1
|
Saini S, Tuli HS, Saini RV, Saini AK, Sak K, Kaur D, Shahwan M, Chauhan R, Chauhan A. Flavonoid-Mediated Suppression of Tumor Angiogenesis: Roles of Ang-Tie/PI3K/AKT. PATHOPHYSIOLOGY 2024; 31:596-607. [PMID: 39449525 PMCID: PMC11503374 DOI: 10.3390/pathophysiology31040043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/03/2024] [Accepted: 10/09/2024] [Indexed: 10/26/2024] Open
Abstract
Angiogenesis is a process involved in the formation of new blood capillaries from pre-existing ones. It is regulated by several anti-angiogenic molecules involved in tumor growth and metastasis. The endothelial angiopoietin Ang-Tie/PI3K/AKT growth receptor pathway is necessary for healthy vascular development. The activation of AKT is controlled by a multistep process involving phosphoinositide 3-kinase (PI3K). This article aims to provide an overview of the role and mechanism of the Ang-Tie/PI3K/AKT signaling pathways and the potential of flavonoids as anti-angiogenic drugs. Flavonoids have shown great potential in preventing angiogenesis by targeting signaling pathways and exhibit additional anti-cancer properties. Research studies have revealed that the currently available anti-angiogenic drugs do not meet the safety and efficacy standards for treating tumor growth. Phytocompounds have long been a valuable resource for the development of novel therapeutic drugs. This article explores recent findings explaining the role and mechanism of the Ang-Tie/PI3K/AKT signaling pathways, as well as the interaction of flavonoids with angiogenic signaling pathways as a novel therapeutic approach. Several investigations have shown that synergistic studies of natural phytocompounds have great potential to target these pathways to inhibit tumor growth. Therefore, flavonoid-based medications may offer a more effective synergistic strategy to treat cancer.
Collapse
Affiliation(s)
- Shallu Saini
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala 133207, India; (R.V.S.); (A.K.S.)
| | - Hardeep Singh Tuli
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala 133207, India; (R.V.S.); (A.K.S.)
| | - Reena V. Saini
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala 133207, India; (R.V.S.); (A.K.S.)
| | - Adesh K. Saini
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala 133207, India; (R.V.S.); (A.K.S.)
| | | | - Damandeep Kaur
- University Centre for Research and Development, Chandigarh University, Mohali 140413, India;
| | - Moyad Shahwan
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman 4184, United Arab Emirates;
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman 346, United Arab Emirates
| | - Ritu Chauhan
- Department of Biotechnology, Graphic Era Deemed to Be University, Dehradun 248002, India;
| | - Abhishek Chauhan
- Amity Institute of Environmental Toxicology, Safety and Management, Amity University, Sector-125, Noida 201303, India;
| |
Collapse
|
2
|
Dallavalasa S, Tulimilli SV, Bettada VG, Karnik M, Uthaiah CA, Anantharaju PG, Nataraj SM, Ramashetty R, Sukocheva OA, Tse E, Salimath PV, Madhunapantula SV. Vitamin D in Cancer Prevention and Treatment: A Review of Epidemiological, Preclinical, and Cellular Studies. Cancers (Basel) 2024; 16:3211. [PMID: 39335182 PMCID: PMC11430526 DOI: 10.3390/cancers16183211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/12/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Inhibition of human carcinomas has previously been linked to vitamin D due to its effects on cancer cell proliferation, migration, angiogenesis, and apoptosis induction. The anticancer activity of vitamin D has been confirmed by several studies, which have shown that increased cancer incidence is associated with decreased vitamin D and that dietary supplementation of vitamin D slows down the growth of xenografted tumors in mice. Vitamin D inhibits the growth of cancer cells by the induction of apoptosis as well as by arresting the cells at the G0/G1 (or) G2/M phase of the cell cycle. Aim and Key Scientific Concepts of the Review: The purpose of this article is to thoroughly review the existing information and discuss and debate to conclude whether vitamin D could be used as an agent to prevent/treat cancers. The existing empirical data have demonstrated that vitamin D can also work in the absence of vitamin D receptors (VDRs), indicating the presence of multiple mechanisms of action for this sunshine vitamin. Polymorphism in the VDR is known to play a key role in tumor cell metastasis and drug resistance. Although there is evidence that vitamin D has both therapeutic and cancer-preventive properties, numerous uncertainties and concerns regarding its use in cancer treatment still exist. These include (a) increased calcium levels in individuals receiving therapeutic doses of vitamin D to suppress the growth of cancer cells; (b) hyperglycemia induction in certain vitamin D-treated study participants; (c) a dearth of evidence showing preventive or therapeutic benefits of cancer in clinical trials; (d) very weak support from proof-of-principle studies; and (e) the inability of vitamin D alone to treat advanced cancers. Addressing these concerns, more potent and less toxic vitamin D analogs have been created, and these are presently undergoing clinical trial evaluation. To provide key information regarding the functions of vitamin D and VDRs, this review provided details of significant advancements in the functional analysis of vitamin D and its analogs and VDR polymorphisms associated with cancers.
Collapse
Affiliation(s)
- Siva Dallavalasa
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR) Laboratory (DST-FIST Supported Center and ICMR Collaborating Center of Excellence-ICMR-CCoE), Department of Biochemistry (DST-FIST Supported Department), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570015, Karnataka, India
| | - SubbaRao V Tulimilli
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR) Laboratory (DST-FIST Supported Center and ICMR Collaborating Center of Excellence-ICMR-CCoE), Department of Biochemistry (DST-FIST Supported Department), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570015, Karnataka, India
| | - Vidya G Bettada
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR) Laboratory (DST-FIST Supported Center and ICMR Collaborating Center of Excellence-ICMR-CCoE), Department of Biochemistry (DST-FIST Supported Department), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570015, Karnataka, India
| | - Medha Karnik
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR) Laboratory (DST-FIST Supported Center and ICMR Collaborating Center of Excellence-ICMR-CCoE), Department of Biochemistry (DST-FIST Supported Department), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570015, Karnataka, India
| | - Chinnappa A Uthaiah
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR) Laboratory (DST-FIST Supported Center and ICMR Collaborating Center of Excellence-ICMR-CCoE), Department of Biochemistry (DST-FIST Supported Department), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570015, Karnataka, India
| | - Preethi G Anantharaju
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR) Laboratory (DST-FIST Supported Center and ICMR Collaborating Center of Excellence-ICMR-CCoE), Department of Biochemistry (DST-FIST Supported Department), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570015, Karnataka, India
| | - Suma M Nataraj
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR) Laboratory (DST-FIST Supported Center and ICMR Collaborating Center of Excellence-ICMR-CCoE), Department of Biochemistry (DST-FIST Supported Department), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570015, Karnataka, India
| | - Rajalakshmi Ramashetty
- Department of Physiology, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570015, Karnataka, India
| | - Olga A Sukocheva
- Department of Hepatology, Royal Adelaide Hospital, Port Rd., Adelaide, SA 5000, Australia
| | - Edmund Tse
- Department of Hepatology, Royal Adelaide Hospital, Port Rd., Adelaide, SA 5000, Australia
| | - Paramahans V Salimath
- JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570015, Karnataka, India
| | - SubbaRao V Madhunapantula
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR) Laboratory (DST-FIST Supported Center and ICMR Collaborating Center of Excellence-ICMR-CCoE), Department of Biochemistry (DST-FIST Supported Department), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570015, Karnataka, India
- Special Interest Group in Cancer Biology and Cancer Stem Cells (SIG-CBCSC), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570015, Karnataka, India
| |
Collapse
|
3
|
Albini A, Noonan DM, Corradino P, Magnoni F, Corso G. The Past and Future of Angiogenesis as a Target for Cancer Therapy and Prevention. Cancer Prev Res (Phila) 2024; 17:289-303. [PMID: 38714356 DOI: 10.1158/1940-6207.capr-24-0085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/04/2024] [Accepted: 05/03/2024] [Indexed: 05/09/2024]
Abstract
Cancer growth is dependent on angiogenesis, the formation of new blood vessels, which represents a hallmark of cancer. After this concept was established in the 1970s, inhibition of tumor development and metastases by blocking the neoangiogenic process has been an important approach to the treatment of tumors. However, antiangiogenic therapies are often administered when cancer has already progressed. The key to reducing the cancer burden is prevention. We noticed 20 years ago that a series of possible cancer chemopreventive agents showed antiangiogenic properties when tested in experimental models. This article reviews the relevant advances in the understanding of the rationale for targeting angiogenesis for cancer therapy, prevention, and interception and recently investigated substances with antiangiogenic activity that may be suitable for such strategies. Many compounds, either dietary derivatives or repurposed drugs, with antiangiogenic activity are possible tools for cancer angioprevention. Such molecules have a favorable safety profile and are likely to allow the prolonged duration necessary for an efficient preventive strategy. Recent evidence on mechanisms and possible use is described here for food derivatives, including flavonoids, retinoids, triterpenoids, omega fatty acids, and carotenoids from marine microorganisms. As examples, a number of compounds, including epigallocatechin, resveratrol, xanthohumol, hydroxytyrosol, curcumin, fenretinide, lycopene, fucoxanthin, and repurposed drugs, such as aspirin, β blockers, renin-angiotensin-aldosterone inhibitors, carnitines, and biguanides, are reviewed.
Collapse
Affiliation(s)
- Adriana Albini
- European Institute of Oncologi IEO, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Douglas M Noonan
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
- IRCCS MultiMedica, Milan, Italy
| | - Paola Corradino
- European Institute of Oncologi IEO, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Francesca Magnoni
- European Institute of Oncologi IEO, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Giovanni Corso
- European Institute of Oncologi IEO, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
4
|
Marrero AD, Cárdenas C, Castilla L, Ortega-Vidal J, Quesada AR, Martínez-Poveda B, Medina MÁ. Antiangiogenic Potential of an Olive Oil Extract: Insights from a Proteomic Study. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:13023-13038. [PMID: 38809962 PMCID: PMC11181319 DOI: 10.1021/acs.jafc.3c08851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 05/31/2024]
Abstract
Extra virgin olive oil (EVOO), a staple of the Mediterranean diet, is rich in phenolic compounds recognized for their potent bioactive effects, including anticancer and anti-inflammatory properties. However, its effects on vascular health remain relatively unexplored. In this study, we examined the impact of a "picual" EVOO extract from Jaén, Spain, on endothelial cells. Proteomic analysis revealed the modulation of angiogenesis-related processes. In subsequent in vitro experiments, the EVOO extract inhibited endothelial cell migration, adhesion, invasion, ECM degradation, and tube formation while inducing apoptosis. These results provide robust evidence of the extract's antiangiogenic potential. Our findings highlight the potential of EVOO extracts in mitigating angiogenesis-related pathologies, such as cancer, macular degeneration, and diabetic retinopathy.
Collapse
Affiliation(s)
- Ana Dácil Marrero
- Departamento
de Biología Molecular y Bioquímica, Facultad de Ciencias,
Andalucía Tech, Universidad de Málaga, E-29071 Málaga, Spain
- Instituto
de Investigación Biomédica y Plataforma en Nanomedicina-IBIMA
Plataforma BIONAND (Biomedical Research Institute of Málaga), E-29071 Málaga, Spain
- CIBER
de Enfermedades Raras (CIBERER), Instituto
de Salud Carlos III, E-28029 Madrid, Spain
| | - Casimiro Cárdenas
- Departamento
de Biología Molecular y Bioquímica, Facultad de Ciencias,
Andalucía Tech, Universidad de Málaga, E-29071 Málaga, Spain
- Servicios
Centrales de Apoyo a la Investigación (SCAI), Universidad de Málaga, E-29071 Málaga, Spain
| | - Laura Castilla
- Departamento
de Biología Molecular y Bioquímica, Facultad de Ciencias,
Andalucía Tech, Universidad de Málaga, E-29071 Málaga, Spain
- Instituto
de Investigación Biomédica y Plataforma en Nanomedicina-IBIMA
Plataforma BIONAND (Biomedical Research Institute of Málaga), E-29071 Málaga, Spain
| | - Juan Ortega-Vidal
- Departamento
de Química Inorgánica y Orgánica, Campus de Excelencia
Internacional Agroalimentaria ceiA3, Universidad
de Jaén, Jaén E- 23071, Spain
| | - Ana R. Quesada
- Departamento
de Biología Molecular y Bioquímica, Facultad de Ciencias,
Andalucía Tech, Universidad de Málaga, E-29071 Málaga, Spain
- Instituto
de Investigación Biomédica y Plataforma en Nanomedicina-IBIMA
Plataforma BIONAND (Biomedical Research Institute of Málaga), E-29071 Málaga, Spain
- CIBER
de Enfermedades Raras (CIBERER), Instituto
de Salud Carlos III, E-28029 Madrid, Spain
| | - Beatriz Martínez-Poveda
- Departamento
de Biología Molecular y Bioquímica, Facultad de Ciencias,
Andalucía Tech, Universidad de Málaga, E-29071 Málaga, Spain
- Instituto
de Investigación Biomédica y Plataforma en Nanomedicina-IBIMA
Plataforma BIONAND (Biomedical Research Institute of Málaga), E-29071 Málaga, Spain
- CIBER
de
Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Miguel Ángel Medina
- Departamento
de Biología Molecular y Bioquímica, Facultad de Ciencias,
Andalucía Tech, Universidad de Málaga, E-29071 Málaga, Spain
- Instituto
de Investigación Biomédica y Plataforma en Nanomedicina-IBIMA
Plataforma BIONAND (Biomedical Research Institute of Málaga), E-29071 Málaga, Spain
- CIBER
de Enfermedades Raras (CIBERER), Instituto
de Salud Carlos III, E-28029 Madrid, Spain
| |
Collapse
|
5
|
Shah R, Phatak N, Choudhary A, Gadewar S, Ajazuddin, Bhattacharya S. Exploring the Theranostic Applications and Prospects of Nanobubbles. Curr Pharm Biotechnol 2024; 25:1167-1181. [PMID: 37861011 DOI: 10.2174/0113892010248189231010085827] [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: 02/08/2023] [Revised: 09/01/2023] [Accepted: 09/07/2023] [Indexed: 10/21/2023]
Abstract
Anticancer medications as well as additional therapeutic compounds, have poor clinical effectiveness due to their diverse distribution, non-selectivity for malignant cells, and undesirable off-target side effects. As a result, ultrasound-based targeted delivery of therapeutic compounds carried in sophisticated nanocarriers has grown in favor of cancer therapy and control. Nanobubbles are nanoscale bubbles that exhibit unique physiochemical properties in both their inner core and outer shell. Manufacturing nanobubbles primarily aims to enhance therapeutic agents' bioavailability, stability, and targeted delivery. The small size of nanobubbles allows for their extravasation from blood vessels into surrounding tissues and site-specific release through ultrasound targeting. Ultrasound technology is widely utilized for therapy due to its speed, safety, and cost-effectiveness, and micro/nanobubbles, as ultrasound contrast agents, have numerous potential applications in disease treatment. Thus, combining ultrasound applications with NBs has recently demonstrated increased localization of anticancer molecules in tumor tissues with triggered release behavior. Consequently, an effective therapeutic concentration of drugs/genes is achieved in target tumor tissues with ultimately increased therapeutic efficacy and minimal side effects on other non-cancerous tissues. This paper provides a brief overview of the production processes for nanobubbles, along with their key characteristics and potential therapeutic uses.
Collapse
Affiliation(s)
- Rahul Shah
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
| | - Niraj Phatak
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
| | - Ashok Choudhary
- Department of Quality Assurance, School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
| | - Sakshi Gadewar
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
| | - Ajazuddin
- Department of Pharmaceutics, Rungta College of Pharmaceutical Sciences & Research, Khoka-Kurud Road, Bhilai, Chhattisgarh, 490024, India
| | - Sankha Bhattacharya
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
| |
Collapse
|
6
|
Kamal MV, Damerla RR, Dikhit PS, Kumar NAN. Prostaglandin-endoperoxide synthase 2 (PTGS2) gene expression and its association with genes regulating the VEGF signaling pathway in head and neck squamous cell carcinoma. J Oral Biol Craniofac Res 2023; 13:567-574. [PMID: 37559688 PMCID: PMC10407435 DOI: 10.1016/j.jobcr.2023.07.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/30/2023] [Accepted: 07/20/2023] [Indexed: 08/11/2023] Open
Abstract
Introduction The PTGS2 gene codes for the cyclooxygenase-2 (COX-2) enzyme that catalyzes the committed step in prostaglandin (PG) synthesis. Various in-vivo and in-vitro data suggest that prostaglandin E2 mediates as a signaling molecule for activating the VEGF signaling pathway (VSP), forming an association between COX-2 and VSP. Several chemotherapy regimens increasingly rely on preventing the synthesis of PGs. The targeted and metronomic chemotherapy agents, which suppress the COX-2 enzymes, have a major role in suppressing the oral cancer cascade. Hence, this study was designed to understand the pattern of PTGS2 expression and genes regulating VSP in head and neck cancers. Methods PTGS2 expression was analyzed in the TCGA database computationally with the help of the UALCAN web-server. The expression of VEGF signaling pathway genes was mined, and their expression pattern was determined. Co-expression analysis was done to elucidate the association between VEGF signaling genes and PTGS2. The ShineyGo web server was used for gene set enrichment. Results Significantly high PTGS2 expression was observed in tumor samples. Further genes regulating VEGF signaling were significantly overexpressed in tumor samples. Co-expression analysis results showed a significant positive correlation between PTGS2 and angiogenesis-regulating genes. The majority of the genes were enriched for angiogenesis pathways. Conclusion PTGS2 was significantly expressed in head and neck cancer, and its expression was associated with genes regulating angiogenesis.
Collapse
Affiliation(s)
- Mehta Vedant Kamal
- Department of Surgical Oncology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Rama Rao Damerla
- Department of Medical Genetics, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Punit Singh Dikhit
- Department of Surgical Oncology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Naveena AN Kumar
- Department of Surgical Oncology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| |
Collapse
|
7
|
Rocchetti MT, Bellanti F, Zadorozhna M, Fiocco D, Mangieri D. Multi-Faceted Role of Luteolin in Cancer Metastasis: EMT, Angiogenesis, ECM Degradation and Apoptosis. Int J Mol Sci 2023; 24:ijms24108824. [PMID: 37240168 DOI: 10.3390/ijms24108824] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/09/2023] [Accepted: 05/13/2023] [Indexed: 05/28/2023] Open
Abstract
Luteolin (3',4',5,7-tetrahydroxyflavone), a member of the flavonoid family derived from plants and fruits, shows a wide range of biomedical applications. In fact, due to its anti-inflammatory, antioxidant and immunomodulatory activities, Asian medicine has been using luteolin for centuries to treat several human diseases, including arthritis, rheumatism, hypertension, neurodegenerative disorders and various infections. Of note, luteolin displays many anti-cancer/anti-metastatic properties. Thus, the purpose of this review consists in highlighting the relevant mechanisms by which luteolin inhibits tumor progression in metastasis, i.e., affecting epithelial-mesenchymal transition (EMT), repressing angiogenesis and lysis of extracellular matrix (ECM), as well as inducing apoptosis.
Collapse
Affiliation(s)
- Maria Teresa Rocchetti
- Department of Clinical and Experimental Medicine, University of Foggia, Via Pinto 1, 71122 Foggia, Italy
| | - Francesco Bellanti
- Department of Medical and Surgical Sciences, University of Foggia, Via Pinto 1, 71122 Foggia, Italy
| | - Mariia Zadorozhna
- Medical Genetics Unit, Department of Molecular Medicine, University of Pavia, Via Forlanini 14, 27100 Pavia, Italy
| | - Daniela Fiocco
- Department of Clinical and Experimental Medicine, University of Foggia, Via Pinto 1, 71122 Foggia, Italy
| | - Domenica Mangieri
- Department of Clinical and Experimental Medicine, University of Foggia, Via Pinto 1, 71122 Foggia, Italy
| |
Collapse
|
8
|
He J, Liu Z, Zhu X, Xia H, Gao H, Lu J. Ultrasonic Microbubble Cavitation Enhanced Tissue Permeability and Drug Diffusion in Solid Tumor Therapy. Pharmaceutics 2022; 14:1642. [PMID: 36015267 PMCID: PMC9414228 DOI: 10.3390/pharmaceutics14081642] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/31/2022] [Accepted: 08/04/2022] [Indexed: 01/21/2023] Open
Abstract
Chemotherapy has an essential role not only in advanced solid tumor therapy intervention but also in society's health at large. Chemoresistance, however, seriously restricts the efficiency and sensitivity of chemotherapeutic agents, representing a significant threat to patients' quality of life and life expectancy. How to reverse chemoresistance, improve efficacy sensitization response, and reduce adverse side effects need to be tackled urgently. Recently, studies on the effect of ultrasonic microbubble cavitation on enhanced tissue permeability and retention (EPR) have attracted the attention of researchers. Compared with the traditional targeted drug delivery regimen, the microbubble cavitation effect, which can be used to enhance the EPR effect, has the advantages of less trauma, low cost, and good sensitization effect, and has significant application prospects. This article reviews the research progress of ultrasound-mediated microbubble cavitation in the treatment of solid tumors and discusses its mechanism of action to provide new ideas for better treatment strategies.
Collapse
Affiliation(s)
- Jide He
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| | - Zenan Liu
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| | - Xuehua Zhu
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| | - Haizhui Xia
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China
| | - Jian Lu
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
9
|
Azab AE, Alesawy MS, Eldehna WM, Elwan A, Eissa IH. New [1,2,4]triazolo[4,3-c]quinazoline derivatives as vascular endothelial growth factor receptor-2 inhibitors and apoptosis inducers: Design, synthesis, docking, and antiproliferative evaluation. Arch Pharm (Weinheim) 2022; 355:e2200133. [PMID: 35822666 DOI: 10.1002/ardp.202200133] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/16/2022] [Accepted: 06/20/2022] [Indexed: 11/10/2022]
Abstract
In continuation of our previous efforts in the field of design and synthesis of vascular endothelial growth factor receptor (VEGFR)-2 inhibitors, a new series of [1,2,4]triazolo[4,3-c]quinazoline derivatives were designed and synthesized as modified analogs of some reported VEGFR-2 inhibitors. The synthesized compounds were designed to have the essential pharmacophoric features of VEGFR-2 inhibitors. Antiproliferative activities of the synthesized compounds were investigated against two tumor cell lines (HepG2 and HCT-116) using sorafenib as a positive control. Compound 10k emerged as the most promising antiproliferative agent with IC50 values of 4.88 and 5.21 µM against HepG2 and HCT-116 cells, respectively. Also, it showed the highest inhibitory activity against VEGFR-2 with an IC50 value of 53.81 nM compared to sorafenib (IC50 = 44.34 nM). Cell cycle analysis revealed that compound 10k can arrest HepG2 cells at both the S and G2/M phases. In addition, this compound produced a tenfold increase in apoptotic cells compared to the control. Furthermore, the effect of compound 10k on the expression level of BAX, Bcl-2, and caspase-3 was assessed. This compound caused a 3.35-fold increase in BAX expression levels and a 1.25-fold reduction in Bcl-2 expression levels. The BAX/Bcl-2 ratio was calculated to be 4.57, indicating a promising apoptotic effect. It also showed a significant increase in the level of caspase-3 (4.12-fold) compared to the control cells. In silico docking, absorption, distribution, metabolism, excretion, and toxicity, and toxicity studies were performed for the synthesized compounds to investigate their binding patterns against the proposed biological target (VEGFR-2) and to assess the drug-likeness characters.
Collapse
Affiliation(s)
- Ahmed E Azab
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Mohamed S Alesawy
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Wagdy M Eldehna
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt.,School of Biotechnology, Badr University in Cairo, Cairo, Egypt
| | - Alaa Elwan
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Ibrahim H Eissa
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| |
Collapse
|
10
|
Motallebzadeh E, Suliman Maashi M, Z.Mahmoud M, Aliasgharzedeh A, Vakili Z, Talaei SA, Mohseni M. Radioprotective effects of N-acetylcysteine on rats’ brainstem following megavoltage X-irradiations. Appl Radiat Isot 2022; 187:110348. [PMID: 35779304 DOI: 10.1016/j.apradiso.2022.110348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/25/2022] [Accepted: 06/23/2022] [Indexed: 11/24/2022]
|
11
|
Lu S, Zhao P, Deng Y, Liu Y. Mechanistic Insights and Therapeutic Delivery through Micro/Nanobubble-Assisted Ultrasound. Pharmaceutics 2022; 14:pharmaceutics14030480. [PMID: 35335857 PMCID: PMC8954263 DOI: 10.3390/pharmaceutics14030480] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/12/2022] [Accepted: 02/19/2022] [Indexed: 02/05/2023] Open
Abstract
Ultrasound with low frequency (20–100 kHz) assisted drug delivery has been widely investigated as a non-invasive method to enhance the permeability and retention effect of drugs. The functional micro/nanobubble loaded with drugs could provide an unprecedented opportunity for targeted delivery. Then, ultrasound with higher intensity would locally burst bubbles and release agents, thus avoiding side effects associated with systemic administration. Furthermore, ultrasound-mediated destruction of micro/nanobubbles can effectively increase the permeability of vascular membranes and cell membranes, thereby not only increasing the distribution concentration of drugs in the interstitial space of target tissues but also promoting the penetration of drugs through cell membranes into the cytoplasm. These advancements have transformed ultrasound from a purely diagnostic utility into a promising theragnostic tool. In this review, we first discuss the structure and generation of micro/nanobubbles. Second, ultrasound parameters and mechanisms of therapeutic delivery are discussed. Third, potential biomedical applications of micro/nanobubble-assisted ultrasound are summarized. Finally, we discuss the challenges and future directions of ultrasound combined with micro/nanobubbles.
Collapse
|
12
|
iASPP is essential for HIF-1α stabilization to promote angiogenesis and glycolysis via attenuating VHL-mediated protein degradation. Oncogene 2022; 41:1944-1958. [PMID: 35169254 DOI: 10.1038/s41388-022-02234-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 01/24/2022] [Accepted: 02/04/2022] [Indexed: 12/15/2022]
Abstract
Hypoxia-inducible factor-1α (HIF-1α) plays central roles in the hypoxia response. It is highly expressed in multiple cancers, but not always correlated with hypoxia. Mutation of the von Hippel-Lindau (VHL) gene, which encodes an E3 ligase, contributes to the constructive activation of HIF-1α in specific tumor types, as exemplified by renal cell carcinoma; but how VHL wild-type tumors acquire this ability is not completely understood. Here, we found that the oncogene iASPP (inhibitor of apoptosis-simulating protein of p53) plays essential roles in such a context. Genetic inhibition of iASPP reduced tumor growth, accompanied by impaired angiogenesis, increased areas of tumor necrosis, and reduced glycolysis that was HIF-1α-dependent. These abilities of iASPP were validated by in vitro assays. Mechanistically, iASPP directly binds VHL at its β domain, a region also involved in HIF-1α binding, therefore blocking VHL's binding and the subsequent degradation of HIF-1α protein under normoxia. iASPP levels correlate with HIF-1α protein and vascular endothelial growth factor (VEGF) and the glucose transporter protein type 1(GLUT1), representative HIF-1α target genes, in human colon cancer tissues. Furthermore, inhibition of iASPP expression synergizes with low toxic dose of the HIF-1α inhibitor YC-1 to inhibit HIF-1α expression and tumor growth. Our findings suggest that iASPP contributes to HIF-1α activation in cancers, and that iASPP-mediated HIF-1α stabilization has potential as a therapeutic approach against cancer.
Collapse
|
13
|
García-Caballero M, Torres-Vargas JA, Marrero AD, Martínez-Poveda B, Medina MÁ, Quesada AR. Angioprevention of Urologic Cancers by Plant-Derived Foods. Pharmaceutics 2022; 14:pharmaceutics14020256. [PMID: 35213989 PMCID: PMC8875200 DOI: 10.3390/pharmaceutics14020256] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 02/05/2023] Open
Abstract
The number of cancer cases worldwide keeps growing unstoppably, despite the undeniable advances achieved by basic research and clinical practice. Urologic tumors, including some as prevalent as prostate, bladder or kidney tumors, are no exceptions to this rule. Moreover, the fact that many of these tumors are detected in early stages lengthens the duration of their treatment, with a significant increase in health care costs. In this scenario, prevention offers the most cost-effective long-term strategy for the global control of these diseases. Although specialized diets are not the only way to decrease the chances to develop cancer, epidemiological evidence support the role of certain plant-derived foods in the prevention of urologic cancer. In many cases, these plants are rich in antiangiogenic phytochemicals, which could be responsible for their protective or angiopreventive properties. Angiogenesis inhibition may contribute to slow down the progression of the tumor at very different stages and, for this reason, angiopreventive strategies could be implemented at different levels of chemoprevention, depending on the targeted population. In this review, epidemiological evidence supporting the role of certain plant-derived foods in urologic cancer prevention are presented, with particular emphasis on their content in bioactive phytochemicals that could be used in the angioprevention of cancer.
Collapse
Affiliation(s)
- Melissa García-Caballero
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
| | - José Antonio Torres-Vargas
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
| | - Ana Dácil Marrero
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
| | - Beatriz Martínez-Poveda
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), E-28019 Madrid, Spain
| | - Miguel Ángel Medina
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
- CIBER de Enfermedades Raras (CIBERER), E-29071 Malaga, Spain
| | - Ana R. Quesada
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
- CIBER de Enfermedades Raras (CIBERER), E-29071 Malaga, Spain
- Correspondence:
| |
Collapse
|
14
|
Guc Z, Guc H. Sunlight: Friend or foe? A natural source of vitamin D or a risk factor for cutaneous malignancy? TURKISH JOURNAL OF PLASTIC SURGERY 2022. [DOI: 10.4103/tjps.tjps_66_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
15
|
Dong S, Chen Z, Wang L, Liu Y, Stagos D, Lin X, Liu M. Marine Bromophenol Bis(2,3,6-Tribromo-4,5-Dihydroxybenzyl)ether Inhibits Angiogenesis in Human Umbilical Vein Endothelial Cells and Reduces Vasculogenic Mimicry in Human Lung Cancer A549 Cells. Mar Drugs 2021; 19:641. [PMID: 34822512 PMCID: PMC8617710 DOI: 10.3390/md19110641] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/08/2021] [Accepted: 11/12/2021] [Indexed: 12/11/2022] Open
Abstract
Angiogenesis, including the growth of new capillary blood vessels from existing ones and the malignant tumors cells formed vasculogenic mimicry, is quite important for the tumor metastasis. Anti-angiogenesis is one of the significant therapies in tumor treatment, while the clinical angiogenesis inhibitors usually exhibit endothelial cells dysfunction and drug resistance. Bis(2,3,6-tribromo-4,5-dihydroxybenzyl)ether (BTDE), a marine algae-derived bromophenol compound, has shown various biological activities, however, its anti-angiogenesis function remains unknown. The present study illustrated that BTDE had anti-angiogenesis effect in vitro through inhibiting human umbilical vein endothelial cells migration, invasion, tube formation, and the activity of matrix metalloproteinases 9 (MMP9), and in vivo BTDE also blocked intersegmental vessel formation in zebrafish embryos. Moreover, BTDE inhibited the migration, invasion, and vasculogenic mimicry formation of lung cancer cell A549. All these results indicated that BTDE could be used as a potential candidate in anti-angiogenesis for the treatment of cancer.
Collapse
Affiliation(s)
- Songtao Dong
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; (S.D.); (Z.C.); (L.W.); (Y.L.)
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| | - Zhongyuan Chen
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; (S.D.); (Z.C.); (L.W.); (Y.L.)
| | - Li Wang
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; (S.D.); (Z.C.); (L.W.); (Y.L.)
| | - Yankai Liu
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; (S.D.); (Z.C.); (L.W.); (Y.L.)
| | - Dimitrios Stagos
- Department of Biochemistry and Biotechnology, School of Health Sciences, University of Thessaly, Biopolis, 41500 Larissa, Greece;
| | - Xiukun Lin
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, 319 Zhongshan Road, Jiangyang, Luzhou 646000, China;
| | - Ming Liu
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; (S.D.); (Z.C.); (L.W.); (Y.L.)
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| |
Collapse
|
16
|
Rampino A, Annese T, Margari A, Tamma R, Ribatti D. Nutraceuticals and their role in tumor angiogenesis. Exp Cell Res 2021; 408:112859. [PMID: 34637764 DOI: 10.1016/j.yexcr.2021.112859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/28/2021] [Accepted: 10/02/2021] [Indexed: 01/15/2023]
Abstract
Angiogenesis plays a pivotal role in cancer initiation, maintenance, and progression. Diet may inhibit, retard or reverse these processes affecting angiogenesis (angioprevention). Nutraceuticals, such as omega-3 fatty acids, amino acids, proteins, vitamins, minerals, fibers, and phenolic compounds, improve health benefits as they are a source of bioactive compounds that, among other effects, can regulate angiogenesis. The literature concerning the pro-angiogenic and/or anti-angiogenic nutraceuticals and the possible activated pathways in cancer and other non-neoplastic diseases by in vivo and in vitro experiments are reviewed.
Collapse
Affiliation(s)
- Antonio Rampino
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Tiziana Annese
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Anna Margari
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Roberto Tamma
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy.
| |
Collapse
|
17
|
Tseng FC, Huang TC. Using data mining technology to explore homocysteine at low levels. Medicine (Baltimore) 2021; 100:e26893. [PMID: 34414944 PMCID: PMC8376364 DOI: 10.1097/md.0000000000026893] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 07/19/2021] [Indexed: 01/04/2023] Open
Abstract
A high homocysteine level is known to be an independent risk factor for cardiovascular diseases; however, whether or not low homocysteine level contributes to any damage to the body has not been extensively studied. Furthermore, acquiring healthy subject databases from domestic studies on homocysteine is not trivial. Therefore, we aimed to investigate the causality between serum homocysteine levels and health status and lifestyle factors, particularly with a focus on low serum homocysteine levels. Additionally, we discussed a systematic methodical platform for data collection and statistical analysis, using the descriptive analysis of the chi-square test, t test, multivariate analysis of variance, and logistic regression.This study was a cross-sectional analysis of 5864 subjects (i.e., clients of a health examination clinic) in Taipei, Taiwan during a general health check-up in 2017. The patients' personal information and associated links were excluded. A sample group was selected as per the health criteria defined for this research whose data were processed using SPSS for descriptive statistical analysis using chi-square test, t test, multivariate analysis of variance, and logistic regression analysis.Those working for >12 hours/day had a higher homocysteine level than those working for <12 hours/day (P < .001). The average serum homocysteine level was 7.9 and 8.6 mol/L for people with poor sleep quality and good sleep quality, respectively (P = .003). The homocysteine value of people known to have cancer was analyzed using the logistic regression analysis, revealing a Δodds value of 0.898. The percentage of subjects with a homocysteine value of ≤6.3 μmol/L, who perceived their health status as "not very good" or "very bad," was higher than those with a higher homocysteine level. The number of subjects who perceived their health as poor was higher than expected.The results suggest that the homocysteine level could be an effective health management indicator. We conclude that normal homocysteine level should not be ≤6.3 μmol/L. Moreover, homocysteine should not be considered as harmful and its fluctuations from the normal range could be utilized to infer a person's physical status for health management.
Collapse
Affiliation(s)
- Fei-Ching Tseng
- The Lianan Wellness Center of the Lianan Preventative Medicine Institution, Songshan District, Taipei City
| | - Tin-Chung Huang
- Ching-Kuo Institute of Management and Health – Graduate School of Health Industry, Zhongshan District, Keelung City, Taiwan
| |
Collapse
|
18
|
Aggarwal N, Yadav J, Chhakara S, Janjua D, Tripathi T, Chaudhary A, Chhokar A, Thakur K, Singh T, Bharti AC. Phytochemicals as Potential Chemopreventive and Chemotherapeutic Agents for Emerging Human Papillomavirus-Driven Head and Neck Cancer: Current Evidence and Future Prospects. Front Pharmacol 2021; 12:699044. [PMID: 34354591 PMCID: PMC8329252 DOI: 10.3389/fphar.2021.699044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/17/2021] [Indexed: 12/20/2022] Open
Abstract
Head and neck cancer (HNC) usually arises from squamous cells of the upper aerodigestive tract that line the mucosal surface in the head and neck region. In India, HNC is common in males, and it is the sixth most common cancer globally. Conventionally, HNC attributes to the use of alcohol or chewing tobacco. Over the past four decades, portions of human papillomavirus (HPV)-positive HNC are increasing at an alarming rate. Identification based on the etiological factors and molecular signatures demonstrates that these neoplastic lesions belong to a distinct category that differs in pathological characteristics and therapeutic response. Slow development in HNC therapeutics has resulted in a low 5-year survival rate in the last two decades. Interestingly, HPV-positive HNC has shown better outcomes following conservative treatments and immunotherapies. This raises demand to have a pre-therapy assessment of HPV status to decide the treatment strategy. Moreover, there is no HPV-specific treatment for HPV-positive HNC patients. Accumulating evidence suggests that phytochemicals are promising leads against HNC and show potential as adjuvants to chemoradiotherapy in HNC. However, only a few of these phytochemicals target HPV. The aim of the present article was to collate data on various leading phytochemicals that have shown promising results in the prevention and treatment of HNC in general and HPV-driven HNC. The review explores the possibility of using these leads against HPV-positive tumors as some of the signaling pathways are common. The review also addresses various challenges in the field that prevent their use in clinical settings.
Collapse
Affiliation(s)
- Nikita Aggarwal
- Molecular Oncology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, India
| | - Joni Yadav
- Molecular Oncology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, India
| | - Suhail Chhakara
- Molecular Oncology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, India
| | - Divya Janjua
- Molecular Oncology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, India
| | - Tanya Tripathi
- Molecular Oncology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, India
| | - Apoorva Chaudhary
- Molecular Oncology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, India
| | - Arun Chhokar
- Molecular Oncology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, India
| | - Kulbhushan Thakur
- Molecular Oncology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, India
| | - Tejveer Singh
- Molecular Oncology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, India
| | - Alok Chandra Bharti
- Molecular Oncology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, India
| |
Collapse
|
19
|
Wang JL, Lan YW, Tsai YT, Chen YC, Staniczek T, Tsou YA, Yen CC, Chen CM. Additive Antiproliferative and Antiangiogenic Effects of Metformin and Pemetrexed in a Non-Small-Cell Lung Cancer Xenograft Model. Front Cell Dev Biol 2021; 9:688062. [PMID: 34235153 PMCID: PMC8255984 DOI: 10.3389/fcell.2021.688062] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/31/2021] [Indexed: 12/16/2022] Open
Abstract
Lung cancer is heterogeneous and challenging to cope with once it has progressed. Chemotherapy is the first step once no active driver mutation has been discovered. Non-antitumor drugs have been found to be beneficial when used as adjuvants to chemotherapy. In this study, the additive effect and mechanism of metformin combined with pemetrexed in non-small-cell lung cancer (NSCLC) cells were elucidated. Three NSCLC cell lines, A549, H1975, and HCC827, were used to analyze tumor cell proliferation, colony formation and the cell cycle in vitro when exposed to metformin alone, pemetrexed alone or their combination. We found that combination treatment in three cell lines exerted antiproliferative effects through cell cycle arrest in the S phase. An ex vivo chicken chorioallantoic membrane (CAM) assay was used to examine the antiangiogenic effect of metformin combined with pemetrexed on vascular structure formation. We further created an A549 orthotopic xenograft model with an in vivo imaging system (IVIS) and explored the associated indicators involved in the tumorigenic process. The in vitro results showed that the combination of metformin and pemetrexed exhibited an antiproliferative effect in reducing cell viability and colony formation, the downregulation of cyclin D1 and A2 and the upregulation of CDKN1B, which are involved in the G1/S phase. For antiangiogenic effects, the combination therapy inhibited the vascular structure, as proven by the CAM assay. We elucidated that combination therapy could target VEGFA and Endoglin by RT-qPCR, ELISA and histopathological findings in an A549 orthotopic NSCLC xenograft model. Our research demonstrated the additive antiproliferative and antiangiogenic effects of the combination of metformin with pemetrexed in NSCLC and could be applied to clinical lung cancer therapy.
Collapse
Affiliation(s)
- Jiun-Long Wang
- Ph.D. Program in Translational Medicine, Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan.,Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Ying-Wei Lan
- Ph.D. Program in Translational Medicine, Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Yi-Ting Tsai
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Ying-Cheng Chen
- Ph.D. Program in Translational Medicine, Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Theresa Staniczek
- Department of Dermatology, Venereology and Allergology, Center of Excellence in Dermatology, University Medical Center and Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Yung-An Tsou
- Department of Otolaryngology-Head and Neck Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Chih-Ching Yen
- Department of Internal Medicine, China Medical University Hospital, College of Health Care, China Medical University, Taichung, Taiwan
| | - Chuan-Mu Chen
- Ph.D. Program in Translational Medicine, Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan.,The iEGG and Animal Biotechnology Center, Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
20
|
The Anticancer Effects of Flavonoids through miRNAs Modulations in Triple-Negative Breast Cancer. Nutrients 2021; 13:nu13041212. [PMID: 33916931 PMCID: PMC8067583 DOI: 10.3390/nu13041212] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 03/31/2021] [Accepted: 04/02/2021] [Indexed: 12/31/2022] Open
Abstract
Triple- negative breast cancer (TNBC) incidence rate has regularly risen over the last decades and is expected to increase in the future. Finding novel treatment options with minimum or no toxicity is of great importance in treating or preventing TNBC. Flavonoids are new attractive molecules that might fulfill this promising therapeutic option. Flavonoids have shown many biological activities, including antioxidant, anti-inflammatory, and anticancer effects. In addition to their anticancer effects by arresting the cell cycle, inducing apoptosis, and suppressing cancer cell proliferation, flavonoids can modulate non-coding microRNAs (miRNAs) function. Several preclinical and epidemiological studies indicate the possible therapeutic potential of these compounds. Flavonoids display a unique ability to change miRNAs' levels via different mechanisms, either by suppressing oncogenic miRNAs or activating oncosuppressor miRNAs or affecting transcriptional, epigenetic miRNA processing in TNBC. Flavonoids are not only involved in the regulation of miRNA-mediated cancer initiation, growth, proliferation, differentiation, invasion, metastasis, and epithelial-to-mesenchymal transition (EMT), but also control miRNAs-mediated biological processes that significantly impact TNBC, such as cell cycle, immune system, mitochondrial dysregulation, modulating signaling pathways, inflammation, and angiogenesis. In this review, we highlighted the role of miRNAs in TNBC cancer progression and the effect of flavonoids on miRNA regulation, emphasizing their anticipated role in the prevention and treatment of TNBC.
Collapse
|
21
|
Bang HJ, Ahn MR. Antiangiogenic effect of pinobanksin on human umbilical vein endothelial cells. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
22
|
Multi-Therapeutic Potential of Naringenin (4',5,7-Trihydroxyflavonone): Experimental Evidence and Mechanisms. PLANTS 2020; 9:plants9121784. [PMID: 33339267 PMCID: PMC7766900 DOI: 10.3390/plants9121784] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/26/2020] [Accepted: 11/26/2020] [Indexed: 12/12/2022]
Abstract
Extensive research has been carried out during the last few decades, providing a detailed account of thousands of discovered phytochemicals and their biological activities that have the potential to be exploited for a wide variety of medicinal purposes. These phytochemicals, which are pharmacologically important for clinical use, primarily consist of polyphenols, followed by terpenoids and alkaloids. There are numerous published reports indicating the primary role of phytochemicals proven to possess therapeutic potential against several diseases. However, not all phytochemicals possess significant medicinal properties, and only some of them exhibit viable biological effects. Naringenin, a flavanone found in citrus fruits, is known to improve immunity, repair DNA damage, and scavenge free radicals. Despite the very low bioavailability of naringenin, it is known to exhibit various promising biological properties of medicinal importance, including anti-inflammatory and antioxidant activities. This review focuses on the various aspects related to naringenin, particularly its physicochemical, pharmacokinetic, and pharmacodynamic properties. Furthermore, various pharmacological activities of naringenin, such as anticancer, antidiabetic, hepatoprotective, neuroprotective, cardioprotective, nephroprotective, and gastroprotective effects, have been discussed along with their mechanisms of action.
Collapse
|
23
|
Hu Z, Zhao G, Gou W, Cheng H. Myricitrin inhibits vascular endothelial growth factor-induced angiogenesis of human umbilical vein endothelial cells and mice. Biomed Pharmacother 2020; 130:110726. [PMID: 34321178 DOI: 10.1016/j.biopha.2020.110726] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 08/31/2020] [Accepted: 09/02/2020] [Indexed: 11/25/2022] Open
Abstract
In the present study, the protective effects of myricitrin against vascular endothelial growth factor (VEGF)-induced angiogenesis of vascular endothelial cells were characterized. Cells were induced with 50 ng/mL VEGF in the presence or absence of various concentrations of myricitrin for 24 h. Myricitrin treatment significantly reduced cell proliferation by more than 50 %. Cells treated with myricitrin showed significantly increased caspase 3/7 activity and apoptosis in a dose-dependent manner. Treatment with 1, 10, or 100 μM myricitrin significantly reduced matrix metalloproteinase (MMP) activity by 23.3 %, 46.2 %, or 64.3 %, respectively. Myricitrin significantly reduced MMP1 and MMP2 mRNA expression. Similarly, treatment with 1, 10, or 100 μM myricitrin reduced MMP1 protein expression by 10.5 %, 31.6 %, or 52.6 %, respectively, and MMP2 protein expression by 10.9 %, 28.2 %, or 43.5 %, respectively. Cells treated with myricitrin showed significant inhibition of cell migration as well as capillary tube and sprouting formation. Myricitrin treatment significantly reduced the VEGF level. Immune-deficient nude mice bearing U251 xenograft tumors were used to investigate the antiangiogenic effects of myricitrin in vivo. The results demonstrated that myricitrin treatment in vivo significantly inhibited U251 cell xenograft tumor growth, as confirmed by the decreases in tumor volume and tumor weight. VEGF expression is a key proangiogenic factor. Myricitrin treatment significantly reduced mRNA and protein VEGF expression. Taken together, these results indicate that myricitrin is a potential inhibitor of VEGF-induced angiogenesis.
Collapse
Affiliation(s)
- Zhipeng Hu
- Department of Vascular Surgery, The General Hospital of NingXia Medical University, Yinchuan, 750004, China
| | - Gang Zhao
- Department of Vascular Surgery, The General Hospital of NingXia Medical University, Yinchuan, 750004, China
| | - Wei Gou
- Department of Vascular Surgery, The General Hospital of NingXia Medical University, Yinchuan, 750004, China
| | - Hua Cheng
- Department of Medical Cardiology, The General Hospital of NingXia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
24
|
Robustaflavone induces G0/G1 cell cycle arrest and apoptosis in human umbilical vein endothelial cells and exhibits anti-angiogenic effects in vivo. Sci Rep 2020; 10:11070. [PMID: 32632123 PMCID: PMC7338547 DOI: 10.1038/s41598-020-67993-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 06/15/2020] [Indexed: 11/25/2022] Open
Abstract
We investigated the anti-angiogenic and pro-apoptotic effects of robustaflavone (RF), a naturally occurring biflavonoid, on human umbilical vein endothelial cells (HUVECs). RF inhibited HUVEC proliferation and showed cytotoxicity that inhibited HUVEC viability. RF-induced apoptosis was characterized by flow cytometry and caspase 3 analysis. We found that RF increased the number of sub-G1 cells and terminal deoxynucleotidyl transferase dUTP nick end-labeled cells. Additionally, RF induced caspase 3 and poly (ADP-ribose) polymerase activation. Potential molecular targets were identified using a human apoptosis antibody array. RF upregulated Bax, Bad, cleaved caspase 3, p21, and phosphorylated p53 levels. RF induced mitochondrial membrane potential loss and the release of cytochrome c and apoptosis-inducing factor. Cell cycle arrest at G0/G1 phase and the downregulation of Cdk4, Cdk6, and cyclin D1 expression were induced by RF. In vivo anti-angiogenic effects were investigated using a tumor allograft animal model and a Matrigel plug assay. RF reduced the volumes and weights of CT-26 cell-derived tumors. The blood vessel density was significantly decreased in RF-treated tumors. RF also inhibited VEGF-A-stimulated blood vessel formation in vivo in Matrigel plugs. These results suggest that RF can potentially inhibit angiogenesis-dependent tumor growth and metastasis.
Collapse
|
25
|
Wang L, Chen N, Cheng H. Fisetin inhibits vascular endothelial growth factor-induced angiogenesis in retinoblastoma cells. Oncol Lett 2020; 20:1239-1244. [PMID: 32724364 PMCID: PMC7377090 DOI: 10.3892/ol.2020.11679] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 05/01/2019] [Indexed: 12/24/2022] Open
Abstract
Fisetin is a small phytochemical molecule with antitumor activity. Angiogenesis is a basic process that occurs during tumor growth and metastasis. The vascular endothelial growth factor (VEGF)/VEGF receptor (VEGFR) pathway is a key regulator of angiogenesis. The aim of the present study was to evaluate whether fisetin affects angiogenesis through the VEGFR pathway. In the present study, Y79 cells were treated with 100 ng/ml VEGF in the presence of fisetin at concentrations of 0, 25, 50 and 100 µM. A Cell Counting Kit-8 assay was used to detect proliferation and the Transwell and Matrigel assays were used to assess cell migration and invasion, respectively. Reverse transcription-quantitative polymerase chain reaction analysis was applied to measure the expression level of VEGFR mRNA and western blot analysis was used to measure the protein expression of VEGFR. An immunofluorescence assay was used to detect the expression of VEGFR. Angiogenesis in vitro was assessed by a tube formation assay. The results demonstrated that fisetin significantly inhibited the proliferation of Y79 cells in a time- and dose-dependent manner. Fisetin also inhibited the migration and invasion of Y79 cells in a dose-dependent manner. Furthermore, fisetin inhibited the expression of VEGFR in Y79 cells in a dose-dependent manner and tumor angiogenesis in vitro. Thus, fisetin was found to inhibit angiogenesis via inhibition of the VEGF/VEGFR signaling pathway, and could be used as a candidate drug to inhibit angiogenesis in retinoblastoma.
Collapse
Affiliation(s)
- Liangjun Wang
- Department of Ophthalmology, Yantai Mountain Hospital, Yantai, Shandong 264001, P.R. China
| | - Ning Chen
- Department of Ophthalmology, Yuhuangding Hospital, Yantai, Shandong 264000, P.R. China
| | - Hongxia Cheng
- Department of Ophthalmology, Yuhuangding Hospital, Yantai, Shandong 264000, P.R. China
| |
Collapse
|
26
|
Nutraceutical Boom in Cancer: Inside the Labyrinth of Reactive Oxygen Species. Int J Mol Sci 2020; 21:ijms21061936. [PMID: 32178382 PMCID: PMC7139678 DOI: 10.3390/ijms21061936] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/05/2020] [Accepted: 03/10/2020] [Indexed: 01/02/2023] Open
Abstract
In recent years, epidemiological studies have shown that food is a very powerful means for maintaining a state of well-being and for health prevention. Many degenerative, autoimmune and neoplastic diseases are related to nutrition and the nutrient-organism interaction could define the balance between health and disease. Nutrients and dietary components influence epigenetic phenomena and modify drugs response; therefore, these food-host interactions can influence the individual predisposition to disease and its potential therapeutic response. Do nutraceuticals have positive or negative effects during chemotherapy? The use of nutraceutical supplements in cancer patients is a controversial debate without a definitive conclusion to date. During cancer treatment, patients take nutraceuticals to alleviate drug toxicity and improve long-term results. Some nutraceuticals may potentiate the effect of cytotoxic chemotherapy by inducing cell growth arrest, cell differentiation, and alteration of the redox state of cells, but in some cases, high levels of them may interfere with the effectiveness of chemotherapy, making cancer cells less reactive to chemotherapy. In this review, we highlighted the emerging opinions and data on the pros and cons on the use of nutraceutical supplements during chemotherapy.
Collapse
|
27
|
Duan L, Yang L, Jin J, Yang F, Liu D, Hu K, Wang Q, Yue Y, Gu N. Micro/nano-bubble-assisted ultrasound to enhance the EPR effect and potential theranostic applications. Theranostics 2020; 10:462-483. [PMID: 31903132 PMCID: PMC6929974 DOI: 10.7150/thno.37593] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 10/11/2019] [Indexed: 12/23/2022] Open
Abstract
Drug delivery for tumor theranostics involves the extensive use of the enhanced permeability and retention (EPR) effect. Previously, various types of nanomedicines have been demonstrated to accumulate in solid tumors via the EPR effect. However, EPR is a highly variable phenomenon because of tumor heterogeneity, resulting in low drug delivery efficacy in clinical trials. Because ultrasonication using micro/nanobubbles as contrast agents can disrupt blood vessels and enhance the specific delivery of drugs, it is an effective approach to improve the EPR effect for the passive targeting of tumors. In this review, the basic thermal effect, acoustic streaming, and cavitation mechanisms of ultrasound, which are characteristics that can be utilized to enhance the EPR effect, are briefly introduced. Second, micro/nanobubble-enhanced ultrasound imaging is discussed to understand the validity and variability of the EPR effect. Third, because the tumor microenvironment is complicated owing to elevated interstitial fluid pressure and the deregulated extracellular matrix components, which may be unfavorable for the EPR effect, few new trends in smart bubble drug delivery systems, which may improve the accuracy of EPR-mediated passive drug targeting, are summarized. Finally, the challenging and major concerns that should be considered in the next generation of micro/nanobubble-contrast-enhanced ultrasound theranostics for EPR-mediated passive drug targeting are also discussed.
Collapse
Affiliation(s)
- Lei Duan
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Li Yang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China
| | - Juan Jin
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China
| | - Fang Yang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China
| | - Dong Liu
- West Anhui University, Lu'an, P.R. China
- Anhui Engineering Laboratory for Conservation and Sustainable Utilization of Traditional Chinese Medicine Resources, P. R. China
| | - Ke Hu
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Qinxin Wang
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Yuanbin Yue
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Ning Gu
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, 211166, P. R. China
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China
| |
Collapse
|
28
|
Okamura N, Ohta T, Kunimasa K, Uto Y, Kumazawa S. Antiangiogenic Activity of Flavonols in Chorioallantoic Membrane (CAM) Assay. FOOD SCIENCE AND TECHNOLOGY RESEARCH 2020. [DOI: 10.3136/fstr.26.891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Naoki Okamura
- Department of Food and Nutritional Sciences, University of Shizuoka
| | - Toshiro Ohta
- Department of Food and Nutritional Sciences, University of Shizuoka
| | | | - Yoshihiro Uto
- Graduate School of Technology, Industrial and Social Science, Tokushima University
| | | |
Collapse
|
29
|
The Mediterranean Diet, a Rich Source of Angiopreventive Compounds in Cancer. Nutrients 2019; 11:nu11092036. [PMID: 31480406 PMCID: PMC6769787 DOI: 10.3390/nu11092036] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 08/19/2019] [Accepted: 08/25/2019] [Indexed: 12/12/2022] Open
Abstract
Diet-based chemoprevention of cancer has emerged as an interesting approach to evade the disease or even target its early phases, reducing its incidence or slowing down tumor progression. In its basis in the essential role of angiogenesis for tumor growth and metastasis, angioprevention proposes the use of inhibitors of angiogenesis in cancer prevention. The anti-angiogenic potential exhibited by many natural compounds contained in many Mediterranean diet constituents makes this dietary pattern especially interesting as a source of chemopreventive agents, defined within the angioprevention strategy. In this review, we focus on natural bioactive compounds derived from the main foods included in the Mediterranean diet that display anti-angiogenic activity, as well as their possible use as angiopreventive agents.
Collapse
|
30
|
Ardisia crispa root hexane fraction suppressed angiogenesis in human umbilical vein endothelial cells (HUVECs) and in vivo zebrafish embryo model. Biomed Pharmacother 2019; 118:109221. [PMID: 31545225 DOI: 10.1016/j.biopha.2019.109221] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 07/08/2019] [Accepted: 07/10/2019] [Indexed: 01/08/2023] Open
Abstract
Ardisia crispa Thunb. A. DC. (Primulaceae) has been used extensively as folk-lore medicine in South East Asia including China and Japan to treat various inflammatory related diseases. Ardisia crispa root hexane fraction (ACRH) has been thoroughly studied by our group and it has been shown to exhibit anti-inflammatory, anti-hyperalgesic, anti-arthritic, anti-ulcer, chemoprevention and suppression against inflammation-induced angiogenesis in various animal model. Nevertheless, its effect against human endothelial cells in vitro has not been reported yet. Hence, the aim of the study is to investigate the potential antiangiogenic property of ACRH in human umbilical vein endothelial cells (HUVECs) and zebrafish embryo model. ACRH was separated from the crude ethanolic extract of the plant's root in prior to experimental studies. MTT assay revealed that ACRH exerted a concentration-dependent antiproliferative effect on HUVEC with the IC50 of 2.49 ± 0.04 μg/mL. At higher concentration (10 μg/mL), apoptosis was induced without affecting the cell cycle distribution. Angiogenic properties including migration, invasion and differentiation of HUVECs, evaluated via wound healing, trans-well invasion and tube formation assay respectively, were significantly suppressed by ACRH in a concentration-dependent manner. Noteworthily, significant antiangiogenic effects were observed even at the lowest concentration used (0.1 μg/mL). Expression of proMMP-2, vascular endothelial growth factor (VEGF)-C, VEGF-D, Angiopoietin-2, fibroblast growth factor (FGF)-1, FGF-2, Follistatin, and hepatocyte growth factor (HGF) were significantly reduced in various degrees by ACRH. The ISV formation in zebrafish embryo was significantly suppressed by ACRH at the concentration of 5 μg/mL. These findings revealed the potential of ACRH as antiangiogenic agent by suppressing multiple proangiogenic proteins. Thus, it can be further verified via the transcription of these proteins from their respective DNA, in elucidating their exact pathways.
Collapse
|
31
|
Koss-Mikołajczyk I, Baranowska M, Todorovic V, Albini A, Sansone C, Andreoletti P, Cherkaoui-Malki M, Lizard G, Noonan D, Sobajic S, Bartoszek A. Prophylaxis of Non-communicable Diseases: Why Fruits and Vegetables may be Better Chemopreventive Agents than Dietary Supplements Based on Isolated Phytochemicals? Curr Pharm Des 2019; 25:1847-1860. [DOI: 10.2174/1381612825666190702093301] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 06/20/2019] [Indexed: 12/23/2022]
Abstract
The World Health Organization (WHO) report from 2014 documented that non-communicable socalled civilization diseases such as cardiovascular disease, chronic respiratory diseases, cancer or type 2 diabetes are responsible for over 50% of all premature deaths in the world. Research carried out over the past 20 years has provided data suggesting that diet is an essential factor influencing the risk of development of these diseases. The increasing knowledge on chemopreventive properties of certain food ingredients, in particular, those of plant origin, opened the discussion on the possibility to use edible plants or their active components in the prevention of these chronic diseases. Health-promoting properties of plant foods are associated with the presence of secondary metabolites that can affect many biological mechanisms of critical importance to the proper functioning of the human organism. Particularly, there have been numerous investigations indicating strong physiological effects of bioactive plant phenols belonging to the flavonoid family. These observations initiated mass production of dietary supplements containing flavonoids commercialized under the name antioxidants, even if their chemical properties did not justify such a term. However, epidemiological studies revealed that isolated bioactive phytochemicals are not as effective as fruits and vegetables containing these substances whereas they are of interest of the functional food industry. In this paper, the critical assessment of reasons for this turn of events has been attempted and the concept of food synergy has been suggested as a future strategy of dietary chemoprevention.
Collapse
Affiliation(s)
- Izabela Koss-Mikołajczyk
- Department of Food Chemistry, Technology and Biotechnology, Faculty of Chemistry, Gdańsk University of Technology, Gdansk, Poland
| | - Monika Baranowska
- Department of Food Chemistry, Technology and Biotechnology, Faculty of Chemistry, Gdańsk University of Technology, Gdansk, Poland
| | - Vanja Todorovic
- Department of Bromatology, Faculty of Pharmacy, University of Belgrade, Studentski trg 1, Beograd, GabrielaNarutowicza 11/12, 80-233, Gdanski, Serbia
| | - Adriana Albini
- Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | | | | | | | - Gérard Lizard
- BioPeroxIL Laboratory, Universite de Bourgogne-Franche Comte, France
| | | | - Sladjana Sobajic
- Department of Bromatology, Faculty of Pharmacy, University of Belgrade, Studentski trg 1, Beograd, GabrielaNarutowicza 11/12, 80-233, Gdanski, Serbia
| | - Agnieszka Bartoszek
- Department of Food Chemistry, Technology and Biotechnology, Faculty of Chemistry, Gdańsk University of Technology, Gdansk, Poland
| |
Collapse
|
32
|
Ince B, Yildirim MEC, Dadaci M. Assessing the Effect of Vitamin D Replacement on Basal Cell Carcinoma Occurrence and Recurrence Rates in Patients with Vitamin D Deficiency. Discov Oncol 2019; 10:145-149. [PMID: 31254251 DOI: 10.1007/s12672-019-00365-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 06/13/2019] [Indexed: 12/18/2022] Open
Abstract
The study aimed to determine whether 25-OH vitamin D3 deficiency is present in patients with diagnosed BCC, and the effect of vitamin D replacement on the rates of BCC recurrence in patients with 25-OH vitamin D3 deficiency. In this prospective study, between 2012 and 2017, in the first stage, 25-OH vitamin D3 levels of all patients diagnosed with BCC between 2012 and 2013 were evaluated. In the second stage between 2014 and 2015, we evaluated the 25-OH vitamin D3 level of patients who had 25-OH vitamin D3 level < 25 ng/mL. All the patients included in the second stage had BCC recurrence. In the third stage, the patients who were diagnosed 25-OH vitamin D3 deficiency with BCC, between 2015 and 2017, were studied. The mean 25-OH vitamin D3 level of the patients in the second stage was 10.12 ng/mL. Recurrence was observed in 9.64% of the patients in the second stage. The mean level of serum 25-OH vitamin D3 in the third stage was 40.1 ng/mL, and 3.49% of these patients presented with recurrence. In all the patients as the initial diagnosis and following the 25-OH vitamin D3 level in all the patients with BCC recurrence, maintaining 25-OH vitamin D3 levels above 25 ng/mL can significantly reduce the recurrence rate.
Collapse
Affiliation(s)
- Bilsev Ince
- Department of Plastic & Reconstructive and Aesthetic Surgery, Meram Faculty of Medicine, Necmettin Erbakan University, 42080, Meram, Konya, Turkey
| | - Mehmet Emin Cem Yildirim
- Department of Plastic & Reconstructive and Aesthetic Surgery, Meram Faculty of Medicine, Necmettin Erbakan University, 42080, Meram, Konya, Turkey.
| | - Mehmet Dadaci
- Department of Plastic & Reconstructive and Aesthetic Surgery, Meram Faculty of Medicine, Necmettin Erbakan University, 42080, Meram, Konya, Turkey
| |
Collapse
|
33
|
Andrews LE, Chan MH, Liu RS. Nano-lipospheres as acoustically active ultrasound contrast agents: evolving tumor imaging and therapy technique. NANOTECHNOLOGY 2019; 30:182001. [PMID: 30645984 DOI: 10.1088/1361-6528/aafeb9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Applying nanobubbles (NBs) for contrast-enhanced ultrasound imaging has received increased attention. NBs are biocompatible, multifunctional, theranostic agents. Their properties of high echogenicity and stability create an agent suitable for ultrasonography diagnosis. Their favorable properties of size, in vivo stability, and ease of modification are being exploited to implement a theranostic platform for cancer treatment. The considerable development offers the potential to overcome drug resistance and adverse side effects that are associated with traditional chemotherapy. This review outlines the principles of ultrasonography and angiogenesis. Microbubbles and micelles are also discussed to underline the superior capabilities of NBs for the application. NBs could passively accumulate to tumor tissue by enhanced permeability and retention effect. In addition, it can also achieve the active transportation by surface modification. Active targeting modalities and stimuli-responsive drug delivery modifications generate a therapeutic vehicle. The cytotoxicity of NBs formulations, multimodal imaging capability, active targeting mechanisms, and drug delivery methods are highlighted to confirm the NB as a vehicle for targeted treatment and enhanced ultrasound imaging.
Collapse
Affiliation(s)
- Laura Emma Andrews
- Department of Chemistry, National Taiwan University, Taiwan. School of Chemistry, The University of Edinburgh, United Kingdom
| | | | | |
Collapse
|
34
|
Chandrasekharan JA, Sharma-Walia N. Arachidonic Acid Derived Lipid Mediators Influence Kaposi's Sarcoma-Associated Herpesvirus Infection and Pathogenesis. Front Microbiol 2019; 10:358. [PMID: 30915039 PMCID: PMC6422901 DOI: 10.3389/fmicb.2019.00358] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 02/11/2019] [Indexed: 12/30/2022] Open
Abstract
Kaposi’s sarcoma-associated herpesvirus (KSHV) infection, particularly latent infection is often associated with inflammation. The arachidonic acid pathway, the home of several inflammation and resolution associated lipid mediators, is widely altered upon viral infections. Several in vitro studies show that these lipid mediators help in the progression of viral pathogenesis. This review summarizes the findings related to human herpesvirus KSHV infection and arachidonic acid pathway metabolites. KSHV infection has been shown to promote inflammation by upregulating cyclooxygenase-2 (COX-2), 5 lipoxygenase (5LO), and their respective metabolites prostaglandin E2 (PGE2) and leukotriene B4 (LTB4) to promote latency and an inflammatory microenvironment. Interestingly, the anti-inflammatory lipid mediator lipoxin is downregulated during KSHV infection to facilitate infected cell survival. These studies aid in understanding the role of arachidonic acid pathway metabolites in the progression of viral infection, the host inflammatory response, and pathogenesis. With limited therapeutic options to treat KSHV infection, use of inhibitors to these inflammatory metabolites and their synthetic pathways or supplementing anti-inflammatory lipid mediators could be an effective alternative therapeutic.
Collapse
Affiliation(s)
- Jayashree A Chandrasekharan
- Department of Microbiology and Immunology, H.M. Bligh Cancer Research Laboratories, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Neelam Sharma-Walia
- Department of Microbiology and Immunology, H.M. Bligh Cancer Research Laboratories, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| |
Collapse
|
35
|
Afsharzadeh M, Abnous K, Yazdian–Robati R, Ataranzadeh A, Ramezani M, Hashemi M. Formulation and evaluation of anticancer and antiangiogenesis efficiency of PLA–PEG nanoparticles loaded with galbanic acid in C26 colon carcinoma, in vitro and in vivo. J Cell Physiol 2018; 234:6099-6107. [DOI: 10.1002/jcp.27346] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 08/13/2018] [Indexed: 01/15/2023]
Affiliation(s)
- Maryam Afsharzadeh
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences Mashhad Iran
| | - Khalil Abnous
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences Mashhad Iran
| | - Rezvan Yazdian–Robati
- Department of Pharmaceutical Biotechnology School of Pharmacy, Mashhad University of Medical Sciences Mashhad Iran
| | - Armin Ataranzadeh
- Department of Molecular Pathologist & Cytogenetic Mashhad University of Medical Sciences, Imam Reza Hospital Mashhad Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences Mashhad Iran
- Department of Pharmaceutical Biotechnology School of Pharmacy, Mashhad University of Medical Sciences Mashhad Iran
| | - Maryam Hashemi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|
36
|
Chellappan DK, Leng KH, Jia LJ, Aziz NABA, Hoong WC, Qian YC, Ling FY, Wei GS, Ying T, Chellian J, Gupta G, Dua K. The role of bevacizumab on tumour angiogenesis and in the management of gynaecological cancers: A review. Biomed Pharmacother 2018; 102:1127-1144. [DOI: 10.1016/j.biopha.2018.03.061] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 03/09/2018] [Accepted: 03/11/2018] [Indexed: 02/06/2023] Open
|
37
|
Tosetti F, Benelli R, Albini A. The Angiogenic Switch in Solid Tumors: Clinical Implications. TUMORI JOURNAL 2018; 1:S9-11. [PMID: 12658894 DOI: 10.1177/03008916020016s103] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Francesca Tosetti
- Laboratory of Molecular Biology, National Cancer Research Institute, Genoa, Italy
| | | | | |
Collapse
|
38
|
Zomorrod MS, Kouhkan F, Soleimani M, Aliyan A, Tasharrofi N. Overexpression of miR-133 decrease primary endothelial cells proliferation and migration via FGFR1 targeting. Exp Cell Res 2018; 369:11-16. [PMID: 29608916 DOI: 10.1016/j.yexcr.2018.02.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 02/02/2018] [Accepted: 02/17/2018] [Indexed: 02/07/2023]
Abstract
Angiogenesis is one of the essential hallmarks of cancer that is controlled by the balance between positive and negative regulators. FGFR1 signaling is crucial for the execution of bFGF-induced proliferation, migration, and tube formation of endothelial cells (ECs) and onset of angiogenesis on tumors. The purpose of this study is to identify whether or not miR-133 regulates FGFR1 expression and accordingly hypothesize if it plays a crucial role in modulating bFGF/FGFR1 activity in ECs and blocking tumor angiogenesis through targeting FGFR1. The influences of miR-133 overexpression on bFGF stimulated endothelial cells were assessed by cell growth curve, MTT assaying, tube formation, and migration assays. Forced expression of miR-133 caused significant reductions in bFGF-induced proliferation and migratory ability of ECs. MiR-133 Expression was negatively correlated with both mRNA and protein levels of FGFR1 in the transfected ECs isolated from peripheral blood. Moreover, overexpression of miR-133 drastically reduced the rate of cell division and disturbed capillary network formation of transfected ECs. These findings suggest that miR-133 plays an important function in bFGF-induced angiogenesis processes in ECs and provides a rationale for new therapeutic approaches to suppress tumor angiogenesis and cancer.
Collapse
Affiliation(s)
- Mina Soufi Zomorrod
- Department of Hematology and Cell Therapy, Faculty of Medical Science, Tarbiat Modares University, Tehran, lran.
| | | | - Masoud Soleimani
- Department of Hematology and Cell Therapy, Faculty of Medical Science, Tarbiat Modares University, Tehran, lran.
| | - Amir Aliyan
- Department of Hematology and Cell Therapy, Faculty of Medical Science, Tarbiat Modares University, Tehran, lran; Department of Chemistry, Rice University, Houston, TX 77054, USA.
| | - Nooshin Tasharrofi
- Stem Cell Technology Research Center, Tehran, Iran; Faculty of pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran.
| |
Collapse
|
39
|
Gallo C, Dallaglio K, Bassani B, Rossi T, Rossello A, Noonan DM, D'Uva G, Bruno A, Albini A. Hop derived flavonoid xanthohumol inhibits endothelial cell functions via AMPK activation. Oncotarget 2018; 7:59917-59931. [PMID: 27494895 PMCID: PMC5312358 DOI: 10.18632/oncotarget.10990] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 06/06/2016] [Indexed: 12/25/2022] Open
Abstract
Angiogenesis, a process characterized by the formation of new blood vessels from pre-existing ones, is a crucial step in tumor growth and dissemination. Recently, increased attention has been addressed to the ability of flavonoids to prevent cancer by suppressing angiogenesis, strategy that we named "angioprevention". Several natural compounds exert their anti-tumor properties by activating 5' adenosine monophosphate-activated protein kinase (AMPK), a key regulator of metabolism in cancer cells. Drugs with angiopreventive activities, in particular metformin, regulate AMPK in endothelial cells. Here we investigated the involvement of AMPK in the anti-angiogenic effects of xanthohumol (XN), the major prenylated flavonoid of the hop plant, and mechanisms of action. The anti-angiogenic activity of XN was more potent than epigallocatechin-3-gallate (EGCG). Treatment of endothelial cells with XN led to increased AMPK phosphorylation and activity. Functional studies using biochemical approaches confirmed that AMPK mediates XN anti-angiogenic activity. AMPK activation by XN was mediated by CAMMKβ, but not LKB1. Analysis of the downstream mechanisms showed that XN-induced AMPK activation reduced nitric oxide (NO) levels in endothelial cells by decreasing eNOS phosphorylation. Finally, AKT pathway was inactivated by XN as part of its anti-angiogenic activity, but independently from AMPK, suggesting that these two signaling pathways proceed autonomously. Our study dissects the molecular mechanism by which XN exerts its potent anti-angiogenic activity, pointing out AMPK as a crucial signal transducer.
Collapse
Affiliation(s)
- Cristina Gallo
- IRCCS "Istituto in Tecnologie Avanzate e Modelli Assistenziali in Oncologia" Arcispedale S. Maria Nuova, Reggio Emilia, Italy
| | - Katiuscia Dallaglio
- IRCCS "Istituto in Tecnologie Avanzate e Modelli Assistenziali in Oncologia" Arcispedale S. Maria Nuova, Reggio Emilia, Italy
| | - Barbara Bassani
- Scientific and Technology Pole, IRCCS MultiMedica, Milan, Italy
| | - Teresa Rossi
- IRCCS "Istituto in Tecnologie Avanzate e Modelli Assistenziali in Oncologia" Arcispedale S. Maria Nuova, Reggio Emilia, Italy
| | | | - Douglas M Noonan
- Department of Biotechnologies and Life Sciencies, University of Insubria, Varese, Italy
| | - Gabriele D'Uva
- Scientific and Technology Pole, IRCCS MultiMedica, Milan, Italy
| | - Antonino Bruno
- Scientific and Technology Pole, IRCCS MultiMedica, Milan, Italy
| | - Adriana Albini
- Scientific and Technology Pole, IRCCS MultiMedica, Milan, Italy
| |
Collapse
|
40
|
Pfeffer U, Ferrari N, Morini M, Benelli R, Noonan DM, Albini A. Antiangiogenic Activity of Chemopreventive Drugs. Int J Biol Markers 2018; 18:70-4. [PMID: 12699068 DOI: 10.1177/172460080301800113] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Tumors growing within the host form dynamic aberrant tissue that consists of host components, including the stroma, an expanding vasculature and often chronic inflammation, in addition to the tumor cells themselves. These host components can contribute to, rather than limit, tumor expansion, whereas deprivation of vessel formation has the potential to confine tumors in small, clinically silent foci. Therapeutic inhibition of vessel formation could be best suited to preventive strategies aimed at the suppression of angiogenesis in primary tumors in subjects at risk, or of micrometastases after surgical removal of a primary tumor. Our analysis of potential cancer chemopreventive molecules including N-acetylcysteine, green tea flavonoids and 4-hydroxyphenyl-retinamide has identified antiangiogenic activities that could account -at least in part - for the tumor prevention effects observed with these compounds. These drugs appear to target common mechanisms of tumor angiogenesis that may permit identification of critical targets for antiangiogenic therapy and antiangiogenic chemoprevention.
Collapse
Affiliation(s)
- U Pfeffer
- Laboratory of Molecular Oncology, National Cancer Research Institute, Genoa, Italy.
| | | | | | | | | | | |
Collapse
|
41
|
Patil VM, Masand N. Anticancer Potential of Flavonoids: Chemistry, Biological Activities, and Future Perspectives. STUDIES IN NATURAL PRODUCTS CHEMISTRY 2018. [DOI: 10.1016/b978-0-444-64179-3.00012-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
42
|
Kim GD. Kaempferol Inhibits Angiogenesis by Suppressing HIF-1α and VEGFR2 Activation via ERK/p38 MAPK and PI3K/Akt/mTOR Signaling Pathways in Endothelial Cells. Prev Nutr Food Sci 2017; 22:320-326. [PMID: 29333385 PMCID: PMC5758096 DOI: 10.3746/pnf.2017.22.4.320] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 11/27/2017] [Indexed: 11/06/2022] Open
Abstract
Kaempferol has been shown to inhibit vascular formation in endothelial cells. However, the underlying mechanisms are not fully understood. In the present study, we evaluated whether kaempferol exerts antiangiogenic effects by targeting extracellular signal-regulated kinase (ERK)/p38 mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K)/Akt/mechanistic target of rapamycin (mTOR) signaling pathways in endothelial cells. Endothelial cells were treated with various concentrations of kaempferol for 24 h. Cell viability was determined by the 3- (4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide assay; vascular formation was analyzed by tube formation, wound healing, and mouse aortic ring assays. Activation of hypoxia-inducible factor-1α (HIF-1α), vascular endothelial growth factor receptor 2 (VEGFR2), ERK/p38 MAPK, and PI3K/Akt/mTOR was analyzed by Western blotting. Kaempferol significantly inhibited cell migration and tube formation in endothelial cells, and suppressed microvessel sprouting in the mouse aortic ring assay. Moreover, kaempferol suppressed the activation of HIF-1α, VEGFR2, and other markers of ERK/p38 MAPK and PI3K/Akt/mTOR signaling pathways in endothelial cells. These results suggest that kaempferol inhibits angiogenesis by suppressing HIF-1α and VEGFR2 activation via ERK/p38 MAPK and PI3K/Akt/mTOR signaling in endothelial cells.
Collapse
Affiliation(s)
- Gi Dae Kim
- Department of Food, Nutrition and Biotechnology, Kyungnam University, Gyeongnam 51767, Korea
| |
Collapse
|
43
|
Cancer chemoprevention revisited: Cytochrome P450 family 1B1 as a target in the tumor and the microenvironment. Cancer Treat Rev 2017; 63:1-18. [PMID: 29197745 DOI: 10.1016/j.ctrv.2017.10.013] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 10/30/2017] [Accepted: 10/31/2017] [Indexed: 02/08/2023]
Abstract
Cancer chemoprevention is the use of synthetic, natural or biological agents to prevent or delay the development or progression of malignancies. Intriguingly, many phytochemicals with anti-inflammatory and anti-angiogenic effects, recently proposed as chemoprevention strategies, are inhibitors of Cytochrome P450 family 1B1 (CYP1B1), an enzyme overexpressed in a wide variety of tumors and associated with angiogenesis. In turn, pro-inflammatory cytokines were reported to boost CYP1B1 expression, suggesting a key role of CYP1B1 in a positive loop of inflammatory angiogenesis. Other well-known pro-tumorigenic activities of CYP1B1 rely on metabolic bioactivation of xenobiotics and steroid hormones into their carcinogenic derivatives. In contrast to initial in vitro observations, in vivo studies demonstrated a protecting role against cancer for the other CYP1 family members (CYP1A1 and CYP1A2), suggesting that the specificity of CYP1 family inhibitors should be carefully taken into account for developing potential chemoprevention strategies. Recent studies also proposed a role of CYP1B1 in multiple cell types found within the tumor microenvironment, including fibroblasts, endothelial and immune cells. Overall, our review of the current literature suggests a positive loop between inflammatory cytokines and CYP1B1, which in turn may play a key role in cancer angiogenesis, acting on both cancer cells and the tumor microenvironment. Strategies aiming at specific CYP1B1 inhibition in multiple cell types may translate into clinical chemoprevention and angioprevention approaches.
Collapse
|
44
|
Feng C, Ho Y, Sun C, Xia G, Ding Q, Gu B. Epigallocatechin gallate inhibits the growth and promotes the apoptosis of bladder cancer cells. Exp Ther Med 2017; 14:3513-3518. [PMID: 29042941 PMCID: PMC5639296 DOI: 10.3892/etm.2017.4981] [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: 09/23/2016] [Accepted: 06/15/2017] [Indexed: 11/20/2022] Open
Abstract
Epigallocatechin gallate (EGCG) has been revealed to inhibit the proliferation and induce the apoptosis of several types of tumor, in addition to inhibiting DNA methyltransferase activity, leading to CpG demethylation. Tissue factor pathway inhibitor 2 (TFPI-2) expression is downregulated in bladder cancer. The present study revealed that this downregulation was partly due to hypermethylation of the TFPI-2 gene promoter, which was decreased by EGCG treatment. In addition, the present study demonstrated that EGCG could inhibit the viability and invasion, and induce the apoptosis, of bladder cancer T24 cells. Furthermore, western blot analysis and reverse transcription-quantitative polymerase chain reaction analyses demonstrated that EGCG could upregulate the expression of TFPI-2. These results suggest that EGCG inhibits the growth and induces the apoptosis of bladder cancer cells through restoring TFPI-2 expression. Thus, EGCG is a potential therapeutic candidate for the treatment of bladder cancer.
Collapse
Affiliation(s)
- Chenchen Feng
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Yatfaat Ho
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Chuanyu Sun
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Guowei Xia
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Qiang Ding
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Bin Gu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| |
Collapse
|
45
|
Fernandes E Silva E, Figueira FDS, Lettnin AP, Carrett-Dias M, Filgueira DDMVB, Kalil S, Trindade GS, Votto APDS. C-Phycocyanin: Cellular targets, mechanisms of action and multi drug resistance in cancer. Pharmacol Rep 2017; 70:75-80. [PMID: 29331790 DOI: 10.1016/j.pharep.2017.07.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 06/30/2017] [Accepted: 07/25/2017] [Indexed: 02/07/2023]
Abstract
C-Phycocyanin (C-PC) has been shown to be promising in cancer treatment; however, although several articles detailing this have been published, its main mechanisms of action and its cellular targets have not yet been defined, nor has a detailed exploration been conducted of its role in the resistance of cancer cells to chemotherapy, rendering clinical use impossible. From our extensive examination of the literature, we have determined as our main hypothesis that C-PC has no one specific target, but rather acts on the membrane, cytoplasm, and nucleus with diverse mechanisms of action. We highlight the cell targets with which C-PC interacts (the MDR1 gene, cytoskeleton proteins, and COX-2 enzyme) that make it capable of killing cells resistant to chemotherapy. We also propose future analyses of the interaction between C-PC and drug extrusion proteins, such as ABCB1 and ABCC1, using in silico and in vitro studies.
Collapse
Affiliation(s)
- Estela Fernandes E Silva
- Programa de Pós-graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande (FURG), Rio Grande, RS, Brazil.
| | | | - Aline Portantiolo Lettnin
- Programa de Pós-graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande (FURG), Rio Grande, RS, Brazil.
| | - Michele Carrett-Dias
- Programa de Pós-graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande (FURG), Rio Grande, RS, Brazil.
| | - Daza de Moraes Vaz Batista Filgueira
- Programa de Pós-graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande (FURG), Rio Grande, RS, Brazil.
| | - Susana Kalil
- Escola de Química e Alimentos, FURG, Rio Grande, RS, Brazil.
| | - Gilma Santos Trindade
- Programa de Pós-graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande (FURG), Rio Grande, RS, Brazil.
| | - Ana Paula de Souza Votto
- Programa de Pós-graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande (FURG), Rio Grande, RS, Brazil.
| |
Collapse
|
46
|
Dettlaff K, Stawny M, Ogrodowczyk M, Jelińska A, Bednarski W, Wątróbska-Świetlikowska D, Keck RW, Khan OA, Mostafa IH, Jankun J. Formulation and characterization of EGCG for the treatment of superficial bladder cancer. Int J Mol Med 2017. [PMID: 28627636 PMCID: PMC5504970 DOI: 10.3892/ijmm.2017.3024] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In the United States, the annual incidence of bladder cancer is approximately 70,000 new cases, with a mortality rate of approximately 15,000/year. The most common subtype (70%) of bladder cancer is superficial, namely hte non-muscle invasive disease form limited to the urothelium. The rate of progression and recurrence is up to 40 and 70%, respectively. Urothelial cell carcinoma of the bladder is typically treated with transurethral resection. The cancerous cells can float onto the adjacent epithelium, increasing the risk of recurrence. The standard of care is to offer adjuvant intravesical agents to reduce the risk of progression and recurrence. Current intravesical treatments are costly and are associated with special biohazard handling protocols. Patients are treated with intravesical therapy with bacillus Calmetter-Guerin (BCG) bacterium, or mitomycin C (MMC) following resection, both of which can cause moderate to severe side-effects which are rarely life-threatening. We previously examined the efficacy of epigallocatechin-3-gallate (EGCG)in comparison with MMC to prevent tumor cell implantation/growth in an animal model of superficial bladder cancer. Experiments revile that EGCG is slightly more effective than MMC at decreasing tumor cell implantation and consequent cancer growth in a bladder. This treatment requires the stringent sterile requirement of EGCG. EGCG can be unstable when sterilized at high temperatures. Thus, we evaluated two low temperature sterilization methods, such as ionizing radiation or the filtration method followed by freeze-drying. Both methods ensure the sterility of the sample; however, infrared and HPLC analysis revealed a slightly better stability of irradiated EGCG over the filtration method. The concentration of stable free radicals following irradiation was low, which are unlikely to exert any damaging effects to EGCG. Therefore, we consider that radiation will be the preferred method of EGCG sterilization, and that this may prove useful for the effective use of EGCG in the treatment of bladder cancer.
Collapse
Affiliation(s)
- Katarzyna Dettlaff
- Department of Pharmaceutical Chemistry, Poznań University of Medical Sciences, 60‑780 Poznań, Poland
| | - Maciej Stawny
- Department of Pharmaceutical Chemistry, Poznań University of Medical Sciences, 60‑780 Poznań, Poland
| | - Magdalena Ogrodowczyk
- Department of Pharmaceutical Chemistry, Poznań University of Medical Sciences, 60‑780 Poznań, Poland
| | - Anna Jelińska
- Department of Pharmaceutical Chemistry, Poznań University of Medical Sciences, 60‑780 Poznań, Poland
| | - Waldemar Bednarski
- Institute of Molecular Physics, Polish Academy of Sciences, 60-179 Poznań, Poland
| | | | - Rick W Keck
- Urology Research Center, Department of Urology, College of Medicine, University of Toledo, Toledo, OH 43614, USA
| | - Omar A Khan
- Urology Research Center, Department of Urology, College of Medicine, University of Toledo, Toledo, OH 43614, USA
| | - Ibrahim H Mostafa
- Urology Research Center, Department of Urology, College of Medicine, University of Toledo, Toledo, OH 43614, USA
| | - Jerzy Jankun
- Urology Research Center, Department of Urology, College of Medicine, University of Toledo, Toledo, OH 43614, USA
| |
Collapse
|
47
|
Lin S, Hu J, Zhou X, Cheung PC. Inhibition of vascular endothelial growth factor-induced angiogenesis by chlorogenic acid via targeting the vascular endothelial growth factor receptor 2-mediated signaling pathway. J Funct Foods 2017; 32:285-295. [DOI: 10.1016/j.jff.2017.03.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
48
|
Kim HJ, Ahn MR. Apigenin Suppresses Angiogenesis by Inhibiting Tube Formation and Inducing Apoptosis. Nat Prod Commun 2016. [DOI: 10.1177/1934578x1601101005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Apigenin has been reported to exert angiogenic and anticancer activities in vitro. The mechanism of inhibition of angiogenesis by apigenin, however, has not been well-established. In this study, we investigated whether apigenin not only inhibited tube formation but also induced apoptosis in human umbilical vein endothelial cells (HUVECs). Furthermore, strong antiangiogenic activity of apigenin was observed in the in vivo assay using chick embryo chorioallantoic membrane (CAM). We also analyzed changes in survival signals and the apoptotic pathway through Western blotting. The results indicate that apigenin exerts its antiangiogenic effects through induction of endothelial apoptosis.
Collapse
Affiliation(s)
- Hyun Ju Kim
- Department of Food Science and Nutrition, Dong-A University, 37 Nakdong-Daero 550 beon-gil, Saha-gu, Busan 49315, Republic of Korea
| | - Mok-Ryeon Ahn
- Department of Food Science and Nutrition, Dong-A University, 37 Nakdong-Daero 550 beon-gil, Saha-gu, Busan 49315, Republic of Korea
- Institute of Convergence Bio-Health, Dong-A University, 32 Daeshingongwon-Ro, Seo-gu, Busan 49210, Republic of Korea
| |
Collapse
|
49
|
Endothelial progenitor cells promote tumor growth and progression by enhancing new vessel formation. Oncol Lett 2016; 12:793-799. [PMID: 27446353 PMCID: PMC4950911 DOI: 10.3892/ol.2016.4733] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 06/02/2016] [Indexed: 12/22/2022] Open
Abstract
Tumor growth and progression require new blood vessel formation to deliver nutrients and oxygen for further cell proliferation and to create a neovascular network exit for tumor cell metastasis. Endothelial progenitor cells (EPCs) are a bone marrow (BM)-derived stem cell population that circulates in the peripheral circulation and homes to the tumor bed to participate in new blood vessel formation. In addition to structural support to nascent vessels, these cells can also regulate the angiogenic process by paracrine secretion of a number of proangiogenic growth factors and cytokines, thus playing a crucial role in tumor neovascularization and development. Inhibition of EPC-mediated new vessel formation may be a promising therapeutic strategy in tumor treatment. EPC-mediated neovascularization is a complex process that includes multiple steps and requires a series of cytokines and modulators, thus understanding the underlying mechanisms may provide anti-neovasculogenesis targets that may be blocked for the prevention of tumor development. The present review stresses the process and contribution of EPCs to the formation of new blood vessels in solid tumors, in an attempt to gain an improved understanding of the underlying cellular and molecular mechanisms involved, and to provide a potential effective therapeutic target for cancer treatment.
Collapse
|
50
|
Puntoni M, Petrera M, Campora S, Garrone E, Defferrari C, Torrisi R, Johansson H, Bruno S, Curotto A, DeCensi A. Prognostic Significance of VEGF after Twenty-Year Follow-up in a Randomized Trial of Fenretinide in Non-Muscle-Invasive Bladder Cancer. Cancer Prev Res (Phila) 2016; 9:437-44. [PMID: 27045034 DOI: 10.1158/1940-6207.capr-15-0345] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 03/25/2016] [Indexed: 11/16/2022]
Abstract
Non-muscle-invasive bladder cancer (NMIBC) may progress to muscle-invasive disease, but no effective preventive treatments are available. In addition, no reliable prognostic biomarkers have been identified. We assessed the long-term effect of the oral retinoid fenretinide and the prognostic value of circulating VEGF levels. We updated through the Tumor Registry the vital status of 99 patients with resected Ta/T1 bladder tumors who were recruited in a randomized trial of 2 years of fenretinide or no treatment in 1993-1994. Serum VEGF levels measured at baseline and 12 months were available in a subgroup of 62 patients. After a median of 20.5 years, 54 subjects died, 35 of any cancer and 14 of bladder cancer. Neither overall survival (OS), nor cancer survival (CS) or bladder cancer survival (BCS) was affected by fenretinide (log-rank P ≥ 0.2). DNA aneuploidy in bladder washing was associated with shorter OS (P = 0.02), CS (P = 0.05), and BCS (P = 0.09). Subjects with baseline VEGF levels in the top quintile (≥350 pg/mL) had a significantly shorter OS (P = 0.01), CS (P = 0.02), and BCS (P = 0.008). The trend across quintiles of VEGF was significant for BCS (P = 0.007). Multivariate analyses showed that, in addition to smoking status, VEGF level in the top quintile was an independent prognostic factor for OS (HR = 2.7; 95% CI, 1.1-6.5), CS (HR = 3.3; 95% CI, 1.1-9.4) and BCS (HR = 8.9; 95% CI,1.3-61). Fenretinide did not affect the long-term outcome of patients with NMIBC. High serum VEGF level was a significant predictor of overall and cancer death and may help to identify high-risk subjects who may benefit from a preventive therapy. Cancer Prev Res; 9(6); 437-44. ©2016 AACR.
Collapse
Affiliation(s)
- Matteo Puntoni
- Clinical Trial Unit, Scientific Direction, E.O. Ospedali Galliera, Genoa, Italy
| | | | - Sara Campora
- Medical Oncology Unit, E.O. Ospedali Galliera, Genoa, Italy
| | - Elsa Garrone
- Liguria Region Mortality Registry, IRCCS AOU San Martino-IST, Genoa, Italy
| | | | - Rosalba Torrisi
- Department of Oncology and Hematology, Humanitas Cancer Center, Humanitas Clinical and Research Hospital, Rozzano (Milan), Italy
| | - Harriet Johansson
- Division of Cancer Prevention and Genetics, European Institute of Oncology, Milan, Italy
| | - Silvia Bruno
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Antonio Curotto
- University Urology Clinic, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Andrea DeCensi
- Medical Oncology Unit, E.O. Ospedali Galliera, Genoa, Italy. Division of Cancer Prevention and Genetics, European Institute of Oncology, Milan, Italy. Wolfson Institute of Preventive Medicine, Queen Mary University of London, UK.
| |
Collapse
|