1
|
Chen W, Cha Z, Huang S, Liu R, Chen J, Kamau PM, Lu X, Li B, Liu D. Recombinant α-Toxin BmK-M9 Inhibits Breast Cancer Progression by Regulating β-Catenin In Vivo. Cell Biochem Biophys 2025:10.1007/s12013-025-01711-8. [PMID: 40080350 DOI: 10.1007/s12013-025-01711-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2025] [Indexed: 03/15/2025]
Abstract
Screening bioactive compounds from natural sources, including animals and plants, is a valuable strategy for identifying novel anti-tumor agents. α-Toxin BmK-M9, a key component of scorpion venom, has received limited attention regarding its potential anti-cancer effects and underlying mechanisms in breast cancer. This study investigates the effects and mechanisms of BmK-M9 in breast cancer using in vitro experiments and a nude mouse model. mRNA sequencing was performed to identify affected signaling pathways, while Western blotting and immunohistochemistry were utilized to analyze the Wnt/β-catenin signaling pathway. The results demonstrated that BmK-M9 significantly inhibited breast cancer cell invasion and migration in vitro and suppressed tumor growth in vivo. Transcriptomic analysis revealed that BmK-M9 influenced cellular processes related to proliferation, apoptosis, motility, and metabolism. Furthermore, BmK-M9 markedly downregulated β-catenin expression in the Wnt/β-catenin pathway. These findings suggest that BmK-M9 exerts anti-tumor effects in breast cancer by modulating Wnt/β-catenin signaling, highlighting its potential as a promising therapeutic candidate.
Collapse
Affiliation(s)
- Wenlin Chen
- Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Kunming, Yunnan, China
| | - Zhuocen Cha
- Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Kunming, Yunnan, China
- Oncology department, Guizhou Hospital of the First Affiliated Hospital, Sun Yat-sen University, Guiyang, Guizhou, China
| | - Saijun Huang
- Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Kunming, Yunnan, China
- Maternal and Child Health Hospital of Changsha County, Changsha, Hunan, China
| | - Ruimin Liu
- Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Kunming, Yunnan, China
| | - Jiayi Chen
- Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Kunming, Yunnan, China
| | - Peter Muiruri Kamau
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province/National & Local Joint Engineering Center of Natural Bioactive Peptides, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- National Resource Center for Non-Human Primates, Kunming Primate Research Center/National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Xingjia Lu
- Department of Breast Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Bowen Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province/National & Local Joint Engineering Center of Natural Bioactive Peptides, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- National Resource Center for Non-Human Primates, Kunming Primate Research Center/National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Dequan Liu
- Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Kunming, Yunnan, China.
| |
Collapse
|
2
|
Wang H, Xu C, Tan M, Su W. Advanced gut-on-chips for assessing carotenoid absorption, metabolism, and transport. Crit Rev Food Sci Nutr 2023; 65:1344-1362. [PMID: 38095598 DOI: 10.1080/10408398.2023.2293250] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Bioengineered strategies enable gut chips to faithfully replicate essential features of intestinal microsystems, encompassing geometric properties, peristalsis, intraluminal fluid flow, oxygen gradients, and the microbiome. This emerging technique serves as a powerful tool for nutrition studies by emulating the absorption and distribution processes in a manner highly relevant to human physiology. It offers unprecedented accessibility for investigating the mechanisms governing nutrition metabolism. While the application of gut-on-chip models in disease modeling and drug screening has been extensively explored, their potential in dietary nutrition research remains relatively unexplored. This comprehensive review provides an overview of the different approaches employed in constructing gut-on-chip platforms using diverse cell sources and niche mimics. Furthermore, it explores the applications and prospects of gut-on-chips in nutrition-related investigations, with a specific focus on carotenoid transport, absorption, and metabolism. Lastly, this review discusses the future development trajectory of this groundbreaking technology paradigm, highlighting its broad applicability in the field of food technology. By harnessing the capabilities of these state-of-the-art techniques within gut chip platforms, researchers can establish a robust scientific foundation for unraveling the intricate mechanisms that govern the behavior and functional properties of carotenoids.
Collapse
Affiliation(s)
- Hui Wang
- Dalian Institute of Chemical Physics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Dalian, China
| | - Cong Xu
- Department of Biomedical Engineering, Columbia University Medical Center, New York, USA
| | - Mingqian Tan
- SKL of Marine Food Processing & Safety Control, Dalian Polytechnic University, Dalian, China
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
- National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, China
| | - Wentao Su
- SKL of Marine Food Processing & Safety Control, Dalian Polytechnic University, Dalian, China
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
- National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, China
| |
Collapse
|
3
|
Bhat Y, Thrishna MR, Banerjee S. Molecular targets and therapeutic strategies for triple-negative breast cancer. Mol Biol Rep 2023; 50:10535-10577. [PMID: 37924450 DOI: 10.1007/s11033-023-08868-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/29/2023] [Indexed: 11/06/2023]
Abstract
Triple-negative breast cancer (TNBC) is known for its heterogeneous complexity and is often difficult to treat. TNBC lacks the expression of major hormonal receptors like estrogen receptor, progesterone receptor, and human epidermal growth factor receptor-2 and is further subdivided into androgen receptor (AR) positive and AR negative. In contrast, AR negative is also known as quadruple-negative breast cancer (QNBC). Compared to AR-positive TNBC, QNBC has a great scarcity of prognostic biomarkers and therapeutic targets. QNBC shows excessive cellular growth and proliferation of tumor cells due to increased expression of growth factors like EGF and various surface proteins. This study briefly reviews the limited data available as protein biomarkers that can be used as molecular targets in treating TNBC as well as QNBC. Targeted therapy and immune checkpoint inhibitors have recently changed cancer treatment. Many studies in medicinal chemistry continue to focus on the synthesis of novel compounds to discover new antiproliferative medicines capable of treating TNBC despite the abundance of treatments currently on the market. Drug repurposing is one of the therapeutic methods for TNBC that has been examined. Moreover, some additional micronutrients, nutraceuticals, and functional foods may be able to lower cancer risk or slow the spread of malignant diseases that have already been diagnosed with cancer. Finally, nanomedicines, or applications of nanotechnology in medicine, introduce nanoparticles with variable chemistry and architecture for the treatment of cancer. This review emphasizes the most recent research on nutraceuticals, medication repositioning, and novel therapeutic strategies for the treatment of TNBC.
Collapse
Affiliation(s)
- Yashasvi Bhat
- School of Bio Science and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - M R Thrishna
- School of Bio Science and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Satarupa Banerjee
- School of Bio Science and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
4
|
Metibemu DS, Ogungbe IV. Carotenoids in Drug Discovery and Medicine: Pathways and Molecular Targets Implicated in Human Diseases. Molecules 2022; 27:6005. [PMID: 36144741 PMCID: PMC9503763 DOI: 10.3390/molecules27186005] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/01/2022] [Accepted: 09/12/2022] [Indexed: 11/23/2022] Open
Abstract
Carotenoids are isoprenoid-derived natural products produced in plants, algae, fungi, and photosynthetic bacteria. Most animals cannot synthesize carotenoids because the biosynthetic machinery to create carotenoids de novo is absent in animals, except arthropods. Carotenoids are biosynthesized from two C20 geranylgeranyl pyrophosphate (GGPP) molecules made from isopentenyl pyrophosphate (IPP) and dimethylallyl pyrophosphate (DMAPP) via the methylerythritol 4-phosphate (MEP) route. Carotenoids can be extracted by a variety of methods, including maceration, Soxhlet extraction, supercritical fluid extraction (SFE), microwave-assisted extraction (MAE), accelerated solvent extraction (ASE), ultrasound-assisted extraction (UAE), pulsed electric field (PEF)-assisted extraction, and enzyme-assisted extraction (EAE). Carotenoids have been reported to exert various biochemical actions, including the inhibition of the Akt/mTOR, Bcl-2, SAPK/JNK, JAK/STAT, MAPK, Nrf2/Keap1, and NF-κB signaling pathways and the ability to increase cholesterol efflux to HDL. Carotenoids are absorbed in the intestine. A handful of carotenoids and carotenoid-based compounds are in clinical trials, while some are currently used as medicines. The application of metabolic engineering techniques for carotenoid production, whole-genome sequencing, and the use of plants as cell factories to produce specialty carotenoids presents a promising future for carotenoid research. In this review, we discussed the biosynthesis and extraction of carotenoids, the roles of carotenoids in human health, the metabolism of carotenoids, and carotenoids as a source of drugs and supplements.
Collapse
Affiliation(s)
| | - Ifedayo Victor Ogungbe
- Department of Chemistry, Physics, and Atmospheric Sciences, Jackson State University, Jackson, MS 39217-0095, USA
| |
Collapse
|
5
|
Countering Triple Negative Breast Cancer via Impeding Wnt/β-Catenin Signaling, a Phytotherapeutic Approach. PLANTS 2022; 11:plants11172191. [PMID: 36079579 PMCID: PMC9460573 DOI: 10.3390/plants11172191] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/18/2022] [Accepted: 08/21/2022] [Indexed: 12/03/2022]
Abstract
Triple negative breast cancer (TNBC) is characterized as a heterogeneous disease with severe malignancy and high mortality. Aberrant Wnt/β-catenin signaling is responsible for self-renewal and mammosphere generation, metastasis and resistance to apoptosis and chemotherapy in TNBC. Nonetheless, in the absence of a targeted therapy, chemotherapy is regarded as the exclusive treatment strategy for the treatment of TNBC. This review aims to provide an unprecedented overview of the plants and herbal derivatives which repress the progression of TNBC through prohibiting the Wnt/β-catenin pathway. Herbal medicine extracts and bioactive compounds (alkaloids, retinoids. flavonoids, terpenes, carotenoids and lignans) alone, in combination with each other and/or with chemotherapy agents could interrupt the various steps of Wnt/β-catenin signaling, i.e., WNT, FZD, LRP, GSK3β, Dsh, APC, β-catenin and TCF/LEF. These phytotherapy agents diminish proliferation, metastasis, breast cancer stem cell self-renewal and induce apoptosis in cell and animal models of TNBC through the down-expression of the downstream target genes of Wnt signaling. Some of the herbal derivatives simultaneously impede Wnt/β-catenin signaling and other overactive pathways in triple negative breast cancer, including: mTORC1; ER stress and SATB1 signaling. The herbal remedies and their bioactive ingredients perform essential roles in the treatment of the very fatal TNBC via repression of Wnt/β-catenin signaling.
Collapse
|
6
|
Hussain Y, Abdullah, Alsharif KF, Aschner M, Theyab A, Khan F, Saso L, Khan H. Therapeutic Role of Carotenoids in Blood Cancer: Mechanistic Insights and Therapeutic Potential. Nutrients 2022; 14:1949. [PMID: 35565917 PMCID: PMC9104383 DOI: 10.3390/nu14091949] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/25/2022] [Accepted: 04/29/2022] [Indexed: 02/07/2023] Open
Abstract
Blood cancers are characterized by pathological disorders causing uncontrolled hematological cell division. Various strategies were previously explored for the treatment of blood cancers, including chemotherapy, Car-T therapy, targeting chimeric antigen receptors, and platelets therapy. However, all these therapies pose serious challenges that limit their use in blood cancer therapy, such as poor metabolism. Furthermore, the solubility and stability of anticancer drugs limit efficacy and bio-distribution and cause toxicity. The isolation and purification of natural killer cells during Car-T cell therapy is a major challenge. To cope with these challenges, treatment strategies from phyto-medicine scaffolds have been evaluated for blood cancer treatments. Carotenoids represent a versatile class of phytochemical that offer therapeutic efficacy in the treatment of cancer, and specifically blood cancer. Carotenoids, through various signaling pathways and mechanisms, such as the activation of AMPK, expression of autophagy biochemical markers (p62/LC3-II), activation of Keap1-Nrf2/EpRE/ARE signaaling pathway, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), increased level of reactive oxygen species, cleaved poly (ADP-ribose) polymerase (c-PARP), c-caspase-3, -7, decreased level of Bcl-xL, cycle arrest at the G0/G1 phase, and decreasing STAT3 expression results in apoptosis induction and inhibition of cancer cell proliferation. This review article focuses the therapeutic potential of carotenoids in blood cancers, addressing various mechanisms and signaling pathways that mediate their therapeutic efficacy.
Collapse
Affiliation(s)
- Yaseen Hussain
- Lab of Controlled Release and Drug Delivery System, College of Pharmaceutical Sciences, Soochow University, Suzhou 215000, China;
- Department of Pharmacy, Bashir Institute of Health Sciences, Bharakahu, Islamabad 44000, Pakistan
| | - Abdullah
- Department of Pharmacy, University of Malakand, Chakdara 18800, Pakistan;
| | - Khalaf F. Alsharif
- Department of Clinical Laboratory, College of Applied Medical Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10463, USA;
| | - Abdulrahman Theyab
- Department of Laboratory and Blood Bank, Security Forces Hospital, P.O. Box 14799, Mecca 21955, Saudi Arabia;
- College of Medicine, Al-Faisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia
| | - Fazlullah Khan
- Faculty of Pharmacy, Capital University of Science & Technology, Islamabad 44000, Pakistan;
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University, 00185 Rome, Italy;
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
| |
Collapse
|
7
|
DU X, Zhang J, Liu L, Xu B, Han H, Dai W, Pei X, Fu X, Hou S. A novel anticancer property of Lycium barbarum polysaccharide in triggering ferroptosis of breast cancer cells. J Zhejiang Univ Sci B 2022; 23:286-299. [PMID: 35403384 PMCID: PMC9002246 DOI: 10.1631/jzus.b2100748] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 12/08/2021] [Indexed: 12/17/2022]
Abstract
Breast cancer is one of the most malignant tumors and is associated with high mortality rates among women. Lycium barbarum polysaccharide (LBP) is an extract from the fruits of the traditional Chinese herb, L. barbarum. LBP is a promising anticancer drug, due to its high activity and low toxicity. Although it has anticancer properties, its mechanisms of action have not been fully established. Ferroptosis, which is a novel anticancer strategy, is a cell death mechanism that relies on iron-dependent lipid reactive oxygen species (ROS) accumulation. In this study, human breast cancer cells (Michigan Cancer Foundation-7 (MCF-7) and MD Anderson-Metastatic Breast-231 (MDA-MB-231)) were treated with LBP. LBP inhibited their viability and proliferation in association with high levels of ferroptosis. Therefore, we aimed to ascertain whether LBP reduced cell viability through ferroptosis. We found that the structure and function of mitochondria, lipid peroxidation, and expression of solute carrier family 7 member 11 (SLC7A11, also known as xCT, the light-chain subunit of cystine/glutamate antiporter system Xc-) and glutathione peroxidase 4 (GPX4) were altered by LBP. Moreover, the ferroptosis inhibitor, Ferrostatin-1 (Fer-1), rescued LBP-induced ferroptosis-associated events including reduced cell viability and glutathione (GSH) production, accumulation of intracellular free divalent iron ions and malondialdehyde (MDA), and down-regulation of the expression of xCT and GPX4. Erastin (xCT inhibitor) and RSL3 (GPX4 inhibitor) inhibited the expression of xCT and GPX4, respectively, which was lower after the co-treatment of LBP with Erastin and RSL3. These results suggest that LBP effectively prevents breast cancer cell proliferation and promotes ferroptosis via the xCT/GPX4 pathway. Therefore, LBP exhibits novel anticancer properties by triggering ferroptosis, and may be a potential therapeutic option for breast cancer.
Collapse
Affiliation(s)
- Xing DU
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Jingjing Zhang
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
- Department of Clinical Pathology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Ling Liu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Bo Xu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Hang Han
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Wenjie Dai
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Xiuying Pei
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Xufeng Fu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China.
| | - Shaozhang Hou
- Department of Clinical Pathology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China. ,
| |
Collapse
|
8
|
Arzi L, Hoshyar R. Saffron anti-metastatic properties, ancient spice novel application. Crit Rev Food Sci Nutr 2021; 62:3939-3950. [PMID: 33653190 DOI: 10.1080/10408398.2020.1871320] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Crocus sativus L. (saffron), was applied as a spice, food colorant and medicine since four millennia ago and has been used as a remedy for various maladies. In the last three decades, the anti-primary tumor properties of saffron and its main carotenoids, crocin and crocetin, have been well explored. Despite the fact that metastasis is the leading cause of death in cancer patients, the anti-metastatic potential of saffron and its carotenoids has been surveyed only this decade. This review aims to provide an unprecedented overview of the anti-metastatic effects of saffron, crocin and crocetin, and the mechanisms underlying these effects. Investigations on various cancers demonstrated the anti-migratory, anti-invasion, anti-angiogenic potentials of saffron and its carotenoids, as well as their effects suppressing cell-ECM adhesion and enhancing cell-cell attachment. Saffron and its carotenoids exert their impact through different mechanisms such as reduction of CD34 and suppression of Wnt/β-catenin, Ras/ERK, P38, DCLK1, EMT, matrix metalloproteinases and urokinases. Crocin displayed more effective anti-metastatic potency, in comparison with saffron extract and crocetin. The bioaccessibility/bioavailability, nontoxicity on normal cells, confirmed anti-tumor efficiency and the recent evidence on the anti-metastatic potential of saffron and its carotenoids, recommends them as a propitious multipotent dietary agent and herbal medicine.
Collapse
Affiliation(s)
- Laleh Arzi
- Department of Microbiology, Shahr-e-Qods Branch, Islamic Azad University, Tehran, Iran
| | - Reyhane Hoshyar
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
9
|
Koklesova L, Liskova A, Samec M, Zhai K, Abotaleb M, Ashrafizadeh M, Brockmueller A, Shakibaei M, Biringer K, Bugos O, Najafi M, Golubnitschaja O, Büsselberg D, Kubatka P. Carotenoids in Cancer Metastasis-Status Quo and Outlook. Biomolecules 2020; 10:E1653. [PMID: 33321708 PMCID: PMC7763577 DOI: 10.3390/biom10121653] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/04/2020] [Accepted: 12/06/2020] [Indexed: 12/11/2022] Open
Abstract
Metastasis represents a major obstacle in cancer treatment and the leading cause of cancer-related deaths. Therefore, the identification of compounds targeting the multi-step and complex process of metastasis could improve outcomes in the management of cancer patients. Carotenoids are naturally occurring pigments with a plethora of biological activities. Carotenoids exert a potent anti-cancer capacity in various cancer models in vitro and in vivo, mediated by the modulation of signaling pathways involved in the migration and invasion of cancer cells and metastatic progression, including key regulators of the epithelial-mesenchymal transition and regulatory molecules, such as matrix metalloproteinases (MMPs), tissue inhibitors of metalloproteinases (TIMPs), urokinase plasminogen activator (uPA) and its receptor (uPAR), hypoxia-inducible factor-1α (HIF-1α), and others. Moreover, carotenoids modulate the expression of genes associated with cancer progression and inflammatory processes as key mediators of the complex process involved in metastasis. Nevertheless, due to the predominantly preclinical nature of the known anti-tumor effects of carotenoids, and unclear results from certain carotenoids in specific cancer types and/or specific parts of the population, a precise analysis of the anti-cancer effects of carotenoids is essential. The identification of carotenoids as effective compounds targeting the complex process of cancer progression could improve the outcomes of advanced cancer patients.
Collapse
Affiliation(s)
- Lenka Koklesova
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (L.K.); (A.L.); (M.S.); (K.B.)
| | - Alena Liskova
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (L.K.); (A.L.); (M.S.); (K.B.)
| | - Marek Samec
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (L.K.); (A.L.); (M.S.); (K.B.)
| | - Kevin Zhai
- Department of Physiology and Biophysics, Weill Cornell Medicine in Qatar, Education City, Qatar Foundation, 24144 Doha, Qatar; (K.Z.); (M.A.)
| | - Mariam Abotaleb
- Department of Physiology and Biophysics, Weill Cornell Medicine in Qatar, Education City, Qatar Foundation, 24144 Doha, Qatar; (K.Z.); (M.A.)
| | - Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956 Istanbul, Turkey;
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956 Istanbul, Turkey
| | - Aranka Brockmueller
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, D-80336 Munich, Germany; (A.B.); (M.S.)
| | - Mehdi Shakibaei
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, D-80336 Munich, Germany; (A.B.); (M.S.)
| | - Kamil Biringer
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (L.K.); (A.L.); (M.S.); (K.B.)
| | | | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah 67146, Iran;
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah 67146, Iran
| | - Olga Golubnitschaja
- Predictive, Preventive, Personalised (3P) Medicine, Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany;
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine in Qatar, Education City, Qatar Foundation, 24144 Doha, Qatar; (K.Z.); (M.A.)
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| |
Collapse
|
10
|
Bhatt T, Patel K. Carotenoids: Potent to Prevent Diseases Review. NATURAL PRODUCTS AND BIOPROSPECTING 2020; 10:109-117. [PMID: 32405969 PMCID: PMC7253555 DOI: 10.1007/s13659-020-00244-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 05/08/2020] [Indexed: 05/05/2023]
Abstract
Carotenoids are the phytochemicals known for their biological activities. They are found in nature in the form of plants, algae, fungi and in microorganisms. This is the major group having two different structure one with oxygen and without oxygen. The Present article aims to present these molecules as a new therapeutic agent, as it has unrealized efficiency to prevent and reduce the symptoms of many diseases like cancer, neurodegenerative diseases such as Alzheimer, cerebral ischemia, diabetes associated with obesity and hypertension, ophthalmic diseases and many more. It can be utilized in the form of dietary supplement as nutraceutical and pharmaceutical compounds. Yet more research and developing test knowledge is needed to make it available to the humans. In this article its sources, biosynthesis, properties, applicability and commercialization of pigments from naturally produced sources are discussed.
Collapse
Affiliation(s)
- Takshma Bhatt
- Department of Biotechnology, President Science College (Affiliated to Gujarat University), Ghatlodia, Ahmedabad, 380061 India
| | - Kirtan Patel
- Department of Biotechnology, President Science College (Affiliated to Gujarat University), Ghatlodia, Ahmedabad, 380061 India
| |
Collapse
|
11
|
Tang C, Gong L, Lvzi Xu, Qiu K, Zhang Z, Wan L. Echinacoside inhibits breast cancer cells by suppressing the Wnt/β-catenin signaling pathway. Biochem Biophys Res Commun 2020; 526:170-175. [PMID: 32201078 DOI: 10.1016/j.bbrc.2020.03.050] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 03/05/2020] [Indexed: 12/11/2022]
Abstract
Echinacoside, a small molecule derived from the natural herbs Cistanche and Echinacea, shows effective anticancer abilities, but the mechanism remains unclear. By using colony formation, scratch, and transwell assays in MDA-MB-231 breast cancer cells, we confirmed the anti-breast cancer ability of Echinacoside in vitro. In addition, we found that Echinacoside can dose-dependently reduce phosho-LRP6, total LRP6, phosho-Dvl2, active β-catenin, and total β-catenin protein expression level in MDA-MB-231 and MDA-MB-468 cells by western blot. We also detected well-known Wnt targets genes, including LEF1, CD44, and cyclin D1 by real-time PCR and western blot, and Echinacoside significantly shows inhibition effect in these two breast cancer cell lines. Furthermore, we investigated its anti-breast cancer ability in an MDA-MB-231 xenograft model in vivo. Echinacoside treatment significantly reduced tumor growth, which was accompanied by a reduction in Wnt/β-catenin signaling. In summary, our results demonstrate that Echinacoside can effectively inhibit Wnt/β-catenin signaling, and therefore, it may be a promising therapeutic target to treat breast cancer.
Collapse
Affiliation(s)
- Chenghuai Tang
- Department of Forensic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Liuping Gong
- Department of Forensic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Lvzi Xu
- Forensic Identification Center, College of Criminal Investigation, Southwest University of Political Science and Law, Chongqing, 401120, China
| | - Kaijin Qiu
- Department of Forensic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Zhong Zhang
- Department of Forensic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Lihua Wan
- Department of Forensic Medicine, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|