1
|
Cheng Y, Zhang W, Sun Q, Wang X, Shang Q, Liu J, Zhang Y, Liu R, Sun C. Probing the biological efficacy and mechanistic pathways of natural compounds in breast cancer therapy via the Hedgehog signaling pathway. J Pharm Anal 2025; 15:101143. [PMID: 40291019 PMCID: PMC12023894 DOI: 10.1016/j.jpha.2024.101143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 10/25/2024] [Accepted: 11/05/2024] [Indexed: 04/30/2025] Open
Abstract
Breast cancer (BC) is one of the most prevalent malignant tumors affecting women worldwide, with its incidence rate continuously increasing. As a result, treatment strategies for this disease have received considerable attention. Research has highlighted the crucial role of the Hedgehog (Hh) signaling pathway in the initiation and progression of BC, particularly in promoting tumor growth and metastasis. Therefore, molecular targets within this pathway represent promising opportunities for the development of novel BC therapies. This study aims to elucidate the therapeutic mechanisms by which natural compounds modulate the Hh signaling pathway in BC. By conducting a comprehensive review of various natural compounds, including polyphenols, terpenes, and alkaloids, we reveal both common and unique regulatory mechanisms that influence this pathway. This investigation represents the first comprehensive analysis of five distinct mechanisms through which natural compounds modulate key molecules within the Hh pathway and their impact on the aggressive behaviors of BC. Furthermore, by exploring the structure-activity relationships between these compounds and their molecular targets, we shed light on the specific structural features that enable natural compounds to interact with various components of the Hh pathway. These novel insights contribute to advancing the development and clinical application of natural compound-based therapeutics. Our thorough review not only lays the groundwork for exploring innovative BC treatments but also opens new avenues for leveraging natural compounds in cancer therapy.
Collapse
Affiliation(s)
- Yining Cheng
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Wenfeng Zhang
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Qi Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Xue Wang
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Qihang Shang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Jingyang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, and Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, 999078, China
| | - Yubao Zhang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Ruijuan Liu
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong, 261000, China
| | - Changgang Sun
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong, 261000, China
| |
Collapse
|
2
|
Malla R, Jyosthsna K, Rani G, Purnachandra Nagaraju G. CD44/PD-L1-mediated networks in drug resistance and immune evasion of breast cancer stem cells: Promising targets of natural compounds. Int Immunopharmacol 2024; 138:112613. [PMID: 38959542 DOI: 10.1016/j.intimp.2024.112613] [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: 01/29/2024] [Revised: 06/28/2024] [Accepted: 06/30/2024] [Indexed: 07/05/2024]
Abstract
Cancer stem cells (CSCs) significantly interfere with immunotherapy, leading to challenges such as low response rates and acquired resistance. PD-L1 expression is associated with the CSC population's overexpression of CD44. Mounting evidence suggests that the breast cancer stem cell (BCSC) marker CD44 and the immune checkpoint PD-L1 contribute to treatment failure through their networks. Natural compounds can overcome therapy resistance in breast cancer by targeting mechanisms underlying resistance in BCSCs. This review provides an updated insight into the CD44 and PD-L1 networks of BCSCs in mediating metastasis and immune evasion. The review critically examines existing literature, providing a comprehensive understanding of the topic and emphasizing the impact of natural flavones on the signaling pathways of BCSCs. Additionally, the review discusses the potential of natural compounds in targeting CD44 and PD-L1 in breast cancer (BC). Natural compounds consistently show potential in targeting regulatory mechanisms of BCSCs, inducing loss of stemness, and promoting differentiation. They offer a promising approach for developing alternative therapeutic strategies to manage breast cancer.
Collapse
Affiliation(s)
- RamaRao Malla
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, Andhra Pradesh, India; Department of Biochemistry and Bioinformatics, School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, Andhra Pradesh, India.
| | - Kattula Jyosthsna
- Department of Biotechnology, School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, Andhra Pradesh, India
| | - G Rani
- Department of Biotechnology, School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, Andhra Pradesh, India
| | - Ganji Purnachandra Nagaraju
- Department of Hematology and Oncology, Heersink School of Medicine, University of Alabama, Birmingham, AL 35233, USA
| |
Collapse
|
3
|
Bel’skaya LV, Dyachenko EI. Oxidative Stress in Breast Cancer: A Biochemical Map of Reactive Oxygen Species Production. Curr Issues Mol Biol 2024; 46:4646-4687. [PMID: 38785550 PMCID: PMC11120394 DOI: 10.3390/cimb46050282] [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: 04/05/2024] [Revised: 05/08/2024] [Accepted: 05/11/2024] [Indexed: 05/25/2024] Open
Abstract
This review systematizes information about the metabolic features of breast cancer directly related to oxidative stress. It has been shown those redox changes occur at all levels and affect many regulatory systems in the human body. The features of the biochemical processes occurring in breast cancer are described, ranging from nonspecific, at first glance, and strictly biochemical to hormone-induced reactions, genetic and epigenetic regulation, which allows for a broader and deeper understanding of the principles of oncogenesis, as well as maintaining the viability of cancer cells in the mammary gland. Specific pathways of the activation of oxidative stress have been studied as a response to the overproduction of stress hormones and estrogens, and specific ways to reduce its negative impact have been described. The diversity of participants that trigger redox reactions from different sides is considered more fully: glycolytic activity in breast cancer, and the nature of consumption of amino acids and metals. The role of metals in oxidative stress is discussed in detail. They can act as both co-factors and direct participants in oxidative stress, since they are either a trigger mechanism for lipid peroxidation or capable of activating signaling pathways that affect tumorigenesis. Special attention has been paid to the genetic and epigenetic regulation of breast tumors. A complex cascade of mechanisms of epigenetic regulation is explained, which made it possible to reconsider the existing opinion about the triggers and pathways for launching the oncological process, the survival of cancer cells and their ability to localize.
Collapse
Affiliation(s)
- Lyudmila V. Bel’skaya
- Biochemistry Research Laboratory, Omsk State Pedagogical University, 644099 Omsk, Russia;
| | | |
Collapse
|
4
|
Zhou X, Liu X, Wan X, Xu M, Wang R, Yang D, Peng M, Jin T, Tang R, Liu M, Hou Y. Oxidized ATM governs stemness of breast cancer stem cell through regulating ubiquitylation and acetylation switch. Biochem Biophys Res Commun 2024; 691:149243. [PMID: 38016338 DOI: 10.1016/j.bbrc.2023.149243] [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: 08/21/2023] [Revised: 11/05/2023] [Accepted: 11/09/2023] [Indexed: 11/30/2023]
Abstract
Cancer stem cells (CSCs), as parts of tumor initiation cells, play a crucial role to tumorigenesis, development and recurrence. However, the complicated mechanisms of CSCs to adapt to tumor microenvironment and its stemness maintenance remains unclear. Here, we show that oxidized ATM, a hypoxia-activated cytoplasm ATM, acts a novel function to maintain CSC stemness in triple-negative breast cancer cells (BCSCs) via regulating histone H4 acetylation. Mechanistically, oxidized ATM phosphorylates TRIM21 (a E3 ubiquitin ligase) serine 80 and serine 469. Serine 80 phosphorylation of TRIM21 is essential for the ubiquitination activity of TRIM21. TRIM21 binds with SIRT1 (one of deacetylase), resulting in ubiquitylation-mediated degradation of SIRT1. The reduced SIRT1 leads to increase of histone H4 acetylation, thus facilitating CSC-related gene expression. Clinical data verify that high level of ATM in breast tumors is positively correlated with malignant grade, and is closely related with low SIRT1, high p-TRIM21, and high CD44 expression. In conclusion, our study provides a novel mechanism by which oxidized ATM governing BCSCs stemness and reveals an important link among oxidized ATM, histone acetylation, and BCSCs maintenance.
Collapse
Affiliation(s)
- Xinyue Zhou
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Xiaoqi Liu
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Xueying Wan
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Ming Xu
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Rui Wang
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Dan Yang
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Meixi Peng
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Ting Jin
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Rui Tang
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Manran Liu
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Yixuan Hou
- Experimental Teaching Center of Basic Medicine Science, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
5
|
He H, Wang S, Zhang W, Gao S, Guan H, Zhou P. Downregulation of TAB182 promotes cancer stem-like cell properties and therapeutic resistance in triple-negative breast cancer cells. BMC Cancer 2023; 23:1101. [PMID: 37953246 PMCID: PMC10642046 DOI: 10.1186/s12885-023-11552-4] [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: 05/01/2023] [Accepted: 10/20/2023] [Indexed: 11/14/2023] Open
Abstract
TAB182 participates in DNA damage repair and radio-/chemosensitivity regulation in various tumors, but its role in tumorigenesis and therapeutic resistance in breast cancer remains unclear. In the current paper, we observed that triple-negative Breast Cancer (TNBC), a highly aggressive type of breast cancer, exhibits a lower expression of TAB182. TAB182 knockdown stimulates the proliferation, migration, and invasion of TNBC cells. Our study first obtained RNA-seq data to explore the cellular functions mediated by TAB182 at the genome level in TNBC cells. A transcriptome analysis and in vitro experiments enabled us to identify that TAB182 downregulation drives the enhanced properties of cancer stem-like cells (CSCs) in TNBC cells. Furthermore, TAB182 deletion contributes to the resistance of cells to olaparib or cisplatin, which can be rescued by silencing GLI2, a gene downstream of cancer stemness-related signaling pathways. Our results reveal a novel function of TAB182 as a potential negative regulator of cancer stem-like properties and drug sensitivity in TNBC cells, suggesting that TAB182 may be a tumor suppressor gene and is associated with increased therapeutic benefits for TNBC patients.
Collapse
Affiliation(s)
- Huan He
- Department of Radiation Toxicology and Oncology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, People's Republic of China
| | - Shaozheng Wang
- Department of Radiation Toxicology and Oncology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China
| | - Wen Zhang
- Department of Radiation Toxicology and Oncology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China
| | - Shanshan Gao
- Department of Radiation Toxicology and Oncology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China
| | - Hua Guan
- Department of Radiation Toxicology and Oncology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China.
| | - Pingkun Zhou
- Department of Radiation Toxicology and Oncology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China.
| |
Collapse
|
6
|
Ordaz-Ramos A, Tellez-Jimenez O, Vazquez-Santillan K. Signaling pathways governing the maintenance of breast cancer stem cells and their therapeutic implications. Front Cell Dev Biol 2023; 11:1221175. [PMID: 37492224 PMCID: PMC10363614 DOI: 10.3389/fcell.2023.1221175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 06/28/2023] [Indexed: 07/27/2023] Open
Abstract
Breast cancer stem cells (BCSCs) represent a distinct subpopulation of cells with the ability to self-renewal and differentiate into phenotypically diverse tumor cells. The involvement of CSC in treatment resistance and cancer recurrence has been well established. Numerous studies have provided compelling evidence that the self-renewal ability of cancer stem cells is tightly regulated by specific signaling pathways, which exert critical roles to maintain an undifferentiated phenotype and prevent the differentiation of CSCs. Signaling pathways such as Wnt/β-catenin, NF-κB, Notch, Hedgehog, TGF-β, and Hippo have been implicated in the promotion of self-renewal of many normal and cancer stem cells. Given the pivotal role of BCSCs in driving breast cancer aggressiveness, targeting self-renewal signaling pathways holds promise as a viable therapeutic strategy for combating this disease. In this review, we will discuss the main signaling pathways involved in the maintenance of the self-renewal ability of BCSC, while also highlighting current strategies employed to disrupt the signaling molecules associated with stemness.
Collapse
Affiliation(s)
- Alejandro Ordaz-Ramos
- Innovation in Precision Medicine Laboratory, Instituto Nacional de Medicina Genómica, Mexico City, México
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Circuito de Posgrados, Ciudad Universitaria, Coyoacán, México
| | - Olivia Tellez-Jimenez
- Innovation in Precision Medicine Laboratory, Instituto Nacional de Medicina Genómica, Mexico City, México
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Circuito de Posgrados, Ciudad Universitaria, Coyoacán, México
| | - Karla Vazquez-Santillan
- Innovation in Precision Medicine Laboratory, Instituto Nacional de Medicina Genómica, Mexico City, México
| |
Collapse
|
7
|
Lučić I, Kurtović M, Mlinarić M, Piteša N, Čipak Gašparović A, Sabol M, Milković L. Deciphering Common Traits of Breast and Ovarian Cancer Stem Cells and Possible Therapeutic Approaches. Int J Mol Sci 2023; 24:10683. [PMID: 37445860 DOI: 10.3390/ijms241310683] [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: 05/06/2023] [Revised: 06/21/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
Breast cancer (BC) and ovarian cancer (OC) are among the most common and deadly cancers affecting women worldwide. Both are complex diseases with marked heterogeneity. Despite the induction of screening programs that increase the frequency of earlier diagnosis of BC, at a stage when the cancer is more likely to respond to therapy, which does not exist for OC, more than 50% of both cancers are diagnosed at an advanced stage. Initial therapy can put the cancer into remission. However, recurrences occur frequently in both BC and OC, which are highly cancer-subtype dependent. Therapy resistance is mainly attributed to a rare subpopulation of cells, named cancer stem cells (CSC) or tumor-initiating cells, as they are capable of self-renewal, tumor initiation, and regrowth of tumor bulk. In this review, we will discuss the distinctive markers and signaling pathways that characterize CSC, their interactions with the tumor microenvironment, and the strategies they employ to evade immune surveillance. Our focus will be on identifying the common features of breast cancer stem cells (BCSC) and ovarian cancer stem cells (OCSC) and suggesting potential therapeutic approaches.
Collapse
Affiliation(s)
- Ivan Lučić
- Laboratory for Oxidative Stress, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Matea Kurtović
- Laboratory for Hereditary Cancer, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Monika Mlinarić
- Laboratory for Oxidative Stress, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Nikolina Piteša
- Laboratory for Hereditary Cancer, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Ana Čipak Gašparović
- Laboratory for Oxidative Stress, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Maja Sabol
- Laboratory for Hereditary Cancer, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Lidija Milković
- Laboratory for Oxidative Stress, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| |
Collapse
|
8
|
Re-Sensitizing Cancer Stem Cells to Conventional Chemotherapy Agents. Int J Mol Sci 2023; 24:ijms24032122. [PMID: 36768445 PMCID: PMC9917165 DOI: 10.3390/ijms24032122] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/26/2022] [Accepted: 01/04/2023] [Indexed: 01/25/2023] Open
Abstract
Cancer stem cells are found in many cancer types. They comprise a distinct subpopulation of cells within the tumor that exhibit properties of stem cells. They express a number of cell surface markers, such as CD133, CD44, ALDH, and EpCAM, as well as embryonic transcription factors Oct4, Nanog, and SOX2. CSCs are more resistant to conventional chemotherapy and can potentially drive tumor relapse. Therefore, it is essential to understand the molecular mechanisms that drive chemoresistance and to target them with specific therapy effectively. Highly conserved developmental signaling pathways such as Wnt, Hedgehog, and Notch are commonly reported to play a role in CSCs chemoresistance development. Studies show that particular pathway inhibitors combined with conventional therapy may re-establish sensitivity to the conventional therapy. Another significant contributor of chemoresistance is a specific tumor microenvironment. Surrounding stroma in the form of cancer-associated fibroblasts, macrophages, endothelial cells, and extracellular matrix components produce cytokines and other factors, thus creating a favorable environment and decreasing the cytotoxic effects of chemotherapy. Anti-stromal agents may potentially help to overcome these effects. Epigenetic changes and autophagy were also among the commonly reported mechanisms of chemoresistance. This review provides an overview of signaling pathway components involved in the development of chemoresistance of CSCs and gathers evidence from experimental studies in which CSCs can be re-sensitized to conventional chemotherapy agents across different cancer types.
Collapse
|
9
|
Hussen BM, Kheder RK, Abdullah ST, Hidayat HJ, Rahman HS, Salihi A, Taheri M, Ghafouri-Fard S. Functional interplay between long non-coding RNAs and Breast CSCs. Cancer Cell Int 2022; 22:233. [PMID: 35864503 PMCID: PMC9306174 DOI: 10.1186/s12935-022-02653-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 07/12/2022] [Indexed: 12/14/2022] Open
Abstract
Breast cancer (BC) represents aggressive cancer affecting most women’s lives globally. Metastasis and recurrence are the two most common factors in a breast cancer patient's poor prognosis. Cancer stem cells (CSCs) are tumor cells that are able to self-renew and differentiate, which is a significant factor in metastasis and recurrence of cancer. Long non-coding RNAs (lncRNAs) describe a group of RNAs that are longer than 200 nucleotides and do not have the ability to code for proteins. Some of these lncRNAs can be mainly produced in various tissues and tumor forms. In the development and spread of malignancies, lncRNAs have a significant role in influencing multiple signaling pathways positively or negatively, making them promise useful diagnostic and prognostic markers in treating the disease and guiding clinical therapy. However, it is not well known how the interaction of lncRNAs with CSCs will affect cancer development and progression. Here, in this review, we attempt to summarize recent findings that focus on lncRNAs affect cancer stem cell self-renewal and differentiation in breast cancer development and progression, as well as the strategies and challenges for overcoming lncRNA's therapeutic resistance.
Collapse
Affiliation(s)
- Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Erbil , Kurdistan Region, Iraq.,Center of Research and Strategic Studies, Lebanese French University, Erbil, Iraq
| | - Ramiar Kamal Kheder
- Department of Medical Analysis, Faculty of Science, Tishk International University, Erbil, Iraq.,Medical Laboratory Science, College of Science, University of Raparin, Rania, KGR, Iraq
| | - Sara Tharwat Abdullah
- Department of Pharmacology and Toxicology, College of Pharmacy, Hawler Medical University, Erbil, Iraq
| | - Hazha Jamal Hidayat
- Department of Biology, College of Education, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Heshu Sulaiman Rahman
- Department of Physiology, College of Medicine, University of Sulaimani, Sulaimaniyah, Republic of Iraq.,Department of Medical Laboratory Sciences, Komar University of Science and Technology, Sulaimaniyah, Republic of Iraq
| | - Abbas Salihi
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran. .,Institute of Human Genetics, Jena University Hospital, Jena, Germany.
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Saha T, Lukong KE. Breast Cancer Stem-Like Cells in Drug Resistance: A Review of Mechanisms and Novel Therapeutic Strategies to Overcome Drug Resistance. Front Oncol 2022; 12:856974. [PMID: 35392236 PMCID: PMC8979779 DOI: 10.3389/fonc.2022.856974] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is the most frequent type of malignancy in women worldwide, and drug resistance to the available systemic therapies remains a major challenge. At the molecular level, breast cancer is heterogeneous, where the cancer-initiating stem-like cells (bCSCs) comprise a small yet distinct population of cells within the tumor microenvironment (TME) that can differentiate into cells of multiple lineages, displaying varying degrees of cellular differentiation, enhanced metastatic potential, invasiveness, and resistance to radio- and chemotherapy. Based on the expression of estrogen and progesterone hormone receptors, expression of human epidermal growth factor receptor 2 (HER2), and/or BRCA mutations, the breast cancer molecular subtypes are identified as TNBC, HER2 enriched, luminal A, and luminal B. Management of breast cancer primarily involves resection of the tumor, followed by radiotherapy, and systemic therapies including endocrine therapies for hormone-responsive breast cancers; HER2-targeted therapy for HER2-enriched breast cancers; chemotherapy and poly (ADP-ribose) polymerase inhibitors for TNBC, and the recent development of immunotherapy. However, the complex crosstalk between the malignant cells and stromal cells in the breast TME, rewiring of the many different signaling networks, and bCSC-mediated processes, all contribute to overall drug resistance in breast cancer. However, strategically targeting bCSCs to reverse chemoresistance and increase drug sensitivity is an underexplored stream in breast cancer research. The recent identification of dysregulated miRNAs/ncRNAs/mRNAs signatures in bCSCs and their crosstalk with many cellular signaling pathways has uncovered promising molecular leads to be used as potential therapeutic targets in drug-resistant situations. Moreover, therapies that can induce alternate forms of regulated cell death including ferroptosis, pyroptosis, and immunotherapy; drugs targeting bCSC metabolism; and nanoparticle therapy are the upcoming approaches to target the bCSCs overcome drug resistance. Thus, individualizing treatment strategies will eliminate the minimal residual disease, resulting in better pathological and complete response in drug-resistant scenarios. This review summarizes basic understanding of breast cancer subtypes, concept of bCSCs, molecular basis of drug resistance, dysregulated miRNAs/ncRNAs patterns in bCSCs, and future perspective of developing anticancer therapeutics to address breast cancer drug resistance.
Collapse
Affiliation(s)
- Taniya Saha
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Kiven Erique Lukong
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
11
|
Yan Y, He M, Zhao L, Wu H, Zhao Y, Han L, Wei B, Ye D, Lv X, Wang Y, Yao W, Zhao H, Chen B, Jin Z, Wen J, Zhu Y, Yu T, Jin F, Wei M. A novel HIF-2α targeted inhibitor suppresses hypoxia-induced breast cancer stemness via SOD2-mtROS-PDI/GPR78-UPR ER axis. Cell Death Differ 2022; 29:1769-1789. [PMID: 35301432 PMCID: PMC9433403 DOI: 10.1038/s41418-022-00963-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/06/2022] [Accepted: 02/18/2022] [Indexed: 12/24/2022] Open
Abstract
Hypoxic tumor microenvironment (TME) plays critical roles in induction of cancer stem cell-like phenotype in breast cancer and contribute to chemoresistance. However, the mechanism underlying stemness reprogramming of breast cancer cells (BCs) by hypoxic TME remains largely unknown. In the present study, we illustrated that HIF-2α, but not HIF-1α, induces stemness in BCs under hypoxia through SOD2-mtROS-PDI/GRP78-UPRER pathway, linking mitochondrial metabolic state to endoplasmic reticulum (ER) response via mitochondrial reactive oxygen species (mtROS) level. HIF-2α activates endoplasmic reticulum unfolded protein response (UPRER) in drug-sensitive MCF7 and T47D cells to induce drug-resistant stem-like phenotype. Genetic depletion or pharmacological inhibition (YQ-0629) of HIF-2α abolished hypoxia-induced stem-like phenotype in vitro and in vivo. Mechanistically, HIF-2α activates transcription of superoxide dismutase 2 (SOD2) under hypoxia and thereby decreases mtROS level. With less mtROS transported to endoplasmic reticulum, the expression and activity of protein disulfide isomerase (PDI) is suppressed, allowing glucose-regulated protein 78 (GRP78) to dissociate from receptor proteins of UPRER and bind misfolded protein to activate UPRER, which eventually confer chemoresistance and stem-like properties to BCs. Moreover, the increase in mtROS and PDI levels caused by HIF-2α knockdown and the subsequent UPRER inhibition could be substantially rescued by mitoTEMPOL (a mtROS scavenger), 16F16 (a PDI inhibitor), or GRP78 overexpression. Overall, we reported the critical roles of HIF-2α-SOD2-mtROS-PDI/GRP78-UPRER axis in mediating hypoxia-induced stemness in BCs, highlighting the interaction between organelles and providing evidence for further development of targeted HIF-2α inhibitor as a promising therapeutic strategy for chemoresistant breast cancer.
Collapse
Affiliation(s)
- Yuanyuan Yan
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning Province, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, Liaoning Province, China
| | - Miao He
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning Province, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, Liaoning Province, China
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning Province, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, Liaoning Province, China
| | - Huizhe Wu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning Province, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, Liaoning Province, China
| | - Yanyun Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning Province, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, Liaoning Province, China
| | - Li Han
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning Province, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, Liaoning Province, China
| | - Binbin Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning Province, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, Liaoning Province, China
| | - Dongman Ye
- Department of Medical Imaging, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, China
| | - Xuemei Lv
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning Province, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, Liaoning Province, China
| | - Yan Wang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning Province, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, Liaoning Province, China
| | - Weifan Yao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning Province, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, Liaoning Province, China
| | - Haishan Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning Province, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, Liaoning Province, China
| | - Bo Chen
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province, China
| | - Zining Jin
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province, China
| | - Jian Wen
- Department of Breast Surgery, The Fourth Affiliated Hospital of China Medical University, No.4 Chongshan East Road, Shenyang, Liaoning, China
| | - Yan Zhu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning Province, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, Liaoning Province, China
| | - Tao Yu
- Department of Medical Imaging, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, China.
| | - Feng Jin
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning Province, China.
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning Province, China. .,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, Liaoning Province, China. .,Liaoning Medical Diagnosis and Treatment Center, Shenyang, Liaoning Province, China.
| |
Collapse
|
12
|
Qiu S, Zhou Y, Kim JT, Bao C, Lee HJ, Chen J. Amentoflavone inhibits tumor necrosis factor-α-induced migration and invasion through AKT/mTOR/S6k1/hedgehog signaling in human breast cancer. Food Funct 2021; 12:10196-10209. [PMID: 34542136 DOI: 10.1039/d1fo01085a] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Inflammatory cytokine tumor necrosis factor-α (TNFα) has been demonstrated to accelerate the progression and metastasis of various carcinomas. In this study, we investigated the effect of amentoflavone on inhibiting the migration and invasion of TNFα-induced breast cancer cells. Results showed that amentoflavone significantly blocked the cellular migration and invasion of MCF10DCIS.com and MDA-MB-231 cells at a concentration of 10 μM but did not affect the cell viability. The mRNA and protein levels of matrix metalloproteinase (MMP)-9, significantly activated by TNFα, were reversed by amentoflavone treatment in a dose-dependent manner in MCF10DCIS.com cells. Congruent with the protein level, the activity of MMP-9 was significantly suppressed by amentoflavone treatment. Additionally, we found that amentoflavone dampened Gli1-dependent noncanonical hedgehog signaling, which is a key factor in the regulation of migration and invasion in TNFα-induced human breast cancer cells. Further study elucidated that TNFα enhanced Gli1 through the activation of the AKT/mTOR/S6K1 cascade, whereas it receded after amentoflavone treatment in human breast cancer cells. In summary, amentoflavone abrogated Gli1 activation in TNFα-induced mammary tumor cells, resulting in a decrease of invasiveness in human breast cancer cells via mediating AKT/mTOR/S6K1 signaling. Amentoflavone should be considered as a potent food ingredient for the retardation of mammary tumorigenesis.
Collapse
Affiliation(s)
- Shuai Qiu
- Department of Food Science and Technology, Chung-Ang University, Anseong, 17546, South Korea.
| | - Yimeng Zhou
- Department of Food Science and Technology, Chung-Ang University, Anseong, 17546, South Korea.
| | - Jin Tae Kim
- Department of Food Science and Technology, Chung-Ang University, Anseong, 17546, South Korea.
| | - Cheng Bao
- School of Life Science, Ludong University, Yantai, 264025, China
| | - Hong Jin Lee
- Department of Food Science and Technology, Chung-Ang University, Anseong, 17546, South Korea.
| | - Jing Chen
- Institute for Advanced and Applied Chemical Synthesis, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
13
|
Wang H, Zhang H, Zhu Y, Wu Z, Cui C, Cai F. Anticancer Mechanisms of Salinomycin in Breast Cancer and Its Clinical Applications. Front Oncol 2021; 11:654428. [PMID: 34381705 PMCID: PMC8350729 DOI: 10.3389/fonc.2021.654428] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 06/28/2021] [Indexed: 01/11/2023] Open
Abstract
Breast cancer (BC) is the most frequent cancer among women worldwide and is the leading cause of cancer-related deaths in women. Cancer cells with stem cell-like features and tumor-initiating potential contribute to drug resistance, tumor recurrence, and metastasis. To achieve better clinical outcomes, it is crucial to eradicate both bulk BC cells and breast cancer stem cells (BCSCs). Salinomycin, a monocarboxylic polyether antibiotic isolated from Streptomyces albus, can precisely kill cancer stem cells (CSCs), particularly BCSCs, by various mechanisms, including apoptosis, autophagy, and necrosis. There is increasing evidence that salinomycin can inhibit cell proliferation, invasion, and migration in BC and reverse the immune-inhibitory microenvironment to prevent tumor growth and metastasis. Therefore, salinomycin is a promising therapeutic drug for BC. In this review, we summarize established mechanisms by which salinomycin protects against BC and discuss its future clinical applications.
Collapse
Affiliation(s)
- Hui Wang
- Laboratory of Tumor Molecular Biology, School of Basic Medical Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Hongyi Zhang
- Department of Breast Surgery, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yihao Zhu
- Laboratory of Tumor Molecular Biology, School of Basic Medical Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Zhonghang Wu
- Department of Scientific Research, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China.,Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Chunhong Cui
- Laboratory of Tumor Molecular Biology, School of Basic Medical Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, China.,Department of Scientific Research, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Fengfeng Cai
- Department of Breast Surgery, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
14
|
García-Martínez A, Pérez-Balaguer A, Ortiz-Martínez F, Pomares-Navarro E, Sanmartín E, García-Escolano M, Montoyo-Pujol YG, Castellón-Molla E, Peiró G. Hedgehog gene expression patterns among intrinsic subtypes of breast cancer: Prognostic relevance. Pathol Res Pract 2021; 223:153478. [PMID: 34022683 DOI: 10.1016/j.prp.2021.153478] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/04/2021] [Accepted: 05/07/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE Hedgehog (Hh) signaling is a crucial developmental regulatory pathway recognized as a primary oncogenesis driver in various human cancers. However, its role in breast carcinoma (BC) has been underexplored. METHODS We analyzed the expression of several Hh associated genes in a clinical series and breast cancer cell lines. We included 193 BC stratified according to intrinsic immunophenotypes. Gene expression profiling ofBOC, PTCH, SMO, GLI1, GLI2, and GLI3 was performed by qRT-PCR. Results were correlated with clinical-pathological variables and outcome. RESULTS We observed expression ofGLI2 in triple-negative/basal-like (TN/BL) and GLI3 in luminal cells. In samples, BOC, GLI1, GLI2, and GLI3 expression correlated significantly with luminal tumors and good prognostic factors. In contrast, PTCH and SMO correlated with TN/BL phenotype and nodal involvement. Patients whose tumors expressed SMO had a poorer outcome, especially those with HER2 phenotype. Positive lymph-node status and high SMO remained independent poor prognostic factors. CONCLUSION Our results support a differential Hh pathway activation in BC phenotypes.SMO levels stratified patients at risk of recurrence and death in HER2 phenotype, and it showed an independent prognostic value. Therefore, SMO could be a potential therapeutic target for a subset of BC patients.
Collapse
Affiliation(s)
- Araceli García-Martínez
- Research Unit, University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain.
| | - Ariadna Pérez-Balaguer
- Research Unit, University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain
| | - Fernando Ortiz-Martínez
- Research Unit, University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain
| | - Eloy Pomares-Navarro
- Research Unit, University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain
| | - Elena Sanmartín
- Research Unit, University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain
| | - Marta García-Escolano
- Research Unit, University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain
| | - Yoel G Montoyo-Pujol
- Research Unit, University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain
| | - Elena Castellón-Molla
- Pathology Dept., University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain
| | - Gloria Peiró
- Research Unit, University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain; Pathology Dept., University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain
| |
Collapse
|
15
|
Zhang R, Tu J, Liu S. Novel molecular regulators of breast cancer stem cell plasticity and heterogeneity. Semin Cancer Biol 2021; 82:11-25. [PMID: 33737107 DOI: 10.1016/j.semcancer.2021.03.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 11/19/2020] [Accepted: 03/11/2021] [Indexed: 12/12/2022]
Abstract
Tumors consist of heterogeneous cell populations, and tumor heterogeneity plays key roles in regulating tumorigenesis, metastasis, recurrence and resistance to anti-tumor therapies. More and more studies suggest that cancer stem cells (CSCs) promote tumorigenesis, metastasis, recurrence and drug resistance as well as are the major source for heterogeneity of cancer cells. CD24-CD44+ and ALDH+ are the most common markers for breast cancer stem cells (BCSCs). Previous studies showed that different BCSC markers label different BCSC populations, indicating the heterogeneity of BCSCs. Therefore, defining the regulation mechanisms of heterogeneous BCSCs is essential for precisely targeting BCSCs and treating breast cancer. In this review, we summarized the novel regulators existed in BCSCs and their niches for BCSC heterogeneity which has been discovered in recent years, and discussed their regulation mechanisms and the latest corresponding cancer treatments, which will extend our understanding on BCSC heterogeneity and plasticity, and provide better prognosis prediction and more efficient novel therapeutic strategies for breast cancer.
Collapse
Affiliation(s)
- Rui Zhang
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, Cancer Institutes, Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Juchuanli Tu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, Cancer Institutes, Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Suling Liu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, Cancer Institutes, Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
16
|
Zheng Q, Zhang M, Zhou F, Zhang L, Meng X. The Breast Cancer Stem Cells Traits and Drug Resistance. Front Pharmacol 2021; 11:599965. [PMID: 33584277 PMCID: PMC7876385 DOI: 10.3389/fphar.2020.599965] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 11/17/2020] [Indexed: 12/13/2022] Open
Abstract
Drug resistance is a major challenge in breast cancer (BC) treatment at present. Accumulating studies indicate that breast cancer stem cells (BCSCs) are responsible for the BC drugs resistance, causing relapse and metastasis in BC patients. Thus, BCSCs elimination could reverse drug resistance and improve drug efficacy to benefit BC patients. Consequently, mastering the knowledge on the proliferation, resistance mechanisms, and separation of BCSCs in BC therapy is extremely helpful for BCSCs-targeted therapeutic strategies. Herein, we summarize the principal BCSCs surface markers and signaling pathways, and list the BCSCs-related drug resistance mechanisms in chemotherapy (CT), endocrine therapy (ET), and targeted therapy (TT), and display therapeutic strategies for targeting BCSCs to reverse drug resistance in BC. Even more importantly, more attention should be paid to studies on BCSC-targeted strategies to overcome the drug resistant dilemma of clinical therapies in the future.
Collapse
Affiliation(s)
- Qinghui Zheng
- Department of Breast Surgery, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Mengdi Zhang
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Fangfang Zhou
- Institutes of Biology and Medical Science, Soochow University, Suzhou, China
| | - Long Zhang
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Xuli Meng
- Department of Breast Surgery, Zhejiang Provincial People's Hospital, Hangzhou, China
| |
Collapse
|
17
|
Iriana S, Asha K, Repak M, Sharma-Walia N. Hedgehog Signaling: Implications in Cancers and Viral Infections. Int J Mol Sci 2021; 22:1042. [PMID: 33494284 PMCID: PMC7864517 DOI: 10.3390/ijms22031042] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/11/2021] [Accepted: 01/14/2021] [Indexed: 12/14/2022] Open
Abstract
The hedgehog (SHH) signaling pathway is primarily involved in embryonic gut development, smooth muscle differentiation, cell proliferation, adult tissue homeostasis, tissue repair following injury, and tissue polarity during the development of vertebrate and invertebrate organisms. GLIoma-associated oncogene homolog (GLI) family of zinc-finger transcription factors and smoothened (SMO) are the signal transducers of the SHH pathway. Both SHH ligand-dependent and independent mechanisms activate GLI proteins. Various transcriptional mechanisms, posttranslational modifications (phosphorylation, ubiquitination, proteolytic processing, SUMOylation, and acetylation), and nuclear-cytoplasmic shuttling control the activity of SHH signaling pathway proteins. The dysregulated SHH pathway is associated with bone and soft tissue sarcomas, GLIomas, medulloblastomas, leukemias, and tumors of breast, lung, skin, prostate, brain, gastric, and pancreas. While extensively studied in development and sarcomas, GLI family proteins play an essential role in many host-pathogen interactions, including bacterial and viral infections and their associated cancers. Viruses hijack host GLI family transcription factors and their downstream signaling cascades to enhance the viral gene transcription required for replication and pathogenesis. In this review, we discuss a distinct role(s) of GLI proteins in the process of tumorigenesis and host-pathogen interactions in the context of viral infection-associated malignancies and cancers due to other causes. Here, we emphasize the potential of the Hedgehog (HH) pathway targeting as a potential anti-cancer therapeutic approach, which in the future could also be tested in infection-associated fatalities.
Collapse
|
18
|
Shan NL, Shin Y, Yang G, Furmanski P, Suh N. Breast cancer stem cells: A review of their characteristics and the agents that affect them. Mol Carcinog 2021; 60:73-100. [PMID: 33428807 DOI: 10.1002/mc.23277] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/17/2022]
Abstract
The evolving concept that cancer stem cells (CSCs) are the driving element in cancer development, evolution and heterogeneity, has overridden the previous model of a tumor consisting of cells all with similar sequentially acquired mutations and a similar potential for renewal, invasion and metastasis. This paradigm shift has focused attention on therapeutically targeting CSCs directly as a means of eradicating the disease. In breast cancers, CSCs can be identified by cell surface markers and are characterized by their ability to self-renew and differentiate, resist chemotherapy and radiation, and initiate new tumors upon serial transplantation in xenografted mice. These functional properties of CSCs are regulated by both intracellular and extracellular factors including pluripotency-related transcription factors, intracellular signaling pathways and external stimuli. Several classes of natural products and synthesized compounds have been studied to target these regulatory elements and force CSCs to lose stemness and/or terminally differentiate and thereby achieve a therapeutic effect. However, realization of an effective treatment for breast cancers, focused on the biological effects of these agents on breast CSCs, their functions and signaling, has not yet been achieved. In this review, we delineate the intrinsic and extrinsic factors identified to date that control or promote stemness in breast CSCs and provide a comprehensive compilation of potential agents that have been studied to target breast CSCs, transcription factors and stemness-related signaling. Our aim is to stimulate further study of these agents that could become the basis for their use as stand-alone treatments or components of combination therapies effective against breast cancers.
Collapse
Affiliation(s)
- Naing L Shan
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
| | - Yoosub Shin
- Yonsei University, College of Medicine, Seoul, Republic of Korea
| | - Ge Yang
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
| | - Philip Furmanski
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA.,Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Nanjoo Suh
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA.,Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| |
Collapse
|
19
|
Sun X, Lv X, Yan Y, Zhao Y, Ma R, He M, Wei M. Hypoxia-mediated cancer stem cell resistance and targeted therapy. Biomed Pharmacother 2020; 130:110623. [PMID: 32791395 DOI: 10.1016/j.biopha.2020.110623] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/02/2020] [Accepted: 08/05/2020] [Indexed: 02/07/2023] Open
Abstract
Drug resistance is a major obstacle in the treatment of tumors, which easily lead to relapse or poor prognosis. Cancer stem cells (CSCs) are regarded as one of the important targets that mediate tumor resistance. Increasing evidence shows that the tumor hypoxia microenvironment is closely related to the resistance of CSCs to chemotherapy and radiotherapy. In this review, we intend to review the articles that have described how the hypoxic microenvironment affects CSC stemness and mediates tumor resistance and provide new directions and methods in the clinical treatment of tumors. Here, we also discuss the feasibility and development prospects of using hypoxia-inducible factors (HIFs) that regulate the hypoxic microenvironment of tumors as targeted agents to treat tumors, as well as to reduce or even reverse the resistance of tumors to chemotherapy and radiotherapy.
Collapse
Affiliation(s)
- Xiaoyu Sun
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning Province, China; Liaoning Engineering Technology Research Center for the Research, Development and Industrialization of Innovative Peptide Drugs, China Medical University, Shenyang, Liaoning Province, China.
| | - Xuemei Lv
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning Province, China; Liaoning Engineering Technology Research Center for the Research, Development and Industrialization of Innovative Peptide Drugs, China Medical University, Shenyang, Liaoning Province, China.
| | - Yuanyuan Yan
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning Province, China; Liaoning Engineering Technology Research Center for the Research, Development and Industrialization of Innovative Peptide Drugs, China Medical University, Shenyang, Liaoning Province, China.
| | - Yanyun Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning Province, China; Liaoning Engineering Technology Research Center for the Research, Development and Industrialization of Innovative Peptide Drugs, China Medical University, Shenyang, Liaoning Province, China.
| | - Rong Ma
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning Province, China; Liaoning Engineering Technology Research Center for the Research, Development and Industrialization of Innovative Peptide Drugs, China Medical University, Shenyang, Liaoning Province, China.
| | - Miao He
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning Province, China; Liaoning Engineering Technology Research Center for the Research, Development and Industrialization of Innovative Peptide Drugs, China Medical University, Shenyang, Liaoning Province, China.
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning Province, China; Liaoning Engineering Technology Research Center for the Research, Development and Industrialization of Innovative Peptide Drugs, China Medical University, Shenyang, Liaoning Province, China.
| |
Collapse
|
20
|
Borah A, Pillai SC, Rochani AK, Palaninathan V, Nakajima Y, Maekawa T, Kumar DS. GANT61 and curcumin-loaded PLGA nanoparticles for GLI1 and PI3K/Akt-mediated inhibition in breast adenocarcinoma. NANOTECHNOLOGY 2020; 31:185102. [PMID: 31952056 DOI: 10.1088/1361-6528/ab6d20] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Current conventional mono and combination therapeutic strategies often fail to target breast cancer tissue effectively due to tumor heterogeneity comprising cancer stem cells (CSCs) and bulk tumor cells. This is further associated with drug toxicity and resistivity in the long run. A nanomedicine platform incorporating combination anti-cancer treatment might overcome these challenges and generate synergistic anti-cancer effects and also reduce drug toxicity. GANT61 and curcumin were co-delivered via polymeric nanoparticles (NPs) for the first time to elicit enhanced anti-tumor activity against heterogeneous breast cancer cell line MCF-7. We adopted the single-emulsion-solvent evaporation method for the preparation of the therapeutic NPs. The GANT61-curcumin PLGA NPs were characterized for their size, shape and chemical properties, and anti-cancer cell studies were undertaken for the plausible explanation of our hypothesis. The synthesized GANT61-curcumin PLGA NPs had a spherical, smooth surface morphology, and an average size of 347.4 d. nm. The NPs induced cytotoxic effects in breast cancer cells at a mid-minimal dosage followed by cell death via autophagy and apoptosis, reduction in their target protein expression along with compromising the self-renewal property of CSCs as revealed by their in vitro cell studies. The dual-drug NPs thus provide a novel perspective on aiding existing anti-cancer nanomedicine therapies to target a heterogeneous tumor mass effectively.
Collapse
Affiliation(s)
- Ankita Borah
- Bio-Nano Electronics Research Centre, Graduate School of Interdisciplinary Science, Toyo University, 2100, Kujirai, Kawagoe, Saitama 350-8585, Japan
| | | | | | | | | | | | | |
Collapse
|
21
|
Gambogic acid increases the sensitivity to paclitaxel in drug‑resistant triple‑negative breast cancer via the SHH signaling pathway. Mol Med Rep 2019; 20:4515-4522. [PMID: 31545492 PMCID: PMC6797991 DOI: 10.3892/mmr.2019.10697] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Accepted: 09/06/2019] [Indexed: 12/29/2022] Open
Abstract
Paclitaxel is the most frequently used therapy regimen for triple-negative breast cancer (TNBC). However, chemoresistance frequently occurs, leading to enhanced failure rates of chemotherapy in TNBC; therefore, novel biological therapies are urgently needed. Gambogic acid (GA) has potent anticancer effects and inhibits tumor growth in several types of human cancer. However, the effects of GA on paclitaxel-resistant TNBC remain unknown. In the present study, the Cell Counting Kit-8 assay was used to examine the effect of GA and/or paclitaxel on the viability of TNBC cells; flow cytometry was used to examine the effects of GA on cell apoptosis; and western blotting and reverse transcription-quantitative PCR were used to determine the effects of GA on the expression of sonic hedgehog (SHH) signaling pathway target genes. The present results indicated that GA significantly inhibited the viability and enhanced the rate of apoptosis in paclitaxel-resistant MDA-MB-231 cells via activating the SHH signaling pathway. In vivo experiments confirmed that GA treatment enhanced the sensitivity of MDA-MB-231 cells to paclitaxel via the SHH signaling pathway. In conclusion, the combination of GA with paclitaxel may increase the antitumor effects on paclitaxel-resistant TNBC via downregulating the SHH signaling pathway.
Collapse
|
22
|
Chen Q, Deng X, Hu X, Guan S, He M, Wang Y, Wei B, Zhang J, Zhao H, Yao W, Jin F, Liu Y, Chen J, Olapade OI, Wu H, Wei M. Breast Cancer Risk-Associated SNPs in the mTOR Promoter Form De Novo KLF5- and ZEB1-Binding Sites that Influence the Cellular Response to Paclitaxel. Mol Cancer Res 2019; 17:2244-2256. [PMID: 31467112 DOI: 10.1158/1541-7786.mcr-18-1072] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/03/2019] [Accepted: 08/26/2019] [Indexed: 12/24/2022]
Abstract
ZEB1 (a positive enhancer) and KLF5 (a negative silencer) affect transcription factors and play inherently conserved roles in tumorigenesis and multidrug resistance. In humans, the rs2295080T-allele at the mTOR promoter locus has been associated with human cancer risk; however, the 63 bp spacing of another SNP rs2295079 has not been identified. Here, we discovered, for the first time, that rs2295079 (-78C/G) and rs2295080 (-141G/T) formed linkage haplotypes, with Ht1 (-78C/-141G) and Ht2 (-78G/-141T) being dominant, which were associated with distinct susceptibility to breast cancer, response to paclitaxel, and clinical outcomes in breast cancer. At the cellular level, compared with Ht1, Ht2 exhibits a much stronger effect on promoting mTOR expression, leading to enhanced tumor cell growth and strengthened resistance to PTX treatment. Mechanistically, the -141T allele of Ht2 creates a novel ZEB1-binding site; meanwhile, the -78C allele of Ht1 exists as an emerging KLF5-binding site, which synergistically induces promote/inhibit mTOR expression, cell proliferation, and excretion of cytotoxic drugs through the ZEB1/KLF5-mTOR-CCND1/ABCB1 cascade, thereby affecting the response to paclitaxel treatment in vivo and in vitro. Our results suggest the existence of a ZEB1/KLF5-mTOR-CCND1/ABCB1 axis in human cells that could be involved in paclitaxel response pathways and functionally regulate interindividualized breast cancer susceptibility and prognosis. IMPLICATIONS: This study highlights the function of haplotypes of mTOR -78C/-141G and -78G/-141T, in affecting breast cancer susceptibility and paclitaxel response regulated by ZEB1/KLF5-mTOR-CCND1/ABCB1 axis.
Collapse
Affiliation(s)
- Qiuchen Chen
- Department of Pharmacology, School of Pharmacy, Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China
| | - Xiaolan Deng
- Department of Pharmacology, School of Pharmacy, Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China.,Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, California
| | - Xiaoyun Hu
- Department of Pharmacology, School of Pharmacy, Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China
| | - Shu Guan
- Department of Breast Surgery, First Hospital of China Medical University, Shenyang, China
| | - Miao He
- Department of Pharmacology, School of Pharmacy, Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China
| | - Yilin Wang
- Department of Pharmacology, School of Pharmacy, Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China
| | - Binbin Wei
- Department of Pharmacology, School of Pharmacy, Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China
| | - Jing Zhang
- Department of Pharmacology, School of Pharmacy, Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China
| | - Haishan Zhao
- Department of Pharmacology, School of Pharmacy, Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China
| | - Weifan Yao
- Department of Pharmacology, School of Pharmacy, Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China
| | - Feng Jin
- Department of Breast Surgery, First Hospital of China Medical University, Shenyang, China
| | - Yong Liu
- Department of Clinical Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jianjun Chen
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, California
| | | | - Huizhe Wu
- Department of Pharmacology, School of Pharmacy, Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China.
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China.
| |
Collapse
|
23
|
Antoszczak M, Huczyński A. Salinomycin and its derivatives - A new class of multiple-targeted "magic bullets". Eur J Med Chem 2019; 176:208-227. [PMID: 31103901 DOI: 10.1016/j.ejmech.2019.05.031] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 05/06/2019] [Accepted: 05/08/2019] [Indexed: 12/23/2022]
Abstract
The history of drug development clearly shows the scale of painstaking effort leading to a finished product - a highly biologically active agent that would be at the same time no or little toxic to human organism. Moreover, the aim of modern drug discovery can move from "one-molecule one-target" concept to more promising "one-molecule multiple-targets" one, particularly in the context of effective fight against cancer and other complex diseases. Gratifyingly, natural compounds are excellent source of potential drug leads. One of such promising naturally-occurring drug candidates is a polyether ionophore - salinomycin (SAL). This compound should be identified as multi-target agent for two reasons. Firstly, SAL combines a broad spectrum of bioactivity, including antibacterial, antifungal, antiviral, antiparasitic and anticancer activity, with high selectivity of action, proving its significant therapeutic potential. Secondly, the multimodal mechanism of action of SAL has been shown to be related to its interactions with multiple molecular targets and signalling pathways that are synergistic for achieving a therapeutic anticancer effect. On the other hand, according to the Paul Ehrlich's "magic bullet" concept, invariably inspiring the scientists working on design of novel target-selective molecules, a very interesting direction of research is rational chemical modification of SAL. Importantly, many of SAL derivatives have been found to be more promising as chemotherapeutics than the native structure. This concise review article is focused both on the possible role of SAL and its selected analogues in future antimicrobial and/or cancer therapy, and on the potential use of SAL as a new class of multiple-targeted "magic bullet" because of its multimodal mechanism of action.
Collapse
Affiliation(s)
- Michał Antoszczak
- Department of Bioorganic Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Umultowska 89b, 61‒614, Poznań, Poland
| | - Adam Huczyński
- Department of Bioorganic Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Umultowska 89b, 61‒614, Poznań, Poland.
| |
Collapse
|
24
|
Hu C, Li M, Guo T, Wang S, Huang W, Yang K, Liao Z, Wang J, Zhang F, Wang H. Anti-metastasis activity of curcumin against breast cancer via the inhibition of stem cell-like properties and EMT. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 58:152740. [PMID: 31005718 DOI: 10.1016/j.phymed.2018.11.001] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 10/31/2018] [Accepted: 11/03/2018] [Indexed: 05/22/2023]
Abstract
BACKGROUND Curcumin is a polyphenolic compound with potent chemopreventive and anti-cancer efficacy. PURPOSE To explore the potential anti-metastasis efficacy of curcumin in breast cancer stem-like cells (BCSCs), which are increasingly considered to be the origin of the recurrence and metastasis of breast cancer. METHODS A CCK8 assay was performed to evaluate cell viability, and a colony formation assay was conducted to determine cell proliferation in MCF-7 and MDA-MB-231 adherent cells. Transwell and wound healing assays were used to detect the effect of curcumin on cell migration and invasion in MDA-MB-231 cells. Mammospheres were cultured with serum free medium (SFM) for three generations and the BCSC surface marker CD44+CD24-/low subpopulation was measured by flow cytometry. Mammosphere formation and differentiation abilities were determined after cell treatment with curcumin. Then, a reverse transcription-quantitative polymerase chain reaction assay was conducted to detect the relative mRNA level of epithelial-mesenchymal transition (EMT) marker genes and western blot analysis was performed to determine the protein expression of stem cell genes in mammospheres treated with curcumin. RESULTS Curcumin exhibited anti-proliferative and colony formation inhibiting activities in both the MCF-7 and MDA-MB-231 cell lines. It also suppressed the migration and invasion of MDA-MB-231 cells. The CD44+CD24-/low subpopulation was larger in mammospheres when MCF-7 and MDA-MB-231 adherent cells were cultured with SFM. Further studies revealed that curcumin inhibited mammosphere formation and differentiation abilities. Moreover, curcumin down-regulated the mRNA expression of Vimentin, Fibronectin, and β-catenin, and up-regulated E-cadherin mRNA expression levels. Western blot analysis demonstrated that curcumin decreased the protein expression of stem cell genes including Oct4, Nanog and Sox2. CONCLUSION The results of the present study suggest that the inhibitor effects of curcumin on breast cancer cells may be related to resistance to cancer stem-like characters and the EMT process. These data indicate that curcumin could function as a type of anti-metastasis agent for breast cancer.
Collapse
Affiliation(s)
- Chenxia Hu
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Mengjie Li
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tingting Guo
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shaoxi Wang
- School of Software & Microelectronics, Northwestern Polytechnical University, Xi'an, China
| | - Weiping Huang
- The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ke Yang
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhiwei Liao
- Cancer Center of Guangzhou Medical University, Guangzhou, China
| | - Jian Wang
- The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fengxue Zhang
- The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Hongqi Wang
- The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
25
|
Genomic testing, tumor microenvironment and targeted therapy of Hedgehog-related human cancers. Clin Sci (Lond) 2019; 133:953-970. [PMID: 31036756 DOI: 10.1042/cs20180845] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/24/2019] [Accepted: 04/11/2019] [Indexed: 12/12/2022]
Abstract
Hedgehog signals are transduced through Patched receptors to the Smoothened (SMO)-SUFU-GLI and SMO-Gi-RhoA signaling cascades. MTOR-S6K1 and MEK-ERK signals are also transduced to GLI activators through post-translational modifications. The GLI transcription network up-regulates target genes, such as BCL2, FOXA2, FOXE1, FOXF1, FOXL1, FOXM1, GLI1, HHIP, PTCH1 and WNT2B, in a cellular context-dependent manner. Aberrant Hedgehog signaling in tumor cells leads to self-renewal, survival, proliferation and invasion. Paracrine Hedgehog signaling in the tumor microenvironment (TME), which harbors cancer-associated fibroblasts, leads to angiogenesis, fibrosis, immune evasion and neuropathic pain. Hedgehog-related genetic alterations occur frequently in basal cell carcinoma (BCC) (85%) and Sonic Hedgehog (SHH)-subgroup medulloblastoma (87%) and less frequently in breast cancer, colorectal cancer, gastric cancer, pancreatic cancer, non-small-cell lung cancer (NSCLC) and ovarian cancer. Among investigational SMO inhibitors, vismodegib and sonidegib are approved for the treatment of patients with BCC, and glasdegib is approved for the treatment of patients with acute myeloid leukemia (AML). Resistance to SMO inhibitors is caused by acquired SMO mutations, SUFU deletions, GLI2 amplification, other by-passing mechanisms of GLI activation and WNT/β-catenin signaling activation. GLI-DNA-interaction inhibitors (glabrescione B and GANT61), GLI2 destabilizers (arsenic trioxide and pirfenidone) and a GLI-deacetylation inhibitor (4SC-202) were shown to block GLI-dependent transcription and tumorigenesis in preclinical studies. By contrast, SMO inhibitors can remodel the immunosuppressive TME that is dominated by M2-like tumor-associated macrophages (M2-TAMs), myeloid-derived suppressor cells and regulatory T cells, and thus, a Phase I/II clinical trial of the immune checkpoint inhibitor pembrolizumab with or without vismodegib in BCC patients is ongoing.
Collapse
|
26
|
He M, Wu H, Jiang Q, Liu Y, Han L, Yan Y, Wei B, Liu F, Deng X, Chen H, Zhao L, Wang M, Wu X, Yao W, Zhao H, Chen J, Wei M. Hypoxia-inducible factor-2α directly promotes BCRP expression and mediates the resistance of ovarian cancer stem cells to adriamycin. Mol Oncol 2019; 13:403-421. [PMID: 30536571 PMCID: PMC6360369 DOI: 10.1002/1878-0261.12419] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 10/27/2018] [Accepted: 11/05/2018] [Indexed: 12/24/2022] Open
Abstract
Ovarian cancer stem cells (OCSCs) are sources of tumor chemoresistance and recurrence. A hypoxic microenvironment contributes to the chemoresistance of cancer stem cells (CSCs), but the underlying mechanism is not fully understood yet. Here, we show that increased HIF-2α expression is associated with enhanced stemness of OCSCs and poor outcomes in ovarian cancer patients. OVCAR-3 and CAOV-3 sphere-forming (OVCAR-3 S and CAOV-3 S) cells with OCSC-like properties showed strong resistance to adriamycin (ADR). Hypoxia (1% O2 ) induced high expression of both HIF-1α and especially HIF-2α, and increased the resistance of OVCAR-3 S and CAOV-3 S cells to ADR. Notably, treatment with ADR further increased the expression of HIF-2α, but not that of HIF-1α. Knockdown of HIF-2α expression substantially attenuated the resistance of OVCAR-3 S and CAOV-3 S cells to ADR, and the HIF-2α overexpression had the opposite effect. Furthermore, in mouse models xenografted with OCSCs, HIF-2α depletion significantly inhibited tumor growth and sensitized OCSCs to ADR in vivo. Mechanistically, HIF-2α directly promotes transcription/expression of BCRP, a gene encoding a transporter protein responsible for pumping drugs (e.g., ADR) out of cells, which in turn increases drug resistance due to increased drug transportation. Collectively, our studies reveal a novel drug-resistant mechanism in ovarian cancer by which hypoxia (and ADR treatment)-induced HIF-2α overexpression endows OCSCs with resistance to ADR by promoting BCRP expression and ADR transportation. Therefore, targeting the HIF-2α/BCRP axis holds therapeutic potential for treating drug-resistant ovarian cancer.
Collapse
Affiliation(s)
- Miao He
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Shenyang, China
| | - Huizhe Wu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Shenyang, China
| | - Qian Jiang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Shenyang, China
| | - Yinuo Liu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Shenyang, China
| | - Li Han
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Shenyang, China
| | - Yuanyuan Yan
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Shenyang, China
| | - Binbin Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Shenyang, China
| | - Fangxiao Liu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Shenyang, China
| | - Xiaolan Deng
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Shenyang, China
| | - Huiying Chen
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Shenyang, China
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Shenyang, China
| | - Min Wang
- Department of gynaecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xin Wu
- Department of gynaecology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Weifan Yao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Shenyang, China
| | - Haishan Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Shenyang, China
| | - Jianjun Chen
- Department of Cancer Biology, University of Cincinnati, Cincinnati, OH, USA
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Shenyang, China
| |
Collapse
|
27
|
Velasco-Velázquez MA, Velázquez-Quesada I, Vásquez-Bochm LX, Pérez-Tapia SM. Targeting Breast Cancer Stem Cells: A Methodological Perspective. Curr Stem Cell Res Ther 2019; 14:389-397. [PMID: 30147014 DOI: 10.2174/1574888x13666180821155701] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 08/01/2018] [Accepted: 08/09/2018] [Indexed: 02/07/2023]
Abstract
Cancer Stem Cells (CSCs) constitute a subpopulation at the top of the tumor cell hierarchy that contributes to tumor heterogeneity and is uniquely capable of seeding new tumors. Because of their biological properties, CSCs have been pointed out as therapeutic targets for the development of new therapies against breast cancer. The identification of drugs that selectively target breast CSCs requires a clear understanding of their biological functions and the experimental methods to evaluate such hallmarks. Herein, we review the methods to study breast CSCs properties and discuss their value in the preclinical evaluation of CSC-targeting drugs.
Collapse
Affiliation(s)
- Marco A Velasco-Velázquez
- Departamento de Farmacologia, Facultad de Medicina, Universidad Nacional Autonoma de Mexico (UNAM), Ciudad de Mexico, Mexico
- Unidad Periférica de Investigación en Biomedicina Traslacional, Facultad de Medicina, UNAM, Ciudad de México, México
| | - Inés Velázquez-Quesada
- Departamento de Farmacologia, Facultad de Medicina, Universidad Nacional Autonoma de Mexico (UNAM), Ciudad de Mexico, Mexico
- Unidad de Desarrollo e Investigacion en Bioprocesos, ENCB, Instituto Politecnico Nacional, Ciudad de Mexico, Mexico
| | - Luz X Vásquez-Bochm
- Departamento de Farmacologia, Facultad de Medicina, Universidad Nacional Autonoma de Mexico (UNAM), Ciudad de Mexico, Mexico
- Posgrado en Ciencias Químicas, UNAM, Ciudad de México, México
| | - Sonia M Pérez-Tapia
- Unidad de Desarrollo e Investigacion en Bioprocesos, ENCB, Instituto Politecnico Nacional, Ciudad de Mexico, Mexico
| |
Collapse
|
28
|
Yan Y, Liu F, Han L, Zhao L, Chen J, Olopade OI, He M, Wei M. HIF-2α promotes conversion to a stem cell phenotype and induces chemoresistance in breast cancer cells by activating Wnt and Notch pathways. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:256. [PMID: 30340507 PMCID: PMC6194720 DOI: 10.1186/s13046-018-0925-x] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 10/02/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Hypoxic tumor microenvironment and maintenance of stemness contribute to drug resistance in breast cancer. However, whether Hypoxia-inducible factor-2α (HIF-2α) in hypoxic tumor microenvironment mediates conversion to a stem cell phenotype and chemoresistance of breast tumors has not been elucidated. METHODS The mRNA and protein expressions of HIF-1α, HIF-2α, Wnt and Notch pathway were determined using qRT-PCR and western blot. Cell viability and renew ability were assessed by MTT, Flow cytometric analysis and soft agar colony formation. RESULTS In our study, acute hypoxia (6-12 h) briefly increased HIF-1α expression, while chronic hypoxia (48 h) continuously enhanced HIF-2α expression and induced the resistance of breast cancer cells to Paclitaxel (PTX). Furthermore, HIF-2α overexpression induced a stem cell phenotype, the resistance to PTX and enhanced protein expression of stem cell markers, c-Myc, OCT4 and Nanog. Most importantly, Wnt and Notch signaling, but not including Shh, pathways were both activated by HIF-2α overexpression. Dickkopf-1 (DKK-1), a Wnt pathway inhibitor, and L685,458, an inhibitor of the Notch pathway, reversed the resistance to PTX and stem phenotype conversion induced by HIF-2α overexpression. In addition, HIF-2α overexpression enhanced tumorigenicity and resistance of xenograft tumors to PTX, increased activation of the Wnt and Notch pathways and induced a stem cell phenotype in vivo. CONCLUSION In conclusion, HIF-2α promoted stem phenotype conversion and induced resistance to PTX by activating Wnt and Notch pathways.
Collapse
Affiliation(s)
- Yuanyuan Yan
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Shenyang North Area, Shenyang City, 110122, Liaoning, China
| | - Fangxiao Liu
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Shenyang North Area, Shenyang City, 110122, Liaoning, China
| | - Li Han
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Shenyang North Area, Shenyang City, 110122, Liaoning, China
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Shenyang North Area, Shenyang City, 110122, Liaoning, China
| | - Jianjun Chen
- Department of Systems Biology, City of hope, Los Angeles, CA, USA
| | - Olufunmilayo I Olopade
- Center for Clinical Cancer Genetics, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Miao He
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China. .,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Shenyang North Area, Shenyang City, 110122, Liaoning, China.
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China. .,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Shenyang North Area, Shenyang City, 110122, Liaoning, China.
| |
Collapse
|
29
|
Versini A, Saier L, Sindikubwabo F, Müller S, Cañeque T, Rodriguez R. Chemical biology of salinomycin. Tetrahedron 2018. [DOI: 10.1016/j.tet.2018.07.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
30
|
Carranza-Rosales P, Guzmán-Delgado NE, Carranza-Torres IE, Viveros-Valdez E, Morán-Martínez J. Breast Organotypic Cancer Models. Curr Top Microbiol Immunol 2018:199-223. [PMID: 29556825 DOI: 10.1007/82_2018_86] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Breast cancer is the most common cancer type diagnosed in women, it represents a critical public health problem worldwide, with 1,671,149 estimated new cases and nearly 571,000 related deaths. Research on breast cancer has mainly been conducted using two-dimensional (2D) cell cultures and animal models. The usefulness of these models is reflected in the vast knowledge accumulated over the past decades. However, considering that animal models are three-dimensional (3D) in nature, the validity of the studies using 2D cell cultures has recently been questioned. Although animal models are important in cancer research, ethical questions arise about their use and usefulness as there is no clear predictivity of human disease outcome and they are very expensive and take too much time to obtain results. The poor performance or failure of most cancer drugs suggests that preclinical research on cancer has been based on an over-dependence on inadequate animal models. For these reasons, in the last few years development of alternative models has been prioritized to study human breast cancer behavior, while maintaining a 3D microenvironment, and to reduce the number of experiments conducted in animals. One way to achieve this is using organotypic cultures, which are being more frequently explored in cancer research because they mimic tissue architecture in vivo. These characteristics make organotypic cultures a valuable tool in cancer research as an alternative to replace animal models and for predicting risk assessment in humans. This chapter describes the cultures of multicellular spheroids, organoids, 3D bioreactors, and tumor slices, which are the most widely used organotypic models in breast cancer research.
Collapse
Affiliation(s)
- Pilar Carranza-Rosales
- Departamento de Biología Celular y Molecular, Instituto Mexicano del Seguro Social. Centro de Investigación Biomédica del Noreste, Monterrey, Nuevo León, Mexico.
| | - Nancy Elena Guzmán-Delgado
- Unidad Médica de Alta Especialidad # 34, División de Investigación, Instituto Mexicano del Seguro Social, Monterrey, Nuevo León, Mexico
| | - Irma Edith Carranza-Torres
- Departamento de Biología Celular y Molecular, Instituto Mexicano del Seguro Social. Centro de Investigación Biomédica del Noreste, Monterrey, Nuevo León, Mexico
| | - Ezequiel Viveros-Valdez
- Departamento de Química Analítica, Ciudad Universitaria, Universidad Autónoma de Nuevo León, Facultad de Ciencias Biológicas, San Nicolás de los Garza, Nuevo León, Mexico
| | - Javier Morán-Martínez
- Departamento de Biología Celular y Ultraestructura, Universidad Autónoma de Coahuila, Facultad de Medicina. Centro de Investigación Biomédica, Torreón, Coahuila, Mexico
| |
Collapse
|
31
|
Jiang J, Li H, Qaed E, Zhang J, Song Y, Wu R, Bu X, Wang Q, Tang Z. Salinomycin, as an autophagy modulator-- a new avenue to anticancer: a review. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:26. [PMID: 29433536 PMCID: PMC5809980 DOI: 10.1186/s13046-018-0680-z] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 01/15/2018] [Indexed: 12/25/2022]
Abstract
Since Salinomycin (Sal) emerged its ability to target breast cancer stem cells in 2009, numerous experiments have been carried out to test Sal’s anticancer effects. What deserve to be mentioned is that Sal can efficiently induce proliferation inhibition, cell death and metastasis suppression against human cancers from different origins both in vivo and in vitro without causing serious side effects as the conventional chemotherapeutical drugs on the body. There may be novel cell death pathways involving the anticancer effects of Sal except the conventional pathways, such as autophagic pathway. This review is focused on how autophagy involves the effects of Sal, trying to describe clearly and systematically why autophagy plays a vital role in predominant anticancer effects of Sal, including its distinctive characteristic. Based on recent advances, we present evidence that a dual role of Sal involving in autophagy may account for its unique anticancer effects - the preference for cancer cells. Further researches are required to confirm the authenticity of this suppose in order to develop an ideal anticancer drug.
Collapse
Affiliation(s)
- Jiang Jiang
- Department of Pharmacology, Dalian Medical University, 9 west section, south road of Lvshun, Dalian, 116044, China
| | - Hailong Li
- Department of Pharmacology, Dalian Medical University, 9 west section, south road of Lvshun, Dalian, 116044, China
| | - Eskandar Qaed
- Department of Pharmacology, Dalian Medical University, 9 west section, south road of Lvshun, Dalian, 116044, China
| | - Jing Zhang
- Department of Pharmacology, Dalian Medical University, 9 west section, south road of Lvshun, Dalian, 116044, China
| | - Yushu Song
- Department of Pharmacology, Dalian Medical University, 9 west section, south road of Lvshun, Dalian, 116044, China
| | - Rong Wu
- Department of Pharmacology, Dalian Medical University, 9 west section, south road of Lvshun, Dalian, 116044, China
| | - Xinmiao Bu
- Department of Pharmacology, Dalian Medical University, 9 west section, south road of Lvshun, Dalian, 116044, China
| | - Qinyan Wang
- Department of Pharmacology, Dalian Medical University, 9 west section, south road of Lvshun, Dalian, 116044, China
| | - Zeyao Tang
- Department of Pharmacology, Dalian Medical University, 9 west section, south road of Lvshun, Dalian, 116044, China
| |
Collapse
|
32
|
Ye D, Li Z, Wei C. Genistein inhibits the S-phase kinase-associated protein 2 expression in breast cancer cells. Exp Ther Med 2017; 15:1069-1075. [PMID: 29434697 PMCID: PMC5772955 DOI: 10.3892/etm.2017.5489] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 09/14/2017] [Indexed: 12/17/2022] Open
Abstract
Breast cancer is one of the most lethal cancers affecting women worldwide and was estimated to account for ~30% of all new cancer diagnoses in women. Although available evidence has proved the tumor suppressor role of genistein in cancer, the underling mechanisms have remained to be fully elucidated. S-phase kinase-associated protein 2 (Skp2) has been revealed to critically enhance the pathogenesis of multiple human cancers. The present study determined whether genistein exerts its anti-tumor function by suppressing Skp2 in breast cancer cells. Genistein significantly inhibited the proliferation, invasion and migration of breast cancer cells. Furthermore, genistein treatment also induced marked apoptosis and a typical cell cycle arrest in G2/M phase. Mechanistically, genistein treatment was identified to cause a significant downregulation of Skp2. Two crucial tumor suppressors, p21 and p27, were upregulated in genistein-treated breast cancer cells. The present results revealed that genistein exerted its tumor suppressor effect at least partially via inhibition of Skp2 and promotion of its downstream targets p21 and p27. Therefore, inactivation of Skp2 by genistein may be a promising approach for breast cancer treatment.
Collapse
Affiliation(s)
- Dengfeng Ye
- Department of Oncological Surgery, The Second Hospital of Shaoxing, Shaoxing, Zhejiang 312000, P.R. China
| | - Zhian Li
- Department of Oncological Surgery, The Second Hospital of Shaoxing, Shaoxing, Zhejiang 312000, P.R. China
| | - Chunshou Wei
- Department of Oncological Surgery, The Second Hospital of Shaoxing, Shaoxing, Zhejiang 312000, P.R. China
| |
Collapse
|
33
|
Wang B, Yu T, Hu Y, Xiang M, Peng H, Lin Y, Han L, Zhang L. Prognostic role of Gli1 expression in breast cancer: a meta-analysis. Oncotarget 2017; 8:81088-81097. [PMID: 29113369 PMCID: PMC5655264 DOI: 10.18632/oncotarget.19080] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 06/20/2017] [Indexed: 02/05/2023] Open
Abstract
Glioma-associated oncogene 1 (Gli1) is a critical transcriptional factor of Sonic hedgehog pathway which has been proved to participate in the initiation and progression of tumor in mammalians. However, its clinical value in breast cancer remains unknown. Thus, a meta-analysis was performed to clarify the association of Gli1 over-expression, clinic-pathological characteristics, molecular subtypes and prognosis in breast cancer. According to included criteria, 13 eligible studies containing 2816 patients all around the world were selected in this study. Our results indicated no significant association of Gli1 expression and histological grade (RR = 1.20, 95% CI: [0.98, 1.47]), T stage (RR = 1.05, 95% CI: [0.87, 1.27]), clinical stage (RR = 1.04, 95% CI: [0.93, 1.18]) and lymph node metastasis (RR = 1.12, 95% CI: [0.92, 1.37]). In addition, pooled RR showed no correlation of Gli1 expression and progesterone receptor (PR) (RR = 0.92, 95% CI: [0.70, 1.21]), estrogen receptor (ER) (RR = 1.03, 95% CI: [0.74, 1.42]), human epidermal growth factor receptor 2 (HER-2) (RR = 1.12, 95% CI: [0.90, 1.39]). Nonetheless, up-regulated Gli1 expression predicts shorter disease-free survival (DFS) (HR = 1.38, 95% CI: [1.05, 1.81]), 3-year survival (HR = 1.74, 95% CI: [1.28, 2.36]), 5-year survival (HR = 2.04, 95% CI: [1.62, 2.57]) and overall survival (OS) (HR = 2.05, 95% CI: [1.60, 2.64]). In conclusion, over-expression of Gli1 tends to progressive stages and is related to unfavorable prognosis of breast cancer, which may become a potential prognosis indicator and therapy target in breast cancer.
Collapse
Affiliation(s)
- Bilan Wang
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, 610041, PR China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, 610041, PR China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Ting Yu
- Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, PR China
| | - Yuzhu Hu
- Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, PR China
| | - Mengmeng Xiang
- Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, PR China
| | - Haoning Peng
- Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, PR China
| | - Yunzhu Lin
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, 610041, PR China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, 610041, PR China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Lu Han
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, 610041, PR China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, 610041, PR China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Lingli Zhang
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, 610041, PR China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, 610041, PR China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, PR China
| |
Collapse
|
34
|
McCubrey JA, Fitzgerald TL, Yang LV, Lertpiriyapong K, Steelman LS, Abrams SL, Montalto G, Cervello M, Neri LM, Cocco L, Martelli AM, Laidler P, Dulińska-Litewka J, Rakus D, Gizak A, Nicoletti F, Falzone L, Candido S, Libra M. Roles of GSK-3 and microRNAs on epithelial mesenchymal transition and cancer stem cells. Oncotarget 2017; 8:14221-14250. [PMID: 27999207 PMCID: PMC5355173 DOI: 10.18632/oncotarget.13991] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 12/13/2016] [Indexed: 12/12/2022] Open
Abstract
Various signaling pathways exert critical roles in the epithelial to mesenchymal transition (EMT) and cancer stem cells (CSCs). The Wnt/beta-catenin, PI3K/PTEN/Akt/mTORC, Ras/Raf/MEK/ERK, hedgehog (Hh), Notch and TP53 pathways elicit essential regulatory influences on cancer initiation, EMT and progression. A common kinase involved in all these pathways is moon-lighting kinase glycogen synthase kinase-3 (GSK-3). These pathways are also regulated by micro-RNAs (miRs). TP53 and components of these pathways can regulate the expression of miRs. Targeting members of these pathways may improve cancer therapy in those malignancies that display their abnormal regulation. This review will discuss the interactions of the multi-functional GSK-3 enzyme in the Wnt/beta-catenin, PI3K/PTEN/Akt/mTORC, Ras/Raf/MEK/ERK, Hh, Notch and TP53 pathways. The regulation of these pathways by miRs and their effects on CSC generation, EMT, invasion and metastasis will be discussed.
Collapse
Affiliation(s)
- James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, USA
| | - Timothy L Fitzgerald
- Department of Surgery, Brody School of Medicine at East Carolina University, Greenville, NC, USA
| | - Li V Yang
- Department of Internal Medicine, Hematology/Oncology Section, Brody School of Medicine at East Carolina University, Greenville, NC, USA
| | - Kvin Lertpiriyapong
- Department of Comparative Medicine, Brody School of Medicine at East Carolina University, Greenville, NC, USA
| | - Linda S Steelman
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, USA
| | - Stephen L Abrams
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, USA
| | - Giuseppe Montalto
- Biomedical Department of Internal Medicine and Specialties, University of Palermo, Palermo, Italy.,Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Melchiorre Cervello
- Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Luca M Neri
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Lucio Cocco
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Alberto M Martelli
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Piotr Laidler
- Chair of Medical Biochemistry, Jagiellonian University Medical College, Kraków, Poland
| | | | - Dariusz Rakus
- Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University, Wroclaw, Poland
| | - Agnieszka Gizak
- Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University, Wroclaw, Poland
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences - Oncological, Clinical and General Pathology Section, University of Catania, Catania, Italy
| | - Luca Falzone
- Department of Biomedical and Biotechnological Sciences - Oncological, Clinical and General Pathology Section, University of Catania, Catania, Italy
| | - Saverio Candido
- Department of Biomedical and Biotechnological Sciences - Oncological, Clinical and General Pathology Section, University of Catania, Catania, Italy
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences - Oncological, Clinical and General Pathology Section, University of Catania, Catania, Italy
| |
Collapse
|
35
|
Magrath JW, Kim Y. Salinomycin's potential to eliminate glioblastoma stem cells and treat glioblastoma multiforme (Review). Int J Oncol 2017; 51:753-759. [PMID: 28766685 DOI: 10.3892/ijo.2017.4082] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 07/12/2017] [Indexed: 12/09/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and deadliest form of primary brain tumor. Despite treatment with surgery, radiotherapy, and chemotherapy with the drug temozolomide, the expected survival after diagnosis remains low. The median survival is only 14.6 months and the two-year survival is a mere 30%. One reason for this is the heterogeneity of GBM including the presence of glioblastoma cancer stem cells (GSCs). GSCs are a subset of cells with the unique ability to proliferate, differentiate, and create tumors. GSCs are resistant to chemotherapy and radiation and thought to play an important role in recurrence. In order to effectively treat GBM, a drug must be identified that can kill GSCs. The ionophore salinomycin has been shown to kill cancer stem cells and is therefore a promising future treatment for GBM. This study focuses on salinomycin's potential to treat GBM including its ability to reduce the CSC population, its toxicity to normal brain cells, its mechanism of action, and its potential for combination treatment.
Collapse
Affiliation(s)
- Justin W Magrath
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487-0203, USA
| | - Yonghyun Kim
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487-0203, USA
| |
Collapse
|
36
|
HER2 in Breast Cancer Stemness: A Negative Feedback Loop towards Trastuzumab Resistance. Cancers (Basel) 2017; 9:cancers9050040. [PMID: 28445439 PMCID: PMC5447950 DOI: 10.3390/cancers9050040] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 04/10/2017] [Accepted: 04/21/2017] [Indexed: 12/19/2022] Open
Abstract
HER2 receptor tyrosine kinase that is overexpressed in approximately 20% of all breast cancers (BCs) is a poor prognosis factor and a precious target for BC therapy. Trastuzumab is approved by FDA to specifically target HER2 for treating HER2+ BC. However, about 60% of patients with HER2+ breast tumor develop de novo resistance to trastuzumab, partially due to the loss of expression of HER2 extracellular domain on their tumor cells. This is due to shedding/cleavage of HER2 by metalloproteinases (ADAMs and MMPs). HER2 shedding results in the accumulation of intracellular carboxyl-terminal HER2 (p95HER2), which is a common phenomenon in trastuzumab-resistant tumors and is suggested as a predictive marker for trastuzumab resistance. Up-regulation of the metalloproteinases is a poor prognosis factor and is commonly seen in mesenchymal-like cancer stem cells that are risen during epithelial to mesenchymal transition (EMT) of tumor cells. HER2 cleavage during EMT can explain why secondary metastatic tumors with high percentage of mesenchymal-like cancer stem cells are mostly resistant to trastuzumab but still sensitive to lapatinib. Importantly, many studies report HER2 interaction with oncogenic/stemness signaling pathways including TGF-β/Smad, Wnt/β-catenin, Notch, JAK/STAT and Hedgehog. HER2 overexpression promotes EMT and the emergence of cancer stem cell properties in BC. Increased expression and activation of metalloproteinases during EMT leads to proteolytic cleavage and shedding of HER2 receptor, which downregulates HER2 extracellular domain and eventually increases trastuzumab resistance. Here, we review the hypothesis that a negative feedback loop between HER2 and stemness signaling drives resistance of BC to trastuzumab.
Collapse
|
37
|
Dewangan J, Srivastava S, Rath SK. Salinomycin: A new paradigm in cancer therapy. Tumour Biol 2017; 39:1010428317695035. [PMID: 28349817 DOI: 10.1177/1010428317695035] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The primary hurdle in the treatment of cancer is acquisition of resistance by the tumor cells toward multiple drugs and selectively targeting the cancer stem cells. This problem was overcome by the chemotherapeutic property of recently discovered drug salinomycin. Exact mechanism of action of salinomycin is not yet known, but there are multiple pathways by which salinomycin inhibits tumor growth. Salinomycin decreases the expression of adenosine triphosphate-binding cassette transporter in multidrug resistance cells and interferes with Akt signaling pathway, Wnt/β-catenin, Hedgehog, and Notch pathways of cancer progression. Salinomycin selectively targets cancer stem cells. The potential of salinomycin to eliminate both cancer stem cells and therapy-resistant cancer cells may characterize the compound as a novel and an efficient chemotherapeutic drug.
Collapse
Affiliation(s)
- Jayant Dewangan
- Genotoxicity Laboratory, Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Sonal Srivastava
- Genotoxicity Laboratory, Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Srikanta Kumar Rath
- Genotoxicity Laboratory, Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow, India
| |
Collapse
|
38
|
Akinyeke T, Weber SJ, Davenport AT, Baker EJ, Daunais JB, Raber J. Effects of alcohol on c-Myc protein in the brain. Behav Brain Res 2016; 320:356-364. [PMID: 27832980 DOI: 10.1016/j.bbr.2016.11.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 10/28/2016] [Accepted: 11/04/2016] [Indexed: 12/18/2022]
Abstract
Alcoholism is a disorder categorized by significant impairment that is directly related to persistent and extreme use of alcohol. The effects of alcoholism on c-Myc protein expression in the brain have been scarcely studied. This is the first study to investigate the role different characteristics of alcoholism have on c-Myc protein in the brain. We analyzed c-Myc protein in the hypothalamus and amygdala from five different animal models of alcohol abuse. c-Myc protein was increased following acute ethanol exposure in a mouse knockout model and following chronic ethanol consumption in vervet monkeys. We also observed increases in c-Myc protein exposure in animals that are genetically predisposed to alcohol and methamphetamine abuse. Lastly, c-Myc protein was increased in animals that were acutely exposed to methamphetamine when compared to control treated animals. These results suggest that in substance abuse c-Myc plays an important role in the brain's response.
Collapse
Affiliation(s)
- Tunde Akinyeke
- Department of Behavioral Neuroscience, ONPRC, Oregon Health and Science University, Portland, OR 97239, United States
| | - Sydney J Weber
- Department of Behavioral Neuroscience, ONPRC, Oregon Health and Science University, Portland, OR 97239, United States
| | - April T Davenport
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27106, United States
| | - Erich J Baker
- School of Engineering and Department of Computer Science, Baylor University Waco, TX 76978, United States
| | - James B Daunais
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27106, United States
| | - Jacob Raber
- Department of Behavioral Neuroscience, ONPRC, Oregon Health and Science University, Portland, OR 97239, United States; Departments of Neurology and Radiation Medicine, Division of Neuroscience, ONPRC, Oregon Health and Science University, Portland, OR 97239, United States.
| |
Collapse
|
39
|
Pavlopoulou A, Oktay Y, Vougas K, Louka M, Vorgias CE, Georgakilas AG. Determinants of resistance to chemotherapy and ionizing radiation in breast cancer stem cells. Cancer Lett 2016; 380:485-493. [DOI: 10.1016/j.canlet.2016.07.018] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 07/15/2016] [Accepted: 07/18/2016] [Indexed: 12/13/2022]
|
40
|
Cheng J, Gao J, Tao K. Prognostic role of Gli1 expression in solid malignancies: a meta-analysis. Sci Rep 2016; 6:22184. [PMID: 26899488 PMCID: PMC4762019 DOI: 10.1038/srep22184] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 02/02/2016] [Indexed: 02/07/2023] Open
Abstract
Gli1 is a downstream transcriptional factor of Sonic hedgehog pathway in mammalians, and has been recognized as a proliferative indicator of carcinogenesis. However, its actual role in prognosis among solid malignancies remains unclear. Therefore we performed this meta-analysis aiming to discover the correlation between Gli1 positivity and clinical prognosis in patients suffering from diverse carcinomas. A total of 39 studies containing 4496 cases were selected into our quantitative analysis via electronic database search. Original data of 3-year, 5-year, 10-year overall survival and disease-free survival were extracted and calculated using odds ratio and Mantel-Haenszel model. Subgroup analysis was also conducted to clarify the possible confounding factors. P < 0.05 was considered significant in statistics. Gli1 redundancy was associated with worse 3-year, 5-year, 10-year overall survival and disease-free survival in solid malignancies. Different source regions, sample-size, mean-age and detection approaches had no impact on the negative prognostic effect of Gli1 over-expression. Nevertheless, stratified by cancer type and subcellular localization, cytoplasmic Gli1 expression and Gli1 positivity in intracranial tumors was not correlated to poorer 3-year and 5-year prognosis. The over-expression of Gli1 is a credible indicator of poorer prognosis in most of solid malignancies, irrespective of intracranial tumors.
Collapse
Affiliation(s)
- Ji Cheng
- Department of General Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038 China
| | - Jinbo Gao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| |
Collapse
|
41
|
Stadler M, Walter S, Walzl A, Kramer N, Unger C, Scherzer M, Unterleuthner D, Hengstschläger M, Krupitza G, Dolznig H. Increased complexity in carcinomas: Analyzing and modeling the interaction of human cancer cells with their microenvironment. Semin Cancer Biol 2015; 35:107-24. [DOI: 10.1016/j.semcancer.2015.08.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/19/2015] [Accepted: 08/21/2015] [Indexed: 02/08/2023]
|
42
|
Nuclear Gli1 expression is associated with pathological complete response and event-free survival in HER2-positive breast cancer treated with trastuzumab-based neoadjuvant therapy. Tumour Biol 2015; 37:4873-81. [DOI: 10.1007/s13277-015-4325-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 10/26/2015] [Indexed: 02/07/2023] Open
|
43
|
Crabtree JS, Miele L. [Modification of a micromethod for determining leukocyte migration inhibition and its significance in oncological patients]. Biomedicines 1981; 6:biomedicines6030077. [PMID: 30018256 PMCID: PMC6163894 DOI: 10.3390/biomedicines6030077] [Citation(s) in RCA: 52] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 07/13/2018] [Accepted: 07/14/2018] [Indexed: 12/21/2022] Open
Abstract
Breast cancer stem cells (BCSC) have been implicated in tumor initiation, progression, metastasis, recurrence, and resistance to therapy. The origins of BCSCs remain controversial due to tumor heterogeneity and the presence of such small side populations for study, but nonetheless, cell surface markers and their correlation with BCSC functionality continue to be identified. BCSCs are driven by persistent activation of developmental pathways, such as Notch, Wnt, Hippo, and Hedgehog and new treatment strategies that are aimed at these pathways are in preclinical and clinical development.
Collapse
Affiliation(s)
- Judy S Crabtree
- Department of Genetics and the Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.
| | - Lucio Miele
- Department of Genetics and the Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.
| |
Collapse
|