformatics Unit, Department of Life Sciences Core Facilities, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Saptaparna Mukherjee
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Yael Aylon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Giovanni Blandino
- Oncogenomic and Epigenetic Lab., IRCCS Regina Elena National Cancer Institute-IFO, Rome, Italy
| | - Eytan Ruppin
- Cancer Data Science Laboratory, NCI, NIH, Bethesda, Maryland.,Center for Bioinformatics and Computational Biology & Department of Computer Sciences, University of Maryland, College Park, Maryland
| | - Moshe Oren
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
18
|
Bozorgi A, Khazaei S, Khademi A, Khazaei M. Natural and herbal compounds targeting breast cancer, a review based on cancer stem cells. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2020; 23:970-983. [PMID: 32952942 PMCID: PMC7478260 DOI: 10.22038/ijbms.2020.43745.10270] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 03/09/2020] [Indexed: 12/13/2022]
Abstract
Cancer stem cells (CSCs) are known as the major reason for therapy resistance. Recently, natural herbal compounds are suggested to have a significant role in inhibiting the breast cancer stem cells (BCSCs). The aim of this study was to explore the effective natural herbal compounds against BCSCs.This review article was designed based on the BCSCs, mechanisms of therapy resistance and natural herbal compounds effective to inhibit their activity. Therefore, Science direct, PubMed and Scopus databases were explored and related original articles were investigated from 2010 to 2019. BCSCs use different mechanisms including special membrane transporters, anti-apoptotic, pro-survival, and self-renewal- related signaling pathways. Natural herbal compounds could disturb these mechanisms, therefore may inhibit or eradicate the BCSCs. Studies show that a broad range of plants, either as a food or medicine, contain anti-cancer agents that phenolic components and their different derivatives share a large quantity. Natural herbal compounds play a pivotal role in the eradication of BCSCs, through the inhibition of biological activities and induction of apoptosis. Although it is necessary to conduct more clinical investigation.
Collapse
Affiliation(s)
- Azam Bozorgi
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Saber Khazaei
- Dental Research Center, Dental Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Abbasali Khademi
- Dental Research Center, Dental Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mozafar Khazaei
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
19
|
Zhang R, Wang P, Yu S, Hansbro P, Wang H. Computerized screening of G-protein coupled receptors to identify and characterize olfactory receptors. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2020; 83:9-19. [PMID: 32019429 DOI: 10.1080/15287394.2019.1709305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Olfactory receptors (ORs) are a group of G protein coupled receptors (GPCRs) that initiate chemical odorant signals. Although ORs are predominantly located in nasal epithelia to detect smell, these receptors are also present in peripherally in non-nasal organs/tissues. Since the quality of life and cognitive and sensorial features of sense of smell are worsened in multiple chemical sensitivity due to the interaction of ORs with offending compounds, it is important to not only differentiate these receptors from other GPCRs but also characterize these organelles to understand the underlying mechanisms of smelling disorders. The aim of this study was develop computerized programs to differentiate ORs from GPCRs. The computer program was developed on the basis of widely accepted basic algorithms. It is noteworthy that an accuracy of 95.5% was attained, a level not achieved using other established techniques for screening of ORs from GPCRs. The high accuracy rate indicates that this method of differential identification appears reliable. Our findings indicate that this novel method may be considered as a tool for identification and characterization of receptors which might aid in therapeutic approaches to human chemical-mediated sensitization.
Collapse
Affiliation(s)
- Rui Zhang
- Xinjiang Laboratory of Minority Speech and Language Information Processing, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumchi, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Pu Wang
- School of Health Sciences, University of Newcastle, Callaghan, Australia
| | - Shunbang Yu
- School of Health Sciences, University of Newcastle, Callaghan, Australia
| | - Philip Hansbro
- Faculty of Health and Medicine, HMRI, School of Biomedical Sciences and Pharmacy, Callaghan, Australia
| | - He Wang
- School of Health Sciences, University of Newcastle, Callaghan, Australia
| |
Collapse
|
20
|
Zhang R, Wang P, Yu S, Wang H. Computational prediction methods to simulate structure and binding sites of coumarin with olfactory receptor 5P3. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2020; 82:1199-1206. [PMID: 31959074 DOI: 10.1080/15287394.2019.1709313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Olfactory receptor 5P3 is coded by OR5P3 gene in human OR5P3 and recognized as the receptor for coumarin present in 30% of fragrances and might adversely affect human health. OR5P3 are also associated with coumarin-related breast cancer development and acquired tamoxifen resistance in breast cancer cells. Previously various investigators noted that coumarin acts as a ligand for OR5P3 and binding of this molecule to OR5P3 leads to downstream changes in the olfactory bulb and central nervous system. However, the interaction between OR5P3 and its ligands at molecular level is not well understood. The aim of this study was to develop a computerized approach to simulate the binding process of coumarin to OR5P3 and determine the binding sites at a molecular level. It is conceivable that understanding this binding mechanism to a specific OR may aid in identification of receptors responsible olfaction and non-olfaction disorders such as breast cancer. The approach employed in this study may thus be utilized in future studies to simulate bind of specific chemicals to ORs and potentially have therapeutic applications.
Collapse
Affiliation(s)
- Rui Zhang
- Xinjiang Laboratory of Minority Speech and Language Information Processing, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumchi, Xinjiang, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Pu Wang
- School of Health Sciences, University of Newcastle, Newcastle, China
| | - Shunbang Yu
- School of Health Sciences, University of Newcastle, Newcastle, China
| | - He Wang
- School of Health Sciences, University of Newcastle, Newcastle, China
| |
Collapse
|
21
|
Subedi L, Teli MK, Lee JH, Gaire BP, Kim MH, Kim SY. A Stilbenoid Isorhapontigenin as a Potential Anti-Cancer Agent against Breast Cancer through Inhibiting Sphingosine Kinases/Tubulin Stabilization. Cancers (Basel) 2019; 11:cancers11121947. [PMID: 31817453 PMCID: PMC6966567 DOI: 10.3390/cancers11121947] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/28/2019] [Accepted: 12/04/2019] [Indexed: 12/24/2022] Open
Abstract
Isorhapontigenin (ISO), a tetrahydroxylated stilbenoid, is an analog of resveratrol (Rsv). The various biological activities of Rsv and its derivatives have been previously reported in the context of both cancer and inflammation. However, the anti-cancer effect of ISO against breast cancer has not been well established, despite being an orally bioavailable dietary polyphenol. In this study, we determine the anti-cancer effects of ISO against breast cancer using MCF7, T47D, and MDA-MB-231 cell lines. We observed that ISO induces breast cancer cell death, cell cycle arrest, oxidative stress, and the inhibition of cell proliferation. Additionally, sphingosine kinase inhibition by ISO controlled tubulin polymerization and cancer cell growth by regulating MAPK/PI3K-mediated cell cycle arrest in MCF7 cells. Interestingly, SPHK1/2 gene silencing increased oxidative stress, cell death, and tubulin destabilization in MCF7 cells. This suggests that the anti-cancer effect of ISO can be regulated by SPHK/tubulin destabilization pathways. Overall, ISO successfully induced breast cancer cell death and cell growth arrest, suggesting this phytochemical is a better alternative for breast cancer treatment. Further studies in animal models could confirm the potency and usability of ISO over Rsv for targeting breast cancer, potentially posing an alternative candidate for improved therapy in the near future.
Collapse
|
22
|
Li B, Zhu HB, Song GD, Cheng JH, Li CZ, Zhang YZ, Zhao P. Regulating the CCNB1 gene can affect cell proliferation and apoptosis in pituitary adenomas and activate epithelial-to-mesenchymal transition. Oncol Lett 2019; 18:4651-4658. [PMID: 31611974 PMCID: PMC6781518 DOI: 10.3892/ol.2019.10847] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 08/06/2019] [Indexed: 02/06/2023] Open
Abstract
The aim of the present study was to investigate the role and potential regulatory mechanisms of cyclin B1 (CCNB1) in the proliferation, apoptosis and epithelial-to-mesenchymal transition (EMT) in pituitary adenomas. A total of 24 specimens were included in the present study. The expression levels of CCNB1 protein in two normal pituitary and 22 pituitary adenoma tissues were determined by western blotting. CCNB1 was knocked-down by lentiviral-mediated infection of short hairpin RNA (shRNA) in GH3 and MMQ cell lines. The proliferation, cell cycle and apoptosis of GH3 and MMQ cell lines were detected using a Cell Counting Kit-8 and flow cytometer. Reverse transcription-quantitative PCR was utilized to detect the expression level of CCNB1 gene and EMT markers. In the present study, resveratrol (RES) was used as an inhibitor of CCNB1. The protein expression level of CCNB1 in pituitary adenomas was higher than that in normal pituitary tissue, as assessed by western blot analysis. In addition, the expression level of CCNB1 in invasive pituitary adenomas was higher when comparing invasive pituitary adenomas and non-invasive pituitary adenomas. Knockdown of CCNB1 resulted in significant decreases in cell viability and proliferation, arrested cell cycle at the G2/M phase and increased apoptosis. In addition, knockdown of CCNB1 significantly decreased the expression levels of the mesothelial cell marker N-cadherin (P<0.001), but significantly increased the expression levels of the epithelial cell markers E-cadherin (P<0.01) and p120-catenin (P<0.001). Further analyses identified that RES inhibited the expression level of CCNB1, and RES treatment exhibited a similar effect as CCNB1 shRNA infection. The present study suggested that suppressing the expression level of CCNB1 could regulate the proliferation and apoptosis of pituitary tumor cells and alter the expression level of various EMT markers. In addition, RES treatment could be used as an inhibitor of CCNB1. The present study also identified the molecular mechanisms underlying CCNB1 role in EMT.
Collapse
Affiliation(s)
- Bin Li
- Neurosurgical Department, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, P.R. China
| | - Hai-Bo Zhu
- Neurosurgical Department, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, P.R. China
| | - Gui-Dong Song
- Department of Cell and Biology, Beijing Neurosurgical Institute, Beijing 100070, P.R. China
| | - Jian-Hua Cheng
- Neurosurgical Department, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, P.R. China
| | - Chu-Zhong Li
- Department of Cell and Biology, Beijing Neurosurgical Institute, Beijing 100070, P.R. China
| | - Ya-Zhuo Zhang
- Department of Cell and Biology, Beijing Neurosurgical Institute, Beijing 100070, P.R. China
| | - Peng Zhao
- Neurosurgical Department, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, P.R. China
| |
Collapse
|
23
|
Chatterjee B, Ghosh K, Kanade SR. Resveratrol modulates epigenetic regulators of promoter histone methylation and acetylation that restores BRCA1, p53, p21 CIP1 in human breast cancer cell lines. Biofactors 2019; 45:818-829. [PMID: 31317586 DOI: 10.1002/biof.1544] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 06/27/2019] [Indexed: 01/09/2023]
Abstract
The epigenetic enzymes catalyze posttranslational modifications (PTMs) of histones, which functionally determine gene expression at the chromatin level. Resveratrol (RVT) a much studied anti-cancer natural molecule is known for restoration of BRCA1, p53, and p21 in cancer cells. We aimed to investigate the role of histone methylation and acetylation on upregulation of these tumor suppressor genes. Our results suggest RVT significantly increase expression of BRCA1, p53, and p21, while decreased expression of protein arginine methyltransferase 5 (PRMT5) and enhancer of Zeste homolog 2 (EZH2) at a 20 μM concentration by 48 hr in both MCF-7 and MDA-MB-231 breast cancer cells. Also, there was an overall loss of H4R3me2s (catalytic product of PRMT5) and H3K27me3 (catalytic product of PRMT5). In contrast, RVT exposure caused a significant decrease in lysine deacetylase (KDAC) activity and expression of KDAC1-3, whereas the expression of lysine acetyltransferase KAT2A/3B was increased compared to the unexposed cells. As an outcome, RVT increased global level of H3K9ac and H3K27ac marks. The chromatin immunoprecipitation showed 20 μM RVT exposure significantly reduced the enrichment of repressive histone marks (H4R3me2s and H3K27me3) while the abundance of activating histone marks (H3K9/27ac) within the proximal promoter region of BRCA1, p53, and p21 was increased. We hypothesize RVT by affecting the expression and function of methylation and acetylation enzymes altered the epigenetic modifications on promoter histones that restored expression of these critically important tumor suppressor genes.
Collapse
Affiliation(s)
- Biji Chatterjee
- Department of Biochemistry and Molecular Biology, School of Biological Sciences, Central University of Kerala, Kasargod, Kerala, India
| | - Krishna Ghosh
- Department of Biochemistry and Molecular Biology, School of Biological Sciences, Central University of Kerala, Kasargod, Kerala, India
- Department of Plant Sciences, School of Life Sciences, University of Hyderabad, Central University P.O., Hyderabad, Telangana, India
| | - Santosh R Kanade
- Department of Biochemistry and Molecular Biology, School of Biological Sciences, Central University of Kerala, Kasargod, Kerala, India
- Department of Plant Sciences, School of Life Sciences, University of Hyderabad, Central University P.O., Hyderabad, Telangana, India
| |
Collapse
|
24
|
Wang N, He J, Pan C, Wang J, Ma M, Shi X, Xu Z. Resveratrol Activates Autophagy via the AKT/mTOR Signaling Pathway to Improve Cognitive Dysfunction in Rats With Chronic Cerebral Hypoperfusion. Front Neurosci 2019; 13:859. [PMID: 31481868 PMCID: PMC6710371 DOI: 10.3389/fnins.2019.00859] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 07/30/2019] [Indexed: 12/18/2022] Open
Abstract
Chronic cerebral hypoperfusion (CCH) is a main cause of vascular dementia and is also an etiological factor of neurological diseases and mental disorders. However, few treatments are available for CCH, and new medications are needed. In the present study, we employed a rat model of CCH that was based on bilateral common carotid artery occlusion and investigated the therapeutic effects of resveratrol and its detailed mechanism of action. We evaluated neurological deficit scores and performed the Morris water maze test, hematoxylin and eosin staining, TUNEL staining, enzyme-linked immunosorbent assays, and Western blot. Resveratrol reduced neurological deficit scores in CCH rats and reduced pathological damage in the frontal cortex and hippocampus. Resveratrol activated autophagy and inhibited the expression of AKT/mechanistic target of rapamycin (mTOR) signaling pathway-related proteins. Treatment with a phosphoinositide-3 kinase inhibitor reversed the protective effect of resveratrol. These findings suggest that resveratrol improves cognitive function in a rat model of CCH and reduces oxidative stress-induced neuronal damage in the frontal cortex and hippocampus by activating autophagy and inhibiting neuronal apoptosis. These effects may be regulated by the AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Nan Wang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jinting He
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Chengliang Pan
- College of Clinical Medicine, Jilin University, Changchun, China
| | - Jiaoqi Wang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Ming Ma
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xinxiu Shi
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhongxin Xu
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
25
|
de Melo FHM, Oliveira JS, Sartorelli VOB, Montor WR. Cancer Chemoprevention: Classic and Epigenetic Mechanisms Inhibiting Tumorigenesis. What Have We Learned So Far? Front Oncol 2018; 8:644. [PMID: 30627525 PMCID: PMC6309127 DOI: 10.3389/fonc.2018.00644] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 12/07/2018] [Indexed: 12/21/2022] Open
Abstract
Cancers derive from step by step processes which are differentiated by the progressively accumulated mutations. For some tumors there is a clear progressive advancement from benign lesions to malignancy and for these, preventive screening programs exist. In such cases having those benign lesions are a clear indicator of predisposition while for some other cases, familial patterns of cancer incidence and the identification of mutations are the main indicators of higher risk for having the disease. For patients identified as having predisposition, chemoprevention is a goal and in some cases a possibility. Chemoprevention is the use of any compound, either natural or synthetic that abrogates carcinogenesis or tumor progression, through different mechanisms, some of which have already been described. For example, the classic mechanisms may involve activation of free radical scavenging enzymes, control of chronic inflammation, and downregulation of specific signaling pathways. More recently, epigenetics allowed further understanding of the chemopreventive potential of several agents, such as sulforaphane, green tea derived compounds, resveratrol, isoflavones, and others which we exploit in this review article. Throughout the text we discuss the properties compounds should have in order to be classified as chemopreventive ones and the challenges in translational research in this area, as lots of the success achieved in vitro cannot be translated into the clinical settings, due to several different drawbacks, which include toxicity, cost, dose definition, patient adherence, and regimen of use.
Collapse
Affiliation(s)
| | - Julia Salles Oliveira
- Department of Physiological Sciences, Santa Casa de São Paulo School of Medical Sciences (FCMSCSP), São Paulo, Brazil
| | | | - Wagner Ricardo Montor
- Department of Physiological Sciences, Santa Casa de São Paulo School of Medical Sciences (FCMSCSP), São Paulo, Brazil
| |
Collapse
|
26
|
Younas M, Hano C, Giglioli-Guivarc'h N, Abbasi BH. Mechanistic evaluation of phytochemicals in breast cancer remedy: current understanding and future perspectives. RSC Adv 2018; 8:29714-29744. [PMID: 35547279 PMCID: PMC9085387 DOI: 10.1039/c8ra04879g] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/15/2018] [Indexed: 12/30/2022] Open
Abstract
Breast cancer is one of the most commonly diagnosed cancers around the globe and accounts for a large proportion of fatalities in women. Despite the advancement in therapeutic and diagnostic procedures, breast cancer still represents a major challenge. Current anti-breast cancer approaches include surgical removal, radiotherapy, hormonal therapy and the use of various chemotherapeutic drugs. However, drug resistance, associated serious adverse effects, metastasis and recurrence complications still need to be resolved which demand safe and alternative strategies. In this scenario, phytochemicals have recently gained huge attention due to their safety profile and cost-effectiveness. These phytochemicals modulate various genes, gene products and signalling pathways, thereby inhibiting breast cancer cell proliferation, invasion, angiogenesis and metastasis and inducing apoptosis. Moreover, they also target breast cancer stem cells and overcome drug resistance problems in breast carcinomas. Phytochemicals as adjuvants with chemotherapeutic drugs have greatly enhanced their therapeutic efficacy. This review focuses on the recently recognized molecular mechanisms underlying breast cancer chemoprevention with the use of phytochemicals such as curcumin, resveratrol, silibinin, genistein, epigallocatechin gallate, secoisolariciresinol, thymoquinone, kaempferol, quercetin, parthenolide, sulforaphane, ginsenosides, naringenin, isoliquiritigenin, luteolin, benzyl isothiocyanate, α-mangostin, 3,3'-diindolylmethane, pterostilbene, vinca alkaloids and apigenin.
Collapse
Affiliation(s)
- Muhammad Younas
- Department of Biotechnology, Quaid-i-Azam University Islamabad-45320 Pakistan +92-51-90644121 +92-51-90644121 +33-767-97-0619
| | - Christophe Hano
- Laboratoire de Biologie des Ligneux et des Grandes Cultures (LBLGC), Plant Lignans Team, UPRES EA 1207, Université d'Orléans F 28000 Chartres France
| | | | - Bilal Haider Abbasi
- Department of Biotechnology, Quaid-i-Azam University Islamabad-45320 Pakistan +92-51-90644121 +92-51-90644121 +33-767-97-0619
- Laboratoire de Biologie des Ligneux et des Grandes Cultures (LBLGC), Plant Lignans Team, UPRES EA 1207, Université d'Orléans F 28000 Chartres France
- EA2106 Biomolecules et Biotechnologies Vegetales, Universite Francois-Rabelais de Tours Tours France
| |
Collapse
|
27
|
D'Alesio C, Punzi S, Cicalese A, Fornasari L, Furia L, Riva L, Carugo A, Curigliano G, Criscitiello C, Pruneri G, Pelicci PG, Faretta M, Bossi D, Lanfrancone L. RNAi screens identify CHD4 as an essential gene in breast cancer growth. Oncotarget 2018; 7:80901-80915. [PMID: 27779108 PMCID: PMC5348363 DOI: 10.18632/oncotarget.12646] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 09/29/2016] [Indexed: 12/15/2022] Open
Abstract
Epigenetic regulation plays an essential role in tumor development and epigenetic modifiers are considered optimal potential druggable candidates. In order to identify new breast cancer vulnerabilities and improve therapeutic chances for patients, we performed in vivo and in vitro shRNA screens in a human breast cancer cell model (MCF10DCIS.com cell line) using epigenetic libraries. Among the genes identified in our screening, we deeply investigated the role of Chromodomain Helicase DNA binding Protein 4 (CHD4) in breast cancer tumorigenesis. CHD4 silencing significantly reduced tumor growth in vivo and proliferation in vitro of MCF10DCIS.com cells. Similarly, in vivo breast cancer growth was decreased in a spontaneous mouse model of breast carcinoma (MMTV-NeuT system) and in metastatic patient-derived xenograft models. Conversely, no reduction in proliferative ability of non-transformed mammary epithelial cells (MCF10A) was detected. Moreover, we showed that CHD4 depletion arrests proliferation by inducing a G0/G1 block of cell cycle associated with up-regulation of CDKN1A (p21). These results highlight the relevance of genetic screens in the identification of tumor frailties and the role of CHD4 as a potential pharmacological target to inhibit breast cancer growth.
Collapse
Affiliation(s)
- Carolina D'Alesio
- Department of Experimental Oncology, European Institute of Oncology, Milan 20141, Italy
| | - Simona Punzi
- Department of Experimental Oncology, European Institute of Oncology, Milan 20141, Italy
| | - Angelo Cicalese
- Department of Experimental Oncology, European Institute of Oncology, Milan 20141, Italy
| | - Lorenzo Fornasari
- Department of Experimental Oncology, European Institute of Oncology, Milan 20141, Italy
| | - Laura Furia
- Department of Experimental Oncology, European Institute of Oncology, Milan 20141, Italy
| | - Laura Riva
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia, Milan 20139, Italy
| | - Alessandro Carugo
- Department of Experimental Oncology, European Institute of Oncology, Milan 20141, Italy.,Department of Molecular and Cellular Oncology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Giuseppe Curigliano
- Division of Experimental Therapeutics, European Institute of Oncology, Milan 20141, Italy
| | - Carmen Criscitiello
- Division of Experimental Therapeutics, European Institute of Oncology, Milan 20141, Italy
| | - Giancarlo Pruneri
- School of Medicine, University of Milan, Milan 20122, Italy.,Biobank for Translational Medicine Unit, Department of Pathology, European Institute of Oncology, Milan 20141, Italy
| | - Pier Giuseppe Pelicci
- Department of Experimental Oncology, European Institute of Oncology, Milan 20141, Italy.,Department of Oncology, University of Milan, Milan 20139, Italy
| | - Mario Faretta
- Department of Experimental Oncology, European Institute of Oncology, Milan 20141, Italy
| | - Daniela Bossi
- Department of Experimental Oncology, European Institute of Oncology, Milan 20141, Italy
| | - Luisa Lanfrancone
- Department of Experimental Oncology, European Institute of Oncology, Milan 20141, Italy
| |
Collapse
|
28
|
García-Aranda M, Redondo M. Protein Kinase Targets in Breast Cancer. Int J Mol Sci 2017; 18:ijms18122543. [PMID: 29186886 PMCID: PMC5751146 DOI: 10.3390/ijms18122543] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/20/2017] [Accepted: 11/22/2017] [Indexed: 01/10/2023] Open
Abstract
With 1.67 million new cases and 522,000 deaths in the year 2012, breast cancer is the most common type of diagnosed malignancy and the second leading cause of cancer death in women around the world. Despite the success of screening programs and the development of adjuvant therapies, a significant percentage of breast cancer patients will suffer a metastatic disease that, to this day, remains incurable and justifies the research of new therapies to improve their life expectancy. Among the new therapies that have been developed in recent years, the emergence of targeted therapies has been a milestone in the fight against cancer. Over the past decade, many studies have shown a causal role of protein kinase dysregulations or mutations in different human diseases, including cancer. Along these lines, cancer research has demonstrated a key role of many protein kinases during human tumorigenesis and cancer progression, turning these molecules into valid candidates for new targeted therapies. The subsequent discovery and introduction in 2001 of the kinase inhibitor imatinib, as a targeted treatment for chronic myelogenous leukemia, revolutionized cancer genetic pathways research, and lead to the development of multiple small-molecule kinase inhibitors against various malignancies, including breast cancer. In this review, we analyze studies published to date about novel small-molecule kinase inhibitors and evaluate if they would be useful to develop new treatment strategies for breast cancer patients.
Collapse
Affiliation(s)
- Marilina García-Aranda
- Biochemistry Department, Hospital Costa del Sol, Carretera de Cádiz km, 187, 29600 Marbella, Málaga, Spain.
| | - Maximino Redondo
- Biochemistry Department, Hospital Costa del Sol, Carretera de Cádiz km, 187, 29600 Marbella, Málaga, Spain.
- Biochemistry Department, Facultad de Medicina de la Universidad de Málaga, Bulevar Louis Pasteur 32, 29010 Málaga, Spain.
| |
Collapse
|
29
|
Zhou W, Xu S, Ying Y, Zhou R, Chen X. Resveratrol Suppresses Growth and Migration of Myelodysplastic Cells by Inhibiting the Expression of Elevated Cyclin D1 (CCND1). DNA Cell Biol 2017; 36:966-975. [PMID: 29035583 DOI: 10.1089/dna.2017.3846] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Wei Zhou
- Department of Hematology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Shilin Xu
- Department of Hematology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Yi Ying
- Department of Hematology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Ruiqing Zhou
- Department of Hematology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Xiaowei Chen
- Department of Hematology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
30
|
Han Y, Jin X, Zhou H, Liu B. Identification of key genes associated with bladder cancer using gene expression profiles. Oncol Lett 2017; 15:297-303. [PMID: 29375713 PMCID: PMC5766060 DOI: 10.3892/ol.2017.7310] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 08/23/2017] [Indexed: 12/19/2022] Open
Abstract
The aim of the present study was to further investigate the molecular mechanisms of bladder cancer. The microarray data GSE52519 were downloaded from Gene Expression Omnibus, comprising 9 bladder cancer and 3 normal bladder tissue samples. Differentially expressed genes (DEGs) were identified using Limma package analysis. Subsequently, Gene Ontology, Kyoto Encyclopedia of Genes and Genomes and Reactome pathway enrichment analyses were performed for down- and upregulated DEGs. Transcription factors and genes associated with cancer from DEGs were identified. Protein-protein interaction (PPI) networks were constructed using STRING, and pathway enrichment analysis was also conducted for genes in the core sub-network that was identified using BioNet. In total, 420 downregulated and 335 upregulated DEGs were identified. Functional and pathway enrichment analyses identified that a number of DEGs, including AURKA, CCNA2, CCNE1, CDC20 and CCNB2, were enriched in the cell cycle. Furthermore, a total of 12 upregulated proto-oncogenes were identified, including AURKA and CCNA2. In the PPI sub-network, a number of DEGs (e.g., CCNB2, CDC20, CCNA2 and MCM6) with higher degrees were enriched in the KEGG pathway of the cell cycle. In conclusion, the DEGs associated with the cell cycle (e.g., CDC20, CCNA2, CCNB2 and AURKA) may serve pivotal roles in the pathogenesis of bladder cancer.
Collapse
Affiliation(s)
- Yuping Han
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Xuefei Jin
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Hui Zhou
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Bin Liu
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
31
|
Wang W, Zhang L, Chen T, Guo W, Bao X, Wang D, Ren B, Wang H, Li Y, Wang Y, Chen S, Tang B, Yang Q, Chen C. Anticancer Effects of Resveratrol-Loaded Solid Lipid Nanoparticles on Human Breast Cancer Cells. Molecules 2017; 22:molecules22111814. [PMID: 29068422 PMCID: PMC6150230 DOI: 10.3390/molecules22111814] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Accepted: 10/21/2017] [Indexed: 12/13/2022] Open
Abstract
In this study, resveratrol-loaded solid lipid nanoparticles (Res-SLNs) were successfully designed to treat MDA-MB-231 cells. The Res-SLNs were prepared using emulsification and low-temperature solidification method. The Res-SLNs were spherical, with small size, negative charge, and narrow size distribution. Compared with free resveratrol, the Res-SLNs displayed a superior ability in inhibiting the proliferation of MDA-MB-231 cells. In addition, Res-SLNs exhibited much stronger inhibitory effects on the invasion and migration of MDA-MB-231 cells. Western blot analysis revealed that Res-SLNs could promote the ratio of Bax/Bcl-2 but decreased the expression of cyclinD1 and c-Myc. These results indicate that the Res-SLN may have great potential for breast cancer treatment.
Collapse
Affiliation(s)
- Wenrui Wang
- Department of Biotechnology, Bengbu Medical College, Anhui, Bengbu 233030, China.
| | - Lingyu Zhang
- Clinical Testing and Diagnose Experimental Center, Bengbu Medical College, Anhui, Bengbu 233030, China.
| | - Tiantian Chen
- Clinical Testing and Diagnose Experimental Center, Bengbu Medical College, Anhui, Bengbu 233030, China.
| | - Wen Guo
- Department of Biotechnology, Bengbu Medical College, Anhui, Bengbu 233030, China.
| | - Xunxia Bao
- Department of Biotechnology, Bengbu Medical College, Anhui, Bengbu 233030, China.
| | - Dandan Wang
- Department of Biotechnology, Bengbu Medical College, Anhui, Bengbu 233030, China.
| | - Baihui Ren
- Department of Biotechnology, Bengbu Medical College, Anhui, Bengbu 233030, China.
| | - Haifeng Wang
- Clinical Testing and Diagnose Experimental Center, Bengbu Medical College, Anhui, Bengbu 233030, China.
| | - Yu Li
- Clinical Testing and Diagnose Experimental Center, Bengbu Medical College, Anhui, Bengbu 233030, China.
| | - Yueyue Wang
- Clinical Testing and Diagnose Experimental Center, Bengbu Medical College, Anhui, Bengbu 233030, China.
| | - Sulian Chen
- Department of Biochemistry and Molecular Biology, Bengbu Medical College, Anhui, Bengbu 233030, China.
| | - Baoding Tang
- Department of Oncology, Bengbu Medical College, Anhui, Bengbu 233030, China.
| | - Qingling Yang
- Department of Biochemistry and Molecular Biology, Bengbu Medical College, Anhui, Bengbu 233030, China.
| | - Changjie Chen
- Department of Biochemistry and Molecular Biology, Bengbu Medical College, Anhui, Bengbu 233030, China.
| |
Collapse
|
32
|
Li J, Sun P, Yue Z, Zhang D, You K, Wang J. miR-144-3p Induces Cell Cycle Arrest and Apoptosis in Pancreatic Cancer Cells by Targeting Proline-Rich Protein 11 Expression via the Mitogen-Activated Protein Kinase Signaling Pathway. DNA Cell Biol 2017; 36:619-626. [PMID: 28574724 DOI: 10.1089/dna.2017.3656] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- Jian Li
- Department of General Surgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Peisheng Sun
- Department of General Surgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Zhongyi Yue
- Department of General Surgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Dezhong Zhang
- Department of General Surgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Kun You
- Department of General Surgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jianguo Wang
- Department of General Surgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
33
|
Pan J, Shen J, Si W, Du C, Chen D, Xu L, Yao M, Fu P, Fan W. Resveratrol promotes MICA/B expression and natural killer cell lysis of breast cancer cells by suppressing c-Myc/miR-17 pathway. Oncotarget 2017; 8:65743-65758. [PMID: 29029468 PMCID: PMC5630368 DOI: 10.18632/oncotarget.19445] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 04/26/2017] [Indexed: 12/31/2022] Open
Abstract
Major histocompatibility complex class I chain-related proteins A and B (MICA and MICB) are important ligands for recognition of tumor cells by immune effector cells. Here, we report that resveratrol upregulated the protein and mRNA expression of MICA and MICB in breast cancer cells, which in turn promoted breast cancer cell lysis by natural killer (NK) cells in vitro and in vivo. Antibodies against NK group 2 member D blocked this effect. The 3'-untranslated regions of MICA and MICB were found to be direct binding targets of miR-17. MICA and MICB expression increased or decreased in breast cancer cells transfected with a miR-17 inhibitor or mimic, respectively. C-Myc overexpression/knockdown increased/decreased transcription of the miR-17-92 cluster host gene. Resveratrol suppressed c-Myc expression, which inhibited the transcription of miR-17-92 cluster, thereby downregulating miR-17. MiR-17 expression correlated inversely with MICA and MICB expression and overall survival in two sets of breast cancer specimens. Resveratrol thus upregulates MICA and MICB by suppressing the c-Myc/miR-17 pathway in breast cancer cells, and increases the cytolysis of breast cancer cells by NK cells. This suggests resveratrol has the potential to promote antitumor immune responses in breast cancer patients.
Collapse
Affiliation(s)
- Jie Pan
- Program of Innovative Cancer Therapeutics, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang Province, Hangzhou 310003, China
| | - Jiaying Shen
- Program of Innovative Cancer Therapeutics, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang Province, Hangzhou 310003, China
| | - Wengong Si
- Program of Innovative Cancer Therapeutics, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang Province, Hangzhou 310003, China
| | - Chengyong Du
- Breast Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang Province, Hangzhou 310003, China
| | - Danni Chen
- Program of Innovative Cancer Therapeutics, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang Province, Hangzhou 310003, China
| | - Liang Xu
- Program of Innovative Cancer Therapeutics, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang Province, Hangzhou 310003, China.,Clinical Research Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang Province, Hangzhou 310003, China
| | - Minya Yao
- Breast Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang Province, Hangzhou 310003, China
| | - Peifen Fu
- Breast Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang Province, Hangzhou 310003, China
| | - Weimin Fan
- Program of Innovative Cancer Therapeutics, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang Province, Hangzhou 310003, China.,Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
34
|
Medina-Aguilar R, Pérez-Plasencia C, Gariglio P, Marchat LA, Flores-Pérez A, López-Camarillo C, García Mena J. DNA methylation data for identification of epigenetic targets of resveratrol in triple negative breast cancer cells. Data Brief 2017; 11:169-182. [PMID: 28229117 PMCID: PMC5312642 DOI: 10.1016/j.dib.2017.02.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/06/2017] [Accepted: 02/02/2017] [Indexed: 12/01/2022] Open
Abstract
Previous studies revealed that some bioactive food components have anti-cancer effects. However epigenetic effects of dietary compound resveratrol are largely unknown in breast cancer cells (M.A. Dawson, T. Kouzarides, 2012) [1]. Here we provide novel data and comparisons of DNA methylation status of promoter gene regions in response to resveratrol treatment at 24 h and 48 h versus untreated MDA-MB-231 breast cancer cells. DNA methylation changes were measured using Array-PRIMES method (aPRIMES) followed by whole-genome hybridization using human DNA methylation promoter microarray NimbleGen HG18 Refseq Promoter 3×720 K array. Our data were associated to corresponding changes in mRNA expression in a set of cancer-related genes. Using gene ontology analysis we also identify cancer-related cellular processes and pathways that can be epigenetically reprogramed by resveratrol. Data in this article are associated to the research articles “Methylation Landscape of Human Breast Cancer Cells in Response to Dietary Compound Resveratrol”. Medina Aguilar et al., PLoS ONE 11(6): e0157866. doi:10.1371/journal.pone.0157866 2016 (A.R. Medina, P.C. Pérez, L.A. Marchat, P. Gariglio, M.J. García, C.S. Rodríguez, G.E. Ruíz, et al., 2016) [2]; and “Resveratrol inhibits cell cycle progression by targeting Aurora kinase A and Polo-like kinase 1 in breast cancer cells” in Oncology Reports. Medina Aguilar et al., 2016 Jun; 35(6):3696-704. doi: 10.3892/or.2016.4728 (A.R. Medina, P. Gariglio, M.J. García, O.E. Arechaga, S.N. Villegas, C.M. Martínez et al., 2016) [3].
Collapse
Affiliation(s)
- Rubiceli Medina-Aguilar
- Departmento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), México City, México
| | | | - Patricio Gariglio
- Departmento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), México City, México
| | - Laurence A Marchat
- Escuela Nacional de Medicina y Homeopatía. Red de Biotecnología. Instituto Politécnico Nacional, México City, México
| | - Ali Flores-Pérez
- Laboratorio de Oncogenómica y Proteómica del Cáncer, Universidad Autónoma de la Ciudad de México, México City, México
| | - César López-Camarillo
- Laboratorio de Oncogenómica y Proteómica del Cáncer, Universidad Autónoma de la Ciudad de México, México City, México
| | - Jaime García Mena
- Departmento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), México City, México
| |
Collapse
|
35
|
Kamaleddin MA. The paradoxical pro- and antiangiogenic actions of resveratrol: therapeutic applications in cancer and diabetes. Ann N Y Acad Sci 2016; 1386:3-15. [PMID: 27880855 DOI: 10.1111/nyas.13283] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 09/24/2016] [Accepted: 09/29/2016] [Indexed: 02/06/2023]
Abstract
Resveratrol, a polyphenol found in grapes, peanuts, and red wine, plays different roles in diseases such as cancer and diabetes. Existing information indicates that resveratrol provides cardioprotection, as evidenced by superior postischemic ventricular recovery, reduced myocardial infarct size, and decreased number of apoptotic cardiomyocytes associated with resveratrol treatment in animal models. Cardiovascular benefits are experienced in humans with routine but not acute consumption of red wine. In this concise review, the paradoxical pro- and antiangiogenic effects of resveratrol are described, and different roles for resveratrol in the formation of new blood vessels are explained through different mechanisms. It is hypothesized that the effects of resveratrol on different cell types are not only dependent on its concentration but also on the physical and chemical conditions surrounding cells. The findings discussed herein shed light on potential therapeutic proapoptotic and antiangiogenic applications of low-dose resveratrol treatment in the prevention and treatment of different diseases.
Collapse
|