1
|
Farahani N, Maghsoodlou A, Akbari M, Tahmasebi S, Daneshi S, Ramezani Farani M, Yusefi AR, Rahimzadeh P, Taheriazam A, Entezari M, Hashemi M. Translating preclinical insights into clinical strategies: Targeting cancer stem cells and stemness in prostate cancer. Pathol Res Pract 2025; 269:155934. [PMID: 40186890 DOI: 10.1016/j.prp.2025.155934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 03/18/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025]
Abstract
CSCs represent a unique group within the tumor microenvironment (TME) elevating the tumorigenesis. The cause of cancer recurrence can also be investigated in the function of CSCs possessing self-renewing capabilities and differentiation into various types of cells. Prostate cancer (PCa) is a malignant disease of the urogenital system characterized by aggressive behavior and heterogeneous nature due to the dysregulation of molecular pathways and the interactions among cells within the TME. The PCa can quickly become resistant to standard chemotherapy and other kinds of therapies such as radiotherapy along with ability to mediate immune evasion. The focus of biology has been on the molecular and cellular alterations in PCa. The CSCs have been recognized as potential biomarkers for predicting the outcome of prostate PCa. Furthermore, a positive correlation exists between CSCs and the metastatic growth and stemness of PCa. The existence of hypoxia enhances the stemness of PCa, and CSCs play a role in dormancy. Genomic and epigenetic elements, including non-coding RNAs, can influence CSCs and the advancement of PCa. Additionally, therapeutic agents and nanotechnology methods aimed at targeting CSCs have been developed to inhibit CSCs in PCa treatment.
Collapse
Affiliation(s)
- Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Amin Maghsoodlou
- Young Researchers and Elite Club, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Mohammadarian Akbari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Safa Tahmasebi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran,Iran
| | - Salman Daneshi
- Department of Public Health, School of Health, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Marzieh Ramezani Farani
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea
| | - Ali Reza Yusefi
- Department of Public Health, Sirjan School of Medical Sciences, Sirjan, Iran
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
2
|
Filippi A, Deculescu-Ioniță T, Hudiță A, Baldasici O, Gălățeanu B, Mocanu MM. Molecular Mechanisms of Dietary Compounds in Cancer Stem Cells from Solid Tumors: Insights into Colorectal, Breast, and Prostate Cancer. Int J Mol Sci 2025; 26:631. [PMID: 39859345 PMCID: PMC11766403 DOI: 10.3390/ijms26020631] [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: 11/21/2024] [Revised: 01/10/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
Cancer stem cells (CSC) are known to be the main source of tumor relapse, metastasis, or multidrug resistance and the mechanisms to counteract or eradicate them and their activity remain elusive. There are different hypotheses that claim that the origin of CSC might be in regular stem cells (SC) and, due to accumulation of mutations, these normal cells become malignant, or the source of CSC might be in any malignant cell that, under certain environmental circumstances, acquires all the qualities to become CSC. Multiple studies indicate that lifestyle and diet might represent a source of wellbeing that can prevent and ameliorate the malignant phenotype of CSC. In this review, after a brief introduction to SC and CSC, we analyze the effects of phenolic and non-phenolic dietary compounds and we highlight the molecular mechanisms that are shown to link diets to CSC activation in colon, breast, and prostate cancer. We focus the analysis on specific markers such as sphere formation, CD surface markers, epithelial-mesenchymal transition (EMT), Oct4, Nanog, Sox2, and aldehyde dehydrogenase 1 (ALDH1) and on the major signaling pathways such as PI3K/Akt/mTOR, NF-κB, Notch, Hedgehog, and Wnt/β-catenin in CSC. In conclusion, a better understanding of how bioactive compounds in our diets influence the dynamics of CSC can raise valuable awareness towards reducing cancer risk.
Collapse
Affiliation(s)
- Alexandru Filippi
- Department of Biochemistry and Biophysics, “Carol Davila” University of Medicine and Pharmacy of Bucharest, 050474 Bucharest, Romania;
| | - Teodora Deculescu-Ioniță
- Department of Pharmacognosy, Phytochemistry and Phytotherapy, “Carol Davila” University of Medicine and Pharmacy of Bucharest, 050474 Bucharest, Romania;
| | - Ariana Hudiță
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania; (A.H.); (B.G.)
| | - Oana Baldasici
- Department of Genetics, Genomics and Experimental Pathology, The Oncology Institute “Prof. Dr. Ion Chiricuță”, 400015 Cluj-Napoca, Romania;
| | - Bianca Gălățeanu
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania; (A.H.); (B.G.)
| | - Maria-Magdalena Mocanu
- Department of Biochemistry and Biophysics, “Carol Davila” University of Medicine and Pharmacy of Bucharest, 050474 Bucharest, Romania;
| |
Collapse
|
3
|
Çınar AK, Serttas R, Çınar AC, Güçlü H, Erdogan S. As shown hesperidin suppresses TGF-β2-induced proliferation and epithelial-mesenchymal transition of retinal pigment epithelial cells. J Mol Histol 2024; 56:10. [PMID: 39612089 DOI: 10.1007/s10735-024-10275-5] [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: 08/20/2024] [Accepted: 11/17/2024] [Indexed: 11/30/2024]
Abstract
This study investigates the potential therapeutic effects and molecular mechanisms of hesperidin treatment on cell migration and epithelial-mesenchymal transition, key stages of proliferative vitreoretinopathy (PVR). Human retinal pigment epithelial cells (ARPE-19) were treated with 10 ng/ml transforming growth factor-beta 2 (TGF-β2) alone or in combination with 1.56 μM hesperidin for 48 h. The impact of treatment on cell migration was evaluated using a wound healing assay. Apoptosis was assessed using DNA staining. mRNA and protein expression were evaluated using real-time PCR and Western blot, respectively. Hesperidin inhibits the proliferation and transformation of the cells by inducing apoptosis and reverses the cell morphology modified by TGF-β2. Hesperidin inhibits cell migration induced by TGF-β2. Upon treatment with hesperidin, the levels of mesenchymal markers upregulated by TGF-β2, such as MMP-1, -2, -9, fibronectin, α-SMA and the transcription factors Snail, Slug and ZEB-1, were downregulated. Conversely, the epithelial marker E-cadherin is upregulated with hesperidin treatment. Additionally, TIMP-1 and TIMP-2 expression levels, which are downregulated, increase with the treatment. These results suggest that hesperidin may inhibit the migration and EMT processes of RPE cells involved in the development of PVR, indicating its potential as a therapeutic agent for treating PVR.
Collapse
Affiliation(s)
- Ayça Küpeli Çınar
- Department of Ophthalmology, School of Medicine, Trakya University, Balkan Campus, Edirne, Türkiye.
| | - Riza Serttas
- Department of Medical Biology, School of Medicine, Trakya University, Balkan Campus, Edirne, Türkiye
| | - Abdulkadir Can Çınar
- Department of Ophthalmology, School of Medicine, Trakya University, Balkan Campus, Edirne, Türkiye
| | - Hande Güçlü
- Department of Ophthalmology, School of Medicine, Trakya University, Balkan Campus, Edirne, Türkiye
| | - Suat Erdogan
- Department of Medical Biology, School of Medicine, Trakya University, Balkan Campus, Edirne, Türkiye
| |
Collapse
|
4
|
Singh A, Singh J, Parween G, Khator R, Monga V. A comprehensive review of apigenin a dietary flavonoid: biological sources, nutraceutical prospects, chemistry and pharmacological insights and health benefits. Crit Rev Food Sci Nutr 2024:1-37. [PMID: 39154213 DOI: 10.1080/10408398.2024.2390550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/19/2024]
Abstract
A multitude of plant-derived bioactive compounds have shown significant promise in preventing chronic illnesses, with flavonoids constituting a substantial class of naturally occurring polyphenolic compounds. Apigenin, a flavone identified as 4',5,7-trihydroxyflavone, holds immense promise as a preventative agent against chronic illnesses. Despite its extensive research and recognized nutraceutical value, its therapeutic application remains underexplored, necessitating further clinical investigations. This review delves into the biological sources, nutraceutical prospects, chemistry, pharmacological insights, and health benefits of apigenin. Through multifaceted analytical studies, we explore its diverse pharmacological profile and potential therapeutic applications across various health domains. The manuscript comprehensively examines apigenin's role as a neuroprotective , anti-inflammatory compound, and a potent antioxidant agent. Additionally, its efficacy in combating cardiovascular diseases, anti-diabetic properties, and anticancer potential has been discussed. Furthermore, the antimicrobial attributes and the challenges surrounding its bioavailability, particularly from herbal supplements have been addressed. Available in diverse forms including tablets, capsules, solid dispersions, co-crystals, inclusion complexes and nano formulations. Additionally, it is prevalent as a nutraceutical supplement in herbal formulations. While strides have been made in overcoming pharmacokinetic hurdles, further research into apigenin's clinical effectiveness and bioavailability from herbal supplements remains imperative for its widespread utilization in preventive medicine.
Collapse
Affiliation(s)
- Abhinav Singh
- Drug Design and Molecular Synthesis Laboratory, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, VPO-Ghudda, Bathinda, India
| | - Jagjit Singh
- Drug Design and Molecular Synthesis Laboratory, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, VPO-Ghudda, Bathinda, India
| | - Gulistan Parween
- Drug Design and Molecular Synthesis Laboratory, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, VPO-Ghudda, Bathinda, India
| | - Rakesh Khator
- Drug Design and Molecular Synthesis Laboratory, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, VPO-Ghudda, Bathinda, India
| | - Vikramdeep Monga
- Drug Design and Molecular Synthesis Laboratory, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, VPO-Ghudda, Bathinda, India
| |
Collapse
|
5
|
Figueira MI, Carvalho TMA, Macário-Monteiro J, Cardoso HJ, Correia S, Vaz CV, Duarte AP, Socorro S. The Pros and Cons of Estrogens in Prostate Cancer: An Update with a Focus on Phytoestrogens. Biomedicines 2024; 12:1636. [PMID: 39200101 PMCID: PMC11351860 DOI: 10.3390/biomedicines12081636] [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] [Received: 06/26/2024] [Revised: 07/14/2024] [Accepted: 07/20/2024] [Indexed: 09/01/2024] Open
Abstract
The role of estrogens in prostate cancer (PCa) is shrouded in mystery, with its actions going from angelic to devilish. The findings by Huggins and Hodges establishing PCa as a hormone-sensitive cancer have provided the basis for using estrogens in therapy. However, despite the clinical efficacy in suppressing tumor growth and the panoply of experimental evidence describing its anticarcinogenic effects, estrogens were abolished from PCa treatment because of the adverse secondary effects. Notwithstanding, research work over the years has continued investigating the effects of estrogens, reporting their pros and cons in prostate carcinogenesis. In contrast with the beneficial therapeutic effects, many reports have implicated estrogens in the disruption of prostate cell fate and tissue homeostasis. On the other hand, epidemiological data demonstrating the lower incidence of PCa in Eastern countries associated with a higher consumption of phytoestrogens support the beneficial role of estrogens in counteracting cancer development. Many studies have investigated the effects of phytoestrogens and the underlying mechanisms of action, which may contribute to developing safe estrogen-based anti-PCa therapies. This review compiles the existing data on the anti- and protumorigenic actions of estrogens and summarizes the anticancer effects of several phytoestrogens, highlighting their promising features in PCa treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Sílvia Socorro
- CICS-UBI, Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal; (M.I.F.)
| |
Collapse
|
6
|
Xu Y, Bai Z, Lan T, Fu C, Cheng P. CD44 and its implication in neoplastic diseases. MedComm (Beijing) 2024; 5:e554. [PMID: 38783892 PMCID: PMC11112461 DOI: 10.1002/mco2.554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 03/20/2024] [Accepted: 04/01/2024] [Indexed: 05/25/2024] Open
Abstract
CD44, a nonkinase single span transmembrane glycoprotein, is a major cell surface receptor for many other extracellular matrix components as well as classic markers of cancer stem cells and immune cells. Through alternative splicing of CD44 gene, CD44 is divided into two isoforms, the standard isoform of CD44 (CD44s) and the variant isoform of CD44 (CD44v). Different isoforms of CD44 participate in regulating various signaling pathways, modulating cancer proliferation, invasion, metastasis, and drug resistance, with its aberrant expression and dysregulation contributing to tumor initiation and progression. However, CD44s and CD44v play overlapping or contradictory roles in tumor initiation and progression, which is not fully understood. Herein, we discuss the present understanding of the functional and structural roles of CD44 in the pathogenic mechanism of multiple cancers. The regulation functions of CD44 in cancers-associated signaling pathways is summarized. Moreover, we provide an overview of the anticancer therapeutic strategies that targeting CD44 and preclinical and clinical trials evaluating the pharmacokinetics, efficacy, and drug-related toxicity about CD44-targeted therapies. This review provides up-to-date information about the roles of CD44 in neoplastic diseases, which may open new perspectives in the field of cancer treatment through targeting CD44.
Collapse
Affiliation(s)
- Yiming Xu
- Department of BiotherapyLaboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Ziyi Bai
- Department of BiotherapyLaboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Tianxia Lan
- Department of BiotherapyLaboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Chenying Fu
- Laboratory of Aging and Geriatric Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Ping Cheng
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
7
|
Erdogan S, Serttas R, Dibirdik I, Turkekul K. Multifaceted impact of adipose conditioned media: Obesity-driven promotion of prostate cancer and cancer stem cell dynamics. Cell Biochem Funct 2024; 42:e3979. [PMID: 38481004 DOI: 10.1002/cbf.3979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 03/22/2024]
Abstract
Obesity is an established risk factor for the development and progression of prostate cancer (PC). This study used adipose conditioned media (ACM) from differentiated adipocytes to assess its effect on PC development and aggressiveness. Due to limited research on ACM's impact on isolated PC stem cells (PCSCs), we also examined CD44+ PCSCs. ACM notably boosted interleukin-1β (IL-1β), IL-6, and IL-8 production in normal prostate epithelial cells and LNCaP cells. It also increased IL-6 and IL-8 production in PC3 and CD44+ LNCaP cells, and IL-1β and IL-6 production in CD44+ PC3 cells. This indicates that ACM induces the production of inflammatory cytokines in both cancer and prostate epithelial cells. Furthermore, ACM promoted proliferation in androgen receptor (AR)-negative PC3 cells, CD44+ PC3 PCSCs, and nonmalignant RWPE cells, without affecting AR-positive LNCaP cells. In addition, ACM-enhanced invasion and migration potential in both PC3 and CD44+ PC3 cells. Western blot analysis indicated the involvement of NF-κB and AKT pathways in ACM-induced proliferation in PC3 cells and NF-κB in PCSCs. In ACM-treated PC3 cells, E-cadherin was downregulated, while N-cadherin, Snail, vimentin, fibronectin, and Twist were upregulated, suggesting ACM-induced invasion via classical epithelial-to-mesenchymal transition (EMT) pathways. In response to ACM, PCSCs exhibited increased expression of E-cadherin, Snail, and vimentin, which are partial EMT markers promoting stemness and resistance to apoptosis. In addition, increased expressions of Nanog, Oct3/4, survivin, and Bcl-2 were observed. Although the molecules we studied have diverse effects on cellular regulation, our data emphasize obesity's multifaceted role in promoting and aggressing PC, notably affecting PCSC populations.
Collapse
Affiliation(s)
- Suat Erdogan
- Department of Medical Biology, School of Medicine, Trakya University, Balkan Campus, Edirne, Türkiye
| | - Riza Serttas
- Department of Medical Biology, School of Medicine, Trakya University, Balkan Campus, Edirne, Türkiye
| | - Ilker Dibirdik
- Department of Medical Biochemistry, School of Medicine, Trakya University, Balkan Campus, Edirne, Türkiye
| | - Kader Turkekul
- Department of Medical Biology, School of Medicine, Trakya University, Balkan Campus, Edirne, Türkiye
| |
Collapse
|
8
|
Al-Rashidi RR, Noraldeen SAM, Kareem AK, Mahmoud AK, Kadhum WR, Ramírez-Coronel AA, Iswanto AH, Obaid RF, Jalil AT, Mustafa YF, Nabavi N, Wang Y, Wang L. Malignant function of nuclear factor-kappaB axis in prostate cancer: Molecular interactions and regulation by non-coding RNAs. Pharmacol Res 2023; 194:106775. [PMID: 37075872 DOI: 10.1016/j.phrs.2023.106775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/09/2023] [Accepted: 04/16/2023] [Indexed: 04/21/2023]
Abstract
Prostate carcinoma is a malignant situation that arises from genomic alterations in the prostate, leading to changes in tumorigenesis. The NF-κB pathway modulates various biological mechanisms, including inflammation and immune responses. Dysregulation of NF-κB promotes carcinogenesis, including increased proliferation, invasion, and therapy resistance. As an incurable disease globally, prostate cancer is a significant health concern, and research into genetic mutations and NF-κB function has the efficacy to facilitate the introduction of novel therapies. NF-κB upregulation is observed during prostate cancer progression, resulting in increased cell cycle progression and proliferation rates. Additionally, NF-κB endorses resistance to cell death and enhances the capacity for metastasis, particularly bone metastasis. Overexpression of NF-κB triggers chemoresistance and radio-resistance, and inhibition of NF-κB by anti-tumor compounds can reduce cancer progression. Interestingly, non-coding RNA transcripts can regulate NF-κB level and its nuclear transfer, offering a potential avenue for modulating prostate cancer progression.
Collapse
Affiliation(s)
| | | | - Ali Kamil Kareem
- Biomedical Engineering Department, Al-Mustaqbal University College, 51001, Hillah, Iraq
| | | | - Wesam R Kadhum
- Department of Pharmacy, Kut University College, Kut 52001, Wasit, Iraq
| | - Andrés Alexis Ramírez-Coronel
- Azogues Campus Nursing Career, Health and Behavior Research Group (HBR), Psychometry and Ethology Laboratory, Catholic University of Cuenca, Ecuador; University of Palermo, Buenos Aires, Argentina; Epidemiology and Biostatistics Research Group, CES University, Colombia
| | - Acim Heri Iswanto
- Department of Public Health, Faculty of Health Science, University of Pembangunan Nasional Veteran Jakarta, Jakarta, Indonesia
| | - Rasha Fadhel Obaid
- Department of Biomedical Engineering, Al-Mustaqbal University College, Babylon, Iraq
| | - Abduladheem Turki Jalil
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul 41001, Iraq
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6 Vancouver, BC, Canada.
| | - Yuzhuo Wang
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6 Vancouver, BC, Canada; Department of Experimental Therapeutics, BC Cancer Research Institute, V5Z1L3 Vancouver, BC, Canada.
| | - Lin Wang
- Department of Geriatrics, Xijing Hospital, The Air Force Military Medical University, Xi'an 710032, China.
| |
Collapse
|
9
|
Anticancer Potential of Apigenin and Isovitexin with Focus on Oncogenic Metabolism in Cancer Stem Cells. Metabolites 2023; 13:metabo13030404. [PMID: 36984844 PMCID: PMC10051376 DOI: 10.3390/metabo13030404] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 02/23/2023] [Accepted: 03/02/2023] [Indexed: 03/11/2023] Open
Abstract
It has been demonstrated that cancer stem cells (CSCs) go through metabolic changes that differentiate them from non-CSCs. The altered metabolism of CSCs plays a vital role in tumor initiation, progression, immunosuppression, and resistance to conventional therapy. Therefore, defining the role of CSC metabolism in carcinogenesis has emerged as a main focus in cancer research. Two natural flavonoids, apigenin and isovitexin, have been shown to act synergistically with conventional chemotherapeutic drugs by sensitizing CSCs, ultimately leading to improved therapeutic efficacy. The aim of this study is to present a critical and broad evaluation of the anti-CSC capability of apigenin and isovitexin in different cancers as novel and untapped natural compounds for developing drugs. A thorough review of the included literature supports a strong association between anti-CSC activity and treatment with apigenin or isovitexin. Additionally, it has been shown that apigenin or isovitexin affected CSC metabolism and reduced CSCs through various mechanisms, including the suppression of the Wnt/β-catenin signaling pathway, the inhibition of nuclear factor-κB protein expression, and the downregulation of the cell cycle via upregulation of p21 and cyclin-dependent kinases. The findings of this study demonstrate that apigenin and isovitexin are potent candidates for treating cancer due to their antagonistic effects on CSC metabolism.
Collapse
|
10
|
Pandey P, Khan F, Upadhyay TK. Deciphering the modulatory role of apigenin targeting oncogenic pathways in human cancers. Chem Biol Drug Des 2023; 101:1446-1458. [PMID: 36746671 DOI: 10.1111/cbdd.14206] [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: 10/28/2022] [Revised: 12/14/2022] [Accepted: 02/01/2023] [Indexed: 02/08/2023]
Abstract
Cancer is a complicated malignancy controlled by numerous intrinsic and extrinsic pathways. There has been a significant increase in interest in recent years in the elucidation of cancer treatments based on natural extracts that have fewer side effects. Numerous natural product-derived chemicals have been investigated for their anticancer effects in the search for an efficient chemotherapeutic method. Therefore, the rationale behind this review is to provide a detailed insights about the anticancerous potential of apigenin via modulating numerous cell signaling pathways. An ingestible plant-derived flavonoid called apigenin has been linked to numerous anticancerous potential in numerous experimental and biological studies. Apigenin has been reported to induce cell growth arrest and apoptotic induction by modulating multiple cell signaling pathways in a wider range of human tumors including those of the breast, lung, liver, skin, blood, colon, prostate, pancreatic, cervical, oral, and stomach. Oncogenic protein networks, abnormal cell signaling, and modulation of the apoptotic machinery are only a few examples of diverse molecular interactions and processes that have not yet been thoroughly addressed by scientific research. Thus, keeping this fact in mind, we tried to focus our review towards summarizing the apigenin-mediated modulation of oncogenic pathways in various malignancies that can be further utilized to develop a potent therapeutic alternative for the treatment of various cancers.
Collapse
Affiliation(s)
- Pratibha Pandey
- Department of Biotechnology, Noida Institute of Engineering and Technology, Greater Noida, Uttar Pradesh, India
| | - Fahad Khan
- Department of Biotechnology, Noida Institute of Engineering and Technology, Greater Noida, Uttar Pradesh, India
| | - Tarun Kumar Upadhyay
- Department of Biotechnology, Parul Institute of Applied Sciences and Centre of Research for Development, Parul University, Vadodara, Gujarat, India
| |
Collapse
|
11
|
Moslehi M, Rezaei S, Talebzadeh P, Ansari MJ, Jawad MA, Jalil AT, Rastegar-Pouyani N, Jafarzadeh E, Taeb S, Najafi M. Apigenin in cancer therapy: Prevention of genomic instability and anticancer mechanisms. Clin Exp Pharmacol Physiol 2023; 50:3-18. [PMID: 36111951 DOI: 10.1111/1440-1681.13725] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/18/2022] [Accepted: 09/14/2022] [Indexed: 12/13/2022]
Abstract
The incidence of cancer has been growing worldwide. Better survival rates following the administration of novel drugs and new combination therapies may concomitantly cause concern regarding the long-term adverse effects of cancer therapy, for example, second primary malignancies. Moreover, overcoming tumour resistance to anticancer agents has been long considered as a critical challenge in cancer research. Some low toxic adjuvants such as herb-derived molecules may be of interest for chemoprevention and overcoming the resistance of malignancies to cancer therapy. Apigenin is a plant-derived molecule with attractive properties for chemoprevention, for instance, promising anti-tumour effects, which may make it a desirable adjuvant to reduce genomic instability and the risks of second malignancies among normal tissues. Moreover, it may improve the efficiency of anticancer modalities. This paper aims to review various effects of apigenin in both normal tissues and malignancies. In addition, we explain how apigenin may have the ability to protect usual cells against the genotoxic repercussions following radiotherapy and chemotherapy. Furthermore, the inhibitory effects of apigenin on tumours will be discussed.
Collapse
Affiliation(s)
- Masoud Moslehi
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sepideh Rezaei
- Department of Chemistry, University of Houston, Houston, Texas, USA
| | - Pourya Talebzadeh
- Student Research Committee, Tehran Medical Faculty, Islamic Azad University, Tehran, Iran
| | - Mohammad Javed Ansari
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-kharj, Saudi Arabia
| | | | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, Iraq
| | - Nima Rastegar-Pouyani
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Emad Jafarzadeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahram Taeb
- Department of Radiology, School of Paramedical Sciences, Guilan University of Medical Sciences, Rasht, Iran.,Medical Biotechnology Research Center, School of Paramedical Sciences, Guilan University of Medical Sciences, Rasht, Iran
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
12
|
Moczulski M, Artelska A, Albrecht Ł, Albrecht A. Photocatalytic Activity of Triphenylphosphine and Potassium Iodide system in the Decarboxylative Alkylation of 3‐Cyanochromones. European J Org Chem 2022. [DOI: 10.1002/ejoc.202200630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Marek Moczulski
- Lodz University of Technology: Politechnika Lodzka Chemistry Żeromskiego 116 90-924 Łódź POLAND
| | - Angelika Artelska
- Lodz University of Technology: Politechnika Lodzka Chemistry Żeromskiego 116 90-924 Łódź POLAND
| | - Łukasz Albrecht
- Lodz University of Technology: Politechnika Lodzka Chemistry Żeromskiego 116 90-924 Łódź POLAND
| | - Anna Albrecht
- Lodz University of Technology Chemistry Zeromskiego 116 90-924 Lodz POLAND
| |
Collapse
|
13
|
Tossing G, Livernoche R, Maios C, Bretonneau C, Labarre A, Parker JA. Genetic and pharmacological PARP inhibition reduces axonal degeneration in C. elegans models of ALS. Hum Mol Genet 2022; 31:3313-3324. [PMID: 35594544 DOI: 10.1093/hmg/ddac116] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 11/12/2022] Open
Abstract
Axonal degeneration is observed in early stages of several neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). This degeneration generally precedes apoptosis and therefore may be a promising therapeutic target. An increasing number of genes have been identified to actively regulate axonal degeneration and regeneration, however, only a few potential therapeutic targets have been identified in the context of neurodegenerative diseases. Here we investigate DLK-1, a major axonal regeneration pathway and its contribution to axonal degeneration phenotypes in several C. elegans ALS models. From this pathway, we identified the PAR polymerases (PARP) PARP-1 and PARP-2 as the most consistent modifiers of axonal degeneration in our models of ALS. Genetic and pharmacological inhibition of PARP-1 and PARP-2 reduces axonal degeneration and improves related motor phenotypes.
Collapse
Affiliation(s)
- Gilles Tossing
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada.,Department of Neuroscience, Université de Montréal, Montreal, Canada
| | | | - Claudia Maios
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada
| | - Constantin Bretonneau
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada.,Department of Neuroscience, Université de Montréal, Montreal, Canada
| | - Audrey Labarre
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada.,Department of Neuroscience, Université de Montréal, Montreal, Canada
| | - J Alex Parker
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada.,Department of Neuroscience, Université de Montréal, Montreal, Canada
| |
Collapse
|
14
|
Zhang L, Song L, Xu Y, Xu Y, Zheng M, Zhang P, Wang Q. Midkine promotes breast cancer cell proliferation and migration by upregulating NR3C1 expression and activating the NF-κB pathway. Mol Biol Rep 2022; 49:2953-2961. [PMID: 35028860 DOI: 10.1007/s11033-022-07116-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 01/04/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND Breast cancer (BC) is the most common malignancy in females and is the second leading cause of cancer-related death among women worldwide. Midkine (MDK) is a heparin-binding growth factor that is abnormally expressed at high levels in various human malignancies. We aimed to uncover the biological function and molecular mechanism of MDK in BC cells. METHODS AND RESULTS MDA-MB-231-shMDK and T47D-shMDK BC cells were established. The in vitro biological functions of MDK were demonstrated by CCK-8 assays, Transwell assays and Western blotting, whereas qPCR pathway arrays were implemented to explore the mechanism of MDK in BC cells. Functionally, we verified that silencing MDK significantly suppressed BC cell proliferation and migration by inhibiting the activation of the nuclear factor kappa B (NF-κB) pathway and the nuclear distribution of NF-κB. Meanwhile, Ingenuity Pathway Analysis (IPA) and a qPCR pathway array revealed that silencing MDK decreased the expression of NR3C1, a potential downstream target of the NF-κB pathway. We also confirmed that treatment with an NF-κB inhibitor suppressed NR3C1 expression in BC cells. Finally, we demonstrated that silencing NR3C1 repressed BC cell proliferation and migration. CONCLUSIONS Our findings highlight a novel mechanism by which MDK influences BC progression via regulation of the NF-κB-NR3C1 pathway.
Collapse
Affiliation(s)
- Lin Zhang
- Jiangsu Province Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.,Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.,Department of Pathology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Li Song
- Department of Pathology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yanyan Xu
- Department of Pathology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yuting Xu
- Department of Pathology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Maojin Zheng
- Department of Pathology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Peng Zhang
- Jiangsu Province Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Qingling Wang
- Jiangsu Province Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China. .,Department of Pathology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
15
|
Targeting Cancer Stem Cells by Dietary Agents: An Important Therapeutic Strategy against Human Malignancies. Int J Mol Sci 2021; 22:ijms222111669. [PMID: 34769099 PMCID: PMC8584029 DOI: 10.3390/ijms222111669] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/23/2021] [Accepted: 10/23/2021] [Indexed: 02/07/2023] Open
Abstract
As a multifactorial disease, treatment of cancer depends on understanding unique mechanisms involved in its progression. The cancer stem cells (CSCs) are responsible for tumor stemness and by enhancing colony formation, proliferation as well as metastasis, and these cells can also mediate resistance to therapy. Furthermore, the presence of CSCs leads to cancer recurrence and therefore their complete eradication can have immense therapeutic benefits. The present review focuses on targeting CSCs by natural products in cancer therapy. The growth and colony formation capacities of CSCs have been reported can be attenuated by the dietary agents. These compounds can induce apoptosis in CSCs and reduce tumor migration and invasion via EMT inhibition. A variety of molecular pathways including STAT3, Wnt/β-catenin, Sonic Hedgehog, Gli1 and NF-κB undergo down-regulation by dietary agents in suppressing CSC features. Upon exposure to natural agents, a significant decrease occurs in levels of CSC markers including CD44, CD133, ALDH1, Oct4 and Nanog to impair cancer stemness. Furthermore, CSC suppression by dietary agents can enhance sensitivity of tumors to chemotherapy and radiotherapy. In addition to in vitro studies, as well as experiments on the different preclinical models have shown capacity of natural products in suppressing cancer stemness. Furthermore, use of nanostructures for improving therapeutic impact of dietary agents is recommended to rapidly translate preclinical findings for clinical use.
Collapse
|
16
|
Targeting cancer stem cells by nutraceuticals for cancer therapy. Semin Cancer Biol 2021; 85:234-245. [PMID: 34273521 DOI: 10.1016/j.semcancer.2021.07.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 02/06/2023]
Abstract
Accumulating evidence has demonstrated that cancer stem cells (CSCs) play an essential role in tumor progression and reoccurrence and drug resistance. Multiple signaling pathways have been revealed to be critically participated in CSC development and maintenance. Emerging evidence indicates that numerous chemopreventive compounds, also known as nutraceuticals, could eliminate CSCs in part via regulating several signaling pathways. Therefore, in this review, we will describe the some natural chemopreventive agents that target CSCs in a variety of human malignancies, including soy isoflavone, curcumin, resveratrol, tea polyphenols, sulforaphane, quercetin, indole-3-carbinol, 3,3'-diindolylmethane, withaferin A, apigenin, etc. Moreover, we discuss that eliminating CSCs by nutraceuticals might be a promising strategy for treating human cancer via overcoming drug resistance and reducing tumor reoccurrence.
Collapse
|
17
|
Cinar AK, Ozal SA, Serttas R, Erdogan S. Eupatilin attenuates TGF-β2-induced proliferation and epithelial-mesenchymal transition of retinal pigment epithelial cells. Cutan Ocul Toxicol 2021; 40:103-114. [PMID: 33719768 DOI: 10.1080/15569527.2021.1902343] [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] [Indexed: 12/12/2022]
Abstract
PURPOSE The main characteristic of proliferative vitreoretinopathy (PVR) is migration, adhesion, and epithelial-mesenchymal transition (EMT) of retinal pigment epithelial cells (RPE). Eupatilin is a naturally occurring flavone that has the potential to inhibit cell proliferation and EMT. However, its efficacy on the PVR model induced by transforming growth factor-2 (TGF-β2) is unknown. In this study, the potential effect of eupatilin on proliferation and EMT in the treatment of RPE was investigated. METHODS Serum starved human RPE cells (ARPE-19) were treated with 10 ng/ml TGF-β2 alone or co-treated with 25 μM eupatilin for 48 h. Quantitative real-time PCR and Western blot analysis were used to assess targets at the mRNA and protein expression level, respectively. Apoptosis and cell cycle progression was assessed by image-based cytometry. The effect of treatment on cell migration was evaluated by wound healing assay. RESULTS Eupatilin inhibited TGF-β2-induced RPE cell proliferation via regulating the cell cycle and inducing apoptosis. TGF-β2 upregulated mRNA expression of mesenchymal markers fibronectin and vimentin was significantly downregulated by the treatment, while the epithelial markers E-cadherin and occludin expression was upregulated. The therapy significantly suppressed TGF-β2 encouraged cell migration through downregulating the expression of transcription factors Twist, Snail, and ZEB1 induced by TGF-β2. Furthermore, eupatilin significantly inhibited the expression of MMP-1, -7, and -9, and suppressed NF-κB signalling. CONCLUSION These results suggest that eupatilin could inhibit the proliferation and transformation into fibroblast-like cells of RPE cells; thus the agent may be a potential therapeutic value in treating PVR.
Collapse
Affiliation(s)
- Ayca Kupeli Cinar
- Department of Ophthalmology, School of Medicine, Trakya University - Balkan Campus, Edirne, Turkey
| | - S Altan Ozal
- Department of Ophthalmology, School of Medicine, Trakya University - Balkan Campus, Edirne, Turkey
| | - Riza Serttas
- Department of Medical Biology, School of Medicine, Trakya University - Balkan Campus, Edirne, Turkey
| | - Suat Erdogan
- Department of Medical Biology, School of Medicine, Trakya University - Balkan Campus, Edirne, Turkey
| |
Collapse
|
18
|
Serttas R, Koroglu C, Erdogan S. Eupatilin Inhibits the Proliferation and Migration of Prostate Cancer Cells through Modulation of PTEN and NF-κB Signaling. Anticancer Agents Med Chem 2021; 21:372-382. [PMID: 32781972 DOI: 10.2174/1871520620666200811113549] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/29/2020] [Accepted: 07/09/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Despite advances in the treatment of prostate cancer, side effects and the risks of developing drug resistance require new therapeutic agents. Eupatilin is a secondary metabolite of Artemisia asiatica and has shown potential anti-tumor activity in some cancers, but its potential in prostate cancer treatment has not yet been evaluated. OBJECTIVE The aim of the study was to investigate the effectiveness of eupatilin on prostate cancer cell proliferation and migration. METHODS Human prostate cancer PC3 and LNCaP cells were exposed to eupatilin and its efficacy on cell survival was determined by the MTT test. Apoptosis and cell cycle phases were evaluated by an image-based cytometer. Cell migration and invasion were evaluated by wound healing and matrigel migration assays; the expression of mRNA and protein was assessed by RT-qPCR and Western blot, respectively. RESULTS Eupatilin time- and dose-dependently reduced the viability of prostate cancer cells. Exposure of PC3 cells to 12.5μM-50μM eupatilin resulted in apoptosis by upregulating the expression of caspase 3, Bax and cytochrome c. Annexin V assessment also confirmed that eupatilin causes apoptosis. The treatment significantly upregulated the mRNA expression of p53, p21, and p27, causing cell cycle arrest in the G1 phase. Administration of eupatilin inhibited migration and invasion of the cells by downregulating the expression of Twist, Slug and MMP-2, -7. In addition, the agent increased protein expression of tumor suppressor PTEN, while transcription factor NF-κB expression was reduced. CONCLUSION Eupatilin strongly prevents the proliferation of prostate cancer cells, and suppresses migration and invasion. Due to its therapeutic potential, the clinical use of eupatilin in prostate cancer should also be supported by in vivo studies.
Collapse
Affiliation(s)
- Riza Serttas
- Department of Medical Biology, School of Medicine, Trakya University, Balkan Campus, Edirne, Turkey
| | - Cagla Koroglu
- Department of Medical Biology, School of Medicine, Trakya University, Balkan Campus, Edirne, Turkey
| | - Suat Erdogan
- Department of Medical Biology, School of Medicine, Trakya University, Balkan Campus, Edirne, Turkey
| |
Collapse
|
19
|
Cardoso HJ, Carvalho TMA, Fonseca LRS, Figueira MI, Vaz CV, Socorro S. Revisiting prostate cancer metabolism: From metabolites to disease and therapy. Med Res Rev 2020; 41:1499-1538. [PMID: 33274768 DOI: 10.1002/med.21766] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 10/24/2020] [Accepted: 11/22/2020] [Indexed: 12/24/2022]
Abstract
Prostate cancer (PCa), one of the most commonly diagnosed cancers worldwide, still presents important unmet clinical needs concerning treatment. In the last years, the metabolic reprogramming and the specificities of tumor cells emerged as an exciting field for cancer therapy. The unique features of PCa cells metabolism, and the activation of specific metabolic pathways, propelled the use of metabolic inhibitors for treatment. The present work revises the knowledge of PCa metabolism and the metabolic alterations that underlie the development and progression of the disease. A focus is given to the role of bioenergetic sources, namely, glucose, lipids, and glutamine sustaining PCa cell survival and growth. Moreover, it is described as the action of oncogenes/tumor suppressors and sex steroid hormones in the metabolic reprogramming of PCa. Finally, the status of PCa treatment based on the inhibition of metabolic pathways is presented. Globally, this review updates the landscape of PCa metabolism, highlighting the critical metabolic alterations that could have a clinical and therapeutic interest.
Collapse
Affiliation(s)
- Henrique J Cardoso
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Tiago M A Carvalho
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Lara R S Fonseca
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Marília I Figueira
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Cátia V Vaz
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Sílvia Socorro
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| |
Collapse
|
20
|
Ince T, Serttas R, Demir B, Atabey H, Seferoglu N, Erdogan S, Sahin E, Erat S, Nural Y. Polysubstituted pyrrolidines linked to 1,2,3-triazoles: Synthesis, crystal structure, DFT studies, acid dissociation constant, drug-likeness, and anti-proliferative activity. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2020.128400] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
21
|
Beken B, Serttas R, Yazicioglu M, Turkekul K, Erdogan S. Quercetin Improves Inflammation, Oxidative Stress, and Impaired Wound Healing in Atopic Dermatitis Model of Human Keratinocytes. PEDIATRIC ALLERGY IMMUNOLOGY AND PULMONOLOGY 2020; 33:69-79. [PMID: 34678092 DOI: 10.1089/ped.2019.1137] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background: Atopic dermatitis (AD) is a common inflammatory skin disease with complex pathogenesis. Natural flavonoids exhibit strong anti-inflammatory and antioxidant properties in many human diseases. In this study, the potential bioactive effect of quercetin, a polyphenolic plant-derived flavonoid, on the AD model of human keratinocytes was evaluated. Methods: Immortalized human HaCaT keratinocytes were treated with interleukin (IL) -4, -13, and tumor necrosis factor-α to mimic AD features in vitro. Then effects of quercetin on inflammation, oxidative stress, and wound healing were assessed. Results: Pretreatment of the cells with 1.5 μM of quercetin significantly reduced the expression of AD-induced IL-1β, IL-6, IL-8, and thymic stromal lymphopoietin, while it strongly enhanced the expression of superoxide dismutase-1 (SOD1), SOD2, catalase, glutathione peroxidase, and IL-10. Quercetin promoted wound healing by inducing epithelial-mesenchymal transition, which was supported by the upregulation of Twist and Snail mRNA expression. Unexpectedly, quercetin pretreatment of AD-induced cells upregulated the mRNA expression of occludin and E-cadherin, while downregulating matrix metalloproteinase 1 (MMP1), MMP2, and MMP9 expression. The pretreatment inhibited AD-induced phosphorylation of extracellular signal-regulated kinase 1/2/mitogen-activated protein kinase (ERK1/2 MAPK) and the expression of nuclear factor-kappa B (NF-κB), but it did not alter signal transducer and activator of transcription 6 (STAT6) phosphorylation. Conclusion: Quercetin may serve as a potential bioactive substance for atopic dermatitis-related symptoms through anti-inflammatory and antioxidant activities along with its acceleration of wound healing via ERK1/2 MAPK and NF-κB pathways.
Collapse
Affiliation(s)
- Burcin Beken
- Department of Pediatric Allergy and Immunology, School of Medicine, Trakya University, Edirne, Turkey
| | - Riza Serttas
- Department of Medical Biology, School of Medicine, Trakya University, Edirne, Turkey
| | - Mehtap Yazicioglu
- Department of Pediatric Allergy and Immunology, School of Medicine, Trakya University, Edirne, Turkey
| | - Kader Turkekul
- Department of Medical Biology, School of Medicine, Trakya University, Edirne, Turkey
| | - Suat Erdogan
- Department of Medical Biology, School of Medicine, Trakya University, Edirne, Turkey
| |
Collapse
|
22
|
Erdogan S, Turkekul K. Neferine inhibits proliferation and migration of human prostate cancer stem cells through p38 MAPK/JNK activation. J Food Biochem 2020; 44:e13253. [PMID: 32394497 DOI: 10.1111/jfbc.13253] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 03/04/2020] [Accepted: 04/04/2020] [Indexed: 12/16/2022]
Abstract
Cancer stem cells (CSCs) are one of the significant causes of cancer treatment failure and metastasis, as they have significant chemo-and radio-resistance leading to tumor recurrence. Here we investigated the possible anticancer properties of neferine, a natural alkaloid, on human prostate cancer (PCa) cells and their stem cells. CD44+ CSCs were isolated from androgen-insensitive PC3 cells by magnetic-activated cell sorting system (MACS). Neferine dose-and time-dependently inhibited the viability of PC3 and CSCs as well as androgen-sensitive LNCaP cells through inducing apoptosis and cell cycle arrest at G1 phase. Neferine was shown to downregulate the expression of Bcl-2 and CDK4, and upregulate caspase 3, clePARP, p21, p27, and p53. The treatment significantly inhibits the migration of CSCs. Neferine induces JNK and p38 MAPK phosphorylation, and downregulates PI3K and NF-ĸβ signaling. In conclusion, neferine may have a therapeutic effect inhibiting the PCa cell proliferation as well as by eliminating CSCs. PRACTICAL APPLICATIONS: Neferine is an alkaloid found in the seed embryo of Nelumbo nucifera and has recently been shown to have anticancer effects on various human cancer cells. More than 90% of cancer-related deaths develop after metastasis, and CSCs are considered to be largely responsible for the cell migration and invasion. It has been shown that treatment of neferine kills not only PCa cells but also CSCs, and may contribute to the prevention of progression of PCa and metastasis by inhibiting cell proliferation and migration.
Collapse
Affiliation(s)
- Suat Erdogan
- Department of Medical Biology, School of Medicine, Trakya University, Edirne, Turkey
| | - Kader Turkekul
- Department of Medical Biology, School of Medicine, Trakya University, Edirne, Turkey
| |
Collapse
|
23
|
Aghajani M, Mokhtarzadeh A, Aghebati-Maleki L, Mansoori B, Mohammadi A, Safaei S, Asadzadeh Z, Hajiasgharzadeh K, Khaze Shahgoli V, Baradaran B. CD133 suppression increases the sensitivity of prostate cancer cells to paclitaxel. Mol Biol Rep 2020; 47:3691-3703. [PMID: 32246247 DOI: 10.1007/s11033-020-05411-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/26/2020] [Indexed: 12/18/2022]
Abstract
One of the major barriers in cancer therapy is the resistance to conventional therapies and cancer stem cells (CSCs) are among the main causes of this problem. CD133 as a CSC marker displays stem cell-like properties, tumorigenic capacity, and drug resistance in various cancers. However, the molecular mechanism behind CD133 function in prostate cancer (PC) still remains unclear. This research aimed to illustrate the probabilistic mechanism of CD133-siRNA and paclitaxel in the reduction of chemoresistance in PC cells. To measure the cell viability, migratory capacity, CSCs properties, invasive potential, apoptosis and cell cycle progression of the cells, the MTT, wound healing, spheroid assay, colony formation assay, DAPI staining and flow cytometry assays were applied in the LNCaP cell line, respectively. Also, quantitative real-time PCR (qRT-PCR) and western blot method were used for measuring the expression of CD133 and the effects of CD133 silencing on the AKT/mTOR/c-myc axis and pro-metastatic genes expression. We showed that the CD133-siRNA considerably decreased the CD133 expression. Moreover, CD133-siRNA and paclitaxel treatment significantly decreased cell proliferation and also inhibited the ability of cell migration and invasion and reduced pro-metastatic genes expression. Additionally, we found that the simultaneous use of CD133-siRNA and paclitaxel increased the paclitaxel-induced apoptosis. Our results confirmed that CD133 silencing combined with paclitaxel synergistically could suppress cell migration, invasion, and proliferation and enhance the chemosensitivity compared with mono treatment. Therefore, CD133 silencing therapy could be viewed as a promising and efficient strategy in PC targeted therapies.
Collapse
Affiliation(s)
- Marjan Aghajani
- Immunology Research Center, Tabriz University of Medical Sciences, Daneshghah Ave, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Daneshghah Ave, Tabriz, Iran
| | - Leili Aghebati-Maleki
- Immunology Research Center, Tabriz University of Medical Sciences, Daneshghah Ave, Tabriz, Iran
| | - Behzad Mansoori
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Ali Mohammadi
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Sahar Safaei
- Immunology Research Center, Tabriz University of Medical Sciences, Daneshghah Ave, Tabriz, Iran
| | - Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Daneshghah Ave, Tabriz, Iran
| | - Khalil Hajiasgharzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Daneshghah Ave, Tabriz, Iran
| | - Vahid Khaze Shahgoli
- Immunology Research Center, Tabriz University of Medical Sciences, Daneshghah Ave, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Daneshghah Ave, Tabriz, Iran.
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
24
|
Ozal SA, Gurlu V, Turkekul K, Guclu H, Erdogan S. Neferine inhibits epidermal growth factor-induced proliferation and migration of retinal pigment epithelial cells through downregulating p38 MAPK and PI3K/AKT signalling. Cutan Ocul Toxicol 2020; 39:97-105. [DOI: 10.1080/15569527.2020.1730882] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Sadik Altan Ozal
- Department of Ophthalmology, School of Medicine, Trakya University, Edirne, Turkey
| | - Vuslat Gurlu
- Department of Ophthalmology, School of Medicine, Trakya University, Edirne, Turkey
| | - Kader Turkekul
- Department of Medical Biology, School of Medicine, Trakya University, Edirne, Turkey
| | - Hande Guclu
- Department of Ophthalmology, School of Medicine, Trakya University, Edirne, Turkey
| | - Suat Erdogan
- Department of Medical Biology, Trakya University, Edirne, Turkey
| |
Collapse
|
25
|
Fontana F, Raimondi M, Marzagalli M, Di Domizio A, Limonta P. Natural Compounds in Prostate Cancer Prevention and Treatment: Mechanisms of Action and Molecular Targets. Cells 2020; 9:cells9020460. [PMID: 32085497 PMCID: PMC7072821 DOI: 10.3390/cells9020460] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/10/2020] [Accepted: 02/15/2020] [Indexed: 02/06/2023] Open
Abstract
Prostate cancer (PCa) represents a major cause of cancer mortality among men in developed countries. Patients with recurrent disease initially respond to androgen-deprivation therapy, but the tumor eventually progresses into castration-resistant PCa; in this condition, tumor cells acquire the ability to escape cell death and develop resistance to current therapies. Thus, new therapeutic approaches for PCa management are urgently needed. In this setting, natural products have been extensively studied for their anti-PCa activities, such as tumor growth suppression, cell death induction, and inhibition of metastasis and angiogenesis. Additionally, numerous studies have shown that phytochemicals can specifically target the androgen receptor (AR) signaling, as well as the PCa stem cells (PCSCs). Interestingly, many clinical trials have been conducted to test the efficacy of nutraceuticals in human subjects, and they have partially confirmed the promising results obtained in vitro and in preclinical models. This article summarizes the anti-cancer mechanisms and therapeutic potentials of different natural compounds in the context of PCa prevention and treatment.
Collapse
Affiliation(s)
- Fabrizio Fontana
- Department of Pharmacological and Biomolecular Sciences, University of Milano, 20133 Milano, Italy; (F.F.); (M.R.); (M.M.); (A.D.D.)
| | - Michela Raimondi
- Department of Pharmacological and Biomolecular Sciences, University of Milano, 20133 Milano, Italy; (F.F.); (M.R.); (M.M.); (A.D.D.)
| | - Monica Marzagalli
- Department of Pharmacological and Biomolecular Sciences, University of Milano, 20133 Milano, Italy; (F.F.); (M.R.); (M.M.); (A.D.D.)
| | - Alessandro Di Domizio
- Department of Pharmacological and Biomolecular Sciences, University of Milano, 20133 Milano, Italy; (F.F.); (M.R.); (M.M.); (A.D.D.)
- SPILLOproject, 20037 Paderno Dugnano, Italy
| | - Patrizia Limonta
- Department of Pharmacological and Biomolecular Sciences, University of Milano, 20133 Milano, Italy; (F.F.); (M.R.); (M.M.); (A.D.D.)
- Correspondence: ; Tel.: +39-0250318213
| |
Collapse
|
26
|
Imran M, Aslam Gondal T, Atif M, Shahbaz M, Batool Qaisarani T, Hanif Mughal M, Salehi B, Martorell M, Sharifi-Rad J. Apigenin as an anticancer agent. Phytother Res 2020; 34:1812-1828. [PMID: 32059077 DOI: 10.1002/ptr.6647] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/03/2020] [Accepted: 01/31/2020] [Indexed: 12/26/2022]
Abstract
Apigenin is an edible plant-derived flavonoid that has been reported as an anticancer agent in several experimental and biological studies. It exhibits cell growth arrest and apoptosis in different types of tumors such as breast, lung, liver, skin, blood, colon, prostate, pancreatic, cervical, oral, and stomach, by modulating several signaling pathways. Apigenin induces apoptosis by the activation of extrinsic caspase-dependent pathway by upregulating the mRNA expressions of caspase-3, caspase-8, and TNF-α. It induces intrinsic apoptosis pathway as evidenced by the induction of cytochrome c, Bax, and caspase-3, while caspase-8, TNF-α, and B-cell lymphoma 2 levels remained unchanged in human prostate cancer PC-3 cells. Apigenin treatment leads to significant downregulation of matrix metallopeptidases-2, -9, Snail, and Slug, suppressing invasion. The expressions of NF-κB p105/p50, PI3K, Akt, and the phosphorylation of p-Akt decreases after treatment with apigenin. However, apigenin-mediated treatment significantly reduces pluripotency marker Oct3/4 protein expression which might be associated with the downregulation of PI3K/Akt/NF-κB signaling.
Collapse
Affiliation(s)
- Muhammad Imran
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health Sciences, The University of Lahore-Lahore, Lahore, Pakistan
| | - Tanweer Aslam Gondal
- School of Exercise and Nutrition, Deakin University, Melbourne, Victoria, Australia
| | - Muhammad Atif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Muhammad Shahbaz
- Department of Food Science and Technology, MNS-University of Agriculture Multan, Multan, Pakistan
| | - Tahira Batool Qaisarani
- Department of Agricultural Engineering and Technology, Ghazi University, Dera Ghazi Khan, Pakistan
| | - Muhammad Hanif Mughal
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health Sciences, The University of Lahore-Lahore, Lahore, Pakistan
| | - Bahare Salehi
- Student Research Committee, School of Medicine, Bam University of Medical Sciences, Bam, Iran
| | - Miquel Martorell
- Department of Nutrition and Dietetics, Faculty of Pharmacy, University of Concepción, Concepción, Chile.,Unidad de Desarrollo Tecnológico, UDT, Universidad de Concepción, Concepción, Chile
| | - Javad Sharifi-Rad
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
27
|
Li Q, Zhuang C, Wang D, Zhang W, Jia R, Sun F, Zhang Y, Du Y. Construction of trisubstituted chromone skeletons carrying electron-withdrawing groups via PhIO-mediated dehydrogenation and its application to the synthesis of frutinone A. Beilstein J Org Chem 2020; 15:2958-2965. [PMID: 31921367 PMCID: PMC6941426 DOI: 10.3762/bjoc.15.291] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 11/27/2019] [Indexed: 12/20/2022] Open
Abstract
The construction of the biologically interesting chromone skeleton was realized by PhIO-mediated dehydrogenation of chromanones under mild conditions. Interestingly, this method also found application in the synthesis of the naturally occurring frutinone A.
Collapse
Affiliation(s)
- Qiao Li
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Chen Zhuang
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Donghua Wang
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Wei Zhang
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Rongxuan Jia
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Fengxia Sun
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology; Hebei Research Center of Pharmaceutical and Chemical Engineering, Shijiazhuang 050018, China
| | - Yilin Zhang
- C. Eugene Bennett Department of Chemistry, West Virginia University, Morgantown, WV 26506-6045, United States
| | - Yunfei Du
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| |
Collapse
|