1
|
Wang YW, Tuan YL, Wang JY, Chang HY, Chu CA, Chen YL, Chen HW, Ho CL, Lee CT, Chow NH. Potential of epithelial membrane protein 3 as a novel therapeutic target for human breast cancer. Oncol Rep 2025; 53:16. [PMID: 39611484 PMCID: PMC11632653 DOI: 10.3892/or.2024.8849] [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: 07/23/2024] [Accepted: 10/18/2024] [Indexed: 11/30/2024] Open
Abstract
Amplification of human epidermal growth factor 2 receptor (HER2) and overexpression of estrogen receptor (ER) and/or progesterone receptor (PR) are key determinants in the treatment planning for human breast cancer (BC). Currently, targeted therapies for BC are focused mainly on these biomarkers. However, development of resistance to targeted drugs is almost unavoidable, emphasizing the importance of biochemical and pharmaceutical advances to improve treatment outcomes. To the best of our knowledge, the present study is the first to show functional crosstalk in vitro between HER2 and epithelial membrane protein 3 (EMP3), a tetraspan membrane protein, in human BC. EMP3 overexpression significantly promoted BC cell proliferation, invasion and migration by Transwell assays via epithelial-mesenchymal transition and transactivated the HER family, resulting in increased ER and PR expression in vitro. Knocking down EMP3 notably suppressed cell proliferation and migration and was accompanied by decreased expression of HER1‑HER3 and p‑SRC proteins. Suppression of EMP3 expression enhanced sensitivity of BC cells to trastuzumab in vitro. Xenograft experiments revealed decreased expression of HER1 and HER2 in stable EMP3‑knockdown cells, resulting in decreased tumor weight and size. In patients with BC, EMP3 overexpression was detected in 72 of 166 cases (43.4%), with 18 of 43 (41.9%) HER2‑amplified BC samples co‑expressing EMP3. Co‑expression of EMP3 and HER2 was positively associated with ER expression (P=0.028) and tended to be associated with nodal metastasis (P=0.085), however this was not significant. Taken together, the present results supported the potential of targeting EMP3 as a novel therapeutic strategy for human BC via co‑expression of HER2 and EMP3.
Collapse
Affiliation(s)
- Yi-Wen Wang
- Department of Food Safety Hygiene and Risk Management, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan, R.O.C
| | - Yih-Lin Tuan
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan, R.O.C
| | - Jiu-Yao Wang
- Center for Allergy, Immunology, and Microbiome, China Medical University Hospital, China Medical University, Taichung 404327, Taiwan, R.O.C
- Department of Allergy, Immunology, and Rheumatology, China Medical University Children's Hospital, China Medical University, Taichung 404327, Taiwan, R.O.C
| | - Hong-Yi Chang
- Department of Anatomy, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan, R.O.C
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807378, Taiwan, R.O.C
| | - Chien-An Chu
- Department of Food Safety Hygiene and Risk Management, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan, R.O.C
| | - Yi-Lin Chen
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan, R.O.C
- Department of Pathology, National Cheng Kung University Hospital, Tainan 701401, Taiwan, R.O.C
| | - Hui-Wen Chen
- Department of Pathology, National Cheng Kung University Hospital, Tainan 701401, Taiwan, R.O.C
| | - Chung-Liang Ho
- Department of Food Safety Hygiene and Risk Management, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan, R.O.C
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan, R.O.C
- Department of Pathology, National Cheng Kung University Hospital, Tainan 701401, Taiwan, R.O.C
| | - Chung-Ta Lee
- Department of Food Safety Hygiene and Risk Management, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan, R.O.C
- Department of Pathology, National Cheng Kung University Hospital, Tainan 701401, Taiwan, R.O.C
| | - Nan-Haw Chow
- Department of Food Safety Hygiene and Risk Management, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan, R.O.C
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan, R.O.C
- Center for Precision Medicine, China Medical University Hospital, China Medical University, Taichung 404327, Taiwan, R.O.C
| |
Collapse
|
2
|
Irdianto SA, Fadhillah F, Lestari R, Fadilah F, Bowolaksono A, Dwiranti A. Extrachromosomal DNA in Breast Cancer Cell Lines: Detection and Characterization. Microsc Res Tech 2024. [PMID: 39733336 DOI: 10.1002/jemt.24780] [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: 07/28/2024] [Revised: 11/14/2024] [Accepted: 12/15/2024] [Indexed: 12/31/2024]
Abstract
This study delves into the intriguing world of extrachromosomal DNA (ecDNA) in breast cancer, uncovering its pivotal role in cancer's aggressiveness and genetic variability. ecDNA, a form of circular DNA found outside chromosomes, is known to play a significant role in cancer progression by increasing oncogene expression. Focusing on two contrasting cell lines, MDA-MB-231 (triple-negative) and MCF-7 (Luminal-A), we utilized advanced microscopy and fluorescence techniques to detect and characterize ecDNA. Our findings reveal a stark difference: MDA-MB-231 cells, known for their high metastatic potential, exhibit a striking abundance of ecDNA, manifested as double minutes and single form with intense fluorescence signals. In contrast, the less aggressive MCF-7 cells harbor significantly fewer ecDNA. This disparity highlights the potential of ecDNA as a key player in cancer progression and a promising target for novel therapies. This research sheds light on the unseen genetic forces driving breast cancer and opens the door to new strategies in cancer treatment. Further research is necessary to understand the mechanisms of ecDNA formation and its role in different breast cancer subtypes.
Collapse
Affiliation(s)
- Shadira Anindieta Irdianto
- Cellular and Molecular Mechanisms in Biological System (CEMBIOS) Research Group, Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Indonesia, Depok, Indonesia
| | - Fadhillah Fadhillah
- Cellular and Molecular Mechanisms in Biological System (CEMBIOS) Research Group, Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Indonesia, Depok, Indonesia
| | - Retno Lestari
- Cellular and Molecular Mechanisms in Biological System (CEMBIOS) Research Group, Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Indonesia, Depok, Indonesia
| | - Fadilah Fadilah
- Department of Medical Chemistry, Faculty of Medicine, Universitas Indonesia, Central Jakarta, Indonesia
| | - Anom Bowolaksono
- Cellular and Molecular Mechanisms in Biological System (CEMBIOS) Research Group, Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Indonesia, Depok, Indonesia
| | - Astari Dwiranti
- Cellular and Molecular Mechanisms in Biological System (CEMBIOS) Research Group, Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Indonesia, Depok, Indonesia
| |
Collapse
|
3
|
Sharma D, Dhobi M, Lather V, Pandita D. An insight into the therapeutic effects of isoliquiritigenin in breast cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:9503-9519. [PMID: 39007925 DOI: 10.1007/s00210-024-03282-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024]
Abstract
Breast cancer ranks as the most widespread malignant condition in women, emerging as a primary contributor to mortality. The primary challenges in cancer treatments involve undesirable side effects. Therefore, exploring natural compounds as additional therapy could provide valuable insights. Isoliquiritigenin (ILN), an isoflavonoid featuring a chalcone moiety primarily sourced from Glycyrrhiza species, has garnered increasing interest in breast cancer research. This review aims to provide a comprehensive understanding of ILN's mechanisms of action in breast cancer, drawing from a range of in vitro and in vivo studies. ILN primarily acts by inhibiting angiogenesis, aromatase, inflammation, and cell proliferation, and preventing invasion and metastasis. Mechanistically, it downregulates miR-374a, phosphoinositide-3-kinase-protein kinase B/Akt, maternal embryonic leucine zipper kinase, vascular endothelial growth factor, and estrogen receptor protein levels, and causes enhancement of Wnt inhibitory factor-1, and Unc-51-like kinase 1 expression to treat breast cancer. ILN emerges as a promising natural option, offering therapeutic advantages with minimal side effects. However, it is important to note that current research on ILN is primarily limited to preclinical models, underscoring the need for further investigation to validate its potential efficacy.
Collapse
Affiliation(s)
- Divya Sharma
- School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, Sector-III, Pushp Vihar, Government of NCT of Delhi, New Delhi, 110017, India
| | - Mahaveer Dhobi
- School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, Sector-III, Pushp Vihar, Government of NCT of Delhi, New Delhi, 110017, India.
| | - Viney Lather
- Amity Institute of Pharmacy, Amity University Uttar Pradesh, Sector 125, Noida, 201313, India.
| | - Deepti Pandita
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences & Research (DIPSAR) Delhi Pharmaceutical Sciences and Research University, Sector-III, Pushp Vihar, Government of NCT of Delhi, New Delhi, 110017, India.
- Centre for Advanced Formulation Technology (CAFT), Delhi Pharmaceutical Sciences and Research University, Sector-III, Pushp Vihar, Government of NCT of Delhi, New Delhi, 110017, India.
| |
Collapse
|
4
|
Liu W, Luo G. CAV1 inhibits Xc - system through IFNGR1 to promote ferroptosis to inhibit stemness and improves anti-PD-1 efficacy in breast cancer. Transl Oncol 2024; 50:102149. [PMID: 39395272 DOI: 10.1016/j.tranon.2024.102149] [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: 06/11/2024] [Revised: 09/18/2024] [Accepted: 09/28/2024] [Indexed: 10/14/2024] Open
Abstract
Breast cancer is the most prevalent malignancy among women worldwide, with breast cancer stem cells (BCSCs) being the primary drivers of metastasis and recurrence. Numerous studies have elucidated the relationship between ferroptosis and cellular stemness, identifying the Xc- system as a key regulatory mechanism governing ferroptosis. However, the interplay between CAV1 and ferroptosis, along with its implications for stemness in breast cancer, remains inadequately understood. This gap in knowledge impedes advancements in targeted therapies for breast cancer. We employed immunohistochemistry and bioinformatics analyses to demonstrate the downregulation of CAV1 in breast cancer tissues. Additionally, we utilized CCK-8 assays, EDU staining, and Transwell assays to assess cell proliferation, migration, and invasion capabilities. Furthermore, we evaluated indicators associated with ferroptosis while examining markers related to stemness through sphere culture experiments and flow cytometry techniques. Our findings indicate that CAV1 expression can induce cell death via ferroptosis while simultaneously inhibiting both cell proliferation and features of stemness by upregulating IFNGR1 and promoting ferroptosis. Moreover, our in vivo experiments revealed that overexpression of CAV1 enhances the efficacy of anti-PD-1 therapy. In conclusion, our study elucidates the regulatory role of CAV1 on ferroptosis within breast cancer contexts; it suppresses BCSC characteristics while positioning CAV1 as a promising therapeutic target for combating this disease.
Collapse
Affiliation(s)
- Wenhong Liu
- Department of Radiology, First Affiliated Hospital of University of South China, 69 Chuanshan Avenue, Hengyang City, Hunan, 421001, China
| | - Guanghua Luo
- Department of Radiology, First Affiliated Hospital of University of South China, 69 Chuanshan Avenue, Hengyang City, Hunan, 421001, China.
| |
Collapse
|
5
|
Sherwani S, Khan MWA, Khan WA, Rajendrasozhan S, Al-Motair K, Khan H, Ahmad S. Estrogenized HSA induced high-affinity autoantibodies in breast cancer - Novel biomarker for early detection. Front Oncol 2024; 14:1493320. [PMID: 39664179 PMCID: PMC11631743 DOI: 10.3389/fonc.2024.1493320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 10/29/2024] [Indexed: 12/13/2024] Open
Abstract
Objective Breast cancer (BC) is the second most prevalent cancer worldwide. Estrogen has been increasingly recognized as a major contributor to the development of BC, playing a more critical role than previously understood. Estrogen derived nucleic acid and protein adducts have been shown to play significant roles in BC development and progression. However, the alterations in molecular mechanism(s) and immune pathways arising as a result of estrogenization still remain elusive. Patients and methods 4-hydroxyestradiol (4-OHE2) was used for adduct formation with protein human serum albumin (HSA) (4-OHE2-HSA). The affinity of antibodies for 4-OHE2-HSA was evaluated in breast cancer patients. Immunoassays (direct binding ELISA, inhibition ELISA, and quantitative precipitin titration assay) were used to assess autoantibodies against estrogenized HSA in BC patients (n = 85) and healthy controls (n = 45). Results Estrogenization of HSA altered both its structure and function and compromised its interactions with various HSA-binding proteins. BC patients demonstrated high-affinity antibodies against 4-OHE2-HSA as compared to HSA (p < 0.05). Additionally, cytokines Interleukin (IL)-1, IL-6 and tumor necrosis factor-alpha (TNF-α) were significantly elevated in BC patients as compared to the control group. Several factors, such as chemotherapy, estrogen receptors (ERs), and combination of surgery and chemotherapy, influenced the production of antibodies in cancer patients. The affinity constant for estrogenized HSA was 1.31 × 10-7 M, while for HSA and 4-OHE2, it was 1.68 × 10-6 M and 1.36 × 10-6 M, respectively. Conclusions Estrogenized HSA is highly immunogenic, resulting in functional alterations. High affinity antibodies were detected in BC patients against 4-OHE2-HSA. Consequently, 4-OHE2-HSA may serve as a novel molecular target for potential cancer therapeutics. Furthermore, autoantibodies against 4-OHE2-HSA could serve as a potential biomarker for early detection of BC.
Collapse
Affiliation(s)
- Subuhi Sherwani
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
- Medical and Diagnostic Research Center, University of Hail, Hail, Saudi Arabia
| | - Mohd Wajid Ali Khan
- Medical and Diagnostic Research Center, University of Hail, Hail, Saudi Arabia
- Department of Chemistry, College of Science, University of Hail, Hail, Saudi Arabia
| | - Wahid Ali Khan
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Saravanan Rajendrasozhan
- Medical and Diagnostic Research Center, University of Hail, Hail, Saudi Arabia
- Department of Chemistry, College of Science, University of Hail, Hail, Saudi Arabia
| | - Khalid Al-Motair
- Medical and Diagnostic Research Center, University of Hail, Hail, Saudi Arabia
| | - Hamda Khan
- Department of Biochemistry, J.N. Medical College, Aligarh Muslim University, Aligarh, India
- IIRC-1, Department of Biosciences, Integral University, Lucknow, India
| | - Saheem Ahmad
- Medical and Diagnostic Research Center, University of Hail, Hail, Saudi Arabia
- Department of Medical Laboratories, College of Applied Medical Sciences, University of Hail, Hail, Saudi Arabia
| |
Collapse
|
6
|
Kumar S, Chaudhri S. Recent update on IGF-1/IGF-1R signaling axis as a promising therapeutic target for triple-negative breast cancer. Pathol Res Pract 2024; 263:155620. [PMID: 39357179 DOI: 10.1016/j.prp.2024.155620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 09/10/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Abstract
Insulin-like growth factor 1/Insulin-like growth factor 1-receptor (IGF-1/IGF-1R) pathway is highly breast cancer subtype context-dependent. Triple-negative breast cancer (TNBC) is an aggressive, highly metastatic cancer showing early recurrence and poor prognosis. High expression of IGF-1 and its receptor IGF-1R, their interaction, autophosphorylation, and activation of intracellular signaling cascades have been significantly associated with TNBC pathophysiology. In the last five to seven years, marvelous work has been done to explore the role of IGF-1/IGF-1R axis in TNBC. In the present review, starting from the general introduction to IGF-1/IGF-1R pathway an up-to-date discussion was focused on its role in TNBC pathophysiology. Further we discussed the up/down stream molecular events of IGF-1/IGF-1R axis, clinical relevance of IGF-1 and IGF-1R levels in TNBC patients, anti-TNBC therapy and possible way-out for IGF-1/IGF-1R axis mediate therapy resistance in TNBC. Combination therapy strategy has been researched to overcome direct IGF-1/IGF-1R pathway inhibition mediated therapy resistance and produced promising results in the management of TNBC. The understanding of up/downstream of the IGF-1/IGF-1R axis provide immense focus on the pathway as a therapeutic target. It is expected within the next decade to determine its potentiality, or lack thereof, for TNBC treatment.
Collapse
Affiliation(s)
- Shashank Kumar
- Molecular Signaling & Drug Discovery Laboratory, Department of Biochemistry, Central University of Punjab, Guddha, Bathinda, Punjab 151401, India.
| | - Smriti Chaudhri
- Molecular Signaling & Drug Discovery Laboratory, Department of Biochemistry, Central University of Punjab, Guddha, Bathinda, Punjab 151401, India
| |
Collapse
|
7
|
Korucu EN, Aydemir S, Menevse E, Erkoc Kaya D, Azzawri AA. Gene expression of MTATP6 and cytochrome P450 in MCF-7 and MDA-MB -231 breast cancer cell lines with juglone and curcumin supplemented. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2024:1-17. [PMID: 39437263 DOI: 10.1080/15257770.2024.2418907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 10/09/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024]
Abstract
It is aimed to determine the effects of naphthoquinones as juglone and curcumin application on cell viability and expression analyzes of CYP3A4 and MTATP6 genes in MCF-7 and MDA-MB-231 human breast cancer cell lines. MCF-7 and MDA-MB-231 cells were incubated, were replaced with containing various concentrations of 5, 10, 15 μM curcumin and 5, 10, 15 μM juglone for MCF-7 and 1, 5, 10 μM curcumin and 1, 2, 3 μM juglone for MDA-MB-231 for 24 h. CYP3A4 and MTATP6 gene expression levels in both cell lines were determined by quantitative real-time polymerase chain reaction (qPCR) method and western blot method. IC50 values for 24 h were found as 22.41 μM for curcumin, and 16.27 μM for juglone in MCF-7, and 10.43 μM for curcumin, and 3.42 μM for juglone in MDA-MB-231 cells. Curcumin showed anti-proliferative, and antioxidant effects. CYP3A4 and MTATP6 gene expressions were decreased in MCF-7 breast cancer cell line when the cells treated with juglone or curcumin. CYP3A4 and MTATP6 gene expressions were decreased at all application doses of juglone in MDA-MB-231 cells whereas CYP3A4 and MTATP6 protein levels were only decreased at 10 μM curcumin compared with the control group.
Collapse
Affiliation(s)
- Emine Nedime Korucu
- Department of Molecular Biology and Genetics, Faculty of Science, Necmettin Erbakan University, Konya, Turkey
| | - Saliha Aydemir
- Department of Molecular Biology and Genetics, Faculty of Science, Necmettin Erbakan University, Konya, Turkey
| | - Esma Menevse
- Department of Medical Biochemistry, Faculty of Medicine, Selcuk University, Konya, Turkey
| | - Dudu Erkoc Kaya
- Department of Medical Biology, Faculty of Medicine, Selcuk University, Konya, Turkey
| | - Ali Ahmed Azzawri
- Department of Medical Genetics, Faculty of Medicine, Selcuk University, Konya, Turkey
| |
Collapse
|
8
|
Santos TMR, Tavares CA, da Cunha EFF, Ramalho TC. Vanadium complex as a potential modulator of the autophagic mechanism through proteins PI3K and ULK1: development, validation and biological implications of a specific force field for [VO(bpy) 2Cl]. J Biomol Struct Dyn 2024; 42:9118-9132. [PMID: 37608540 DOI: 10.1080/07391102.2023.2250453] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 08/12/2023] [Indexed: 08/24/2023]
Abstract
The modulation of autophagy has been presented as a very useful strategy in anticancer treatments. In this sense, the vanadium complex (VC) bis(2,2'-bipyridine)chlorooxovanadium(IV), [VO(bpy)2Cl], is known for its ability to induce autophagy in triple-negative breast cancer cells (TNBC). An excellent resource to investigate the role of VC in the induction of autophagy is to make use of Molecular Dynamics (MD) simulations. However, until now, the scarcity of force field parameters for the VC prevented a reliable analysis. The autophagy signaling pathway starts with the PI3K protein and ends with ULK1. Therefore, in the first stage of this work, we developed a new AMBER force field for the VC (VCFF) from a quantum structure, obtained by DFT calculations. In the second stage, the VCFF was validated through structural analyses. From this, it was possible to investigate, through docking and MD (200 ns), the performance of the PI3K-VC and ULK1-VC systems (third stage). The analyses of this last stage involved RMSD, hydrogen bonds, RMSF and two pathways for the modulation of autophagy. In general, this work fills in the absence of force field parameters (FF) for VC by proposing an efficient and new FF, in addition to investigating, at the molecular level, how VC is able to induce autophagy in TNBC cells. This study encourages new parameterizations of metallic complexes and contributes to the understanding of the duality of autophagic processes.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Taináh M R Santos
- Laboratory of Molecular Modelling, Department of Chemistry, Federal University of Lavras, Lavras, MG, Brazil
| | - Camila A Tavares
- Laboratory of Molecular Modelling, Department of Chemistry, Federal University of Lavras, Lavras, MG, Brazil
| | - Elaine F F da Cunha
- Laboratory of Molecular Modelling, Department of Chemistry, Federal University of Lavras, Lavras, MG, Brazil
| | - Teodorico C Ramalho
- Laboratory of Molecular Modelling, Department of Chemistry, Federal University of Lavras, Lavras, MG, Brazil
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
9
|
Tian Y, He X, Yuan Y, Zhang S, Wang C, Dong J, Liu Z, Jing H. TME-Responsive Nanoplatform with Glutathione Depletion for Enhanced Tumor-Specific Mild Photothermal/Gene/Ferroptosis Synergistic Therapy. Int J Nanomedicine 2024; 19:9145-9160. [PMID: 39258005 PMCID: PMC11386068 DOI: 10.2147/ijn.s475698] [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: 06/25/2024] [Accepted: 08/28/2024] [Indexed: 09/12/2024] Open
Abstract
Background Triple negative breast cancer (TNBC) is one of the worst prognosis types of breast cancer that urgently needs effective therapy methods. However, cancer is a complicated disease that usually requires multiple treatment modalities. Methods A tumor microenvironment (TME)-responsive PFC/TRIM37@Fe-TA@HA (abbreviated as PTFTH) nanoplatform was constructed by coating Fe3+ and tannic acid (TA) on the surface of TRIM37-siRNA loaded phase-transition perfluorocarbon (PFC) nanodroplets and further modifying them with hyaluronic acid (HA) to achieve tumor-specific mild photothermal/gene/ferroptosis synergistic therapy (MPTT/GT/ Ferroptosis) in vitro. Once internalized into tumor cells through CD44 receptor-mediated active targeting, the HA shell of PTFTH would be preliminarily disassembled by hyaluronidase (HAase) to expose the Fe-TA metal-phenolic networks (MPNs), which would further degrade in response to an acidic lysosomal environment, leading to HAase/pH dual-responsive release of Fe3+ and PFC/TRIM37. Results PTFTH showed good biocompatibility in vitro. On the one hand, the released Fe3+ could deplete the overexpressed glutathione (GSH) through redox reactions and produce Fe2+, which in turn converts endogenous H2O2 into highly cytotoxic hydroxyl radicals (•OH) for chemodynamic therapy (CDT). On the other hand, the local hyperthermia generated by PTFTH under 808 nm laser irradiation could not only improve CDT efficacy through accelerating the Fe2+-mediated Fenton reaction, but also enhance TRIM37-siRNA delivery for gene therapy (GT). The consumption of GSH and accumulation of •OH synergistically augmented intracellular oxidative stress, resulting in substantial tumor cell ferroptosis. Moreover, PTFTH possessed outstanding contrast enhanced ultrasound (CEUS), photoacoustic imaging (PAI) and magnetic resonance imaging (MRI) ability. Conclusion This PTFTH based multiple-mode therapeutic strategy has successfully achieved a synergistic anticancer effect in vitro and has the potential to be translated into clinical application for tumor therapy in future.
Collapse
Affiliation(s)
- Yuhang Tian
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People's Republic of China
| | - Xiang He
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People's Republic of China
| | - Yanchi Yuan
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People's Republic of China
| | - Shijie Zhang
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, People's Republic of China
| | - Chunyue Wang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People's Republic of China
| | - Jialin Dong
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People's Republic of China
| | - Zhao Liu
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People's Republic of China
| | - Hui Jing
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People's Republic of China
| |
Collapse
|
10
|
Yu B, Xing Z, Tian X, Feng R. A Prognostic Risk Signature of Two Autophagy-Related Genes for Predicting Triple-Negative Breast Cancer Outcomes. BREAST CANCER (DOVE MEDICAL PRESS) 2024; 16:529-544. [PMID: 39246674 PMCID: PMC11379032 DOI: 10.2147/bctt.s475007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 08/17/2024] [Indexed: 09/10/2024]
Abstract
Background Triple-negative breast cancer (TNBC) is recognized as the most aggressive molecular subtype of breast cancer. Recent studies have highlighted the complex role of autophagy in the pathogenesis of TNBC. Methods In this study, we evaluated 18,330 genes, including 1111 autophagy-related genes, (ARGs), across 579 TNBC samples from online databases. Differentially expressed ARGs in TNBC were identified using high-throughput RNA-seq data from the Cancer Genome Atlas (TCGA). Prognostic factors were examined through Cox regression and multivariate Cox analyses, with predictive efficacy assessed using receiver operating characteristic (ROC) curves. A nomogram integrating the risk signature with clinicopathological factors, such as TNM stage, was developed. Immunohistochemical analysis of clinical samples was also conducted. Results EIF4EBP1 and NPAS3 were significantly correlated with prognostic outcomes in patients with TNBC. Multivariate Cox regression analysis demonstrated that the expression levels of these two genes were accurate predictors of disease progression in TNBC samples from TCGA and the GSE31519 dataset. The efficacy of this predictive model was validated using ROC curve analysis and calibration plots, confirming its ability to accurately estimate the 1-, 2-, and 3-year survival rates for individuals with TNBC. Additionally, EIF4EBP1 and NPAS3 expression influenced drug sensitivity in TNBC cell lines, with notably lower NPAS3 expression in TNBC tissues, particularly in Stage III cases. This study is the first to report NPAS3 expression in patients with TNBC. Conclusion The autophagy-related genes EIF4EBP1 and NPAS3 may serve as independent prognostic factors for individuals with TNBC.
Collapse
Affiliation(s)
- Bing Yu
- Department of Breast Surgery, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, 300100, People's Republic of China
| | - Zhimei Xing
- State Key Laboratory of Component‑Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, People's Republic of China
| | - Xiaoxuan Tian
- State Key Laboratory of Component‑Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, People's Republic of China
| | - Rui Feng
- Department of Breast Surgery, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, 300100, People's Republic of China
| |
Collapse
|
11
|
Banerjee R, Maitra I, Bhattacharya T, Banerjee M, Ramanathan G, Rayala SK, Venkatraman G, Rajeswari D. Next-generation biomarkers for prognostic and potential therapeutic enhancement in Triple negative breast cancer. Crit Rev Oncol Hematol 2024; 201:104417. [PMID: 38901639 DOI: 10.1016/j.critrevonc.2024.104417] [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: 05/01/2024] [Revised: 06/04/2024] [Accepted: 06/07/2024] [Indexed: 06/22/2024] Open
Abstract
Triple-negative breast carcinoma (TNBC) is one of the most challenging subtypes of breast carcinoma and it has very limited therapeutic options as it is highly aggressive. The prognostic biomarkers are crucial for early diagnosis of the tumor, it also helps in anticipating the trajectory of the illness and optimizing the therapy options. Several therapeutic biomarkers are being used. Among them, the next-generation biomarkers that include Circulating tumor (ct) DNA, glycogen, lipid, and exosome biomarkers provide intriguing opportunities for enhancing the prognosis of TNBC. Lipid and glycogen biomarkers serve as essential details on the development of the tumor along with the efficacy of the treatment, as it exhibits metabolic alteration linked to TNBC. Several types of biomarkers have predictive abilities in TNBC. Elevated levels are associated with worse outcomes. ctDNA being a noninvasive biomarker reveals the genetic composition of the tumor, as well as helps to monitor the progression of the disease. Traditional therapies are ineffective in TNBC due to a lack of receptors, targeted drug delivery provides a tailored approach to overcome drug resistance and site-specific action by minimizing the side effects in TNBC treatment. This enhances therapeutic outcomes against the aggressive nature of breast cancer. This paper includes all the recent biomarkers which has been researched so far in TNBC and the state of art for TNBC which is explored.
Collapse
Affiliation(s)
- Risav Banerjee
- Department of Biomedical Genetics, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Indrajit Maitra
- Department of Biomedical Genetics, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Trisha Bhattacharya
- Department of Biotechnology, Indian Academy Degree College, Autonomous, Hennur cross, Kalyan Nagar, Bengaluru, Karnataka 560043, India
| | - Manosi Banerjee
- Department of Biomedical Genetics, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Gnanasambandan Ramanathan
- Department of Biomedical Genetics, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Suresh Kumar Rayala
- Department of Biotechnology, Indian Institute of Technology, Madras, Tamil Nadu 600036, India
| | - Ganesh Venkatraman
- Department of Biomedical Genetics, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India.
| | - Devi Rajeswari
- Department of Biomedical Genetics, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India.
| |
Collapse
|
12
|
Wang Y, Zhang Y, Qi X. EP300 promotes tumor stemness via epigenetic activation of CRISP3 leading to lobaplatin resistance in triple-negative breast cancer. Hum Cell 2024; 37:1475-1488. [PMID: 38879857 DOI: 10.1007/s13577-024-01091-w] [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: 03/11/2024] [Accepted: 06/02/2024] [Indexed: 08/23/2024]
Abstract
Lobaplatin shows antitumor activity against a wide range of tumors, including triple-negative breast cancer (TNBC), and has been linked to cancer stem cell pool. Here, we investigated the molecular mechanisms behind lobaplatin resistance and stemness in vitro and in vivo. Two chemoresistance-related GEO data sets (GSE70690 and GSE103115) were included to screen out relevant genes. Cysteine-rich secretory protein 3 (CRISP3) was found to be overexpressed in lobaplatin-resistant TNBC and related to poor diagnosis. CRISP3 expression was significantly correlated with tumor stemness markers in lobaplatin-resistant cells. E1A-associated protein p300 (EP300) regulated CRISP3 expression by affecting the H3K27ac modification of the CRISP3 promoter. In addition, knocking down EP300 curbed the malignant biological behavior of lobaplatin-resistant cells, which was antagonized by CRISP3 overexpression. Collectively, our results highlight the EP300/CRISP3 axis as a key driver of lobaplatin resistance in TNBC and suggest that therapeutic targeting of this axis may be an effective strategy for enhancing platinum sensitivity in TNBC.
Collapse
Affiliation(s)
- Yan Wang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Yi Zhang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, 400038, People's Republic of China.
| | - Xiaowei Qi
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, 400038, People's Republic of China.
| |
Collapse
|
13
|
Hung CC, Tsai CL, Chin YT, Wang YC, Liu CH, Lin ML, Chen SS, He JL, Tsai CW, Su CH, Bau DAT, Chang WS. Impacts of Matrix Metalloproteinase-2 Promoter Genotypes on Breast Cancer Risk. Cancer Genomics Proteomics 2024; 21:502-510. [PMID: 39191502 PMCID: PMC11363925 DOI: 10.21873/cgp.20467] [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: 07/04/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND/AIM Matrix metalloproteinase-2 (MMP-2) has been implicated in the pathogenesis of breast cancer (BC). However, there is limited research on the role of MMP-2 genotypes in BC risk. This study aimed to investigate the associations between two MMP-2 promoter polymorphisms, rs243865 and rs2285053, and BC risk. MATERIALS AND METHODS MMP-2 genotypes were analyzed using PCR-based RFLP methodology in a cohort comprising 1,232 BC cases and 1,232 controls. RESULTS Genotypic frequencies of MMP-2 rs243865 and rs2285053 in controls were consistent with Hardy-Weinberg equilibrium (p=0.3702 and 0.2036, respectively). There were no significant differences in the distribution of rs243865 and rs2285053 genotypes between BC cases and controls (p for trend=0.1602 and 0.2170, respectively). Variant genotypes at rs243865 and rs2285053 appeared to confer a protective effect, although not statistically significant (all p>0.05). Similarly, the variant T allele at rs243865 and rs2285053 showed a non-significant trend towards decreased BC risk (OR=0.84 and 0.89, 95%CI=0.69-1.02 and 0.78-1.02, p=0.0811 and 0.1043, respectively). There was no interaction observed between MMP-2 rs243865 or rs2285053 genotypes and age. Stratified analysis did not reveal significant associations between MMP-2 rs243865 or rs2285053 genotypes and triple-negative breast cancer (TNBC) (p=0.6458 and 0.8745, respectively). Among both TNBC and non-TNBC cases, none of the variant genotypes at rs243865 or rs2285053 showed significant associations with TNBC (all p>0.05). CONCLUSION MMP-2 rs243865 and rs2285053 genotypes appear to have a minimal impact on individual susceptibility to BC or TNBC.
Collapse
Affiliation(s)
- Chih-Chiang Hung
- Division of Breast Surgery, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan, R.O.C
| | - Chung-Lin Tsai
- Division of Cardiac and Vascular Surgery, Cardiovascular Center, Taichung Veterans General Hospital, Taichung, Taiwan, R.O.C
| | - Yu-Ting Chin
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Yun-Chi Wang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Chia-Hua Liu
- Division of Breast Surgery, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan, R.O.C
| | - Meng-Liang Lin
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan, R.O.C
| | - Shih-Shun Chen
- Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan, R.O.C
| | - Jie-Long He
- Department of Post-Baccalaureate Veterinary Medicine, Asia University, Taichung, Taiwan, R.O.C
| | - Chia-Wen Tsai
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Chen-Hsien Su
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - DA-Tian Bau
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C.;
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan, R.O.C
| | - Wen-Shin Chang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C.;
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| |
Collapse
|
14
|
Abdulmalek SA, Saleh AM, Shahin YR, El Azab EF. Functionalized siRNA-chitosan nanoformulations promote triple-negative breast cancer cell death via blocking the miRNA-21/AKT/ERK signaling axis: in-silico and in vitro studies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6941-6962. [PMID: 38592437 PMCID: PMC11422444 DOI: 10.1007/s00210-024-03068-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 03/22/2024] [Indexed: 04/10/2024]
Abstract
Oncogenic microRNA (miRNA), especially miRNA-21 upregulation in triple-negative breast cancer (TNBC), suggests a new class of therapeutic targets. In this study, we aimed to create GE11 peptide-conjugated small interfering RNA-loaded chitosan nanoparticles (GE11-siRNA-CSNPs) for the targeting of EGFR overexpressed TNBC and selectively inhibit miRNA-21 expression. A variety of in-silico and in vitro cellular and molecular studies were conducted to investigate the binding affinities of specific targets used as well as the anticancer efficacies and mechanisms of GE11-siRNA-CSNPs in TNBC cells. An in-silico assessment reveals a distinct binding affinity of miRNA-21 with siRNA as well as between the extracellular domain of EGFR and synthesized peptides. Notably, the in vitro results showed that GE11-siRNA-CSNPs were revealed to have better cytotoxicity against TNBC cells. It significantly inhibits miRNA-21 expression, cell migration, and colony formation. The results also indicated that GE11-siRNA-CSNPs impeded cell cycle progression. It induces cell death by reducing the expression of the antiapoptotic gene Bcl-2 and increasing the expression of the proapoptotic genes Bax, Caspase 3, and Caspase 9. Additionally, the docking analysis and immunoblot investigations verified that GE1-siRNA-CSNPs, which specifically target TNBC cells and suppress miRNA-21, can prevent the effects of miRNA-21 on the proliferation of TNBC cells via controlling EGFR and subsequently inhibiting the PI3K/AKT and ERK1/2 signaling axis. The GE11-siRNA-CSNPs design, which specifically targets TNBC cells, offers a novel approach for the treatment of breast cancer with improved effectiveness. This study suggests that GE11-siRNA-CSNPs could be a promising candidate for further assessment as an additional strategy in the treatment of TNBC.
Collapse
Affiliation(s)
- Shaymaa A Abdulmalek
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, 21511, Egypt.
| | - Abdulrahman M Saleh
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El‑Aini Street, Cairo, 11562, Egypt
- Aweash El-Hagar Family Medicine Center, Epidemiological Surveillance Unit, MOHP, Mansoura, 35711, Egypt
| | - Yasmin R Shahin
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, 21511, Egypt
| | - Eman Fawzy El Azab
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, 21511, Egypt
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences at Al-Qurayyat, Jouf University, Al-Qurayyat, 77454, Saudi Arabia
| |
Collapse
|
15
|
Zanini E, Forster-Gross N, Bachmann F, Brüngger A, McSheehy P, Litherland K, Burger K, Groner AC, Roceri M, Bury L, Stieger M, Willemsen-Seegers N, de Man J, Vu-Pham D, van Riel HWE, Zaman GJR, Buijsman RC, Kellenberger L, Lane HA. Dual TTK/PLK1 inhibition has potent anticancer activity in TNBC as monotherapy and in combination. Front Oncol 2024; 14:1447807. [PMID: 39184047 PMCID: PMC11341980 DOI: 10.3389/fonc.2024.1447807] [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: 06/12/2024] [Accepted: 07/23/2024] [Indexed: 08/27/2024] Open
Abstract
Background Threonine tyrosine kinase (TTK) and polo-like kinase 1 (PLK1) are common essential kinases that collaborate in activating the spindle assembly checkpoint (SAC) at the kinetochore, ensuring appropriate chromosome alignment and segregation prior to mitotic exit. Targeting of either TTK or PLK1 has been clinically evaluated in cancer patients; however, dual inhibitors have not yet been pursued. Here we present the in vitro and in vivo characterization of a first in class, dual TTK/PLK1 inhibitor (BAL0891). Methods Mechanism of action studies utilized biochemical kinase and proteomics-based target-engagement assays. Cellular end-point assays included immunoblot- and flow cytometry-based cell cycle analyses and SAC integrity evaluation using immunoprecipitation and immunofluorescence approaches. Anticancer activity was assessed in vitro using cell growth assays and efficacy was evaluated, alone and in combination with paclitaxel and carboplatin, using mouse models of triple negative breast cancer (TNBC). Results BAL0891 elicits a prolonged effect on TTK, with a transient activity on PLK1. This unique profile potentiates SAC disruption, forcing tumor cells to aberrantly exit mitosis with faster kinetics than observed with a TTK-specific inhibitor. Broad anti-proliferative activity was demonstrated across solid tumor cell lines in vitro. Moreover, intermittent intravenous single-agent BAL0891 treatment of the MDA-MB-231 mouse model of TNBC induced profound tumor regressions associated with prolonged TTK and transient PLK1 in-tumor target occupancy. Furthermore, differential tumor responses across a panel of thirteen TNBC patient-derived xenograft models indicated profound anticancer activity in a subset (~40%). Using a flexible dosing approach, pathologically confirmed cures were observed in combination with paclitaxel, whereas synergy with carboplatin was schedule dependent. Conclusions Dual TTK/PLK1 inhibition represents a novel approach for the treatment of human cancer, including TNBC patients, with a potential for potent anticancer activity and a favorable therapeutic index. Moreover, combination approaches may provide an avenue to expand responsive patient populations.
Collapse
Affiliation(s)
- Elisa Zanini
- Basilea Pharmaceutica International Ltd, Allschwil, Switzerland
| | | | - Felix Bachmann
- Basilea Pharmaceutica International Ltd, Allschwil, Switzerland
| | - Adrian Brüngger
- Basilea Pharmaceutica International Ltd, Allschwil, Switzerland
| | - Paul McSheehy
- Basilea Pharmaceutica International Ltd, Allschwil, Switzerland
| | | | - Karin Burger
- Basilea Pharmaceutica International Ltd, Allschwil, Switzerland
| | - Anna C. Groner
- Basilea Pharmaceutica International Ltd, Allschwil, Switzerland
| | - Mila Roceri
- Basilea Pharmaceutica International Ltd, Allschwil, Switzerland
| | - Luc Bury
- Basilea Pharmaceutica International Ltd, Allschwil, Switzerland
| | - Martin Stieger
- Basilea Pharmaceutica International Ltd, Allschwil, Switzerland
| | | | - Jos de Man
- Crossfire Oncology B.V., Oss, Netherlands
| | | | | | | | | | | | - Heidi A. Lane
- Basilea Pharmaceutica International Ltd, Allschwil, Switzerland
| |
Collapse
|
16
|
Zou Z, Luo T, Wang X, Wang B, Li Q. Exploring the interplay between triple-negative breast cancer stem cells and tumor microenvironment for effective therapeutic strategies. J Cell Physiol 2024; 239:e31278. [PMID: 38807378 DOI: 10.1002/jcp.31278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/28/2024] [Accepted: 04/05/2024] [Indexed: 05/30/2024]
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive and metastatic malignancy with poor treatment outcomes. The interaction between the tumor microenvironment (TME) and breast cancer stem cells (BCSCs) plays an important role in the development of TNBC. Owing to their ability of self-renewal and multidirectional differentiation, BCSCs maintain tumor growth, drive metastatic colonization, and facilitate the development of drug resistance. TME is the main factor regulating the phenotype and metastasis of BCSCs. Immune cells, cancer-related fibroblasts (CAFs), cytokines, mesenchymal cells, endothelial cells, and extracellular matrix within the TME form a complex communication network, exert highly selective pressure on the tumor, and provide a conducive environment for the formation of BCSC niches. Tumor growth and metastasis can be controlled by targeting the TME to eliminate BCSC niches or targeting BCSCs to modify the TME. These approaches may improve the treatment outcomes and possess great application potential in clinical settings. In this review, we summarized the relationship between BCSCs and the progression and drug resistance of TNBC, especially focusing on the interaction between BCSCs and TME. In addition, we discussed therapeutic strategies that target the TME to inhibit or eliminate BCSCs, providing valuable insights into the clinical treatment of TNBC.
Collapse
Affiliation(s)
- Zhuoling Zou
- Queen Mary College, Nanchang University, Nanchang, Jiangxi, China
| | - Tinglan Luo
- Department of Oncology, The Seventh People's Hospital of Chongqing (Affiliated Central Hospital of Chongqing University of Technology), Chongqing, China
| | - Xinyuan Wang
- Department of Clinical Medicine, The Second Clinical College of Chongqing Medicine University, Chongqing, China
| | - Bin Wang
- Department of Oncology, The Seventh People's Hospital of Chongqing (Affiliated Central Hospital of Chongqing University of Technology), Chongqing, China
| | - Qing Li
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
17
|
Long L, Xu J, Qi X, Pen Y, Wang C, Jiang W, Peng X, Hu Z, Yi W, Xie L, Lei X, Wang Z, Zhuo L. Discovery of novel small molecules targeting the USP21/JAK2/STAT3 axis for the treatment of triple-negative breast cancer. Eur J Med Chem 2024; 273:116500. [PMID: 38776807 DOI: 10.1016/j.ejmech.2024.116500] [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: 03/12/2024] [Revised: 05/10/2024] [Accepted: 05/12/2024] [Indexed: 05/25/2024]
Abstract
The deficiency in available targeted agents and frequency of chemoresistance are primary challenges in clinical management of triple-negative breast cancer (TNBC). The aberrant expression of USP21 and JAK2 represents a characterized mechanism of TNBC progression and resistance to paclitaxel (PTX). Despite its clear that high expression of USP21-mediated de-ubiquitination leads to increased levels of JAK2 protein, we lack regulator molecules to dissect the mechanisms that the interaction between USP21 and JAK2 contributes to the phenotype and resistance of TNBC. Here, we report a USP21/JAK2/STAT3 axis-targeting regulator 13c featuring a N-anthraniloyl tryptamine scaffold that showed excellent anti-TNBC potency and promising safety profile. Importantly, the therapeutic potential of using 13c in combination with PTX in PTX-resistant TNBC was demonstrated. This study showcases N-anthraniloyl tryptamine derivatives as a novel anti-TNBC chemotype with a pharmacological mode of action targeting the USP21/JAK2/STAT3 axis and provides a potential therapeutic target for the treatment of TNBC.
Collapse
Affiliation(s)
- Lin Long
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China; School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Jiachi Xu
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China; Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Xiaowen Qi
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yan Pen
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Chengkun Wang
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Weifan Jiang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Xue Peng
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Zecheng Hu
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Wenjun Yi
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Liming Xie
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Xiaoyong Lei
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China; School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Zhen Wang
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China; School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China; National Health Commission Key Laboratory of Birth Defect Research and Prevention Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan 410008, China.
| | - Linsheng Zhuo
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China; School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China; National Health Commission Key Laboratory of Birth Defect Research and Prevention Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan 410008, China.
| |
Collapse
|
18
|
Cai X, Liu W, Zhang J, Li Z, Liu M, Hu S, Luo J, Peng K, Ye B, Wang Y, Yan R. Study of Iron Complex Photosensitizer with Hollow Double-Shell Nano Structure Used to Enhance Ferroptosis and Photodynamic Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309086. [PMID: 38321834 DOI: 10.1002/smll.202309086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/07/2023] [Indexed: 02/08/2024]
Abstract
Ferroptosis therapy, which uses ferroptosis inducers to produce lethal lipid peroxides and induce tumor cell death, is considered a promising cancer treatment strategy. However, challenges remain regarding how to increase the accumulation of reactive oxygen species (ROS) in the tumor microenvironment (TME) to enhance antitumor efficacy. In this study, a hyaluronic acid (HA) encapsulated hollow mesoporous manganese dioxide (H-MnO2) with double-shell nanostructure is designed to contain iron coordinated cyanine near-infrared dye IR783 (IR783-Fe) for synergistic ferroptosis photodynamic therapy against tumors. The nano photosensitizer IR783-Fe@MnO2-HA, in which HA actively targets the CD44 receptor, subsequently dissociates and releases Fe3+ and IR783 in acidic TME. First, Fe3+ consumes glutathione to produce Fe2+, which promotes the Fenton reaction in cells to produce hydroxyl free radicals (·OH) and induce ferroptosis of tumor cells. In addition, MnO2 catalyzes the production of O2 from H2O2 and enhances the production of singlet oxygen (1O2) by IR783 under laser irradiation, thus increasing the production and accumulation of ROS to provide photodynamic therapy. The highly biocompatible IR783-Fe@MnO2-HA nano-photosensitizers have exhibited tumor-targeting ability and efficient tumor inhibition in vivo due to the synergistic effect of photodynamic and ferroptosis antitumor therapies.
Collapse
Affiliation(s)
- Xinrui Cai
- Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Weixing Liu
- Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Jiahao Zhang
- Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Zhongrui Li
- Electron Microbeam Analysis Laboratory, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Mengkang Liu
- Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Shuo Hu
- Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Jun Luo
- Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Kai Peng
- Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Baofen Ye
- Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Yue Wang
- Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Ran Yan
- Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing, 211198, China
| |
Collapse
|
19
|
Li M, Kang S, Deng X, Li H, Zhao Y, Tang W, Sheng M. Erianin inhibits the progression of triple-negative breast cancer by suppressing SRC-mediated cholesterol metabolism. Cancer Cell Int 2024; 24:166. [PMID: 38734640 PMCID: PMC11088164 DOI: 10.1186/s12935-024-03332-2] [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: 12/12/2023] [Accepted: 04/18/2024] [Indexed: 05/13/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is highly malignant and lacks effective biotherapeutic targets. The development of efficient anticancer drugs with low toxicity and few side effects is a hotspot in TNBC treatment research. Although erianin is known to have potent antitumor activity, its regulatory mechanism and target in TNBC have not been fully elucidated, hampering further drug development. This study showed that erianin can significantly inhibit TNBC cell proliferation and migration, promote cell apoptosis, and inhibit the growth of transplanted tumors in mice. Mechanistically, through network pharmacology analysis, molecular docking and cellular thermal shift assays, we preliminarily identified SRC as the cellular target of erianin. Erianin potently inhibited the expression of SRC, which mediated the anticancer effect of erianin in TNBC. Moreover, erianin can downregulate the expression of genes related to cholesterol synthesis and uptake by targeting SRC, interfering with cholesterol levels in TNBC, thereby inhibiting the progression of TNBC in vivo and in vitro. Taken together, our results suggest that erianin may inhibit the progression of TNBC by suppressing SRC-mediated cholesterol metabolism, and erianin has the great potential to be an effective treatment for TNBC patients.
Collapse
Affiliation(s)
- Ming Li
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
| | - Shiyao Kang
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
| | - Xuming Deng
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
| | - Huimin Li
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
| | - Yuan Zhao
- Kunming University of Science and Technology Affiliated Puer City People's Hospital, Puer, Yunnan, 665000, China
| | - Wenru Tang
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
| | - Miaomiao Sheng
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China.
| |
Collapse
|
20
|
Cordani M, Dando I, Ambrosini G, González-Menéndez P. Signaling, cancer cell plasticity, and intratumor heterogeneity. Cell Commun Signal 2024; 22:255. [PMID: 38702718 PMCID: PMC11067149 DOI: 10.1186/s12964-024-01643-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2024] Open
Abstract
Cancer's complexity is in part due to the presence of intratumor heterogeneity and the dynamic nature of cancer cell plasticity, which create substantial obstacles in effective cancer management. Variability within a tumor arises from the existence of diverse populations of cancer cells, impacting the progression, spread, and resistance to treatments. At the core of this variability is the concept of cellular plasticity - the intrinsic ability of cancer cells to alter their molecular and cellular identity in reaction to environmental and genetic changes. This adaptability is a cornerstone of cancer's persistence and progression, making it a formidable target for treatments. Emerging studies have emphasized the critical role of such plasticity in fostering tumor diversity, which in turn influences the course of the disease and the effectiveness of therapeutic strategies. The transformative nature of cancer involves a network of signal transduction pathways, notably those that drive the epithelial-to-mesenchymal transition and metabolic remodeling, shaping the evolutionary path of cancer cells. Despite advancements, our understanding of the precise molecular machinations and signaling networks driving these changes is still evolving, underscoring the necessity for further research. This editorial presents a series entitled "Signaling Cancer Cell Plasticity and Intratumor Heterogeneity" in Cell Communication and Signaling, dedicated to unraveling these complex processes and proposing new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Complutense University, Madrid, 28040, Spain.
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, 28040, Spain.
| | - Ilaria Dando
- Department of Neuroscience, Biomedicine and Movement Sciences, Biochemistry Section, University of Verona, Verona, 37134, Italy.
| | - Giulia Ambrosini
- Department of Neuroscience, Biomedicine and Movement Sciences, Biochemistry Section, University of Verona, Verona, 37134, Italy.
| | - Pedro González-Menéndez
- Departamento de Morfología y Biología Celular, School of Medicine, Julián Claveria 6, Oviedo, 33006, Spain.
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, 33006, Spain.
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias (HUCA), Oviedo, 33011, Spain.
| |
Collapse
|
21
|
Ferreira Almeida C, Correia-da-Silva G, Teixeira N, Amaral C. Influence of tumor microenvironment on the different breast cancer subtypes and applied therapies. Biochem Pharmacol 2024; 223:116178. [PMID: 38561089 DOI: 10.1016/j.bcp.2024.116178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/15/2024] [Accepted: 03/28/2024] [Indexed: 04/04/2024]
Abstract
Despite the significant improvements made in breast cancer therapy during the last decades, this disease still has increasing incidence and mortality rates. Different targets involved in general processes, like cell proliferation and survival, have become alternative therapeutic options for this disease, with some of them already used in clinic, like the CDK4/6 inhibitors for luminal A tumors treatment. Nevertheless, there is a demand for novel therapeutic strategies focused not only on tumor cells, but also on their microenvironment. Tumor microenvironment (TME) is a very complex and dynamic system that, more than surrounding and supporting tumor cells, actively participates in tumor development and progression. During the last decades, it has become clear that the cellular and acellular components of TME differ between the various breast cancer subtypes and shape the differences regarding their severity and prognosis. The pivotal role of the TME in controlling tumor growth and influencing responses to therapy represents a potential source for novel targets and therapeutic strategies. In this review, we present a description of the multiple therapeutic options used for different breast cancer subtypes, as well as the influence that the TME may exert on the development of the disease and on the response to the distinct therapies, which in some cases may explain their failure by the occurrence of relapses and resistance. Furthermore, the ongoing studies focused on the use of TME components for developing potential cancer treatments are described.
Collapse
Affiliation(s)
- Cristina Ferreira Almeida
- UCIBIO, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal
| | - Georgina Correia-da-Silva
- UCIBIO, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal.
| | - Natércia Teixeira
- UCIBIO, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal
| | - Cristina Amaral
- UCIBIO, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal.
| |
Collapse
|
22
|
Wasson MCD, Venkatesh J, Cahill HF, McLean ME, Dean CA, Marcato P. LncRNAs exhibit subtype-specific expression, survival associations, and cancer-promoting effects in breast cancer. Gene 2024; 901:148165. [PMID: 38219875 DOI: 10.1016/j.gene.2024.148165] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 12/25/2023] [Accepted: 01/11/2024] [Indexed: 01/16/2024]
Abstract
Long non-coding RNAs (lncRNAs) play important roles in cancer progression, influencing processes such as invasion, metastasis, and drug resistance. Their reported cell type-dependent expression patterns suggest the potential for specialized functions in specific contexts. In breast cancer, lncRNA expression has been associated with different subtypes, highlighting their relevance in disease heterogeneity. However, our understanding of lncRNA function within breast cancer subtypes remains limited, warranting further investigation. We conducted a comprehensive analysis using the TANRIC dataset derived from the TCGA-BRCA cohort, profiling the expression, patient survival associations and immune cell type correlations of 12,727 lncRNAs across subtypes. Our findings revealed subtype-specific associations of lncRNAs with patient survival, tumor infiltrating lymphocytes and other immune cells. Targeting of lncRNAs exhibiting subtype-specific survival associations and expression in a panel of breast cancer cells demonstrated a selective reduction in cell proliferation within their associated subtype, supporting subtype-specific functions of certain lncRNAs. Characterization of HER2 + -specific lncRNA LINC01269 and TNBC-specific lncRNA AL078604.2 showed nuclear localization and altered expression of hundreds of genes enriched in cancer-promoting processes, including apoptosis, cell proliferation and immune cell regulation. This work emphasizes the importance of considering the heterogeneity of breast cancer subtypes and the need for subtype-specific analyses to fully uncover the relevance and potential impact of lncRNAs. Collectively, these findings demonstrate the contribution of lncRNAs to the distinct molecular, prognostic, and cellular composition of breast cancer subtypes.
Collapse
Affiliation(s)
| | | | - Hannah F Cahill
- Department of Pathology, Dalhousie University, Halifax, NS B3H4R2, Canada
| | - Meghan E McLean
- Department of Pathology, Dalhousie University, Halifax, NS B3H4R2, Canada
| | - Cheryl A Dean
- Department of Pathology, Dalhousie University, Halifax, NS B3H4R2, Canada
| | - Paola Marcato
- Department of Pathology, Dalhousie University, Halifax, NS B3H4R2, Canada; Department of Microbiology & Immunology, Dalhousie University, Halifax, NS B3H4R2, Canada; Nova Scotia Health Authority, Halifax, NS B3H1V8, Canada.
| |
Collapse
|
23
|
Zhang Y, Xiang Z, Chen L, Deng X, Liu H, Peng X. PSMA2 promotes glioma proliferation and migration via EMT. Pathol Res Pract 2024; 256:155278. [PMID: 38574629 DOI: 10.1016/j.prp.2024.155278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/15/2024] [Accepted: 03/26/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND Gliomas advance rapidly and are associated with a poor prognosis. Epithelial-mesenchymal transition (EMT) accelerates the progression of gliomas, exerting a pivotal role in glioma development. Proteasome subunit alpha type-2 (PSMA2) exhibits high expression levels in gliomas. however, its specific involvement in glioma progression and its correlation with EMT remain elusive. This study aims to elucidate the role of PSMA2 in glioma progression and its potential association with EMT. METHODS Online tools were employed to analyze the expression patterns and survival curves of PSMA2 in gliomas. The relationship between PSMA2 and various characteristics of glioma patients was investigated using data from the TCGA and CGGA databases. In vitro, cell proliferation and migration were assessed through CCK-8, colony formation, and transwell assays. Furthermore, a tumor xenograft model in nude mice was established to evaluate in vivo tumorigenesis. Protein binding to PSMA2 was scrutinized using co-immunoprecipitation MS (co-IP MS). The potential biological functions and molecular pathways associated with PSMA2 were explored through GO analysis and KEGG analysis, and the correlation between PSMA2 and EMT was validated through correlation analysis and Western blot experiments. RESULTS Bioinformatics analysis revealed a significant upregulation of PSMA2 across various cancers, with particularly heightened expression in gliomas. Moreover, elevated PSMA2 levels were correlated with advanced tumor stages and diminished survival rates among glioma patients. Inhibition of PSMA2 demonstrated a pronounced suppressive effect on glioma cell proliferation, both in vitro and in vivo. Knockdown of PSMA2 also impeded the migratory capacity of glioma cells. GO and KEGG enrichment analyses indicated that PSMA2-binding proteins (identified through Co-IP-MS) were associated with cell adhesion molecule binding and cadherin binding. Western blot results further confirmed the role of PSMA2 in promoting epithelial-mesenchymal transition (EMT) in glioma cells. CONCLUSION Our study provides evidence supporting the role of PSMA2 as a regulatory factor in EMT and suggests its potential as a prognostic biomarker for glioma progression.
Collapse
Affiliation(s)
- Yujun Zhang
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Zijin Xiang
- Department of Pharmacy, Shaodong People's Hospital, Shaodong, Hunan 422800, China
| | - Le Chen
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Xingyan Deng
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Huaizheng Liu
- Department of Emergency, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Xiangdong Peng
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China.
| |
Collapse
|
24
|
Zheng S, Yang B, Li L, Chen M, Zhang L, Chi W, Shao ZM, Xiu B, Chi Y, Wu J. CRTAM promotes antitumor immune response in triple negative breast cancer by enhancing CD8+ T cell infiltration. Int Immunopharmacol 2024; 129:111625. [PMID: 38354509 DOI: 10.1016/j.intimp.2024.111625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/22/2024] [Accepted: 01/29/2024] [Indexed: 02/16/2024]
Abstract
The immunomodulatory (IM) subtype of triple negative breast cancer (TNBC) exhibits high expression of immune cell signaling genes and is more responsive to immunotherapy. However, the specific mechanism underlying this phenomenon remains unclear. One of the potential key genes appears to be the cytotoxic and regulatory T cell molecule (CRTAM). A cohort of 360 previously untreated TNBC patients from Fudan University Shanghai Cancer Center (FUSCC) underwent RNA sequencing analysis of their primary tumor tissue. Combined with three RNA-seq datasets obtained from the GEO database, a LASSO regression analysis was conducted to identify genes specific to the IM type of TNBC. Our findings revealed elevated CRTAM expression in the IM-type TNBC, which correlated with a favorable overall survival and recurrence-free survival in TNBC patients. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis indicated a strong association between CRTAM and immune responses as well as immune system processes. Notably, CRTAM overexpression induced STAT1 phosphorylation and upregulation of interferon-stimulated genes. We also found that CRTAM enhanced tumor-associated immune cell infiltration, especially CD8+ T cells, which may be related to the increased expression of MHC class I molecules caused by CRTAM overexpression. These results suggest that CRTAM may serve as a potential biomarker for predicting the efficacy of immunotherapy in TNBC.
Collapse
Affiliation(s)
- Shuyue Zheng
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Benlong Yang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lun Li
- Department of Breast Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ming Chen
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liyi Zhang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Weiru Chi
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bingqiu Xiu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Yayun Chi
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Jiong Wu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
25
|
Gali A, Bijnsdorp IV, Piersma SR, Pham TV, Gutiérrez-Galindo E, Kühnel F, Tsolakos N, Jimenez CR, Hausser A, Alexopoulos LG. Protein kinase D drives the secretion of invasion mediators in triple-negative breast cancer cell lines. iScience 2024; 27:108958. [PMID: 38323010 PMCID: PMC10844833 DOI: 10.1016/j.isci.2024.108958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 11/28/2023] [Accepted: 01/15/2024] [Indexed: 02/08/2024] Open
Abstract
The protein kinase D (PKD) family members regulate the fission of cargo vesicles at the Golgi complex and play a pro-oncogenic role in triple-negative breast cancer (TNBC). Whether PKD facilitates the secretion of tumor-promoting factors in TNBC, however, is still unknown. Using the pharmacological inhibition of PKD activity and siRNA-mediated depletion of PKD2 and PKD3, we identified the PKD-dependent secretome of the TNBC cell lines MDA-MB-231 and MDA-MB-468. Mass spectrometry-based proteomics and antibody-based assays revealed a significant downregulation of extracellular matrix related proteins and pro-invasive factors such as LIF, MMP-1, MMP-13, IL-11, M-CSF and GM-CSF in PKD-perturbed cells. Notably, secretion of these proteins in MDA-MB-231 cells was predominantly controlled by PKD2 and enhanced spheroid invasion. Consistently, PKD-dependent secretion of pro-invasive factors was more pronounced in metastatic TNBC cell lines. Our study thus uncovers a novel role of PKD2 in releasing a pro-invasive secretome.
Collapse
Affiliation(s)
- Alexia Gali
- Biomedical Systems Laboratory, National Technical University of Athens, 15780 Athens, Greece
- Protavio Ltd, Demokritos Science Park, 15341 Athens, Greece
| | - Irene V. Bijnsdorp
- Department of Urology, Cancer Center Amsterdam, Cancer Center Amsterdam, Amsterdam UMC, de Boelelaan 1117, Amsterdam 1081 HV, the Netherlands
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, OncoProteomics Laboratory, de Boelelaan 1117, , Amsterdam 1081 HV, the Netherlands
| | - Sander R. Piersma
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, OncoProteomics Laboratory, de Boelelaan 1117, , Amsterdam 1081 HV, the Netherlands
| | - Thang V. Pham
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, OncoProteomics Laboratory, de Boelelaan 1117, , Amsterdam 1081 HV, the Netherlands
| | | | - Fiona Kühnel
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569 Stuttgart, Germany
| | - Nikos Tsolakos
- Protavio Ltd, Demokritos Science Park, 15341 Athens, Greece
| | - Connie R. Jimenez
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, OncoProteomics Laboratory, de Boelelaan 1117, , Amsterdam 1081 HV, the Netherlands
| | - Angelika Hausser
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569 Stuttgart, Germany
- Stuttgart Research Center for Systems Biology, University of Stuttgart, 70569 Stuttgart, Germany
| | - Leonidas G. Alexopoulos
- Biomedical Systems Laboratory, National Technical University of Athens, 15780 Athens, Greece
- Protavio Ltd, Demokritos Science Park, 15341 Athens, Greece
| |
Collapse
|
26
|
Li M, Zhao Y, Li H, Kang S, Deng X, Sheng M. Mechanism of Erianin anti-triple negative breast cancer based on transcriptomics methods and network pharmacology. Aging (Albany NY) 2024; 16:2848-2865. [PMID: 38329441 PMCID: PMC10911376 DOI: 10.18632/aging.205516] [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: 09/04/2023] [Accepted: 12/13/2023] [Indexed: 02/09/2024]
Abstract
Triple negative breast cancer (TNBC) is a highly aggressive illness that lacks effective targeted treatments. Although Erianin has shown potential antitumor properties, its precise mechanism of action and target in TNBC remain unclear, hampering the development of drugs. The present study investigated the underlying mechanism of action of Erianin in treating TNBC by using transcriptomics and network pharmacology approaches. We evaluated Erianin's bioactivity in TNBC cell lines and xenograft tumor models. The results showed that Erianin significantly inhibited TNBC cell proliferation and impeded tumor growth. A subsequent analysis of transcriptomic and network pharmacological data identified 51 mutual targets. Analysis of protein-protein interactions identified eight hub targets. Furthermore, molecular docking indicated that the PPARA binding energy was the lowest for Erianin among the hub targets, followed by ROCK2, PDGFRB, CCND1, MUC1, and CDK1. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes functional enrichment analysis showed that the common targets were associated with multiple cancer-related signaling pathways, including focal adhesion, PI3K-Akt signaling pathway, Rap1 signaling pathway, microRNAs in cancer, and human papillomavirus infection. The results of the Western blot and immunohistochemistry experiment further showed that Erianin could suppress PI3K/Akt signaling pathway activation. After co-incubation with SC79, the cell inhibition rate of Erianin was decreased, which further confirmed that Erianin inhibits TNBC progression via the PI3K-AKT signaling pathway. In conclusion, our results indicated that Erianin has the potential to inhibit the proliferation of TNBC by downregulating the PI3K/AKT signaling pathway by transcriptomics and network pharmacology. Therefore, Erianin appears to be a promising compound for the effective treatment of TNBC.
Collapse
Affiliation(s)
- Ming Li
- Laboratory of Molecular Genetics of Aging and Tumour, Medical School, Kunming University of Science and Technology, Chenggong Campus, Kunming, Yunnan 650500, China
| | - Yuan Zhao
- Kunming University of Science and Technology Affiliated Puer City People’s Hospital, Puer, Yunnan 665000, China
| | - Huimin Li
- Laboratory of Molecular Genetics of Aging and Tumour, Medical School, Kunming University of Science and Technology, Chenggong Campus, Kunming, Yunnan 650500, China
| | - Shiyao Kang
- Laboratory of Molecular Genetics of Aging and Tumour, Medical School, Kunming University of Science and Technology, Chenggong Campus, Kunming, Yunnan 650500, China
| | - Xuming Deng
- Laboratory of Molecular Genetics of Aging and Tumour, Medical School, Kunming University of Science and Technology, Chenggong Campus, Kunming, Yunnan 650500, China
| | - Miaomiao Sheng
- Laboratory of Molecular Genetics of Aging and Tumour, Medical School, Kunming University of Science and Technology, Chenggong Campus, Kunming, Yunnan 650500, China
| |
Collapse
|
27
|
Xiao G, Lu W, Yuan J, Liu Z, Wang P, Fan H. Fbxw7 suppresses carcinogenesis and stemness in triple-negative breast cancer through CHD4 degradation and Wnt/β-catenin pathway inhibition. J Transl Med 2024; 22:99. [PMID: 38268032 PMCID: PMC10809768 DOI: 10.1186/s12967-024-04897-2] [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: 11/28/2023] [Accepted: 01/14/2024] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND Cancer stem cells (CSCs) are a small population of cells in tumor tissues that can drive tumor initiation and promote tumor progression. A small number of previous studies indirectly mentioned the role of F-box and WD repeat domain-containing 7 (FBXW7) as a tumor suppressor in Triple-negative breast cancer (TNBC). However, few studies have focused on the function of FBXW7 in cancer stemness in TNBC and the related mechanism. METHODS We detected FBXW7 by immunohistochemistry (IHC) in 80 TNBC patients. FBXW7 knockdown and overexpression in MD-MBA-231 and HCC1937 cell models were constructed. The effect of FBXW7 on malignant phenotype and stemness was assessed by colony assays, flow cytometry, transwell assays, western blot, and sphere formation assays. Immunoprecipitation-Mass Spectrometry (IP-MS) and ubiquitination experiments were used to find and verify potential downstream substrate proteins of FBXW7. Animal experiments were constructed to examine the effect of FBXW7 on tumorigenic potential and cancer stemness of TNBC cells in vivo. RESULTS The results showed that FBXW7 was expressed at low levels in TNBC tissues and positively correlated with prognosis of TNBC patients. In vitro, FBXW7 significantly inhibited colony formation, cell cycle progression, cell migration, EMT process, cancer stemness and promotes apoptosis. Further experiments confirmed that chromodomain-helicase-DNA-binding protein 4 (CHD4) is a novel downstream target of FBXW7 and is downregulated by FBXW7 via proteasomal degradation. Moreover, CHD4 could promote the nuclear translocation of β-catenin and reverse the inhibitory effect of FBXW7 on β-catenin, and ultimately activate the Wnt/β-catenin pathway. Rescue experiments confirmed that the FBXW7-CHD4-Wnt/β-catenin axis was involved in regulating the maintenance of CSC in TNBC cells. In animal experiments, FBXW7 reduced CSC marker expression and suppressed TNBC cell tumorigenesis in vivo. CONCLUSIONS Taken together, these results highlight that FBXW7 degrades CHD4 protein through ubiquitination, thereby blocking the activation of the Wnt/β-catenin pathway to inhibit the stemness of TNBC cells. Thus, targeting FBXW7 may be a promising strategy for therapeutic intervention against TNBC.
Collapse
Affiliation(s)
- Guodong Xiao
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Dong Road, ErQi District, Zhengzhou, 450052, Henan, China
| | - Weiping Lu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Dong Road, ErQi District, Zhengzhou, 450052, Henan, China
| | - Jing Yuan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Dong Road, ErQi District, Zhengzhou, 450052, Henan, China
| | - Zuyue Liu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Dong Road, ErQi District, Zhengzhou, 450052, Henan, China
| | - Peili Wang
- Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, No 127 Dongming Road, Jinshui District, Zhengzhou, 450008, Henan, China
| | - Huijie Fan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Dong Road, ErQi District, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
28
|
Yang R, Cheng S, Xiao J, Pei Y, Zhu Z, Zhang J, Feng J, Li J. GLS and GOT2 as prognostic biomarkers associated with dendritic cell and immunotherapy response in breast cancer. Heliyon 2024; 10:e24163. [PMID: 38234908 PMCID: PMC10792574 DOI: 10.1016/j.heliyon.2024.e24163] [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: 08/16/2023] [Revised: 11/28/2023] [Accepted: 01/04/2024] [Indexed: 01/19/2024] Open
Abstract
Breast cancer is the females' most common cancer. Targeting the immune microenvironment is a new and promising treatment method for breast cancer. Nevertheless, only a small section of patients can profit by immunotherapy, and improving the ability to accurately predict the potential for immunotherapy response is still awaiting further exploration. In this study, we found that the key factors of glutamine metabolism, glutaminase 1 (GLS) and mitochondrial aspartate transaminase (GOT2), showed opposite expression patterns in breast cancer samples. Based on the expression level of GLS and GOT2, we divided the breast cancer samples into two clusters: Cluster 2 showed GLS expressed higher and GOT2 expressed lower, whereas Cluster 1 showed GOT2 expressed higher and GLS expressed lower. GSEA showed that the clusters were related to pathways of immunity. Further analysis showed that Cluster 2 was positively associated with immunity infiltration. Through WGCNA, we identified a module strongly correlated with glutamine metabolism and immunity and identified 11 dendritic cell-associated genes involved in dendritic cell development, maturation, activation and other functions. In addition, Cluster 2 also showed higher immune checkpoint gene expression, which suggest the Cluster 2 had even better response to immunotherapy. The validation dataset could also be clustered into two groups. Cluster 2 (GLS expressed higher and GOT2 expressed lower) of the validation dataset was also positively associated with dendritic cells and a better immunotherapy response. Thus, these data indicate that GLS and GOT2 are prognostic biomarkers which closely related to dendritic cells and better reacted to immunotherapy in breast cancer.
Collapse
Affiliation(s)
- Ruifang Yang
- Anhui University of Science and Technology Affiliated Fengxian Hospital, Shanghai, 201499, China
| | - Shuo Cheng
- Anhui University of Science and Technology Affiliated Fengxian Hospital, Shanghai, 201499, China
| | - Jie Xiao
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China
| | - Yujie Pei
- Anhui University of Science and Technology Affiliated Fengxian Hospital, Shanghai, 201499, China
| | - Zhonglin Zhu
- Anhui University of Science and Technology Affiliated Fengxian Hospital, Shanghai, 201499, China
| | - Jifa Zhang
- Anhui University of Science and Technology Affiliated Fengxian Hospital, Shanghai, 201499, China
| | - Jing Feng
- Anhui University of Science and Technology Affiliated Fengxian Hospital, Shanghai, 201499, China
- The Second Affiliated Hospital, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Jing Li
- Anhui University of Science and Technology Affiliated Fengxian Hospital, Shanghai, 201499, China
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
29
|
Wang Y, Sun Y, Wang F, Wang H, Hu J. Ferroptosis induction via targeting metabolic alterations in triple-negative breast cancer. Biomed Pharmacother 2023; 169:115866. [PMID: 37951026 DOI: 10.1016/j.biopha.2023.115866] [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: 09/13/2023] [Revised: 11/04/2023] [Accepted: 11/07/2023] [Indexed: 11/13/2023] Open
Abstract
Triple-negative breast cancer (TNBC), the most aggressive form of breast cancer, presents severe threats to women's health. Therefore, it is critical to find novel treatment approaches. Ferroptosis, a newly identified form of programmed cell death, is marked by the buildup of lipid reactive oxygen species (ROS) and high iron concentrations. According to previous studies, ferroptosis sensitivity can be controlled by a number of metabolic events in cells, such as amino acid metabolism, iron metabolism, and lipid metabolism. Given that TNBC tumors are rich in iron and lipids, inducing ferroptosis in these tumors is a potential approach for TNBC treatment. Notably, the metabolic adaptability of cancer cells allows them to coordinate an attack on one or more metabolic pathways to initiate ferroptosis, offering a novel perspective to improve the high drug resistance and clinical therapy of TNBC. However, a clear picture of ferroptosis in TNBC still needs to be completely revealed. In this review, we provide an overview of recent advancements regarding the connection between ferroptosis and amino acid, iron, and lipid metabolism in TNBC. We also discuss the probable significance of ferroptosis as an innovative target for chemotherapy, radiotherapy, immunotherapy, nanotherapy and natural product therapy in TNBC, highlighting its therapeutic potential and application prospects.
Collapse
Affiliation(s)
- Yaru Wang
- The Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Yue Sun
- The Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Feiran Wang
- The Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Hongyi Wang
- The Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Jing Hu
- The Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China.
| |
Collapse
|
30
|
Paba C, Dorigo V, Senigagliesi B, Tormena N, Parisse P, Voitchovsky K, Casalis L. Lipid bilayer fluidity and degree of order regulates small EVs adsorption on model cell membrane. J Colloid Interface Sci 2023; 652:1937-1943. [PMID: 37690301 DOI: 10.1016/j.jcis.2023.08.117] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/09/2023] [Accepted: 08/19/2023] [Indexed: 09/12/2023]
Abstract
Small extracellular vesicles (sEVs) are known to play an important role in the communication between distant cells and to deliver biological information throughout the body. To date, many studies have focused on the role of sEVs characteristics such as cell origin, surface composition, and molecular cargo on the resulting uptake by the recipient cell. Yet, a full understanding of the sEV fusion process with recipient cells and in particular the role of cell membrane physical properties on the uptake are still lacking. Here we explore this problem using sEVs from a cellular model of triple-negative breast cancer fusing to a range of synthetic planar lipid bilayers both with and without cholesterol, and designed to mimic the formation of 'raft'-like nanodomains in cell membranes. Using time-resolved Atomic Force Microscopy we were able to track the sEVs interaction with the different model membranes, showing the process to be strongly dependent on the local membrane fluidity. The strongest interaction and fusion is observed over the less fluid regions, with sEVs even able to disrupt ordered domains at sufficiently high cholesterol concentration. Our findings suggest the biophysical characteristics of recipient cell membranes to be crucial for sEVs uptake regulation.
Collapse
Affiliation(s)
- Carolina Paba
- Department of Physics, University of Trieste, 34127 Trieste, Italy
| | | | | | - Nicolò Tormena
- Department of Physics, University of Durham, Durham DH1 3LE, United Kingdom
| | - Pietro Parisse
- Elettra Sincrotrone Trieste, 34149 Basovizza TS, Italy; IOM-CNR, 34149 Basovizza TS, Italy.
| | - Kislon Voitchovsky
- Department of Physics, University of Durham, Durham DH1 3LE, United Kingdom.
| | | |
Collapse
|
31
|
El-Shafey ES, Elsherbiny ES. Cytotoxic effect of Ziziphus Spina-Christi extract alone and in combination with doxorubicin on breast cancer cells. UKRAINIAN BIOCHEMICAL JOURNAL 2023; 95:50-63. [DOI: 10.15407/ubj95.06.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
Ziziphus Spina-Christi (L.) (ZSC) is a traditional Arabian medicinal plant used to treat inflammatory symptoms, swellings and pain since long. Triple negative breast cancer (TNBC) is a form of cancer with a poor prognosis owing to the paucity of therapy alternatives. Two of the most critical pathways of TNBC development are Wnt/β-catenin signaling and autophagy. In the present study, we intended to identify the possible mechanisms of the cytotoxic effects mediated by ZSC extract on MDA-MB-231 breast cancer cells and to improve the efficacy of DOX in combination with ZSC. The MTT test was used to estimate cell viability and IC50 values. Apoptosis was detected using AnnexinV-FITC detection kit. ELISA was used to measure caspase-3 levels. Cell cycle and the level of autophagosome marker LC3-II were analysed using flow cytometry. Acidic vesicular organelle (AVOs) formation was observed by fluorescence microscopy. Real-time PCR was used to monitor changes in gene expression of β-catenin and autophagic adapter NBR1. It was shown that ZSC treatment dose-dependently inhibited MDA-MB-231 cell viability and induced apoptosis with accompanying elevation of caspase-3 level. Besides ZSC caused a significant elevation in LC3II level and downregulation of NBR1 gene expression with subsequent downregulation of β-catenin gene expression, indicating the inhibition of the oncogenic Wnt pathway. ZSC and DOX combination had synergistic cytotoxic effect by more effective suppression of Wnt pathway and induction of apoptosis and autosis. Keywords: apoptosis, autophagic adapter NBR1, autophagosome marker LC3-II, breast cancer cells, DOX, Wnt/β-catenin signaling, Ziziphus Spina-Christi
Collapse
|
32
|
Qiao L, Dong C, Jia W, Ma B. NAA20 recruits Rin2 and promotes triple-negative breast cancer progression by regulating Rab5A-mediated activation of EGFR signaling. Cell Signal 2023; 112:110922. [PMID: 37827343 DOI: 10.1016/j.cellsig.2023.110922] [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: 07/05/2023] [Revised: 09/25/2023] [Accepted: 10/09/2023] [Indexed: 10/14/2023]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype with poor prognosis and high mortality. To improve the prognosis and survival of TNBC patients, it is necessary to explore new targets and signaling pathways to develop novel therapies for TNBC treatment. N-α-acetyltransferase 20 (NAA20) is one of the catalytic subunits of N-terminal acetyltransferase (NatB). It has been reported that NAA20 played a critical role in cancer progression. In this study, we found that NAA20 expression was markedly higher in TNBC tissues than in paracancerous normal tissues using The Cancer Genome Atlas (TCGA) analysis. This result was further confirmed by qRT-PCR and immunohistochemistry (IHC). Knockdown of NAA20 significantly inhibited TNBC cell viability by CCK8 and colony formation assays and cell migration and invasion by Transwell assays. Additionally, NAA20 knockdown decreased the expression of EGFR in TNBC cells. Upon stimulation with EGF and knockdown of NAA20, EGFR internalization and degradation were observed by confocal microscopy. The western blot results showed that NAA20 knockdown down-regulated PI3K, AKT, and mTOR phosphorylation. Next, we further explored the underlying molecular mechanisms of NAA20 by co-immunoprecipitation (Co-IP). The results suggested that there was an interacting relationship between NAA20 and Rab5A. Over-expression of NAA20 could potentiate the expression of Rab5A. Furthermore, the knockdown of Rab5A inhibited EGFR expression and the phosphorylation of downstream signaling targets. NAA20 over-expression offset the knockdown effect of Rab5A and activated EGFR signaling. Finally, we constructed a xenograft mouse model transfected TNBC cells to investigate the role of NAA20 in vivo. NAA20 knockdown markedly suppressed tumor growth and decreased tumor volume and weight. In conclusion, our study demonstrated that NAA20, a novel target of TNBC, could promote TNBC progression by regulating Rab5A-mediated activation of EGFR signaling.
Collapse
Affiliation(s)
- Lei Qiao
- Department of Breast and Thyroid Surgery, Xinjiang Medical University affiliated Tumor Hospital, Urumqi, Xinjiang Uygur Autonomous Region 830000, China
| | - Chao Dong
- Department of Breast and Thyroid Surgery, Xinjiang Medical University affiliated Tumor Hospital, Urumqi, Xinjiang Uygur Autonomous Region 830000, China
| | - Wenlei Jia
- Department of Breast and Thyroid Surgery, Xinjiang Medical University affiliated Tumor Hospital, Urumqi, Xinjiang Uygur Autonomous Region 830000, China
| | - Binlin Ma
- Department of Breast and Thyroid Surgery, Xinjiang Medical University affiliated Tumor Hospital, Urumqi, Xinjiang Uygur Autonomous Region 830000, China.
| |
Collapse
|
33
|
Lin J, Qu Z, Pu H, Shen LS, Yi X, Lin YS, Gong RH, Chen GQ, Chen S. In Vitro and In Vivo Anti-Cancer Activity of Lasiokaurin in a Triple-Negative Breast Cancer Model. Molecules 2023; 28:7701. [PMID: 38067432 PMCID: PMC10707582 DOI: 10.3390/molecules28237701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/17/2023] [Accepted: 08/17/2023] [Indexed: 12/18/2023] Open
Abstract
Due to its intricate heterogeneity, high invasiveness, and poor prognosis, triple-negative breast cancer (TNBC) stands out as the most formidable subtype of breast cancer. At present, chemotherapy remains the prevailing treatment modality for TNBC, primarily due to its lack of estrogen receptors (ERs), progesterone receptors (PRs), and human epidermal growth receptor 2 (HER2). However, clinical chemotherapy for TNBC is marked by its limited efficacy and a pronounced incidence of adverse effects. Consequently, there is a pressing need for novel drugs to treat TNBC. Given the rich repository of diverse natural compounds in traditional Chinese medicine, identifying potential anti-TNBC agents is a viable strategy. This study investigated lasiokaurin (LAS), a natural diterpenoid abundantly present in Isodon plants, revealing its significant anti-TNBC activity both in vitro and in vivo. Notably, LAS treatment induced cell cycle arrest, apoptosis, and DNA damage in TNBC cells, while concurrently inhibiting cell metastasis. In addition, LAS effectively inhibited the activation of the phosphatidylinositol-3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR) pathway and signal transducer and activator of transcription 3 (STAT3), thus establishing its potential for multitarget therapy against TNBC. Furthermore, LAS demonstrated its ability to reduce tumor growth in a xenograft mouse model without exerting detrimental effects on the body weight or vital organs, confirming its safe applicability for TNBC treatment. Overall, this study shows that LAS is a potent candidate for treating TNBC.
Collapse
Affiliation(s)
- Jinrong Lin
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Zhao Qu
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College, China Three Gorges University, Yichang 443002, China
| | - Huanhuan Pu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Li-Sha Shen
- Chongqing Academy of Chinese Materia Medica, Chongqing 400065, China
| | - Xianguo Yi
- College of Animal Science and Technology, Xinyang Agricultural and Forestry University, Xinyang 464000, China;
| | - Yu-Shan Lin
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
| | - Rui-Hong Gong
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Hong Kong 999077, China
| | - Guo-Qing Chen
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Hong Kong 999077, China
- Research Centre for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hung Hom, Hong Kong 999077, China
| | - Sibao Chen
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Hong Kong 999077, China
- Research Centre for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hung Hom, Hong Kong 999077, China
| |
Collapse
|
34
|
Yang C, Wang M, Gong Y, Deng M, Ling Y, Li Q, Wang J, Zhou Y. Discovery and identification of a novel PI3K inhibitor with enhanced CDK2 inhibition for the treatment of triple negative breast cancer. Bioorg Chem 2023; 140:106779. [PMID: 37579621 DOI: 10.1016/j.bioorg.2023.106779] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 08/16/2023]
Abstract
Blocking the PI3K pathway has been recognized as a promising strategy for cancer therapy. Herein, we report the discovery of novel PI3K inhibitors utilizing 7-azaindole-based fragment-oriented growth. Among them, compound FD2056 stands out as the most promising candidate, maintaining potent inhibitory activity against PI3K and enhanced CDK2 inhibition, and showing moderate selectivity among 108 kinases. In cellular assays, the inhibitor FD2056 demonstrated superior anti-proliferative profiles over reference compounds against TNBC cells and significantly increased apoptosis of MDA-MB-231 cells in a dose-dependent manner. Moreover, FD2056 showed more efficacious anti-TNBC activity than the corresponding drugs BKM120 and CYC202 at an oral dose of 15 mg/kg in the MDA-MB-231 xenograft model, inhibiting tumor growth by 43% with no observable toxic effects. All these results suggest that FD2056 has potential for further development as a promising anticancr compound, and co-targeting PI3K and CDK2 pathways may provide an alternative therapeutic strategy for the treatment of TNBC.
Collapse
Affiliation(s)
- Chengbin Yang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China; Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Department of Chemistry, Fudan University, Shanghai 200433, China
| | - Menghui Wang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yimin Gong
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Department of Chemistry, Fudan University, Shanghai 200433, China
| | - Mingli Deng
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Department of Chemistry, Fudan University, Shanghai 200433, China
| | - Yun Ling
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Department of Chemistry, Fudan University, Shanghai 200433, China
| | - Qingquan Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jianxin Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China.
| | - Yaming Zhou
- Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Department of Chemistry, Fudan University, Shanghai 200433, China.
| |
Collapse
|
35
|
Yang H, Zhang T, Chen C, Chiang C, Chen K, Wu Y, Liu Z, Zhou Y, Zhu L, Zheng D. Laxiflorin B covalently binds the tubulin colchicine-binding site to inhibit triple negative breast cancer proliferation and induce apoptosis. Chem Biol Interact 2023; 383:110681. [PMID: 37648048 DOI: 10.1016/j.cbi.2023.110681] [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: 07/22/2023] [Accepted: 08/26/2023] [Indexed: 09/01/2023]
Abstract
Laxiflorin B is a natural ent-kaurane diterpenoid that can be isolated from the leaves of the Isodon eriocalyx var. laxiflora, a perennial shrub native to parts of China. While this compound has potent cytotoxic activity against various tumor cells, the anti-tumor targets and molecular mechanisms of Laxiflorin B are unclear. Here, we show that Laxiflorin B exhibits strong antiproliferative and proapoptotic effects on triple-negative breast cancer (TNBC) cells. At the mechanistic level, we show that β-tubulin (TUBB) is a cellular target of Laxiflorin B. By covalently binding the Cys239 and C354 residues of the TUBB colchicine-binding site, Laxiflorin B disturbs microtubule integrity and structure in vitro and in vivo. Cytotoxicity analyses also showed that the α, β-unsaturated carbonyl in the D ring of Laxiflorin B is responsible for mediating its covalent binding and anti-tumor activity. To assess the therapeutic effects of Laxiflorin B, we synthesized a Laxiflorin B-ALA pro-drug and delivered it by intraperitoneal injection (10 mg/kg) into a 4T1 orthotopic tumor mouse model. Drug treatment had anti-tumor effects without inducing notable weight loss or organ dysfunction. We conclude that Laxiflorin B is a promising colchicine binding site inhibitor that might be exploited in the context of TNBC treatment in the future.
Collapse
Affiliation(s)
- Heng Yang
- Guangdong Provincial Key Laboratory of Genome Stability and Disease Prevention, International Cancer Center, Department of Cell Biology and Genetics, School of Basic Medical Sciences; School of Pharmacy; Department of Pharmacy, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine); Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China
| | - Tiantian Zhang
- Guangdong Provincial Key Laboratory of Genome Stability and Disease Prevention, International Cancer Center, Department of Cell Biology and Genetics, School of Basic Medical Sciences; School of Pharmacy; Department of Pharmacy, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine); Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China
| | - Chunlan Chen
- Guangdong Provincial Key Laboratory of Genome Stability and Disease Prevention, International Cancer Center, Department of Cell Biology and Genetics, School of Basic Medical Sciences; School of Pharmacy; Department of Pharmacy, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine); Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China
| | - Chengyao Chiang
- Southern University of Science and Technology, Yantian Hospital, Shenzhen, China
| | - Kai Chen
- School of Materials Science and Engineering, Central South University of Forestry and Technology, Changsha, 410004, China
| | - Yan Wu
- Guangdong Provincial Key Laboratory of Genome Stability and Disease Prevention, International Cancer Center, Department of Cell Biology and Genetics, School of Basic Medical Sciences; School of Pharmacy; Department of Pharmacy, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine); Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China
| | - Zhengxin Liu
- Guangdong Provincial Key Laboratory of Genome Stability and Disease Prevention, International Cancer Center, Department of Cell Biology and Genetics, School of Basic Medical Sciences; School of Pharmacy; Department of Pharmacy, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine); Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China
| | - Yajun Zhou
- Guangdong Provincial Key Laboratory of Genome Stability and Disease Prevention, International Cancer Center, Department of Cell Biology and Genetics, School of Basic Medical Sciences; School of Pharmacy; Department of Pharmacy, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine); Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China
| | - Lizhi Zhu
- Guangdong Provincial Key Laboratory of Genome Stability and Disease Prevention, International Cancer Center, Department of Cell Biology and Genetics, School of Basic Medical Sciences; School of Pharmacy; Department of Pharmacy, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine); Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China.
| | - Duo Zheng
- Guangdong Provincial Key Laboratory of Genome Stability and Disease Prevention, International Cancer Center, Department of Cell Biology and Genetics, School of Basic Medical Sciences; School of Pharmacy; Department of Pharmacy, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine); Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China.
| |
Collapse
|
36
|
Zhang Z, Zhang R, Li D. Molecular Biology Mechanisms and Emerging Therapeutics of Triple-Negative Breast Cancer. Biologics 2023; 17:113-128. [PMID: 37767463 PMCID: PMC10520847 DOI: 10.2147/btt.s426392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that is conventionally characterized by the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor-2 (HER2), accounting for approximately 15-20% of all breast cancers. Compared to other molecular phenotypes, TNBC is typically associated with high malignancy and poor prognosis. Cytotoxic agents have been the mainstay of treatment for the past few decades due to the lack of definitive targets and limited therapeutic interventions. However, recent developments have demonstrated that TNBC has peculiar molecular classifications and biomarkers, which provide the possibility of evolving treatment from basic cytotoxic chemotherapy to an expanding domain of targeted therapies. This review presents a framework for understanding the current clinical experience surrounding molecular biology mechanisms in TNBC (Figure 1). Including immunotherapy, polymerase (PARP) and PI3K/AKT pathway inhibitors, antibody-drug conjugates, and androgen receptor (AR) blockade. Additionally, the role of miRNA therapeutics targeting TNBC and potential strategies targeting cancer stem cells (CSCs) are discussed and highlighted. As more and more treatments arise on the horizon, we believe that patients with TNBC will have a new sense of hope.
Collapse
Affiliation(s)
- Zhiying Zhang
- Inner Mongolia Medical University, Department of Thyroid Breast Surgery, Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia, 010050, People’s Republic of China
| | - Rui Zhang
- Inner Mongolia Medical University, Department of Thyroid Breast Surgery, Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia, 010050, People’s Republic of China
| | - Donghai Li
- Inner Mongolia Medical University, Department of Thyroid Breast Surgery, Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia, 010050, People’s Republic of China
| |
Collapse
|
37
|
Caetano S, Garcia AR, Figueira I, Brito MA. MEF2C and miR-194-5p: New Players in Triple Negative Breast Cancer Tumorigenesis. Int J Mol Sci 2023; 24:14297. [PMID: 37762600 PMCID: PMC10531597 DOI: 10.3390/ijms241814297] [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: 08/16/2023] [Revised: 09/08/2023] [Accepted: 09/16/2023] [Indexed: 09/29/2023] Open
Abstract
Among breast cancer (BC) subtypes, the most aggressive is triple negative BC (TNBC), which is prone to metastasis. We previously found that microRNA (miR)-194-5p is downregulated at the early stages of TNBC brain metastasis development. Additionally, the transcription factor myocyte enhancer factor 2 (MEF2)C, a bioinformatically predicted miR-194-5p target, was increasingly expressed throughout TNBC brain metastasis formation and disease severity. However, the contributions of these two players to malignant cells' features remain undetermined. This study aimed at disclosing the role of miR-194-5p and MEF2C in TNBC tumorigenesis. The transfection of 4T1 cells with a silencer for MEF2C or with a pre-miRNA for miR-194-5p was employed to study TNBC cells' phenotypic alterations regarding epithelial and mesenchymal markers, as well as migratory capability alterations. MEF2C-silenced cells presented a decline in both vimentin and cytokeratin expression, whereas the overexpression of miR-194-5p promoted an increase in cytokeratin and a reduction in vimentin, reflecting the acquisition of an epithelial phenotype. Both treatments reduced TNBC cells' migration. These results suggest that MEF2C may determine TNBC cells' invasive properties by partially determining the occurrence of epithelial-mesenchymal transition, while the overexpression of miR-194-5p promotes a decline in TNBC cells' aggressive behavior and reinforces this miRNA's role as a tumor suppressor in TNBC.
Collapse
Affiliation(s)
- Sara Caetano
- iMed—Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal; (S.C.); (A.R.G.); (I.F.)
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Ana Rita Garcia
- iMed—Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal; (S.C.); (A.R.G.); (I.F.)
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Inês Figueira
- iMed—Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal; (S.C.); (A.R.G.); (I.F.)
- Farm-ID—Faculty of Pharmacy Research and Development Association, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Maria Alexandra Brito
- iMed—Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal; (S.C.); (A.R.G.); (I.F.)
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| |
Collapse
|
38
|
Liao YC, Cheng TC, Tu SH, Chang J, Guo P, Chen LC, Ho YS. Tumor targeting and therapeutic assessments of RNA nanoparticles carrying α9-nAChR aptamer and anti-miR-21 in triple-negative breast cancers. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:351-366. [PMID: 37547295 PMCID: PMC10400867 DOI: 10.1016/j.omtn.2023.07.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 07/11/2023] [Indexed: 08/08/2023]
Abstract
Triple-negative breast cancer (TNBC) is highly aggressive with a poor prognosis because of a lack of cell markers as drug targets. α9-Nicotinic acetylcholine receptor (nAChR) is expressed abundantly in TNBC; thus, it is a valuable biomarker for TNBC detection and treatment. In this study, we utilized thermodynamically stable three-way junction (3WJ) packaging RNA (pRNA) as the core to construct RNA nanoparticles with an α9-nAChR RNA aptamer as a targeting ligand and an anti-microRNA-21 (miR-21) as a therapeutic module. We compared the configuration of the two RNA nanoparticles and found that 3WJ-B-α9-nAChR-aptamer fluorescent RNA nanoparticles (3WJ-B-α9-apt-Alexa) exhibited better specificity for α9-nAChR in TNBC cells compared with 3WJ-C-α9-nAChR. Furthermore, 3WJ-B-α9-apt-Alexa bound more efficiently to TNBC patient-derived xenograft (PDX) tumors than 3WJ fluorescent RNA nanoparticles (3WJ-Alexa) with little or no accumulation in healthy organs after systemic injection in mice. Moreover, 3WJ-B-α9-nAChR-aptamer RNA nanoparticles carrying anti-miR-21 (3WJ-B-α9-apt-anti-miR-21) significantly suppressed TNBC-PDX tumor growth and induced cell apoptosis because of reduced miR-21 gene expression and upregulated the phosphatase and tensin homolog (PTEN) and programmed cell death 4 (PDCD4) proteins. In addition, no pathological changes were detected upon toxicity examination of treated mice. In conclusion, the 3WJ-B-α9-nAChR-aptamer RNA nanoparticles established in this study efficiently deliver therapeutic anti-miR-21, indicating their potential as a novel TNBC therapy.
Collapse
Affiliation(s)
- You-Cheng Liao
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110031, Taiwan
| | - Tzu-Chun Cheng
- Institute of Biochemistry and Molecular Biology, College of Life Sciences, China Medical University, Taichung 406040, Taiwan
| | - Shih-Hsin Tu
- Department of Surgery, Taipei Medical University Hospital, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Jungshan Chang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110031, Taiwan
- International Master/PhD Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110031, Taiwan
| | - Peixuan Guo
- Center for RNA Nanobiotechnology and Nanomedicine, The Ohio State University, Columbus, OH, USA
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
- James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- College of Medicine, The Ohio State University, Columbus, OH, USA
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Li-Ching Chen
- Department of Biological Science & Technology, College of Life Sciences, China Medical University, Taichung 406040, Taiwan
| | - Yuan-Soon Ho
- Institute of Biochemistry and Molecular Biology, College of Life Sciences, China Medical University, Taichung 406040, Taiwan
| |
Collapse
|
39
|
sofi S, Jan N, Qayoom H, Alkhanani M, Almilaibary A, Ahmad Mir M. Elucidation of interleukin-19 as a therapeutic target for breast cancer by computational analysis and experimental validation. Saudi J Biol Sci 2023; 30:103774. [PMID: 37675062 PMCID: PMC10477739 DOI: 10.1016/j.sjbs.2023.103774] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 07/30/2023] [Accepted: 08/05/2023] [Indexed: 09/08/2023] Open
Abstract
Interleukin 19 (IL-19) is a cytokine produced by monocytes and belongs to the family of IL-10. The IL-19 protein stimulates fibronectin (FN) expression and assembly, metastasis, and cell division in breast cancer (BC) cells. IL-19, which is connected to breast pathogenesis and has an autocrine action in BC cells, is a key predictor of prognosis for many tumour forms, including breast cancer. Augmented IL-19 expression has been related to poorer clinical outcomes for patients with BC and directly enhances proliferation and migration while also serving as a microenvironment for tumour formation. The main aim of our study was to examine the expression profile, functional role, and prognostic significance of interleukin-19 in BC pathogenesis and also to find out the molecular mechanism of IL-19 in BC. In this work, we used the various computational approach and tools, to evaluate the expression profile and prognostic implication of IL-19 in BC and discover the role of IL-19 in BC pathogenesis. IL-19 was shown to be highly upregulated in BC as compared to other interleukins. Also, its levels were highly overexpressed in liminal BC patients, mostly in 3rd stage groups under the age group of 21-40 years. IL-19 levels were increased in BC and elevated expression of IL-19 was examined to have worse overall survival (OS). The KEGG analysis and gene ontology of IL-19 depict that IL-19 is significantly augmented in cytokine activity and receptor-ligand activity and also in the JAK-STAT signaling pathway. Moreover, IL-19 showed a high correlation with IL20RA, as later is involved with the JAK-STAT signaling pathway. The in-vivo and in-vitro studies have also reflected that upregulation of IL-19 enhances tumor development and affects clinical outcomes in BC patients through several pathways including the JAK TAT signalling pathway. Overall, our study indicates that IL-19 increases tumour growth and that inhibiting it in addition to standard treatments will greatly improve BC patient's therapeutic responses.
Collapse
Affiliation(s)
- Shazia sofi
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, J&K, India
| | - Nusrat Jan
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, J&K, India
| | - Hina Qayoom
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, J&K, India
| | - Mustfa Alkhanani
- Department of Biology, College of Science, Hafr Al Batin, University of Hafr Al-Batin, 31991, Saudi Arabia
| | - Abdullah Almilaibary
- Department of Family and Community Medicine, Faculty of Medicine, Al Baha University, Albaha 65511, Saudi Arabia
| | - Manzoor Ahmad Mir
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, J&K, India
| |
Collapse
|
40
|
Chu M, Meng T, Zhou Y, Jin L, Dai Q, Ma L, Chen H. Molecular mechanism of Ruxian Shuhou prescription in the treatment of triple-negative breast cancer based on network pharmacology. Medicine (Baltimore) 2023; 102:e34763. [PMID: 37657065 PMCID: PMC10476815 DOI: 10.1097/md.0000000000034763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/03/2023] Open
Abstract
We aimed to explore the molecular mechanism of Ruxian Shuhou prescription in the treatment of triple-negative breast cancer (TNBC) by using network pharmacology. The active components and targets of the prescription were obtained by Traditional Chinese medicine systems pharmacology database. Gencards database, online mendelian inheritance in man database, therapeutic target database, and DRUGBANK database were used to search for the TNBC-related targets. The potential targets of Ruxian Shuhou prescription for TNBC were screened out by the intersection of effective ingredient action targets and disease targets. A herb-active ingredient-target network was constructed and analyzed for key ingredients. A protein-protein interaction network was constructed for studying key targets. Furthermore, gene ontology analysis and Kyoto encyclopedia of genes and genomes pathway enrichment analysis were carried out. Finally, the relationship between key ingredients and key genes was evaluated by molecular docking. The key ingredients of Ruxian Shuhou prescription for the treatment of TNBC may be Quercetin, Luteolin and Kaempferol, while the key therapeutic targets may be protein kinase B, interleukin-6, cellular tumor antigen p53, and vascular endothelial growth factor A. The related signaling pathways were mainly involved in tumor, apoptosis and virus infection, among which the PI3K-Akt signaling pathway was the most closely related to TNBC. Molecular docking showed that the key ingredients had high binding activity with the key targets. The molecular mechanisms of Ruxian Shuhou prescription for TNBC are likely to involve multi-ingredient, multi-target and multi-pathway.
Collapse
Affiliation(s)
- Meiling Chu
- Breast Department of TCM, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
41
|
Chamandi G, El-Hajjar L, El Kurdi A, Le Bras M, Nasr R, Lehmann-Che J. ER Negative Breast Cancer and miRNA: There Is More to Decipher Than What the Pathologist Can See! Biomedicines 2023; 11:2300. [PMID: 37626796 PMCID: PMC10452617 DOI: 10.3390/biomedicines11082300] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Breast cancer (BC), the most prevalent cancer in women, is a heterogenous disease. Despite advancements in BC diagnosis, prognosis, and therapeutics, survival rates have drastically decreased in the metastatic setting. Therefore, BC still remains a medical challenge. The evolution of high-throughput technology has highlighted gaps in the classification system of BCs. Of particular interest is the notorious triple negative BC, which was recounted as being heterogenous itself and it overlaps with distinct subtypes, namely molecular apocrine (MA) and luminal androgen (LAR) BCs. These subtypes are, even today, still misdiagnosed and poorly treated. As such, researchers and clinicians have been looking for ways through which to refine BC classification in order to properly understand the initiation, development, progression, and the responses to the treatment of BCs. One tool is biomarkers and, specifically, microRNA (miRNA), which are highly reported as associated with BC carcinogenesis. In this review, the diverse roles of miRNA in estrogen receptor negative (ER-) and androgen receptor positive (AR+) BC are depicted. While highlighting their oncogenic and tumor suppressor functions in tumor progression, we will discuss their diagnostic, prognostic, and predictive biomarker potentials, as well as their drug sensitivity/resistance activity. The association of several miRNAs in the KEGG-reported pathways that are related to ER-BC carcinogenesis is presented. The identification and verification of accurate miRNA panels is a cornerstone for tackling BC classification setbacks, as is also the deciphering of the carcinogenesis regulators of ER - AR + BC.
Collapse
Affiliation(s)
- Ghada Chamandi
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, 11-0236 Beirut, Lebanon; (G.C.); (L.E.-H.)
- Pathophysiology of Breast Cancer Team, INSERM U976, Immunologie Humaine, Pathophysiologie, Immunothérapie (HIPI), Université Paris Cité, 75010 Paris, France;
| | - Layal El-Hajjar
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, 11-0236 Beirut, Lebanon; (G.C.); (L.E.-H.)
- Office of Basic/Translational Research and Graduate Studies, Faculty of Medicine, American University of Beirut, 11-0236 Beirut, Lebanon
| | - Abdallah El Kurdi
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, 11-0236 Beirut, Lebanon;
| | - Morgane Le Bras
- Pathophysiology of Breast Cancer Team, INSERM U976, Immunologie Humaine, Pathophysiologie, Immunothérapie (HIPI), Université Paris Cité, 75010 Paris, France;
| | - Rihab Nasr
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, 11-0236 Beirut, Lebanon; (G.C.); (L.E.-H.)
| | - Jacqueline Lehmann-Che
- Pathophysiology of Breast Cancer Team, INSERM U976, Immunologie Humaine, Pathophysiologie, Immunothérapie (HIPI), Université Paris Cité, 75010 Paris, France;
| |
Collapse
|
42
|
Shoaib TH, Ibraheem W, Abdelrahman M, Osman W, Sherif AE, Ashour A, Ibrahim SRM, Ghazawi KF, Miski SF, Almadani SA, ALsiyud DF, Mohamed GA, Alzain AA. Exploring the potential of approved drugs for triple-negative breast cancer treatment by targeting casein kinase 2: Insights from computational studies. PLoS One 2023; 18:e0289887. [PMID: 37578958 PMCID: PMC10424868 DOI: 10.1371/journal.pone.0289887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/27/2023] [Indexed: 08/16/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive malignancy that requires effective targeted drug therapy. In this study, we employed in silico methods to evaluate the efficacy of seven approved drugs against human ck2 alpha kinase, a significant modulator of TNBC metastasis and invasiveness. Molecular docking revealed that the co-crystallized reference inhibitor 108600 achieved a docking score of (-7.390 kcal/mol). Notably, among the seven approved drugs tested, sunitinib, bazedoxifene, and etravirine exhibited superior docking scores compared to the reference inhibitor. Specifically, their respective docking scores were -10.401, -7.937, and -7.743 kcal/mol. Further analysis using MM/GBSA demonstrated that these three top-ranked drugs possessed better binding energies than the reference ligand. Subsequent molecular dynamics simulations identified etravirine, an FDA-approved antiviral drug, as the only repurposed drug that demonstrated a stable and reliable binding mode with the human ck2 alpha protein, based on various analysis measures including RMSD, RMSF, and radius of gyration. Principal component analysis indicated that etravirine exhibited comparable stability of motion as a complex with human ck2 alpha protein, similar to the co-crystallized inhibitor. Additionally, Density functional theory (DFT) calculations were performed on a complex of etravirine and a representative gold atom positioned at different sites relative to the heteroatoms of etravirine. The results of the DFT calculations revealed low-energy complexes that could potentially serve as guides for experimental trials involving gold nanocarriers of etravirine, enhancing its delivery to malignant cells and introducing a new drug delivery route. Based on the results obtained in this research study, etravirine shows promise as a potential antitumor agent targeting TNBC, warranting further investigation through experimental and clinical assessments.
Collapse
Affiliation(s)
- Tagyedeen H. Shoaib
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, University of Gezira, Gezira, Sudan
| | - Walaa Ibraheem
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, University of Gezira, Gezira, Sudan
| | - Mohammed Abdelrahman
- Faculty of Pharmacy, Department of Pharmaceutics, University of Gezira, Gezira, Sudan
| | - Wadah Osman
- Faculty of Pharmacy, Department of Pharmacognosy, Prince Sattam Bin Abdulaziz University, Al-kharj, Saudi Arabia
- Faculty of Pharmacy, Department of Pharmacognosy, University of Khartoum, Khartoum, Sudan
| | - Asmaa E. Sherif
- Faculty of Pharmacy, Department of Pharmacognosy, Prince Sattam Bin Abdulaziz University, Al-kharj, Saudi Arabia
- Faculty of Pharmacy, Department of Pharmacognosy, Mansoura University, Mansoura, Egypt
| | - Ahmed Ashour
- Faculty of Pharmacy, Department of Pharmacognosy, Prince Sattam Bin Abdulaziz University, Al-kharj, Saudi Arabia
- Faculty of Pharmacy, Department of Pharmacognosy, Mansoura University, Mansoura, Egypt
| | - Sabrin R. M. Ibrahim
- Department of Chemistry, Preparatory Year Program, Batterjee Medical College, Jeddah, Saudi Arabia
- Faculty of Pharmacy, Department of Pharmacognosy, Assiut University, Assiut, Egypt
| | - Kholoud F. Ghazawi
- Clinical Pharmacy Department, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Samar F. Miski
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Al-Madinah Al-Munawwarah, Saudi Arabia
| | - Sara A. Almadani
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Al-Madinah Al-Munawwarah, Saudi Arabia
| | - Duaa Fahad ALsiyud
- Department of Medical Laboratories—Hematology, King Fahd Armed Forces Hospital, Corniche Road, Andalus, Jeddah, Saudi Arabia
| | - Gamal A. Mohamed
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulrahim A. Alzain
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, University of Gezira, Gezira, Sudan
| |
Collapse
|
43
|
Bednarek R, Wojkowska DW, Braun M, Watala C, Salifu MO, Swiatkowska M, Babinska A. Triple negative breast cancer metastasis is hindered by a peptide antagonist of F11R/JAM‑A protein. Cancer Cell Int 2023; 23:160. [PMID: 37563645 PMCID: PMC10416405 DOI: 10.1186/s12935-023-03023-4] [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: 04/12/2023] [Accepted: 08/06/2023] [Indexed: 08/12/2023] Open
Abstract
BACKGROUND The F11R/JAM-A cell adhesion protein was examined as the therapeutic target in triple negative breast cancer (TNBC) with the use of the peptide antagonist to F11R/JAM-A, that previously inhibited the early stages of breast cancer metastasis in vitro. METHODS The online in silico analysis was performed by TNMPlot, UALCAN, and KM plotter. The in vitro experiments were performed to verify the effect of peptide 4D (P4D) on human endothelial cell lines EA.hy926 and HMEC-1 as well as on human TNBC cell line MDA-MB-231. The cell morphology upon P4D treatment was verified by light microscopy, while the cell functions were assessed by colony forming assay, MTT cell viability assay, BrdU cell proliferation assay, and Transepithelial/Endothelial Electrical Resistance measurements. The in vivo experiments on 4T1 murine breast cancer model were followed by histopathological analysis and a series of quantitative analyses of murine tissues. RESULTS By in silico analysis we have found the elevated gene expression in breast cancer with particular emphasis on TNBC. The elevated F11R expression in TNBC was related with poorer survival prognosis. Peptide 4D has altered the morphology and increased the permeability of endothelial monolayers. The colony formation, viability, and proliferation of MDA-MB-231 cells were decreased. P4D inhibited the metastasis in 4T1 breast cancer murine model in a statistically significant manner that was demonstrated by the resampling bootstrap technique. CONCLUSIONS The P4D peptide antagonist to F11R/JAM-A is able to hinder the metastasis in TNBC. This assumption needs to be confirmed by additional 4T1 mouse model study performed on larger group size, before making the decision on human clinical trials.
Collapse
Affiliation(s)
- Radosław Bednarek
- Department of Cytobiology and Proteomics, Chair of Biomedical Sciences, Medical University of Lodz, ul. Mazowiecka 6/8, 92-215, Lodz, Poland.
| | - Dagmara W Wojkowska
- Department of Haemostasis and Haemostatic Disorders, Medical University of Lodz, Lodz, Poland
| | - Marcin Braun
- Department of Pathology, Chair of Oncology, Medical University of Lodz, Lodz, Poland
| | - Cezary Watala
- Department of Haemostasis and Haemostatic Disorders, Medical University of Lodz, Lodz, Poland
| | - Moro O Salifu
- Department of Medicine, Downstate Medical Center, State University of New York, Brooklyn, NY, USA
| | - Maria Swiatkowska
- Department of Cytobiology and Proteomics, Chair of Biomedical Sciences, Medical University of Lodz, ul. Mazowiecka 6/8, 92-215, Lodz, Poland
| | - Anna Babinska
- Department of Medicine, Downstate Medical Center, State University of New York, Brooklyn, NY, USA
| |
Collapse
|
44
|
Bandyopadhayaya S, Yadav P, Sharma A, Dey SK, Nag A, Maheshwari R, Ford BM, Mandal CC. Oncogenic role of an uncharacterized cold-induced zinc finger protein 726 in breast cancer. J Cell Biochem 2023. [PMID: 37192271 DOI: 10.1002/jcb.30417] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 03/29/2023] [Accepted: 04/18/2023] [Indexed: 05/18/2023]
Abstract
The unobtrusive cold environmental temperature can be linked to the development of cancer. This study, for the first time, envisaged cold stress-mediated induction of a zinc finger protein 726 (ZNF726) in breast cancer. However, the role of ZNF726 in tumorigenesis has not been defined. This study investigated the putative role of ZNF726 in breast cancer tumorigenic potency. Gene expression analysis using multifactorial cancer databases predicted overexpression of ZNF726 in various cancers, including breast cancer. Experimental observations found that malignant breast tissues and highly aggressive MDA-MB-231 cells showed an elevated ZNF726 expression as compared to benign and luminal A type (MCF-7), respectively. Furthermore, ZNF726 silencing decreased breast cancer cell proliferation, epithelial-mesenchymal transition, and invasion accompanied by the inhibition of colony-forming ability. Concordantly, ZNF726 overexpression significantly demonstrated opposite outcomes than ZNF726 knockdown. Taken together, our findings propose cold-inducible ZNF726 as a functional oncogene demonstrating its prominent role in facilitating breast tumorigenesis. An inverse correlation between environmental temperature and total serum cholesterol was observed in the previous study. Furthermore, experimental outcomes illustrate that cold stress elevated cholesterol content hinting at the involvement of the cholesterol regulatory pathway in cold-induced ZNF726 gene regulation. This observation was bolstered by a positive correlation between the expression of cholesterol-regulatory genes and ZNF726. Exogenous cholesterol treatment elevated ZNF726 transcript levels while knockdown of ZNF726 decreased the cholesterol content via downregulating various cholesterol regulatory gene expressions (e.g., SREBF1/2, HMGCoR, LDLR). Moreover, an underlying mechanism supporting cold-driven tumorigenesis is proposed through interdependent regulation of cholesterol regulatory pathway and cold-inducible ZNF726 expression.
Collapse
Affiliation(s)
- Shreetama Bandyopadhayaya
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Pooja Yadav
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Ankit Sharma
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Sanjay Kumar Dey
- Dr. B. R. Ambedkar Centre for Biomedical Research (ACBR), University of Delhi, Delhi, India
| | - Alo Nag
- Department of Biochemistry, University of Delhi South Campus, New Delhi, India
| | - Rekha Maheshwari
- Department of General Surgery, JLN Medical College, Ajmer, Rajasthan, India
| | - Bridget M Ford
- Department of Biology, University of the Incarnate Word, San Antonio, Texas, USA
| | - Chandi C Mandal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| |
Collapse
|
45
|
Pantelaiou-Prokaki G, Reinhardt O, Georges NS, Agorku DJ, Hardt O, Prokakis E, Mieczkowska IK, Deppert W, Wegwitz F, Alves F. Basal-like mammary carcinomas stimulate cancer stem cell properties through AXL-signaling to induce chemotherapy resistance. Int J Cancer 2023; 152:1916-1932. [PMID: 36637144 DOI: 10.1002/ijc.34429] [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: 05/26/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 01/14/2023]
Abstract
Basal-like breast cancer (BLBC) is the most aggressive and heterogeneous breast cancer (BC) subtype. Conventional chemotherapies represent next to surgery the most frequently employed treatment options. Unfortunately, resistant tumor phenotypes often develop, resulting in therapeutic failure. To identify the early events occurring upon the first drug application and initiating chemotherapy resistance in BLBC, we leveraged the WAP-T syngeneic mammary carcinoma mouse model and we developed a strategy combining magnetic-activated cell sorting (MACS)-based tumor cell enrichment with high-throughput transcriptome analyses. We discovered that chemotherapy induced a massive gene expression reprogramming toward stemness acquisition to tolerate and survive the cytotoxic treatment in vitro and in vivo. Retransplantation experiments revealed that one single cycle of cytotoxic drug combination therapy (Cyclophosphamide, Adriamycin and 5-Fluorouracil) suffices to induce resistant tumor cell phenotypes in vivo. We identified Axl and its ligand Pros1 as highly induced genes driving cancer stem cell (CSC) properties upon chemotherapy in vivo and in vitro. Furthermore, from our analysis of BLBC patient datasets, we found that AXL expression is also strongly correlated with CSC-gene signatures, a poor response to conventional therapies and worse survival outcomes in those patients. Finally, we demonstrate that AXL inhibition sensitized BLBC-cells to cytotoxic treatment in vitro. Together, our data support AXL as a promising therapeutic target to optimize the efficiency of conventional cytotoxic therapies in BLBC.
Collapse
Affiliation(s)
- Garyfallia Pantelaiou-Prokaki
- Max Planck Institute for Multidisciplinary Sciences, Translational Molecular Imaging, Göttingen, Germany.,Department of Gynecology and Obstetrics, University Medical Center Göttingen, Göttingen, Germany
| | - Oliver Reinhardt
- Max Planck Institute for Multidisciplinary Sciences, Translational Molecular Imaging, Göttingen, Germany
| | - Nadine S Georges
- Department of Gynecology and Obstetrics, University Medical Center Göttingen, Göttingen, Germany
| | - David J Agorku
- Miltenyi Biotec B.V. & Co. KG, R&D Reagents, Bergisch Gladbach, Germany
| | - Olaf Hardt
- Miltenyi Biotec B.V. & Co. KG, R&D Reagents, Bergisch Gladbach, Germany
| | - Evangelos Prokakis
- Department of Gynecology and Obstetrics, University Medical Center Göttingen, Göttingen, Germany
| | - Iga K Mieczkowska
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Wolfgang Deppert
- University Medical Center Hamburg Eppendorf, Institute for Tumor Biology, University of Hamburg, Hamburg, Germany
| | - Florian Wegwitz
- Department of Gynecology and Obstetrics, University Medical Center Göttingen, Göttingen, Germany.,Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Frauke Alves
- Max Planck Institute for Multidisciplinary Sciences, Translational Molecular Imaging, Göttingen, Germany.,Institute for Diagnostic and Interventional Radiology, University Medical Center Göttingen, Göttingen, Germany.,Clinic for Hematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
46
|
Liu H, Yan R, Xiao Z, Huang X, Yao J, Liu J, An G, Ge Y. Targeting DCLK1 attenuates tumor stemness and evokes antitumor immunity in triple-negative breast cancer by inhibiting IL-6/STAT3 signaling. Breast Cancer Res 2023; 25:43. [PMID: 37069669 PMCID: PMC10108533 DOI: 10.1186/s13058-023-01642-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 03/20/2023] [Indexed: 04/19/2023] Open
Abstract
Triple-negative breast cancer (TNBC) exhibits the poorest outcomes among breast cancer subtypes due to the high heterogeneity and a lasting scarcity of effectual treatments. Targeted therapies based on molecular subtypes of TNBC are critical step toward tailoring treatments to improve clinical outcomes. Gastrointestinal cancer stem cell (CSC) marker DCLK1 was reported to be highly expressed in stem cell-rich subtype of TNBC. Here, we firstly explored the impacts of DCLK1 on tumor cells as well as their immune microenvironment in TNBC and potential therapeutic strategies for TNBC patients with high DCLK1 expression. Our results disclosed that DCLK1 overexpression promoted, while knockout of DCLK1 suppressed the CSC-like traits of TNBC cells and resistance to chemotherapeutics. Besides, DCLK1 supported immune escape by inhibiting intratumoral cytotoxic T cell infiltration in TNBC and hence limited immune checkpoint inhibitors efficacy. Mechanistically, bioinformatics analysis revealed that IL-6/STAT3 signaling was significantly enriched in high DCLK1-expressing patients, and our results further revealed that DCLK1 enhanced IL-6 expression and STAT3 activation in TNBC cells, which finally gave rise to upregulated CSC traits and suppressed CD8+ T-cell activity. Inhibiting IL-6/STAT3 pathway by IL-6R antagonist, Tocilizumab or STAT3 inhibitor, S31-201 could abolish DCLK1-promoted malignant phenotypes of TNBC cells. Finally, DCLK1 was identified to be specifically and highly expressed in the mesenchymal-like subtype of TNBC and targeting DCLK1 could improve chemotherapy efficacy and activate antitumor immunity. Overall, our study revealed the potential clinical benefits of targeting DCLK1 in TNBC treatment.
Collapse
Affiliation(s)
- Heshu Liu
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Rui Yan
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Zeru Xiao
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Xuying Huang
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jiannan Yao
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jian Liu
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Guangyu An
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Yang Ge
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
47
|
Jiao Y, Kang G, Pan P, Fan H, Li Q, Li X, Li J, Wang Y, Jia Y, Zhang L, Sun H, Ma X. Acetylcholine promotes chronic stress-induced lung adenocarcinoma progression via α5-nAChR/FHIT pathway. Cell Mol Life Sci 2023; 80:119. [PMID: 37029227 PMCID: PMC11072774 DOI: 10.1007/s00018-023-04742-7] [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: 11/14/2022] [Revised: 02/26/2023] [Accepted: 02/28/2023] [Indexed: 04/09/2023]
Abstract
Chronic stress significantly elevates the expression levels of various neurotransmitters in the tumour microenvironment, thereby promoting the cell growth and metastasis of lung adenocarcinoma (LUAD). However, the role of chronic stress in the progression of LUAD remains unclear. In this study, we found that chronic restraint stress increases the levels of the neurotransmitter acetylcholine (ACh), and the α5-nicotinic acetylcholine receptor (α5-nAChR) and decreased fragile histidine triad (FHIT) expression in vivo. Crucially, the increased ACh levels promoted LUAD cell migration and invasion via modulation of the α5-nAChR/DNA methyltransferase 1 (DNMT1)/FHIT axis. In a chronic unpredictable stress (CUMS) mouse model, chronic stress promotes tumour development, accompanied by changes in α5-nAChR, DNMT1, FHIT, and vimentin. Together, these findings reveal a novel chronic stress-mediated LUAD signalling pathway: chronic stress enforces lung adenocarcinoma cell invasion and migration via the ACh/α5-nAChR/FHIT axis, which could be a potential therapeutic target for chronic stress-related LUAD.
Collapse
Affiliation(s)
- Yang Jiao
- Research Center of Basic Medicine, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250013, People's Republic of China
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, Shandong, People's Republic of China
| | - Guiyu Kang
- Department of Medical Laboratory, Weifang Medical University, Weifang, 261053, Shandong, People's Republic of China
| | - Pan Pan
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, Shandong, People's Republic of China
| | - Huiping Fan
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, Shandong, People's Republic of China
| | - Qiang Li
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, Shandong, People's Republic of China
| | - Xiangying Li
- Research Center of Basic Medicine, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250013, People's Republic of China
| | - Jingtan Li
- Research Center of Basic Medicine, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250013, People's Republic of China
| | - Yan Wang
- Research Center of Basic Medicine, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250013, People's Republic of China
| | - Yanfei Jia
- Research Center of Basic Medicine, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250013, People's Republic of China
| | - Lulu Zhang
- Research Center of Basic Medicine, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250013, People's Republic of China
| | - Haiji Sun
- College of Life Science, Shandong Normal University, Shandong, 250014, People's Republic of China
- Shandong Intelligent Technology Innovation Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiaoli Ma
- Research Center of Basic Medicine, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250013, People's Republic of China.
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, Shandong, People's Republic of China.
- Department of Medical Laboratory, Weifang Medical University, Weifang, 261053, Shandong, People's Republic of China.
- Shandong Intelligent Technology Innovation Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
48
|
Liu F, Xie B, Ye R, Xie Y, Zhong B, Zhu J, Tang Y, Lin Z, Tang H, Wu Z, Li H. Overexpression of tripartite motif-containing 47 (TRIM47) confers sensitivity to PARP inhibition via ubiquitylation of BRCA1 in triple negative breast cancer cells. Oncogenesis 2023; 12:13. [PMID: 36906594 PMCID: PMC10008536 DOI: 10.1038/s41389-023-00453-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 01/30/2023] [Accepted: 02/03/2023] [Indexed: 03/13/2023] Open
Abstract
Triple-negative breast cancers (TNBC) frequently harbor defects in DNA double-strand break repair through homologous recombination (HR), such as BRCA1 dysfunction. However, less than 15% of TNBC patients were found to carry BRCA1 mutation, indicating that there are other mechanisms regulating BRCA1-deficient in TNBC. In the current study, we shown that overexpression of TRIM47 correlates with progression and poor prognosis in triple-negative breast cancer. Moreover, we demonstrated that TRIM47 directly interacts with BRCA1 and induces ubiquitin-ligase-mediated proteasome turnover of BRCA1, subsequently leads to a decrease of BRCA1 protein levels in TNBC. Moreover, the downstream gene expression of BRCA1, such as p53, p27, p21 was significantly reduced in the overexpression of TRIM47 cell lines but increased in TRIM47-deleted cells. Functionally, we found that overexpression of TRIM47 in TNBC cells confers an exquisite sensitivity to olaparib, an inhibitor of poly-(ADP-ribose)-polymerase (PARP), but TRIM47 inhibition significantly confers TNBC cells resistance to olaparib both in vitro and in vivo. Furthermore, we showed that overexpression of BRCA1 significant increase the olaparib resistance in TRIM47-overexpression-induced PARP inhibitions sensitivity. Taken together, our results uncover a novel mechanism for BRCA1-deficient in TNBC and targeting TRIM47/BRCA1 axis may be a promising prognostic factor and a valuable therapeutic target for TNBC.
Collapse
Affiliation(s)
- Fengen Liu
- Department of General Surgery III, the First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, P. R. China
| | - Binhui Xie
- Department of General Surgery I, the First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, P. R. China.,Ganzhou Key Laboratory of Hepatocellular carcinoma, the First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Rong Ye
- Department of General Surgery III, the First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, P. R. China
| | - Yuankang Xie
- Department of General Surgery I, the First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, P. R. China
| | - Baiyin Zhong
- Department of General Surgery I, the First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, P. R. China
| | - Jinrong Zhu
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yao Tang
- Department of Pathology, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510310, China
| | - Zelong Lin
- Department of Pathology, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510310, China
| | - Huiru Tang
- Cheerland Watson Precision Medicine Co. Ltd, Shenzhen, 518036, P. R. China.
| | - Ziqing Wu
- Department of Pathology, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510310, China. .,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, China. .,Department of Pathology, School of Basic Medical Science, Southern Medical University, Guangzhou, 510515, China.
| | - Heping Li
- Department of Medical Oncology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, P. R. China.
| |
Collapse
|
49
|
Wang F, Li N, Wang W, Ma L, Sun Y, Wang H, Zhan J, Yu D. A Multifunctional, Highly Biocompatible, and Double-Triggering Caramelized Nanotheranostic System Loaded with Fe 3O 4 and DOX for Combined Chemo-Photothermal Therapy and Real-Time Magnetic Resonance Imaging Monitoring of Triple Negative Breast Cancer. Int J Nanomedicine 2023; 18:881-897. [PMID: 36844435 PMCID: PMC9948638 DOI: 10.2147/ijn.s393507] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 02/14/2023] [Indexed: 02/20/2023] Open
Abstract
Purpose Owing to lack of specific molecular targets, the current clinical therapeutic strategy for triple negative breast cancer (TNBC) is still limited. In recent years, some nanosystems for malignancy treatment have received considerable attention. In this study, we prepared caramelized nanospheres (CNSs) loaded with doxorubicin (DOX) and Fe3O4 to achieve the synergistic effect of combined therapy and real-time magnetic resonance imaging (MRI) monitoring, so as to improve the diagnosis and therapeutic effect of TNBC. Methods CNSs with biocompatibility and unique optical properties were prepared by hydrothermal method, DOX and Fe3O4 were loaded on it to obtain Fe3O4/DOX@CNSs nanosystem. Characteristics including morphology, hydrodynamic size, zeta potentials and magnetic properties of Fe3O4/DOX@CNSs were evaluated. The DOX release was evaluated by different pH/near-infrared (NIR) light energy. Biosafety, pharmacokinetics, MRI and therapeutic treatment of Fe3O4@CNSs, DOX and Fe3O4/DOX@CNSs were examined in vitro or in vivo. Results Fe3O4/DOX@CNSs has an average particle size of 160 nm and a zeta potential of 27.5mV, it demonstrated that Fe3O4/DOX@CNSs is a stable and homogeneous dispersed system. The hemolysis experiment of Fe3O4/DOX@CNSs proved that it can be used in vivo. Fe3O4/DOX@CNSs displayed high photothermal conversion efficiency, extensive pH/heat-induced DOX release. 70.3% DOX release is observed under the 808 nm laser in the pH = 5 PBS solution, obviously higher than pH = 5 (50.9%) and pH = 7.4 (less than 10%). Pharmacokinetic experiments indicated the t1/2β, and AUC0-t of Fe3O4/DOX@CNSs were 1.96 and 1.31 -fold higher than those of DOX solution, respectively. Additionally, Fe3O4/DOX@CNSs with NIR had the greatest tumor suppression in vitro and in vivo. Moreover, this nanosystem demonstrated distinct contrast enhancement on T2 MRI to achieve real-time imaging monitoring during treatment. Conclusion Fe3O4/DOX@CNSs is a highly biocompatible, double-triggering and improved DOX bioavailability nanosystem that combines chemo-PTT and real-time MRI monitoring to achieve integration of diagnosis and treatment of TNBC.
Collapse
Affiliation(s)
- Fangqing Wang
- Department of Radiology, Qilu Hospital, Shandong University, Affiliated Hospital of Shandong University, Jinan, 250012, People’s Republic of China
| | - Nianlu Li
- Physical and Chemical Laboratory, Shandong Academy of Occupational Health and Occupational Medicine, Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250002, People’s Republic of China
| | - Wenbo Wang
- Department of Radiology, Qilu Hospital, Shandong University, Affiliated Hospital of Shandong University, Jinan, 250012, People’s Republic of China
| | - Long Ma
- The Testing Center of Shandong Bureau of China Metallurgical Geology Bureau, Shandong Normal University, Jinan, 250014, People’s Republic of China
| | - Yaru Sun
- Department of Nuclear Medicine, The Second Hospital of Shandong University, Affiliated Hospital of Shandong University, Jinan, 250033, People’s Republic of China
| | - Hong Wang
- Department of Radiology, Qilu Hospital, Shandong University, Affiliated Hospital of Shandong University, Jinan, 250012, People’s Republic of China
| | - Jinhua Zhan
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, People’s Republic of China,Correspondence: Jinhua Zhan, School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, People’s Republic of China, Email
| | - Dexin Yu
- Department of Radiology, Qilu Hospital, Shandong University, Affiliated Hospital of Shandong University, Jinan, 250012, People’s Republic of China,Dexin Yu, Department of Radiology, Qilu Hospital, Shandong University, Affiliated Hospital of Shandong University, Jinan, 250012, People’s Republic of China, Tel +86-18560081629, Fax +86-531-86927544, Email
| |
Collapse
|
50
|
Application value of circulating LncRNA in diagnosis, treatment, and prognosis of breast cancer. Funct Integr Genomics 2023; 23:61. [PMID: 36792760 DOI: 10.1007/s10142-023-00983-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/17/2023]
Abstract
Breast cancer is the malignant tumor with the highest incidence in women worldwide. It is highly heterogeneous, has a high incidence of drug resistance, recurrence, and metastasis, and is one of the malignant tumors with the highest mortality rate. The early diagnosis, treatment monitoring, and prognosis assessment of breast cancer are the key factors affecting the survival of patients. However, due to the lack of specific biomarkers, breast cancer is still an essential factor affecting women's quality of life and physical and mental health. Long non-coding RNA can regulate various genes and different signaling pathways and plays an essential role in the occurrence and development of tumors. Recent studies have found that the abnormal expression of circulating long non-coding RNA in serum, saliva, and other biological body fluids plays a significant role in early diagnosis, pathological classification, stage, therapeutic effect monitoring, and prognosis evaluation of breast cancer. This article will review the potential application value of circulating lncRNA in breast cancer.
Collapse
|