1
|
Yuan J, Yang L, Zhang H, Beeraka NM, Zhang D, Wang Q, Wang M, Pr HV, Sethi G, Wang G. Decoding tumor microenvironment: EMT modulation in breast cancer metastasis and therapeutic resistance, and implications of novel immune checkpoint blockers. Biomed Pharmacother 2024; 181:117714. [PMID: 39615165 DOI: 10.1016/j.biopha.2024.117714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/15/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024] Open
Abstract
Tumor microenvironment (TME) and epithelial-mesenchymal transition (EMT) play crucial roles in the initiation and progression of tumors. TME is composed of various cell types, such as immune cells, fibroblasts, and endothelial cells, as well as non-cellular components like extracellular matrix (ECM) proteins and soluble factors. These elements interact with tumor cells through a complex network of signaling pathways involving cytokines, growth factors, metabolites, and non-coding RNA-carrying exosomes. Hypoxic conditions within the TME further modulate these interactions, collectively influencing tumor growth, metastatic potential, and response to therapy. EMT represents a dynamic and reversible process where epithelial cells undergo phenotypic changes to adopt mesenchymal characteristics in several cancers, including breast cancers. This transformation enhances cell motility and imparts stem cell-like properties, which are closely associated with increased metastatic capability and resistance to conventional cancer treatments. Thus, understanding the crosstalk between the TME and EMT is essential for unraveling the underlying mechanisms of breast cancer metastasis and therapeutic resistance. This review uniquely examines the intricate interplay between the tumor TME and epithelial-mesenchymal transition EMT in driving breast cancer metastasis and treatment resistance. It explores the therapeutic potential of targeting the TME-EMT axis, specifically through CD73-TGF-β dual-blockade, to improve outcomes in triple-negative breast cancer. Additionally, it underscores new strategies to enhance immune checkpoint blockade (ICB) responses by modulating EMT, thereby offering innovative insights for more effective cancer treatment.
Collapse
Affiliation(s)
- Jie Yuan
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| | - Li Yang
- Department of Clinical Laboratory Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| | - Hua Zhang
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| | - Narasimha M Beeraka
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya Str., Moscow 119991, Russia; Raghavendra Institute of Pharmaceutical Education and Research (RIPER), Chiyyedu, Anantapuramu, Andhra Pradesh 515721, India; Department of Studies in Molecular Biology, Faculty of Science and Technology, University of Mysore, Mysore, Karnataka, 570006, India.
| | - Danfeng Zhang
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| | - Qun Wang
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| | - Minghua Wang
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| | - Hemanth Vikram Pr
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Geng Wang
- Department of Breast, Thyroid and Vascular Surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| |
Collapse
|
2
|
Janus P, Kuś P, Jaksik R, Vydra N, Toma-Jonik A, Gramatyka M, Kurpas M, Kimmel M, Widłak W. Transcriptional responses to direct and indirect TGFB1 stimulation in cancerous and noncancerous mammary epithelial cells. Cell Commun Signal 2024; 22:522. [PMID: 39468555 PMCID: PMC11514872 DOI: 10.1186/s12964-024-01821-5] [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: 03/26/2024] [Accepted: 09/07/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Transforming growth factor beta (TGFβ) is important for the morphogenesis and secretory function of the mammary gland. It is one of the main activators of the epithelial-mesenchymal transition (EMT), a process important for tissue remodeling and regeneration. It also provides cells with the plasticity to form metastases during tumor progression. Noncancerous and cancer cells respond differently to TGFβ. However, knowledge of the cellular signaling cascades triggered by TGFβ in various cell types is still limited. METHODS MCF10A (noncancerous, originating from fibrotic breast tissue) and MCF7 (cancer, estrogen receptor-positive) breast epithelial cells were treated with TGFB1 directly or through conditioned media from stimulated cells. Transcriptional changes (via RNA-seq) were assessed in untreated cells and after 1-6 days of treatment. Differentially expressed genes were detected with DESeq2 and the hallmark collection was selected for gene set enrichment analysis. RESULTS TGFB1 induces EMT in both the MCF10A and MCF7 cell lines but via slightly different mechanisms (signaling through SMAD3 is more active in MCF7 cells). Many EMT-related genes are expressed in MCF10A cells at baseline. Both cell lines respond to TGFB1 by decreasing the expression of genes involved in cell proliferation: through the repression of MYC (and the protein targets) in MCF10A cells and the activation of p63-dependent signaling in MCF7 cells (CDKN1A and CDKN2B, which are responsible for the inhibition of cyclin-dependent kinases, are upregulated). In addition, estrogen receptor signaling is inhibited and caspase-dependent cell death is induced only in MCF7 cells. Direct incubation with TGFB1 and treatment of cells with conditioned media similarly affected transcriptional profiles. However, TGFB1-induced protein secretion is more pronounced in MCF10A cells; therefore, the signaling is propagated through conditioned media (bystander effect) more effectively in MCF10A cells than in MCF7 cells. CONCLUSIONS Estrogen receptor-positive breast cancer patients may benefit from high levels of TGFB1 expression due to the repression of estrogen receptor signaling, inhibition of proliferation, and induction of apoptosis in cancer cells. However, some TGFB1-stimulated cells may undergo EMT, which increases the risk of metastasis.
Collapse
Affiliation(s)
- Patryk Janus
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, Gliwice, 44-102, Poland
| | - Paweł Kuś
- Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka 16, Gliwice, 44-100, Poland
| | - Roman Jaksik
- Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka 16, Gliwice, 44-100, Poland
| | - Natalia Vydra
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, Gliwice, 44-102, Poland
| | - Agnieszka Toma-Jonik
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, Gliwice, 44-102, Poland
| | - Michalina Gramatyka
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, Gliwice, 44-102, Poland
| | - Monika Kurpas
- Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka 16, Gliwice, 44-100, Poland
| | - Marek Kimmel
- Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka 16, Gliwice, 44-100, Poland.
- Departments of Statistics and Bioengineering, Rice University, Houston, TX, USA.
| | - Wiesława Widłak
- Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, Gliwice, 44-102, Poland.
| |
Collapse
|
3
|
Matikhina T, Cohen CJ. Targeting TGFβ with chimeric switch receptor and secreted trap to improve T cells anti-tumor activity. Front Immunol 2024; 15:1460266. [PMID: 39512355 PMCID: PMC11540659 DOI: 10.3389/fimmu.2024.1460266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/23/2024] [Indexed: 11/15/2024] Open
Abstract
Introduction TGFβ is a major immunoinhibitory factor present in the microenvironment of solid tumors. Various cancer types acquire the ability to overexpress TGFβ to escape immune response. Specifically, TGFβ dampens cytotoxic T cell activity, and its presence has been correlated with tumor invasion and poor prognosis. Methods In this study, we developed two approaches to counteract the effects of TGFβ and provide a functional advantage to genetically engineered T cells in the immunoinhibitory tumor milieu. We designed a TGFβRI-based co-stimulatory switch receptor (CSRI), comprising the TGFβ receptor I extracellular binding domain and a 4-1BB co-stimulatory signaling moiety. Additionally, we tested the efficacy of a TGFβ-binding scFv trap produced by T cells. Results We demonstrated that both approaches enhanced tumor-specific T cell cytokine secretion, upregulated activation markers, and reduced inhibition markers upon co-culture with melanoma targets. Furthermore, CSRI and the anti-TGFβ trap exhibited improved anti-tumor function in vivo. Conclusion Overall, we show that targeting the TGFβ pathway can enhance cellular immunotherapy.
Collapse
Affiliation(s)
| | - Cyrille J. Cohen
- The Laboratory of Tumor Immunology and Immunotherapy, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
4
|
Lo HC, Lin TE, Lin CY, Wang WH, Chen YC, Tsai PH, Su JC, Lu MK, Hsu WH, Lin TY. Targeting TGFβ receptor-mediated snail and twist: WSG, a polysaccharide from Ganoderma lucidum, and it-based dissolvable microneedle patch suppress melanoma cells. Carbohydr Polym 2024; 341:122298. [PMID: 38876710 DOI: 10.1016/j.carbpol.2024.122298] [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: 04/18/2024] [Accepted: 05/19/2024] [Indexed: 06/16/2024]
Abstract
Cutaneous melanoma is a lethal skin cancer variant with pronounced aggressiveness and metastatic potential. However, few targeted medications inhibit the progression of melanoma. Ganoderma lucidum, which is a type of mushroom, is widely used as a non-toxic alternative adjunct therapy for cancer patients. This study determines the effect of WSG, which is a water-soluble glucan that is derived from G. lucidum, on melanoma cells. The results show that WSG inhibits cell viability and the mobility of melanoma cells. WSG induces changes in the expression of epithelial-to-mesenchymal transition (EMT)-related markers. WSG also downregulates EMT-related transcription factors, Snail and Twist. Signal transduction assays show that WSG reduces the protein levels in transforming growth factor β receptors (TGFβRs) and consequently inhibits the phosphorylation of intracellular signaling molecules, such as FAK, ERK1/2 and Smad2. An In vivo study shows that WSG suppresses melanoma growth in B16F10-bearing mice. To enhance transdermal drug delivery and prevent oxidation, two highly biocompatible compounds, polyvinyl alcohol (PVA) and polyvinylpyrrolidone (PVP), are used to synthesize a dissolvable microneedle patch that is loaded with WSG (MN-WSG). A functional assay shows that MN-WSG has an effect that is comparable to that of WSG alone. These results show that WSG has significant potential as a therapeutic agent for melanoma treatment. MN-WSG may allow groundbreaking therapeutic approaches and offers a novel method for delivering this potent compound effectively.
Collapse
Affiliation(s)
- Hung-Chih Lo
- Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan; Institute of Traditional Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Tzu-En Lin
- Institute of Biomedical Engineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan; Institute of Applied Mechanics, National Taiwan University, Taipei, Taiwan
| | - Che-Yu Lin
- Institute of Applied Mechanics, National Taiwan University, Taipei, Taiwan
| | - Wei-Hao Wang
- Institute of Traditional Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Chen Chen
- Institute of Biomedical Engineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Pei-Hsien Tsai
- Institute of Biomedical Engineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Jung-Chen Su
- Department of Pharmacy, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Mei-Kuang Lu
- Institute of Traditional Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan; Department of Chinese Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Traditional Chinese Medicine Glycomics Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wei-Hung Hsu
- Institute of Traditional Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; LO-Sheng Hospital Ministry of Health and Welfare, Taipei, Taiwan; School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan; Department of Chinese Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Tung-Yi Lin
- Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan; Institute of Traditional Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Chinese Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Traditional Chinese Medicine Glycomics Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
5
|
Barros AB, Teles FB, Araújo DD, Da Silva DA, Santos LBPD, Aldeman NLS, Cajado AG, Assef ANB, Wilke DV, Lima-Junior RCP, Araújo AJ, Marinho-Filho JDB. Combining cashew gum with cyclophosphamide in murine melanoma model: A strategy for the reduction of side effects. Int J Biol Macromol 2024; 275:133588. [PMID: 38960246 DOI: 10.1016/j.ijbiomac.2024.133588] [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: 03/02/2024] [Revised: 06/28/2024] [Accepted: 06/29/2024] [Indexed: 07/05/2024]
Abstract
The understanding of cancer immunity and antitumor factors generated by natural polysaccharides is not yet fully comprehended. Polysaccharides, like cashew gum (CG), can exhibit immunomodulatory action and may assist in the antitumor process and side effects relieve. This study aimed to determine the antitumor effect of CG alone or in combination with cyclophosphamide (CTX), and its interactions with immune cells, in a murine melanoma model, using the B16-F10 cell line. Tumor growth inhibition, hematological, histopathological, ELISA, flow cytometry, immunofluorescence, and qRT-PCR analyses were performed to elucidate the antitumor potential, involvement of immune cells, and potential toxic effects. CG showed significant tumor growth inhibition, reaching up to 42.9 % alone and 51.4 % in combination with CTX, with mild toxicity to organs. CG enhanced leukocyte count, even in the presence of CTX. Furthermore, CG influenced the activation of tumor-associated macrophages (TAM), characterized by an increase in Il4, as well as a reduction in Ifng, Il1b, Tgfb, and Il6 gene expression. Nevertheless, these effects did not compromise the antitumor activity of CG. In summary, the combination of CG with CTX is a promising approach for leukopenia, one of the most important side effects of cancer treatment and deserves further investigation.
Collapse
Affiliation(s)
- Ayslan Batista Barros
- Universidade Federal do Delta do Parnaíba, Núcleo de Pesquisa e Pós-graduação, 64.202-020 Parnaíba, PI, Brazil
| | - Felipe Barros Teles
- Núcleo de Pesquisa e Desenvolvimento de Medicamentos, Departamento de Fisiologia e Farmacologia, Universidade Federal do Ceará, 60430-160 Fortaleza, Ceará, Brazil
| | - Dakson Douglas Araújo
- Universidade Federal do Delta do Parnaíba, Núcleo de Pesquisa e Pós-graduação, 64.202-020 Parnaíba, PI, Brazil
| | - Durcilene Alves Da Silva
- Universidade Federal do Delta do Parnaíba, Núcleo de Pesquisa e Pós-graduação, 64.202-020 Parnaíba, PI, Brazil
| | | | - Nayze Lucena Sangreman Aldeman
- Universidade Federal do Delta do Parnaíba, Núcleo de Pesquisa e Pós-graduação, 64.202-020 Parnaíba, PI, Brazil; Faculdade de Ciências Humanas, Exatas e da Saúde do Piauí, Instituto de Educação Superior do Vale do Parnaíba, 64212-790 Parnaíba, Brazil
| | - Aurilene Gomes Cajado
- Núcleo de Pesquisa e Desenvolvimento de Medicamentos, Departamento de Fisiologia e Farmacologia, Universidade Federal do Ceará, 60430-160 Fortaleza, Ceará, Brazil
| | - Alexia Nathália Brígido Assef
- Núcleo de Pesquisa e Desenvolvimento de Medicamentos, Departamento de Fisiologia e Farmacologia, Universidade Federal do Ceará, 60430-160 Fortaleza, Ceará, Brazil
| | - Diego Veras Wilke
- Núcleo de Pesquisa e Desenvolvimento de Medicamentos, Departamento de Fisiologia e Farmacologia, Universidade Federal do Ceará, 60430-160 Fortaleza, Ceará, Brazil
| | - Roberto Cesar Pereira Lima-Junior
- Núcleo de Pesquisa e Desenvolvimento de Medicamentos, Departamento de Fisiologia e Farmacologia, Universidade Federal do Ceará, 60430-160 Fortaleza, Ceará, Brazil
| | - Ana Jérsia Araújo
- Universidade Federal do Delta do Parnaíba, Núcleo de Pesquisa e Pós-graduação, 64.202-020 Parnaíba, PI, Brazil
| | | |
Collapse
|
6
|
Tienoue Fotso HM, Mbong Angie MA, Ntentie FR, Makamwe I, Edoun Ebouel FL, Kenjing Ndansack E, Julius Oben E. Aqueous Extract of Leaves and Flowers of Acmella caulirhiza Reduces the Proliferation of Cancer Cells by Underexpressing Some Genes and Activating Caspase-3. Biochem Res Int 2024; 2024:3293305. [PMID: 38371392 PMCID: PMC10874292 DOI: 10.1155/2024/3293305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 10/16/2023] [Accepted: 01/29/2024] [Indexed: 02/20/2024] Open
Abstract
The increasing prevalence of cancers and the multiple side effects of cancer treatments have led researchers to constantly search for plants containing bioactive compounds with cell death properties. This work aimed at evaluating the antiproliferative effect of an Acmella caulirhiza extract. After evaluation of the in vitro antioxidant potential of the three extracts of Acmella caulirhiza (aqueous (AE-Ac), hydroethanolic (HEE-Ac), and ethanolic (EE-Ac)) through the scavenging of DPPH● and NO● radicals, the extract with the best antioxidant activity was selected for bioactive compound assessment and antiproliferative tests. Subsequently, the cytotoxic activity was evaluated on the viability of breast (MCF-7), brain (CT2A, SB-28, and GL-261), colon (MC-38), and skin (YUMM 1.7 and B16-F1) cancer lines using the MTT method. Then, the line where the extract was the most active was selected to evaluate the expression of certain genes involved in cancerogenesis by RT-PCR and the expression of cleaved caspase-3 involved in cell death mechanism by western blot. The AE-Ac showed the best scavenging activity with IC50s of 0.52 and 0.02 for DPPH● and NO●, respectively. This AE-Ac was found to contain alkaloids, flavonoids, and tannins and was more active on YUMM 1.7 cells (IC50 = 149.42 and 31.99 μg/mL for 24 and 48 h, respectively). Results also showed that AE-Ac downregulated the expression of inflammation (IL-1b (p = 0.017) and IL-6 (p = 0.028)), growth factors (PDGF (p = 0.039), IGF (p = 0.034), E2F1(p = 0.038), and E2F2(p = 0.016)), and antiapoptotic protein genes (Bcl-2 (p = 0.028) and Bcl-6 (p = 0.039)). The cleaved caspase-3 was positively modulated by the AE-Ac inducing thus cell death by apoptosis. AE-Ac showed inhibitory effects on the expression of genes involved in cancer progression making it a potential health intervention agent that can be exploited in cancer therapy protocols.
Collapse
Affiliation(s)
| | - Mary-Ann Mbong Angie
- Department of Biochemistry, Faculty of Sciences, University of Yaounde 1, P.O. Box: 812, Yaounde, Cameroon
| | - Françoise Raïssa Ntentie
- Department of Biological Science, Higher Teachers' Training College, University of Yaounde 1, P.O. Box: 47, Yaounde, Cameroon
| | - Inelle Makamwe
- Department of Biochemistry, Faculty of Sciences, University of Yaounde 1, P.O. Box: 812, Yaounde, Cameroon
| | - Ferdinand Lanvin Edoun Ebouel
- Department of Biochemistry, Faculty of Sciences, University of Yaounde 1, P.O. Box: 812, Yaounde, Cameroon
- Centre for Food and Nutrition Research, Institute of Medical Research and Medicinal Plant Studies, MINRESI, P.O. Box: 13033, Yaounde, Cameroon
| | | | - Enyong Julius Oben
- Department of Biochemistry, Faculty of Sciences, University of Yaounde 1, P.O. Box: 812, Yaounde, Cameroon
- Cameroon Nutrition and Dietetics Research Center, J&A Oben Foundation, P.O. Box: 8348, Yaounde, Cameroon
| |
Collapse
|
7
|
Wang P, Zhou R, Zhou R, Feng S, Zhao L, Li W, Lin J, Rajapakse A, Lee CH, Furnari FB, Burgess AW, Gunter JH, Liu G, Ostrikov KK, Richard DJ, Simpson F, Dai X, Thompson EW. Epidermal growth factor potentiates EGFR(Y992/1173)-mediated therapeutic response of triple negative breast cancer cells to cold atmospheric plasma-activated medium. Redox Biol 2024; 69:102976. [PMID: 38052106 PMCID: PMC10746566 DOI: 10.1016/j.redox.2023.102976] [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: 10/26/2023] [Accepted: 11/24/2023] [Indexed: 12/07/2023] Open
Abstract
Cold atmospheric plasma (CAP) holds promise as a cancer-specific treatment that selectively kills various types of malignant cells. We used CAP-activated media (PAM) to utilize a range of the generated short- and long-lived reactive species. Specific antibodies, small molecule inhibitors and CRISPR/Cas9 gene-editing approaches showed an essential role for receptor tyrosine kinases, especially epidermal growth factor (EGF) receptor, in mediating triple negative breast cancer (TNBC) cell responses to PAM. EGF also dramatically enhanced the sensitivity and specificity of PAM against TNBC cells. Site-specific phospho-EGFR analysis, signal transduction inhibitors and reconstitution of EGFR-depleted cells with EGFR-mutants confirmed the role of phospho-tyrosines 992/1173 and phospholipase C gamma signaling in up-regulating levels of reactive oxygen species above the apoptotic threshold. EGF-triggered EGFR activation enhanced the sensitivity and selectivity of PAM effects on TNBC cells. The proposed approach based on the synergy of CAP and EGFR-targeted therapy may provide new opportunities to improve the clinical management of TNBC.
Collapse
Affiliation(s)
- Peiyu Wang
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China; Centre for Genomics and Personalised Health, School of Biomedical Science, Faculty of Health, Queensland University of Technology, Brisbane, Queensland 4059, Australia; Translational Research Institute, Woolloongabba, Queensland 4102, Australia; State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, PR China
| | - Renwu Zhou
- State Key Laboratory of Electrical Insulation and Power Equipment, Centre for Plasma Biomedicine, School of Electrical Engineering, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Rusen Zhou
- School of Chemistry and Physics, Queensland University of Technology, Brisbane, Queensland 4000, Australia
| | - Shuo Feng
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Liqian Zhao
- Department of Neurosurgery, Institute of Brain Disease, Nanfang Hospital of Southern Medical University, Guangzhou 510515, PR China
| | - Wenshao Li
- School of Chemistry and Physics, Queensland University of Technology, Brisbane, Queensland 4000, Australia
| | - Jinyong Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, PR China
| | - Aleksandra Rajapakse
- Centre for Genomics and Personalised Health, School of Biomedical Science, Faculty of Health, Queensland University of Technology, Brisbane, Queensland 4059, Australia; Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| | - Chia-Hwa Lee
- Centre for Genomics and Personalised Health, School of Biomedical Science, Faculty of Health, Queensland University of Technology, Brisbane, Queensland 4059, Australia; Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| | - Frank B Furnari
- Department of Medicine, University of California San Diego, California 92093, USA
| | - Antony W Burgess
- Walter and Elisa Hall Institute, Melbourne, Victoria 3052, Australia
| | - Jennifer H Gunter
- Centre for Genomics and Personalised Health, School of Biomedical Science, Faculty of Health, Queensland University of Technology, Brisbane, Queensland 4059, Australia; Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| | - Gang Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, PR China
| | - Kostya Ken Ostrikov
- School of Chemistry and Physics, Queensland University of Technology, Brisbane, Queensland 4000, Australia
| | - Derek J Richard
- Centre for Genomics and Personalised Health, School of Biomedical Science, Faculty of Health, Queensland University of Technology, Brisbane, Queensland 4059, Australia; Translational Research Institute, Woolloongabba, Queensland 4102, Australia; Cancer and Ageing Research Program, Woolloongabba, Queensland 4102, Australia
| | - Fiona Simpson
- Frazer Institute, The University of Queensland, Brisbane, Queensland 4102, Australia
| | - Xiaofeng Dai
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China; Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China.
| | - Erik W Thompson
- Centre for Genomics and Personalised Health, School of Biomedical Science, Faculty of Health, Queensland University of Technology, Brisbane, Queensland 4059, Australia; Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| |
Collapse
|
8
|
Poursani EM, Mercatelli D, Raninga P, Bell JL, Saletta F, Kohane FV, Neumann DP, Zheng Y, Rouaen JRC, Jue TR, Michniewicz FT, Schadel P, Kasiou E, Tsoli M, Cirillo G, Waters S, Shai-Hee T, Cazzoli R, Brettle M, Slapetova I, Kasherman M, Whan R, Souza-Fonseca-Guimaraes F, Vahdat L, Ziegler D, Lock JG, Giorgi FM, Khanna K, Vittorio O. Copper chelation suppresses epithelial-mesenchymal transition by inhibition of canonical and non-canonical TGF-β signaling pathways in cancer. Cell Biosci 2023; 13:132. [PMID: 37480151 PMCID: PMC10362738 DOI: 10.1186/s13578-023-01083-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 07/10/2023] [Indexed: 07/23/2023] Open
Abstract
BACKGROUND Metastatic cancer cells exploit Epithelial-mesenchymal-transition (EMT) to enhance their migration, invasion, and resistance to treatments. Recent studies highlight that elevated levels of copper are implicated in cancer progression and metastasis. Clinical trials using copper chelators are associated with improved patient survival; however, the molecular mechanisms by which copper depletion inhibits tumor progression and metastasis are poorly understood. This remains a major hurdle to the clinical translation of copper chelators. Here, we propose that copper chelation inhibits metastasis by reducing TGF-β levels and EMT signaling. Given that many drugs targeting TGF-β have failed in clinical trials, partly because of severe side effects arising in patients, we hypothesized that copper chelation therapy might be a less toxic alternative to target the TGF-β/EMT axis. RESULTS Our cytokine array and RNA-seq data suggested a link between copper homeostasis, TGF-β and EMT process. To validate this hypothesis, we performed single-cell imaging, protein assays, and in vivo studies. Here, we used the copper chelating agent TEPA to block copper trafficking. Our in vivo study showed a reduction of TGF-β levels and metastasis to the lung in the TNBC mouse model. Mechanistically, TEPA significantly downregulated canonical (TGF-β/SMAD2&3) and non-canonical (TGF-β/PI3K/AKT, TGF-β/RAS/RAF/MEK/ERK, and TGF-β/WNT/β-catenin) TGF-β signaling pathways. Additionally, EMT markers of MMP-9, MMP-14, Vimentin, β-catenin, ZEB1, and p-SMAD2 were downregulated, and EMT transcription factors of SNAI1, ZEB1, and p-SMAD2 accumulated in the cytoplasm after treatment. CONCLUSIONS Our study suggests that copper chelation therapy represents a potentially effective therapeutic approach for targeting TGF-β and inhibiting EMT in a diverse range of cancers.
Collapse
Affiliation(s)
- Ensieh M Poursani
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Daniele Mercatelli
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Prahlad Raninga
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Jessica L Bell
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Federica Saletta
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Felix V Kohane
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Daniel P Neumann
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Ye Zheng
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Jourdin R C Rouaen
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Toni Rose Jue
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Filip T Michniewicz
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Piper Schadel
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Erin Kasiou
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Maria Tsoli
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
| | - Giuseppe Cirillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Shafagh Waters
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Tyler Shai-Hee
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Riccardo Cazzoli
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Merryn Brettle
- Katharina Gauss Light Microscopy Facility, University of New South Wales, Sydney, NSW, Australia
| | - Iveta Slapetova
- Katharina Gauss Light Microscopy Facility, University of New South Wales, Sydney, NSW, Australia
| | - Maria Kasherman
- Katharina Gauss Light Microscopy Facility, University of New South Wales, Sydney, NSW, Australia
| | - Renee Whan
- Katharina Gauss Light Microscopy Facility, University of New South Wales, Sydney, NSW, Australia
| | | | | | - David Ziegler
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
| | - John G Lock
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Federico M Giorgi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - KumKum Khanna
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Orazio Vittorio
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia.
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
9
|
Lin TY, Lo HC, Qiu WL, Chao CH, Lu MK, Hsu HY. Biochemical characterization and anti-cancer activity of tangential flow filtration system assisted purification of fucoglucan from Laminaria japonica. Int J Biol Macromol 2023; 227:1-9. [PMID: 36528139 DOI: 10.1016/j.ijbiomac.2022.12.078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 11/15/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022]
Abstract
Polysaccharide from Laminaria japonica (LJPS) exhibits multiple biological functions. However, we found that crude LJPS doesn't show good anti-lung cancer activity in this study. We therefore used tangential flow filtration (TFF) system to optimize the anticancer activity of LJPS. We divided the crude LJPS into two fractions by TFF system with a 10 kDa filter and denoted as retentate (10K-R) and filtration (10K-F). The chemical assay revealed that the main molecular mass of 10K-R and 10K-F is about 985 and 3 kDa, respectively. The main components of 10K-R include fucose (19.3 %), and glucose (59.5 %); while glucose (88.6 %) is a major component of 10K-F. Biological functions showed that 10K-R but not 10K-F inhibited the viability and mobility of cancer cells. 10K-R downregulated expressions of transforming growth factor β receptor and Slug, and inhibited intracellular signaling molecules, including FAK, AKT, ERK1/2, and Smad2. This study is the first concept to purify the polysaccharide by TFF system and showed the potential mechanism of 10K-R inhibited cancer cells.
Collapse
Affiliation(s)
- Tung-Yi Lin
- Institute of Traditional Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Biomedical Industry Ph.D. Program, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Hung-Chih Lo
- Institute of Traditional Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wei-Lun Qiu
- Institute of Traditional Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chi-Hsein Chao
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan
| | - Mei-Kuang Lu
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan; Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan.
| | - Hsien-Yeh Hsu
- Institute of Taiwan Fucoidan Development, Taipei, Taiwan; Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
10
|
Influence of tumor cell-derived TGF-β on macrophage phenotype and macrophage-mediated tumor cell invasion. Int J Biochem Cell Biol 2022; 153:106330. [DOI: 10.1016/j.biocel.2022.106330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 11/06/2022]
|
11
|
Das SK, Mishra S, Saha KD, Chandra D, Hara M, Mostafa AA, Bhaumik A. N-Rich, Polyphenolic Porous Organic Polymer and Its In Vitro Anticancer Activity on Colorectal Cancer. Molecules 2022; 27:7326. [PMID: 36364150 PMCID: PMC9657835 DOI: 10.3390/molecules27217326] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/19/2022] [Accepted: 10/24/2022] [Indexed: 08/15/2023] Open
Abstract
N-rich organic materials bearing polyphenolic moieties in their building networks and nanoscale porosities are very demanding in the context of designing efficient biomaterials or drug carriers for the cancer treatment. Here, we report the synthesis of a new triazine-based secondary-amine- and imine-linked polyphenolic porous organic polymer material TrzTFPPOP and explored its potential for in vitro anticancer activity on the human colorectal carcinoma (HCT 116) cell line. This functionalized (-OH, -NH-, -C=N-) organic material displayed an exceptionally high BET surface area of 2140 m2 g-1 along with hierarchical porosity (micropores and mesopores), and it induced apoptotic changes leading to high efficiency in colon cancer cell destruction via p53-regulated DNA damage pathway. The IC30, IC50, and IC70 values obtained from the MTT assay are 1.24, 3.25, and 5.25 μg/mL, respectively.
Collapse
Affiliation(s)
- Sabuj Kanti Das
- School of Materials Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Snehasis Mishra
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata 700032, India
| | - Krishna Das Saha
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata 700032, India
| | - Debraj Chandra
- World Research Hub Initiative, Institute of Innovative Research, Tokyo Institute of Technology, Nagatsuta-cho 4259, Midori-ku, Yokohama 226-8503, Japan
| | - Michikazu Hara
- Laboratory for Materials and Structures, Institute of Innovative Research, Tokyo Institute of Technology, Nagatsuta-cho 4259, Midori-ku, Yokohama 226-8503, Japan
| | - Amany A. Mostafa
- Nanomedicine & Tissue Engineering Laboratory, Department of Ceramic, National Research Centre, El Bohouth St., Dokki, Cairo 12622, Egypt
| | - Asim Bhaumik
- School of Materials Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| |
Collapse
|
12
|
Hypoxia Selectively Increases a SMAD3 Signaling Axis to Promote Cancer Cell Invasion. Cancers (Basel) 2022; 14:cancers14112751. [PMID: 35681731 PMCID: PMC9179584 DOI: 10.3390/cancers14112751] [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: 05/18/2022] [Accepted: 05/30/2022] [Indexed: 01/27/2023] Open
Abstract
Transforming growth factor β (TGFβ) plays a paradoxical role in cancer, first inhibiting then promoting its progression, a duality that poses a real challenge for the development of effective TGFβ-targeted therapies. The major TGFβ downstream effectors, SMAD2 and SMAD3, display both distinct and overlapping functions and accumulating evidence suggests that their activation ratio may contribute to the dual effect of TGFβ. However, the mechanisms responsible for their selective activation remain poorly understood. Here, we provide experimental evidence that hypoxia induces the pro-invasive arm of TGFβ signaling through a selective increase in SMAD3 interaction with SMAD-Anchor for Receptor Activation (SARA). This event relies on HDAC6-dependent SMAD3 bioavailability, as well as increased SARA recruitment to EEA1+ endosomes. A motility gene expression study indicated that SMAD3 selectively increased the expression of ITGB2 and VIM, two genes that were found to be implicated in hypoxia-induced cell invasion and associated with tumor progression and metastasis in cohorts of cancer patients. Furthermore, CAM xenograft assays show the significant benefit of selective inhibition of the SMAD3 signaling pathway as opposed to global TGFβ inhibition in preventing tumor progression. Overall, these results suggest that fine-tuning of the pro-invasive HDAC6-SARA-SMAD3 axis could be a better strategy towards effective cancer treatments.
Collapse
|
13
|
Shi N, Wang Z, Zhu H, Liu W, Zhao M, Jiang X, Zhao J, Ren C, Zhang Y, Luo L. Research progress on drugs targeting the TGF-β signaling pathway in fibrotic diseases. Immunol Res 2022; 70:276-288. [PMID: 35147920 PMCID: PMC9197809 DOI: 10.1007/s12026-022-09267-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/27/2022] [Indexed: 02/07/2023]
Abstract
Tissue fibrosis is a key factor leading to disability and death worldwide; however, thus far, there are no approved treatments for fibrosis. Transforming growth factor (TGF)-β is a major pro-fibrotic cytokine, which is expected to become a target in the treatment of fibrosis; however, since TGF-β has a wide range of biological functions involving a variety of biological processes in the body, a slight change in TGF-β may have a systematic effect. Indiscriminate inhibition of TGF-β can lead to adverse reactions, which can affect the efficacy of treatment. Therefore, it has become very important to explore how both the TGF-β signaling pathway is inhibited and the safe and efficient TGF-β small molecule inhibitors or neutralizing antibodies are designed in the treatment of fibrotic diseases. In this review, we mainly discuss the key role of the TGF-β signaling pathway in fibrotic diseases, as well as the development of fibrotic drugs in recent years, and explore potential targets in the treatment of fibrotic diseases in order to guide subsequent drug development.
Collapse
Affiliation(s)
- Ning Shi
- Department of Neurosurgery, Cancer Research Institute, School of Basic Medical Science, Xiangya Hospital, Central South University, Changsha, 410008, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, 410008, China
| | - Zhihong Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Taiping Road #27, Beijing, 100850, China
| | - Hecheng Zhu
- Changsha Kexin Cancer Hospital, Changsha, 410205, Hunan, China
| | - Weidong Liu
- Department of Neurosurgery, Cancer Research Institute, School of Basic Medical Science, Xiangya Hospital, Central South University, Changsha, 410008, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, 410008, China
| | - Ming Zhao
- Changsha Kexin Cancer Hospital, Changsha, 410205, Hunan, China
| | - Xingjun Jiang
- Department of Neurosurgery, Cancer Research Institute, School of Basic Medical Science, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jin Zhao
- Department of Neurosurgery, Cancer Research Institute, School of Basic Medical Science, Xiangya Hospital, Central South University, Changsha, 410008, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, 410008, China
| | - Caiping Ren
- Department of Neurosurgery, Cancer Research Institute, School of Basic Medical Science, Xiangya Hospital, Central South University, Changsha, 410008, China.
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, 410008, China.
| | - Yan Zhang
- Department of Obstetrics and Gynecology, First Medical Center, General Hospital of Chinese PLA, Beijing, China.
| | - Longlong Luo
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Taiping Road #27, Beijing, 100850, China.
| |
Collapse
|