1
|
Souto EP, Gong P, Landua JD, Rajaram Srinivasan R, Ganesan A, Dobrolecki LE, Purdy SC, Pan X, Zeosky M, Chung A, Yi SS, Ford HL, Lewis MT. Lineage Tracing and Single-Cell RNA Sequencing Reveal a Common Transcriptional State in Breast Cancer Tumor-Initiating Cells Characterized by IFN/STAT1 Activity. Cancer Res 2025; 85:1390-1409. [PMID: 40230213 PMCID: PMC11997551 DOI: 10.1158/0008-5472.can-23-4022] [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: 12/19/2023] [Revised: 09/03/2024] [Accepted: 01/31/2025] [Indexed: 04/16/2025]
Abstract
A tumor cell subpopulation of tumor-initiating cells (TIC) or "cancer stem cells" is associated with therapeutic resistance, as well as both local and distant recurrences. Signal transducer and activator of transcription (STAT) activity is elevated in TICs in claudin-low models of human triple-negative breast cancer, which enables enrichment of TICs using a STAT-responsive reporter. Lineage tracing of TICs as they undergo cell state changes could enable a better understanding of the molecular phenotypes of TIC and uncover strategies to selectively target TICs. In this study, we developed a STAT-responsive lineage-tracing system and used it in conjunction with the original reporter to enrich for cells with enhanced mammosphere-forming potential. This approach was able to detect TICs in some, but not all, basal-like triple-negative breast cancer xenograft models, indicating that STAT signaling has both TIC-related and TIC-independent functions. Single-cell RNA sequencing (RNA-seq) of reporter-tagged xenografts and clinical samples identified a common IFN/STAT1-associated transcriptional state in TICs that was previously linked to inflammation and macrophage differentiation. Surprisingly, most of the identified genes were not present in previously published TIC signatures derived using bulk RNA-seq. Finally, bone marrow stromal cell antigen-2 was identified as a cell surface marker of this state that functionally regulated TIC frequency. These results suggest that TICs may exploit the IFN/STAT1 signaling axis to promote their activity and that targeting this pathway may help eliminate TICs. Significance: Coupling single-cell transcriptomics with tumor-initiating cell enrichment identified IFN response gene expression not previously reported in bulk RNA-sequencing-derived signatures and proposed IFN/STAT1 signaling as a candidate therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Eric P. Souto
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Cancer and Cell Biology Graduate Program, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Ping Gong
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - John D. Landua
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | | | - Abhinaya Ganesan
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Cancer and Cell Biology Graduate Program, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Lacey E. Dobrolecki
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Stephen C. Purdy
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (UC-AMC), Aurora, Colorado
- Pharmacology Graduate Program, UC-AMC, Aurora, Colorado
- University of Colorado Cancer Center, UC-AMC, Aurora, Colorado
| | - Xingxin Pan
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, Texas
| | - Michael Zeosky
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, Texas
| | - Anna Chung
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, Texas
| | - S. Stephen Yi
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, Texas
| | - Heide L. Ford
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (UC-AMC), Aurora, Colorado
- Pharmacology Graduate Program, UC-AMC, Aurora, Colorado
- University of Colorado Cancer Center, UC-AMC, Aurora, Colorado
| | - Michael T. Lewis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Cancer and Cell Biology Graduate Program, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Department of Radiology, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
2
|
Shang T, Jia Z, Li J, Cao H, Xu H, Cong L, Ma D, Wang X, Liu J. Unraveling the triad of hypoxia, cancer cell stemness, and drug resistance. J Hematol Oncol 2025; 18:32. [PMID: 40102937 PMCID: PMC11921735 DOI: 10.1186/s13045-025-01684-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 03/05/2025] [Indexed: 03/20/2025] Open
Abstract
In the domain of addressing cancer resistance, challenges such as limited effectiveness and treatment resistance remain persistent. Hypoxia is a key feature of solid tumors and is strongly associated with poor prognosis in cancer patients. Another significant portion of the development of acquired drug resistance is attributed to tumor stemness. Cancer stem cells (CSCs), a small tumor cell subset with self-renewal and proliferative abilities, are crucial for tumor initiation, metastasis, and intra-tumoral heterogeneity. Studies have shown a significant association between hypoxia and CSCs in the context of tumor resistance. Recent studies reveal a strong link between hypoxia and tumor stemness, which together promote tumor survival and progression during treatment. This review elucidates the interplay between hypoxia and CSCs, as well as their correlation with resistance to therapeutic drugs. Targeting pivotal genes associated with hypoxia and stemness holds promise for the development of novel therapeutics to combat tumor resistance.
Collapse
Affiliation(s)
- Tongxuan Shang
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Ziqi Jia
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jiayi Li
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, 100730, China
| | - Heng Cao
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Hengyi Xu
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lin Cong
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Dongxu Ma
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xiang Wang
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jiaqi Liu
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
3
|
Cai J, Liao W, Wen J, Ye F, Nie Q, Chen W, Zhao C. Algae-derived polysaccharides and polysaccharide-based nanoparticles: A natural frontier in breast cancer therapy. Int J Biol Macromol 2025; 297:139936. [PMID: 39824414 DOI: 10.1016/j.ijbiomac.2025.139936] [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: 09/02/2024] [Revised: 01/01/2025] [Accepted: 01/14/2025] [Indexed: 01/20/2025]
Abstract
Breast cancer is the second leading cause of cancer-related mortality among women worldwide, with its progression closely tied to the tumor microenvironment. To address the limitations and adverse effects of conventional therapies, algal polysaccharides and their nanoparticle derivatives have emerged as promising and effective anti-breast cancer agents. These bioactive compounds, derived from algae, are distinguished by their natural origin, non-toxicity, and significant medical relevance. Notably, algal polysaccharide-based nanoparticles exhibit advantageous properties such as hydrophilicity, biodegradability, prolonged circulation, and selective accumulation in tumor tissues. This review explores the relationship between the structural attributes of algal polysaccharides and their therapeutic efficacy. It further highlights the advantages of algal polysaccharide-based nanoparticles as drug delivery systems, particularly their potential in tumor targeting and overcoming multidrug resistance, thereby providing a theoretical foundation for their application in breast cancer treatment.
Collapse
Affiliation(s)
- Jiaer Cai
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Wei Liao
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Jiahui Wen
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Fangting Ye
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Qing Nie
- College of Marine and Biology Engineering, Yancheng Institute of Technology, Yancheng 224000, China
| | - Weichao Chen
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| | - Chao Zhao
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| |
Collapse
|
4
|
Leck LYW, Abd El-Aziz YS, McKelvey KJ, Park KC, Sahni S, Lane DJR, Skoda J, Jansson PJ. Cancer stem cells: Masters of all traits. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167549. [PMID: 39454969 DOI: 10.1016/j.bbadis.2024.167549] [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: 02/05/2024] [Revised: 10/01/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Abstract
Cancer is a heterogeneous disease, which contributes to its rapid progression and therapeutic failure. Besides interpatient tumor heterogeneity, tumors within a single patient can present with a heterogeneous mix of genetically and phenotypically distinct subclones. These unique subclones can significantly impact the traits of cancer. With the plasticity that intratumoral heterogeneity provides, cancers can easily adapt to changes in their microenvironment and therapeutic exposure. Indeed, tumor cells dynamically shift between a more differentiated, rapidly proliferating state with limited tumorigenic potential and a cancer stem cell (CSC)-like state that resembles undifferentiated cellular precursors and is associated with high tumorigenicity. In this context, CSCs are functionally located at the apex of the tumor hierarchy, contributing to the initiation, maintenance, and progression of tumors, as they also represent the subpopulation of tumor cells most resistant to conventional anti-cancer therapies. Although the CSC model is well established, it is constantly evolving and being reshaped by advancing knowledge on the roles of CSCs in different cancer types. Here, we review the current evidence of how CSCs play a pivotal role in providing the many traits of aggressive tumors while simultaneously evading immunosurveillance and anti-cancer therapy in several cancer types. We discuss the key traits and characteristics of CSCs to provide updated insights into CSC biology and highlight its implications for therapeutic development and improved treatment of aggressive cancers.
Collapse
Affiliation(s)
- Lionel Y W Leck
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Yomna S Abd El-Aziz
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Oral Pathology Department, Faculty of Dentistry, Tanta University, Tanta, Egypt
| | - Kelly J McKelvey
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia
| | - Kyung Chan Park
- Proteina Co., Ltd./Seoul National University, Seoul, South Korea
| | - Sumit Sahni
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia
| | - Darius J R Lane
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience & Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Jan Skoda
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic; International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.
| | - Patric J Jansson
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia.
| |
Collapse
|
5
|
Young KS, Hancock GR, Fink EC, Zigrossi A, Flowers B, Cooper DA, Nguyen VT, Martinez MC, Mon KS, Bosland M, Zak DR, Runde AP, Sharifi MN, Kastrati I, Minh DDL, Kregel S, Fanning SW. Targeting unique ligand binding domain structural features downregulates DKK1 in Y537S ESR1 mutant breast cancer cells. Breast Cancer Res 2025; 27:10. [PMID: 39825366 PMCID: PMC11742495 DOI: 10.1186/s13058-024-01945-z] [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: 06/06/2024] [Accepted: 12/08/2024] [Indexed: 01/20/2025] Open
Abstract
Resistance to endocrine therapies remains a major clinical hurdle in breast cancer. Mutations to estrogen receptor alpha (ERα) arise after continued therapeutic pressure. Next generation selective estrogen receptor modulators and degraders/downregulators (SERMs and SERDs) show clinical efficacy, but responses are often non-durable. A tyrosine to serine point mutation at position 537 in the ERα ligand binding domain (LBD) is among the most common and most pathogenic alteration in this setting. It enables endocrine therapy resistance by superceding intrinsic structural-energetic gatekeepers of ER hormone-dependence, it enhances metastatic burden by enabling neomorphic ER-dependent transcriptional programs, and it resists SERM and SERD inhibiton by reducing their binding affinities and abilities to antagonize transcriptional coregulator binding. However, a subset of SERMs and SERDs can achieve efficacy by adopting poses that force the mutation to engage in a new interaction that favors the therapeutic receptor antagonist conformation. We previously described a chemically unconventional SERM, T6I-29, that demonstrates significant anti-proliferative activities in Y537S ERα breast cancer cells. Here, we use a comprehensive suite of structural-biochemical, in vitro, and in vivo approaches to better T6I-29's activities in breast cancer cells harboring Y537S ERα. RNA sequencing in cells treated with T6I-29 reveals a neomorphic downregulation of DKK1, a secreted glycoprotein known to play oncogenic roles in other cancers. Importantly, we find that DKK1 is significantly enriched in ER + breast cancer plasma compared to healthy controls. This study shows how new SERMs and SERDs can identify new therapeutic pathways in endocrine-resistant ER + breast cancers.
Collapse
Affiliation(s)
- K S Young
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL, 50153, USA
| | - G R Hancock
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL, 50153, USA
| | - E C Fink
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL, 50153, USA
| | - A Zigrossi
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL, 50153, USA
| | - B Flowers
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL, 50153, USA
| | - D A Cooper
- Department of Chemistry, Illinois Institute of Technology, Chicago, IL, 60616, USA
| | - V T Nguyen
- Department of Chemistry, Illinois Institute of Technology, Chicago, IL, 60616, USA
| | - M C Martinez
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL, 50153, USA
| | - K S Mon
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL, 50153, USA
| | - M Bosland
- Department of Pathology, University of Illinois Chicago, Chicago, IL, 60607, USA
| | - D R Zak
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL, 50153, USA
| | - A P Runde
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL, 50153, USA
| | - M N Sharifi
- Department of Medicine, University of Wisconsin, Madison, WI, 53705, USA
| | - I Kastrati
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL, 50153, USA
| | - D D L Minh
- Department of Chemistry, Illinois Institute of Technology, Chicago, IL, 60616, USA
| | - S Kregel
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL, 50153, USA
| | - Sean W Fanning
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL, 50153, USA.
| |
Collapse
|
6
|
Amer H, Kampan NC, Itsiopoulos C, Flanagan KL, Scott CL, Kartikasari AER, Plebanski M. Interleukin-6 Modulation in Ovarian Cancer Necessitates a Targeted Strategy: From the Approved to Emerging Therapies. Cancers (Basel) 2024; 16:4187. [PMID: 39766086 PMCID: PMC11674514 DOI: 10.3390/cancers16244187] [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: 10/16/2024] [Revised: 11/27/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025] Open
Abstract
Despite significant advances in treatments, ovarian cancer (OC) remains one of the most prevalent and lethal gynecological cancers in women. The frequent detection at the advanced stages has contributed to low survival rates, resistance to various treatments, and disease recurrence. Thus, a more effective approach is warranted to combat OC. The cytokine Interleukin-6 (IL6) has been implicated in various stages of OC development. High IL6 levels are also correlated with a lower survival rate in OC patients. In this current review, we summarized the pivotal roles of IL6 in OC, including the initiation, development, invasion, metastasis, and drug resistance mechanisms. This article systematically highlights how targeting IL6 improves OC outcomes by altering various cancer processes and reports the ongoing clinical trials that would further shape the IL6-based targeted therapies. This review also suggests how combining IL6-targeted therapies with other therapeutic strategies could further enhance their efficacy to combat OC.
Collapse
Affiliation(s)
- Hina Amer
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia; (H.A.); (A.E.R.K.)
| | - Nirmala C. Kampan
- Department of Obstetrics and Gynecology, Faculty of Medicine, University Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Catherine Itsiopoulos
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia; (H.A.); (A.E.R.K.)
| | - Katie L. Flanagan
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia; (H.A.); (A.E.R.K.)
- School of Medicine and School of Health Sciences, University of Tasmania, Launceston, TAS 7250, Australia
- Tasmanian Vaccine Trial Centre, Clifford Craig Foundation, Launceston General Hospital, Launceston, TAS 7250, Australia
| | - Clare L. Scott
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Faculty of Medicine, Dentistry, and Health Sciences, The University of Melbourne, Parkville, VIC 3052, Australia
- The Royal Women’s Hospital, Parkville, VIC 3052, Australia
| | - Apriliana E. R. Kartikasari
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia; (H.A.); (A.E.R.K.)
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia; (H.A.); (A.E.R.K.)
| |
Collapse
|
7
|
Panda SK, Robinson N, Desiderio V. Decoding secret role of mesenchymal stem cells in regulating cancer stem cells and drug resistance. Biochim Biophys Acta Rev Cancer 2024; 1879:189205. [PMID: 39481663 DOI: 10.1016/j.bbcan.2024.189205] [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] [Revised: 09/23/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024]
Abstract
Drug resistance caused by the efflux of chemotherapeutic drugs is one of the most challenging obstacles to successful cancer therapy. Several efflux transporters have been identified since the discovery of the P-gp/ABCB1 transporter in 1976. Over the last four decades, researchers have focused on developing efflux transporter inhibitors to overcome drug resistance. However, even with the third-generation inhibitors available, we are still far from effectively inhibiting the efflux transporters. Additionally, Cancer stem cells (CSCs) pose another significant challenge, contributing to cancer recurrence even after successful treatment. The ability of CSCs to enter dormancy and evade detection makes them almost invulnerable to chemotherapeutic drug treatment. In this review, we discuss how Mesenchymal stem cells (MSCs), one of the key components of the Tumor Microenvironment (TME), regulate both the CSCs and efflux transporters. We propose a new approach focusing on MSCs, which can be crucial to successfully address CSCs and efflux transporters.
Collapse
Affiliation(s)
- Sameer Kumar Panda
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples 80138, Italy; Center for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5001, Australia
| | - Nirmal Robinson
- Center for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5001, Australia
| | - Vincenzo Desiderio
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples 80138, Italy.
| |
Collapse
|
8
|
He Y, Song T, Ning J, Wang Z, Yin Z, Jiang P, Yuan Q, Yu W, Cheng F. Lactylation in cancer: Mechanisms in tumour biology and therapeutic potentials. Clin Transl Med 2024; 14:e70070. [PMID: 39456119 PMCID: PMC11511673 DOI: 10.1002/ctm2.70070] [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: 08/06/2024] [Revised: 10/03/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Lactylation, a recently identified form of protein post-translational modification (PTM), has emerged as a key player in cancer biology. The Warburg effect, a hallmark of tumour metabolism, underscores the significance of lactylation in cancer progression. By regulating gene transcription and protein function, lactylation facilitates metabolic reprogramming, enabling tumours to adapt to nutrient limitations and sustain rapid growth. Over the past decade, extensive research has revealed the intricate regulatory network underlying lactylation in tumours. Large-scale sequencing and machine learning have confirmed the widespread occurrence of lactylation sites across the tumour proteome. Targeting lactylation enzymes or metabolic pathways has demonstrated promising anti-tumour effects, highlighting the therapeutic potential of this modification. This review comprehensively explores the mechanisms of lactylation in cancer cells and the tumour microenvironment. We expound on the application of advanced omics technologies for target identification and data modelling within the lactylation field. Additionally, we summarise existing anti-lactylation drugs and discuss their clinical implications. By providing a comprehensive overview of recent advancements, this review aims to stimulate innovative research and accelerate the translation of lactylation-based therapies into clinical practice. KEY POINTS: Lactylation significantly influences tumour metabolism and gene regulation, contributing to cancer progression. Advanced sequencing and machine learning reveal widespread lactylation sites in tumours. Targeting lactylation enzymes shows promise in enhancing anti-tumour drug efficacy and overcoming chemotherapy resistance. This review outlines the clinical implications and future research directions of lactylation in oncology.
Collapse
Affiliation(s)
- Yipeng He
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Tianbao Song
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Jinzhuo Ning
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Zefeng Wang
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Zhen Yin
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Pengcheng Jiang
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Qin Yuan
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Weimin Yu
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Fan Cheng
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| |
Collapse
|
9
|
Kong W, Gao Y, Zhao S, Yang H. Cancer stem cells: advances in the glucose, lipid and amino acid metabolism. Mol Cell Biochem 2024; 479:2545-2563. [PMID: 37882986 DOI: 10.1007/s11010-023-04861-6] [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: 05/20/2023] [Accepted: 09/13/2023] [Indexed: 10/27/2023]
Abstract
Cancer stem cells (CSCs) are a class of cells with self-renewal and multi-directional differentiation potential, which are present in most tumors, particularly in aggressive tumors, and perform a pivotal role in recurrence and metastasis and are expected to be one of the important targets for tumor therapy. Studies of tumor metabolism in recent years have found that the metabolic characteristics of CSCs are distinct from those of differentiated tumor cells, which are unique to CSCs and contribute to the maintenance of the stemness characteristics of CSCs. Moreover, these altered metabolic profiles can drive the transformation between CSCs and non-CSCs, implying that these metabolic alterations are important markers for CSCs to play their biological roles. The identification of metabolic changes in CSCs and their metabolic plasticity mechanisms may provide some new opportunities for tumor therapy. In this paper, we review the metabolism-related mechanisms of CSCs in order to provide a theoretical basis for their potential application in tumor therapy.
Collapse
Affiliation(s)
- Weina Kong
- Department of Obstetrics and Gynecology, Xijing Hospital, Air Forth Military Medical University, 127 Changle West Road, Xincheng District, Xi'an City, Shaanxi Province, China
| | - Yunge Gao
- Department of Obstetrics and Gynecology, Xijing Hospital, Air Forth Military Medical University, 127 Changle West Road, Xincheng District, Xi'an City, Shaanxi Province, China
| | - Shuhua Zhao
- Department of Obstetrics and Gynecology, Xijing Hospital, Air Forth Military Medical University, 127 Changle West Road, Xincheng District, Xi'an City, Shaanxi Province, China
| | - Hong Yang
- Department of Obstetrics and Gynecology, Xijing Hospital, Air Forth Military Medical University, 127 Changle West Road, Xincheng District, Xi'an City, Shaanxi Province, China.
| |
Collapse
|
10
|
Melwani PK, Balla MMS, Bhamani A, Nandha SR, Checker R, Pandey BN. Macrophage-conditioned medium enhances tunneling nanotube formation in breast cancer cells via PKC, Src, NF-κB, and p38 MAPK signaling. Cell Signal 2024; 121:111274. [PMID: 38936787 DOI: 10.1016/j.cellsig.2024.111274] [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: 10/30/2023] [Revised: 06/13/2024] [Accepted: 06/24/2024] [Indexed: 06/29/2024]
Abstract
Tumor-associated macrophages (TAMs) secrete cytokines, chemokines, and growth factors in the tumor microenvironment (TME) to support cancer progression. Higher TAM infiltration in the breast TME is associated with a poor prognosis. Previous studies have demonstrated the role of macrophages in stimulating long-range intercellular bridges referred to as tunneling nanotubes (TNTs) in cancer cells. Intercellular communication between cancer cells via TNTs promotes cancer growth, invasion, metastasis, and therapy resistance. Given the important role of TNTs and macrophages in cancer, the role of macrophage-induced TNTs in chemotherapy drug doxorubicin resistance is not known. Furthermore, the mechanism of macrophage-mediated TNT formation is elusive. In this study, it is shown that the macrophage-conditioned medium (MΦCM) partially mimicked inflammatory TME, induced an EMT phenotype, and increased migration in MCF-7 breast cancer cells. Additionally, secreted proteins in MΦCM induced TNT formation in MCF-7 cells, which led to increased resistance to doxorubicin. Transcriptomic analysis of MΦCM-treated MCF-7 cells showed enrichment of the NF-κB and focal adhesion pathways, as well as upregulation of genes involved in EMT, extracellular remodeling, and actin cytoskeleton reorganization. Interestingly, inhibitors of PKC, Src, NF-κB, and p38 decreased macrophage-induced TNT formation in MCF-7 cells. These results reveal the novel role of PKC and Src in inducing TNT formation in cancer cells and suggest that inhibition of PKC and Src activity may likely contribute to reduced macrophage-breast cancer cell interaction and the potential therapeutic strategy of cancer.
Collapse
Affiliation(s)
- Pooja Kamal Melwani
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Mumbai 400 085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400 094, India.
| | - Murali Mohan Sagar Balla
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Mumbai 400 085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400 094, India
| | - Aman Bhamani
- K. J. Somaiya College of Science and Commerce, Vidyavihar, Mumbai 400077, India
| | - Shivani R Nandha
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Mumbai 400 085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400 094, India
| | - Rahul Checker
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Mumbai 400 085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400 094, India
| | - Badri Narain Pandey
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Mumbai 400 085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400 094, India.
| |
Collapse
|
11
|
Souto EP, Gong P, Landua JD, Srinivasan RR, Ganesan A, Dobrolecki LE, Purdy SC, Pan X, Zeosky M, Chung A, Yi SS, Ford HL, Lewis MT. The interferon/STAT1 signaling axis is a common feature of tumor-initiating cells in breast cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.15.557958. [PMID: 37745510 PMCID: PMC10515955 DOI: 10.1101/2023.09.15.557958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
A tumor cell subpopulation of tumor-initiating cells (TIC), or "cancer stem cells", are associated with therapeutic resistance, as well as both local and distant recurrence. Enriched populations of TIC are identified by markers including aldehyde dehydrogenase (ALDH1) activity, the cell surface marker combination CD44 + /CD24 - , or fluorescent reporters for signaling pathways that regulate TIC function. We showed previously that S ignal T ransducer and A ctivator of T ranscription (STAT)-mediated transcription allows enrichment for TIC in claudin-low models of human triple-negative breast cancer using a STAT-responsive reporter. However, the molecular phenotypes of STAT TIC are not well understood, and there is no existing method to lineage-trace TIC as they undergo cell state changes. Using a new STAT-responsive lineage-tracing (LT) system in conjunction with our original reporter, we enriched for cells with enhanced mammosphere-forming potential in some, but not all, basal-like triple-negative breast cancer (TNBC) xenograft models (TNBC) indicating TIC-related and TIC-independent functions for STAT signaling. Single-cell RNA sequencing (scRNAseq) of reporter-tagged xenografts and clinical samples identified a common interferon (IFN)/STAT1-associated transcriptional state, previously linked to inflammation and macrophage differentiation, in TIC. Surprisingly, most of the genes we identified are not present in previously published TIC signatures derived using bulk RNA sequencing. Finally, we demonstrated that bone marrow stromal cell antigen 2 (BST2), is a cell surface marker of this state, and that it functionally regulates TIC frequency. These results suggest TIC may exploit the IFN/STAT1 signaling axis to promote their activity, and that targeting this pathway may help eliminate TIC. Significance TIC differentially express interferon response genes, which were not previously reported in bulk RNA sequencing-derived TIC signatures, highlighting the importance of coupling single-cell transcriptomics with enrichment to derive TIC signatures.
Collapse
|
12
|
Young K, Hancock G, Fink E, Zigrossi A, Flowers B, Cooper D, Nguyen V, Martinez M, Mon K, Bosland M, Zak D, Runde A, Sharifi M, Kastrati I, Minh D, Kregel S, Fanning S. Targeting Unique Ligand Binding Domain Structural Features Downregulates DKK1 in Y537S ESR1 Mutant Breast Cancer Cells. RESEARCH SQUARE 2024:rs.3.rs-4542467. [PMID: 38978585 PMCID: PMC11230492 DOI: 10.21203/rs.3.rs-4542467/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Resistance to endocrine therapies remains a major clinical hurdle in breast cancer. Mutations to estrogen receptor alpha (ERα) arise after continued therapeutic pressure. Next generation selective estrogen receptor modulators and degraders/downregulators (SERMs and SERDs) show clinical efficacy, but responses are often non-durable. A tyrosine to serine point mutation at position 537 in the ERα ligand binding domain (LBD) is among the most common and most pathogenic alteration in this setting. It enables endocrine therapy resistance by superceding intrinsic structural-energetic gatekeepers of ER hormone-dependence, it enhances metastatic burden by enabling neomorphic ER-dependent transcriptional programs, and it resists SERM and SERD inhibiton by reducing their binding affinities and abilities to antagonize transcriptional coregulator binding. However, a subset of SERMs and SERDs can achieve efficacy by adopting poses that force the mutation to engage in a new interaction that favors the therapeutic receptor antagonist conformation. We previously described a chemically unconventional SERM, T6I-29, that demonstrates significant anti-proliferative activities in Y537S ERα breast cancer cells. Here, we use a comprehensive suite of structural-biochemical, in vitro, and in vivo approaches to better T6I-29's activities in breast cancer cells harboring Y537S ERα. RNA sequencing in cells treated with T6I-29 reveals a neomorphic downregulation of DKK1, a secreted glycoprotein known to play oncogenic roles in other cancers. Importantly, we find that DKK1 is significantly enriched in ER+ breast cancer plasma compared to healthy controls. This study shows how new SERMs and SERDs can identify new therapeutic pathways in endocrine-resistant ER+ breast cancers.
Collapse
Affiliation(s)
- K.S. Young
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 50153
| | - G.R. Hancock
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 50153
| | - E. Fink
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 50153
| | - A. Zigrossi
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 50153
| | - B. Flowers
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 50153
| | - D.A. Cooper
- Department of Chemistry, Illinois Institute of Technology, Chicago, IL 60616
| | - V.T. Nguyen
- Department of Chemistry, Illinois Institute of Technology, Chicago, IL 60616
| | - M. Martinez
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 50153
| | - K.S. Mon
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 50153
| | - M. Bosland
- Department of Pathology, University of Illinois Chicago, Chicago, IL 60607
| | - D. Zak
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 50153
| | - A. Runde
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 50153
| | - M.N. Sharifi
- Department of Medicine, University of Wisconsin, Madison, WI 53705
| | - I. Kastrati
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 50153
| | - D.D.L. Minh
- Department of Chemistry, Illinois Institute of Technology, Chicago, IL 60616
| | - S. Kregel
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 50153
| | - S.W. Fanning
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 50153
| |
Collapse
|
13
|
Young K, Hancock G, Fink E, Zigrossi A, Flowers B, Cooper D, Nguyen V, Martinez M, Mon K, Bosland M, Zak D, Runde A, Sharifi M, Kastrati I, Minh D, Kregel S, Fanning S. Targeting Unique Ligand Binding Domain Structural Features Downregulates DKK1 in Y537S ESR1 Mutant Breast Cancer Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.28.596307. [PMID: 38854123 PMCID: PMC11160638 DOI: 10.1101/2024.05.28.596307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Resistance to endocrine therapies remains a major clinical hurdle in breast cancer. Mutations to estrogen receptor alpha (ERα) arise after continued therapeutic pressure. Next generation selective estrogen receptor modulators and degraders/downregulators (SERMs and SERDs) show clinical efficacy, but responses are often non-durable. A tyrosine to serine point mutation at position 537 in the ERα ligand binding domain (LBD) is among the most common and most pathogenic alteration in this setting. It enables endocrine therapy resistance by superceding intrinsic structural-energetic gatekeepers of ER hormone-dependence, it enhances metastatic burden by enabling neomorphic ER-dependent transcriptional programs, and it resists SERM and SERD inhibiton by reducing their binding affinities and abilities to antagonize transcriptional coregulator binding. However, a subset of SERMs and SERDs can achieve efficacy by adopting poses that force the mutation to engage in a new interaction that favors the therapeutic receptor antagonist conformation. We previously described a chemically unconventional SERM, T6I-29, that demonstrates significant anti-proliferative activities in Y537S ERα breast cancer cells. Here, we use a comprehensive suite of structural-biochemical, in vitro, and in vivo approaches to better T6I-29's activities in breast cancer cells harboring Y537S ERα. RNA sequencing in cells treated with T6I-29 reveals a neomorphic downregulation of DKK1, a secreted glycoprotein known to play oncogenic roles in other cancers. Importantly, we find that DKK1 is significantly enriched in ER+ breast cancer plasma compared to healthy controls. This study shows how new SERMs and SERDs can identify new therapeutic pathways in endocrine-resistant ER+ breast cancers.
Collapse
Affiliation(s)
- K.S. Young
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 50153
| | - G.R. Hancock
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 50153
| | - E. Fink
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 50153
| | - A. Zigrossi
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 50153
| | - B. Flowers
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 50153
| | - D.A. Cooper
- Department of Chemistry, Illinois Institute of Technology, Chicago, IL 60616
| | - V.T. Nguyen
- Department of Chemistry, Illinois Institute of Technology, Chicago, IL 60616
| | - M. Martinez
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 50153
| | - K.S. Mon
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 50153
| | - M. Bosland
- Department of Pathology, University of Illinois Chicago, Chicago, IL 60607
| | - D. Zak
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 50153
| | - A. Runde
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 50153
| | - M.N. Sharifi
- Department of Medicine, University of Wisconsin, Madison, WI 53705
| | - I. Kastrati
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 50153
| | - D.D.L. Minh
- Department of Chemistry, Illinois Institute of Technology, Chicago, IL 60616
| | - S. Kregel
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 50153
| | - S.W. Fanning
- Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 50153
| |
Collapse
|
14
|
Wang J, Zhang N, Ding X, Fu C, Li X, Li B, Ding J, Sun T. Targeted nanostrategies eliminate pre-metastatic niche of cancer. NANO RESEARCH 2024; 17:5358-5373. [DOI: 10.1007/s12274-024-6412-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/08/2023] [Accepted: 12/11/2023] [Indexed: 05/14/2025]
|
15
|
Maroni G, Krishnan I, Alfieri R, Maymi VA, Pandell N, Csizmadia E, Zhang J, Weetall M, Branstrom A, Braccini G, Cabrera San Millán E, Storti B, Bizzarri R, Kocher O, Bassères DS, Welner RS, Magli MC, Merelli I, Clohessy JG, Ali A, Tenen DG, Levantini E. Tumor Microenvironment Landscapes Supporting EGFR-mutant NSCLC Are Modulated at the Single-cell Interaction Level by Unesbulin Treatment. CANCER RESEARCH COMMUNICATIONS 2024; 4:919-937. [PMID: 38546390 PMCID: PMC10964845 DOI: 10.1158/2767-9764.crc-23-0161] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 01/26/2024] [Accepted: 03/12/2024] [Indexed: 11/03/2024]
Abstract
Lung cancer is the leading cause of cancer deaths. Lethal pulmonary adenocarcinomas (ADC) present with frequent mutations in the EGFR. Genetically engineered murine models of lung cancer expedited comprehension of the molecular mechanisms driving tumorigenesis and drug response. Here, we systematically analyzed the evolution of tumor heterogeneity in the context of dynamic interactions occurring with the intermingled tumor microenvironment (TME) by high-resolution transcriptomics. Our effort identified vulnerable tumor-specific epithelial cells, as well as their cross-talk with niche components (endothelial cells, fibroblasts, and tumor-infiltrating immune cells), whose symbiotic interface shapes tumor aggressiveness and is almost completely abolished by treatment with Unesbulin, a tubulin binding agent that reduces B cell-specific Moloney murine leukemia virus integration site 1 (BMI-1) activity. Simultaneous magnetic resonance imaging (MRI) analysis demonstrated decreased tumor growth, setting the stage for future investigations into the potential of novel therapeutic strategies for EGFR-mutant ADCs. SIGNIFICANCE Targeting the TME is an attractive strategy for treatment of solid tumors. Here we revealed how EGFR-mutant landscapes are affected at the single-cell resolution level during Unesbulin treatment. This novel drug, by targeting cancer cells and their interactions with crucial TME components, could be envisioned for future therapeutic advancements.
Collapse
Affiliation(s)
- Giorgia Maroni
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- Harvard Medical School, Boston, Massachusetts
- Institute of Biomedical Technologies, National Research Council (CNR), Pisa, Italy
| | | | - Roberta Alfieri
- Institute of Biomedical Technologies, National Research Council (CNR), Pisa, Italy
| | - Valerie A. Maymi
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Preclinical Murine Pharmacogenetics Core, Beth Israel Deaconess Cancer Center, Dana-Farber/Harvard Cancer Center, Boston, Massachusetts
| | - Nicole Pandell
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Preclinical Murine Pharmacogenetics Core, Beth Israel Deaconess Cancer Center, Dana-Farber/Harvard Cancer Center, Boston, Massachusetts
| | - Eva Csizmadia
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Junyan Zhang
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | | | | | - Giulia Braccini
- Institute of Biomedical Technologies, National Research Council (CNR), Pisa, Italy
| | | | - Barbara Storti
- NEST, Scuola Normale Superiore and Istituto Nanoscienze-CNR, Pisa, Italy
| | - Ranieri Bizzarri
- NEST, Scuola Normale Superiore and Istituto Nanoscienze-CNR, Pisa, Italy
- Department of Surgical, Medical and Molecular Pathology, and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Olivier Kocher
- Harvard Medical School, Boston, Massachusetts
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Daniela S. Bassères
- Biochemistry Department, Chemistry Institute, University of Sao Paulo, Sao Paulo, Brazil
| | - Robert S. Welner
- Department of Medicine, Hemathology/Oncology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Maria Cristina Magli
- Institute of Biomedical Technologies, National Research Council (CNR), Pisa, Italy
| | - Ivan Merelli
- Institute of Biomedical Technologies, National Research Council (CNR), Pisa, Italy
| | - John G. Clohessy
- Harvard Medical School, Boston, Massachusetts
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Preclinical Murine Pharmacogenetics Core, Beth Israel Deaconess Cancer Center, Dana-Farber/Harvard Cancer Center, Boston, Massachusetts
| | - Azhar Ali
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Daniel G. Tenen
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- Harvard Medical School, Boston, Massachusetts
- Harvard Stem Cell Institute, Cambridge, Massachusetts
| | - Elena Levantini
- Harvard Medical School, Boston, Massachusetts
- Institute of Biomedical Technologies, National Research Council (CNR), Pisa, Italy
- Harvard Stem Cell Institute, Cambridge, Massachusetts
| |
Collapse
|
16
|
Yu T, Rui L, Jiumei Z, Ziwei L, Ying H. Advances in the study of autophagy in breast cancer. Breast Cancer 2024; 31:195-204. [PMID: 38315272 PMCID: PMC10901946 DOI: 10.1007/s12282-023-01541-7] [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: 11/13/2023] [Accepted: 12/25/2023] [Indexed: 02/07/2024]
Abstract
Breast cancer is the most prevalent malignant tumor among women, with a high incidence and mortality rate all year round, which seriously affects women's health. Autophagy, a well-conserved cellular process inherent in eukaryotic organisms, plays a pivotal role in degrading damaged proteins and organelles, recycling their breakdown products to aid cells in navigating stress and gradually restoring homeostatic equilibrium. Recent studies have unveiled the intricate connection between autophagy and breast cancer. Autophagy is a double-edged sword in breast cancer, demonstrating a dual role: restraining its onset and progression on one hand, while promoting its metastasis and advancement on the other. It is also because of this interrelationship between the two that regulation of autophagy in the treatment of breast cancer is now an important strategy in clinical treatment. In this article, we systematically survey the recent research findings, elucidating the multifaceted role of autophagy in breast cancer and its underlying mechanisms, with the aim of contributing new references to the clinical management of breast cancer.
Collapse
Affiliation(s)
- Tang Yu
- The Third Affiliated Hospital of Kunming Medical University, Kunming, China
- The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liu Rui
- The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhao Jiumei
- Chongqing Nanchuan District People's Hospital, Chongqing, China
| | - Li Ziwei
- Chongqing Health Center for Women and Children, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Hu Ying
- The Second Affiliatied Hospital of Kunming Medical University and Department of Clinical Larboratory, Kunming, China.
| |
Collapse
|
17
|
Bimonte VM, Catanzaro G, Po A, Trocchianesi S, Besharat ZM, Spinello Z, Curreli M, Fabi A, Bei R, Milella M, Vacca A, Ferretti E, Migliaccio S. The endocrine disruptor cadmium modulates the androgen-estrogen receptors ratio and induces inflammatory cytokines in luminal (A) cell models of breast cancer. Endocrine 2024; 83:798-809. [PMID: 37979099 PMCID: PMC10902028 DOI: 10.1007/s12020-023-03594-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 10/30/2023] [Indexed: 11/19/2023]
Abstract
PURPOSE Breast cancer (BC) is the most common malignancy that affects women, and it is, to date, their leading cause of death. Luminal A molecular subtype accounts for 40% of BC and is characterized by hormone receptors positive/human epidermal growth factor 2 expression and current treatment consists of surgery plus aromatase inhibitor therapy. Interestingly, several studies demonstrated that the heavy metal cadmium (Cd), classified as a group 1 human carcinogen and widely spread in the environment, exerts estrogen-like activities in several tissues and suggested an intriguing relationship between increased Cd exposure and BC incidence. Thus, aim of this study was to evaluate effects of Cd on Luminal A BC estrogen receptor (ER) positive/progesterone receptor positive cell models in vitro to characterize the mechanism(s) involved in breast cell homeostasis disruption. METHODS T47D and MCF7 were exposed to Cd (0.5-1 µM) for 6-24 h to evaluate potential alterations in: cells viability, steroid receptors and intracellular signaling by western blot. Moreover, we evaluated the expression of inflammatory cytokines interleukin by RT-PCR. RESULTS Our results showed a significant induction of androgen receptor (AR) and an increased AR/ER ratio. Further, Cd exposure increased pro-inflammatory cytokines interleukin (IL)6, IL8 and tumor necrosis factor α levels. Finally, as previously demonstrated by our group, Cd alters pathways such as mitogen-activated protein kinase family and protein kinase B. CONCLUSION In conclusion, our study demonstrates that Cd modifies the expression and pattern of ERs and AR in BC cell lines, suggesting an alteration of BC cells homeostasis, likely predisposing to a carcinogenetic microenvironment.
Collapse
Affiliation(s)
- Viviana M Bimonte
- Department of Movement, Human and Health Sciences, University of Foro Italico, 00195, Rome, Italy
| | - Giuseppina Catanzaro
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161, Rome, Italy
| | - Agnese Po
- Department of Molecular Medicine, "Sapienza" University of Rome, 00161, Rome, Italy
| | - Sofia Trocchianesi
- Department of Molecular Medicine, "Sapienza" University of Rome, 00161, Rome, Italy
| | - Zein Mersini Besharat
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161, Rome, Italy
| | - Zaira Spinello
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161, Rome, Italy
| | - Mariaignazia Curreli
- Department of Movement, Human and Health Sciences, University of Foro Italico, 00195, Rome, Italy
| | - Alessandra Fabi
- Precision Medicine in Senology Unit, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168, Rome, Italy
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - Michele Milella
- Department of Oncology, University of Verona, 37134, Verona, Italy
| | - Alessandra Vacca
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161, Rome, Italy
| | - Elisabetta Ferretti
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161, Rome, Italy
| | - Silvia Migliaccio
- Department of Movement, Human and Health Sciences, University of Foro Italico, 00195, Rome, Italy.
| |
Collapse
|
18
|
McWhorter R, Bonavida B. The Role of TAMs in the Regulation of Tumor Cell Resistance to Chemotherapy. Crit Rev Oncog 2024; 29:97-125. [PMID: 38989740 DOI: 10.1615/critrevoncog.2024053667] [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: 07/12/2024]
Abstract
Tumor-associated macrophages (TAMs) are the predominant cell infiltrate in the immunosuppressive tumor microenvironment (TME). TAMs are central to fostering pro-inflammatory conditions, tumor growth, metastasis, and inhibiting therapy responses. Many cancer patients are innately refractory to chemotherapy and or develop resistance following initial treatments. There is a clinical correlation between the level of TAMs in the TME and chemoresistance. Hence, the pivotal role of TAMs in contributing to chemoresistance has garnered significant attention toward targeting TAMs to reverse this resistance. A prerequisite for such an approach requires a thorough understanding of the various underlying mechanisms by which TAMs inhibit response to chemotherapeutic drugs. Such mechanisms include enhancing drug efflux, regulating drug metabolism and detoxification, supporting cancer stem cell (CSCs) resistance, promoting epithelial-mesenchymal transition (EMT), inhibiting drug penetration and its metabolism, stimulating angiogenesis, impacting inhibitory STAT3/NF-κB survival pathways, and releasing specific inhibitory cytokines including TGF-β and IL-10. Accordingly, several strategies have been developed to overcome TAM-modulated chemoresistance. These include novel therapies that aim to deplete TAMs, repolarize them toward the anti-tumor M1-like phenotype, or block recruitment of monocytes into the TME. Current results from TAM-targeted treatments have been unimpressive; however, the use of TAM-targeted therapies in combination appears promising These include targeting TAMs with radiotherapy, chemotherapy, chemokine receptor inhibitors, immunotherapy, and loaded nanoparticles. The clinical limitations of these strategies are discussed.
Collapse
Affiliation(s)
| | - Benjamin Bonavida
- Department of Microbiology, Immunology, & Molecular Genetics, David Geffen School of Medicine at UCLA, Johnson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90025-1747, USA
| |
Collapse
|
19
|
Jan N, Sofi S, Qayoom H, Haq BU, Shabir A, Mir MA. Targeting breast cancer stem cells through retinoids: A new hope for treatment. Crit Rev Oncol Hematol 2023; 192:104156. [PMID: 37827439 DOI: 10.1016/j.critrevonc.2023.104156] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 09/09/2023] [Accepted: 10/06/2023] [Indexed: 10/14/2023] Open
Abstract
Breast cancer is a complex and diverse disease accounting for nearly 30% of all cancers diagnosed in females. But unfortunately, patients develop resistance to the existing chemotherapeutic regimen, resulting in approximately 90% treatment failure. With over half a million deaths annually, it is imperative to explore new therapeutic approaches to combat the disease. Within a breast tumor, a small sub-population of heterogeneous cells, with a unique ability of self-renew and differentiation and responsible for tumor formation, initiation, and recurrence are referred to as breast cancer stem cells (BCSCs). These BCSCs have been identified as one of the main contributors to chemoresistance in breast cancer, making them an attractive target for developing novel therapeutic strategies. These cells exhibit surface biomarkers such as CD44+, CD24-/LOW, ALDH, CD133, and CD49f phenotypes. Higher expression of CD44+ and ALDH activity has been associated with the formation of tumors in various cancers. Moreover, the abnormal regulation of signaling pathways, including Hedgehog, Notch, β-catenin, JAK/STAT, and P13K/AKT/mTOR, leads to the formation of cancer stem cells, resulting in the development of tumors. The growing drug resistance in BC is a significant challenge, highlighting the need for new therapeutic strategies to combat this dreadful disease. Retinoids, a large group of synthetic derivatives of vitamin A, have been studied as chemopreventive agents in clinical trials and have been shown to regulate various crucial biological functions including vision, development, inflammation, and metabolism. On a cellular level, the retinoid activity has been well characterized and translated and is known to induce differentiation and apoptosis, which play important roles in the outcome of the transformation of tissues into malignant. Retinoids have been investigated extensively for their use in the treatment and prevention of cancer due to their high receptor-binding affinity to directly modulate gene expression programs. Therefore, in this study, we aim to summarize the current understanding of BCSCs, their biomarkers, and the associated signaling pathways. Retinoids, such as Adapalene, a third-generation retinoid, have shown promising anti-cancer potential and may serve as therapeutic agents to target BCSCs.
Collapse
Affiliation(s)
- Nusrat Jan
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, India
| | - Shazia Sofi
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, India
| | - Hina Qayoom
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, India
| | - Burhan Ul Haq
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, India
| | - Aisha Shabir
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, India
| | - Manzoor Ahmad Mir
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, India.
| |
Collapse
|
20
|
Zaarour RF, Ribeiro M, Azzarone B, Kapoor S, Chouaib S. Tumor microenvironment-induced tumor cell plasticity: relationship with hypoxic stress and impact on tumor resistance. Front Oncol 2023; 13:1222575. [PMID: 37886168 PMCID: PMC10598765 DOI: 10.3389/fonc.2023.1222575] [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: 05/14/2023] [Accepted: 09/27/2023] [Indexed: 10/28/2023] Open
Abstract
The role of tumor interaction with stromal components during carcinogenesis is crucial for the design of efficient cancer treatment approaches. It is widely admitted that tumor hypoxic stress is associated with tumor aggressiveness and thus impacts susceptibility and resistance to different types of treatments. Notable biological processes that hypoxia functions in include its regulation of tumor heterogeneity and plasticity. While hypoxia has been reported as a major player in tumor survival and dissemination regulation, the significance of hypoxia inducible factors in cancer stem cell development remains poorly understood. Several reports indicate that the emergence of cancer stem cells in addition to their phenotype and function within a hypoxic tumor microenvironment impacts cancer progression. In this respect, evidence showed that cancer stem cells are key elements of intratumoral heterogeneity and more importantly are responsible for tumor relapse and escape to treatments. This paper briefly reviews our current knowledge of the interaction between tumor hypoxic stress and its role in stemness acquisition and maintenance. Our review extensively covers the influence of hypoxia on the formation and maintenance of cancer stem cells and discusses the potential of targeting hypoxia-induced alterations in the expression and function of the so far known stem cell markers in cancer therapy approaches. We believe that a better and integrated understanding of the effect of hypoxia on stemness during carcinogenesis might lead to new strategies for exploiting hypoxia-associated pathways and their targeting in the clinical setting in order to overcome resistance mechanisms. More importantly, at the present time, efforts are oriented towards the design of innovative therapeutical approaches that specifically target cancer stem cells.
Collapse
Affiliation(s)
- RF. Zaarour
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - M. Ribeiro
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - B. Azzarone
- Tumor Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - S. Kapoor
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - S. Chouaib
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Faculty of Medicine, University Paris-Saclay, Villejuif, France
| |
Collapse
|
21
|
Guyot B, Clément F, Drouet Y, Schmidt X, Lefort S, Delay E, Treilleux I, Foy JP, Jeanpierre S, Thomas E, Kielbassa J, Tonon L, Zhu HH, Saintigny P, Gao WQ, de la Fouchardiere A, Tirode F, Viari A, Blay JY, Maguer-Satta V. An Early Neoplasia Index (ENI10), Based on Molecular Identity of CD10 Cells and Associated Stemness Biomarkers, is a Predictor of Patient Outcome in Many Cancers. CANCER RESEARCH COMMUNICATIONS 2023; 3:1966-1980. [PMID: 37707389 PMCID: PMC10540743 DOI: 10.1158/2767-9764.crc-23-0196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/01/2023] [Accepted: 09/08/2023] [Indexed: 09/15/2023]
Abstract
An accurate estimate of patient survival at diagnosis is critical to plan efficient therapeutic options. A simple and multiapplication tool is needed to move forward the precision medicine era. Taking advantage of the broad and high CD10 expression in stem and cancers cells, we evaluated the molecular identity of aggressive cancer cells. We used epithelial primary cells and developed a breast cancer stem cell–based progressive model. The superiority of the early-transformed isolated molecular index was evaluated by large-scale analysis in solid cancers. BMP2-driven cell transformation increases CD10 expression which preserves stemness properties. Our model identified a unique set of 159 genes enriched in G2–M cell-cycle phases and spindle assembly complex. Using samples predisposed to transformation, we confirmed the value of an early neoplasia index associated to CD10 (ENI10) to discriminate premalignant status of a human tissue. Using a stratified Cox model, a large-scale analysis (>10,000 samples, The Cancer Genome Atlas Pan-Cancer) validated a strong risk gradient (HRs reaching HR = 5.15; 95% confidence interval: 4.00–6.64) for high ENI10 levels. Through different databases, Cox regression model analyses highlighted an association between ENI10 and poor progression-free intervals for more than 50% of cancer subtypes tested, and the potential of ENI10 to predict drug efficacy. The ENI10 index constitutes a robust tool to detect pretransformed tissues and identify high-risk patients at diagnosis. Owing to its biological link with refractory cancer stem cells, the ENI10 index constitutes a unique way of identifying effective treatments to improve clinical care. SIGNIFICANCE We identified a molecular signature called ENI10 which, owing to its biological link with stem cell properties, predicts patient outcome and drugs efficiency in breast and several other cancers. ENI10 should allow early and optimized clinical management of a broad number of cancers, regardless of the stage of tumor progression.
Collapse
Affiliation(s)
- Boris Guyot
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Department of Cancer Initiation and Tumor cell Identity, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Universite Claude Bernard Lyon 1, CRCL, Lyon, France
| | - Flora Clément
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Department of Cancer Initiation and Tumor cell Identity, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Universite Claude Bernard Lyon 1, CRCL, Lyon, France
| | | | - Xenia Schmidt
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Department of Cancer Initiation and Tumor cell Identity, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Universite Claude Bernard Lyon 1, CRCL, Lyon, France
| | - Sylvain Lefort
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Department of Cancer Initiation and Tumor cell Identity, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Universite Claude Bernard Lyon 1, CRCL, Lyon, France
| | - Emmanuel Delay
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Department of Cancer Initiation and Tumor cell Identity, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Universite Claude Bernard Lyon 1, CRCL, Lyon, France
- Centre Léon Bérard, Lyon, France
| | | | - Jean-Philippe Foy
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Department of Cancer Initiation and Tumor cell Identity, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Department of Tumor Escape Resistance and Immunity, CRCL, Lyon, France
| | - Sandrine Jeanpierre
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Department of Cancer Initiation and Tumor cell Identity, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Universite Claude Bernard Lyon 1, CRCL, Lyon, France
- Centre Léon Bérard, Lyon, France
| | - Emilie Thomas
- Bioinformatics Platform, Synergie Lyon Cancer Foundation, Lyon, France
| | - Janice Kielbassa
- Bioinformatics Platform, Synergie Lyon Cancer Foundation, Lyon, France
| | - Laurie Tonon
- Bioinformatics Platform, Synergie Lyon Cancer Foundation, Lyon, France
| | - Helen He Zhu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Stem Cell Research Center, Shanghai Cancer Institute and Department of Urology, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Pierre Saintigny
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Department of Cancer Initiation and Tumor cell Identity, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Centre Léon Bérard, Lyon, France
- Department of Tumor Escape Resistance and Immunity, CRCL, Lyon, France
| | - Wei-Qiang Gao
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Stem Cell Research Center, Shanghai Cancer Institute and Department of Urology, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, P.R. China
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Arnaud de la Fouchardiere
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Department of Cancer Initiation and Tumor cell Identity, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Centre Léon Bérard, Lyon, France
- Department of Tumor Escape Resistance and Immunity, CRCL, Lyon, France
| | - Franck Tirode
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Department of Cancer Initiation and Tumor cell Identity, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Universite Claude Bernard Lyon 1, CRCL, Lyon, France
- Centre Léon Bérard, Lyon, France
| | - Alain Viari
- Bioinformatics Platform, Synergie Lyon Cancer Foundation, Lyon, France
| | - Jean-Yves Blay
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Department of Cancer Initiation and Tumor cell Identity, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Centre Léon Bérard, Lyon, France
- Department of Tumor Escape Resistance and Immunity, CRCL, Lyon, France
| | - Véronique Maguer-Satta
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Department of Cancer Initiation and Tumor cell Identity, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Universite Claude Bernard Lyon 1, CRCL, Lyon, France
- Centre Léon Bérard, Lyon, France
| |
Collapse
|
22
|
Lv J, Liu X, Zhou Y, Cheng F, Chen H, Li S, Wang D, Zhou L, Wang Z, Zhou N, Chen J, Huang B. YAP Inactivation by Soft Mechanotransduction Relieves MAFG for Tumor Cell Dedifferentiation. RESEARCH (WASHINGTON, D.C.) 2023; 6:0215. [PMID: 37614365 PMCID: PMC10443527 DOI: 10.34133/research.0215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 08/09/2023] [Indexed: 08/25/2023]
Abstract
Solid tumor cells live in a highly dynamic mechanical microenvironment. How the extracellular-matrix-generated mechanotransduction regulates tumor cell development and differentiation remains an enigma. Here, we show that a low mechanical force generated from the soft matrix induces dedifferentiation of moderately stiff tumor cells to soft stem-cell-like cells. Mechanistically, integrin β8 was identified to transduce mechano-signaling to trigger tumor cell dedifferentiation by recruiting RhoGDI1 to inactivate RhoA and subsequently Yes-associated protein (YAP). YAP inactivation relieved the inhibition of v-maf avian musculoaponeurotic fibrosarcoma oncogene homolog G (MAFG), allowing MAFG to transactivate the stemness genes NANOG, SOX2, and NESTIN. Inactivation also restored β8 expression, thereby forming a closed mechanical loop. Importantly, MAFG expression is correlated with worse prognosis. Our findings provide mechanical insights into the regulation of tumor cell dedifferentiation, which has therapeutic implications for exploring innovative strategies to attack malignancies.
Collapse
Affiliation(s)
- Jiadi Lv
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases,
Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College (PUMC), Beijing, 100005, China
| | - Xiaohan Liu
- Department of Histology and Embryology, Basic Medical College,
China Medical University, Shenyang, Liaoning 110122, China
| | - Yabo Zhou
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases,
Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College (PUMC), Beijing, 100005, China
| | - Feiran Cheng
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases,
Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College (PUMC), Beijing, 100005, China
| | - Haoran Chen
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases,
Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College (PUMC), Beijing, 100005, China
| | - Shunshun Li
- Department of Immunology, Basic Medical College,
China Medical University, Shenyang, Liaoning 110122, China
| | - Dianheng Wang
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases,
Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College (PUMC), Beijing, 100005, China
| | - Li Zhou
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases,
Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College (PUMC), Beijing, 100005, China
| | - Zhenfeng Wang
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases,
Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College (PUMC), Beijing, 100005, China
| | - Nannan Zhou
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases,
Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College (PUMC), Beijing, 100005, China
| | - Jie Chen
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases,
Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College (PUMC), Beijing, 100005, China
| | - Bo Huang
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases,
Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College (PUMC), Beijing, 100005, China
- Department of Biochemistry & Molecular Biology, Tongji Medical College,
Huazhong University of Science & Technology, Wuhan 430030, China
| |
Collapse
|
23
|
Wang X, Golino JL, Hawk NV, Xie C. Reciprocal Interaction of Cancer Stem Cells of Cholangiocarcinoma with Macrophage. Stem Cell Rev Rep 2023; 19:2013-2023. [PMID: 37249733 PMCID: PMC10390592 DOI: 10.1007/s12015-023-10557-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/08/2023] [Indexed: 05/31/2023]
Abstract
Cholangiocarcinoma is an aggressive type of liver cancer with few effective treatment options. Therefore, there is great need to better understand the biology of this malignancy to further development of novel treatment options. Cancer stem cells (CSCs) are thought to the underlying reason for cancer initiation, metastasis, and relapse. However, due to their elusive character and differences in identification among different types of cancer, it remains a challenge to study such cells. Additionally, characterization of the tumor microenvironment such as interactions with immune cells remain largely unknown. Here, we employ a fluorescent reporter system to track and isolate stem-like cancer cells of cholangiocarcinoma cell lines. Following verification of a stem-like signature (upregulated expression of stemness markers, resistance to chemotherapy, increased spheroid formation, and tumorigenesis capabilities despite inoculation of a small number of cells), we analyzed the interaction of these cells with macrophages via direct and indirect coculture assays. We noted direct coculturing increased stemness among CSC populations and induced both M1 (CD80 and HLA-DR) and M2 (CD163) tumor associated macrophage polarization. These studies suggest that there is a bi-directional crosstalk between macrophages and CSCs that promotes stemness renewal and tumor associated macrophage polarization.
Collapse
Affiliation(s)
- Xin Wang
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Jihye L Golino
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Nga Voong Hawk
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Changqing Xie
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20814, USA.
- NCI CCR Liver Cancer Program, National Institutes of Health, 10 Center Drive, Building 10 3B43, Bethesda, MD, 20892, USA.
| |
Collapse
|
24
|
Fan M, Shi Y, Zhao J, Li L. Cancer stem cell fate determination: mito-nuclear communication. Cell Commun Signal 2023; 21:159. [PMID: 37370081 PMCID: PMC10294499 DOI: 10.1186/s12964-023-01160-x] [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: 02/12/2023] [Accepted: 05/06/2023] [Indexed: 06/29/2023] Open
Abstract
Cancer stem cells (CSCs) are considered to be responsible for tumor recurrence and metastasis. Therefore, clarification of the mechanisms involved in CSC stemness maintenance and cell fate determination would provide a new strategy for cancer therapy. Unregulated cellular energetics has been accepted as one of the hallmarks of cancer cells, but recent studies have revealed that mitochondrial metabolism can also actively determine CSC fate by affecting nuclear stemness gene expression. Herein, from the perspective of mito-nuclear communication, we review recent progress on the influence of mitochondria on CSC potential from four aspects: metabolism, dynamics, mitochondrial homeostasis, and reactive oxygen species (ROS). Video Abstract.
Collapse
Affiliation(s)
- Mengchen Fan
- School of Basic Medical Sciences, Medical College of Yan’an University, Yanan, 716000 China
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi’an, 710032 China
| | - Ying Shi
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi’an, 710032 China
| | - Jumei Zhao
- School of Basic Medical Sciences, Medical College of Yan’an University, Yanan, 716000 China
| | - Ling Li
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi’an, 710032 China
| |
Collapse
|
25
|
Haidar Ahmad S, El Baba R, Herbein G. Polyploid giant cancer cells, cytokines and cytomegalovirus in breast cancer progression. Cancer Cell Int 2023; 23:119. [PMID: 37340387 DOI: 10.1186/s12935-023-02971-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 06/12/2023] [Indexed: 06/22/2023] Open
Abstract
BACKGROUND Breast cancer is the most common cancer among women. Accumulated evidence over the past decades indicates a very high prevalence of human cytomegalovirus (HCMV) in breast cancer. High-risk HCMV strains possess a direct oncogenic effect displayed by cellular stress, polyploid giant cancer cells (PGCCs) generation, stemness, and epithelial-to-mesenchymal transition (EMT) leading to cancer of aggressive phenotype. Breast cancer development and progression have been regulated by several cytokines where the latter can promote cancer cell survival, help in tumor immune evasion, and initiate the EMT process, thereby resulting in invasion, angiogenesis, and breast cancer metastasis. In the present study, we screened cytokines expression in cytomegalovirus-transformed HMECs (CTH cells) cultures infected with HCMV high-risk strains namely, HCMV-DB and BL, as well as breast cancer biopsies, and analyzed the association between cytokines production, PGCCs count, and HCMV presence in vitro and in vivo. METHODS In CTH cultures and breast cancer biopsies, HCMV load was quantified by real-time qPCR. PGCCs count in CTH cultures and breast cancer biopsies was identified based on cell morphology and hematoxylin and eosin staining, respectively. CTH supernatants were evaluated for the production of TGF-β, IL-6, IL1-β, and IL-10 by ELISA assays. The above-mentioned cytokines expression was assessed in breast cancer biopsies using reverse transcription-qPCR. The correlation analyses were performed using Pearson correlation test. RESULTS The revealed PGCCs/cytokine profile in our in vitro CTH model matched that of the breast cancer biopsies, in vivo. Pronounced cytokine expression and PGCCs count were detected in particularly CTH-DB cultures and basal-like breast cancer biopsies. CONCLUSIONS The analysis of cytokine profiles in PGCCs present mostly in basal-like breast cancer biopsies and derived from CTH cells chronically infected with the high-risk HCMV strains might have the potential to provide novel therapies such as cytokine-based immunotherapy which is a promising field in cancer treatments.
Collapse
Affiliation(s)
- Sandy Haidar Ahmad
- Department Pathogens and Inflammation-EPILAB, EA4266, University of France-Comté, 16 Route de Gray, 25030, Besançon Cedex, France
| | - Ranim El Baba
- Department Pathogens and Inflammation-EPILAB, EA4266, University of France-Comté, 16 Route de Gray, 25030, Besançon Cedex, France
| | - Georges Herbein
- Department Pathogens and Inflammation-EPILAB, EA4266, University of France-Comté, 16 Route de Gray, 25030, Besançon Cedex, France.
- Department of Virology, CHRU Besancon, Besancon, France.
| |
Collapse
|
26
|
Beziaud L, Young CM, Alonso AM, Norkin M, Minafra AR, Huelsken J. IFNγ-induced stem-like state of cancer cells as a driver of metastatic progression following immunotherapy. Cell Stem Cell 2023; 30:818-831.e6. [PMID: 37267916 DOI: 10.1016/j.stem.2023.05.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/29/2023] [Accepted: 05/08/2023] [Indexed: 06/04/2023]
Abstract
Despite the remarkable success of immune checkpoint blockade (ICB) therapy, most cancer patients still do not respond. We now find that immunotherapy can induce stem-like properties in tumors. Using mouse models of breast cancer, we observe that cancer stem cells (CSCs) show not only enhanced resistance to T cell cytotoxicity, but that interferon gamma (IFNγ) produced by activated T cells directly converts non-CSCs to CSCs. IFNγ enhances several CSC phenotypes, such as resistance to chemo- and radiotherapy and metastasis formation. We identified the branched-chain amino acid aminotransaminase 1 (BCAT1) as a downstream mediator of IFNγ-induced CSC plasticity. Targeting BCAT1 in vivo improved cancer vaccination and ICB therapy by preventing IFNγ-induced metastasis formation. Breast cancer patients treated with ICB exhibited a similar increase in CSC markers expression indicating comparable responses to immune activation in humans. Collectively, we discover an unexpected, pro-tumoral role for IFNγ that may contribute to cancer immunotherapy failure.
Collapse
Affiliation(s)
- Laurent Beziaud
- Ecole Polytechnique Fédérale de Lausanne (EPFL), ISREC (Swiss Institute for Experimental Cancer Research), 1015 Lausanne, Switzerland; Agora Translational Cancer Research Center, 1005 Lausanne, Switzerland
| | - C Megan Young
- Ecole Polytechnique Fédérale de Lausanne (EPFL), ISREC (Swiss Institute for Experimental Cancer Research), 1015 Lausanne, Switzerland; Agora Translational Cancer Research Center, 1005 Lausanne, Switzerland
| | - Angela Madurga Alonso
- Ecole Polytechnique Fédérale de Lausanne (EPFL), ISREC (Swiss Institute for Experimental Cancer Research), 1015 Lausanne, Switzerland; Agora Translational Cancer Research Center, 1005 Lausanne, Switzerland
| | - Maxim Norkin
- Ecole Polytechnique Fédérale de Lausanne (EPFL), ISREC (Swiss Institute for Experimental Cancer Research), 1015 Lausanne, Switzerland; Agora Translational Cancer Research Center, 1005 Lausanne, Switzerland
| | - Anna Rita Minafra
- Ecole Polytechnique Fédérale de Lausanne (EPFL), ISREC (Swiss Institute for Experimental Cancer Research), 1015 Lausanne, Switzerland
| | - Joerg Huelsken
- Ecole Polytechnique Fédérale de Lausanne (EPFL), ISREC (Swiss Institute for Experimental Cancer Research), 1015 Lausanne, Switzerland; Agora Translational Cancer Research Center, 1005 Lausanne, Switzerland.
| |
Collapse
|
27
|
González-Callejo P, Gener P, Díaz-Riascos ZV, Conti S, Cámara-Sánchez P, Riera R, Mancilla S, García-Gabilondo M, Peg V, Arango D, Rosell A, Labernadie A, Trepat X, Albertazzi L, Schwartz S, Seras-Franzoso J, Abasolo I. Extracellular vesicles secreted by triple-negative breast cancer stem cells trigger premetastatic niche remodeling and metastatic growth in the lungs. Int J Cancer 2023; 152:2153-2165. [PMID: 36705298 DOI: 10.1002/ijc.34447] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 11/11/2022] [Accepted: 12/13/2022] [Indexed: 01/28/2023]
Abstract
Tumor secreted extracellular vesicles (EVs) are potent intercellular signaling platforms. They are responsible for the accommodation of the premetastatic niche (PMN) to support cancer cell engraftment and metastatic growth. However, complex cancer cell composition within the tumor increases also the heterogeneity among cancer secreted EVs subsets, a functional diversity that has been poorly explored. This phenomenon is particularly relevant in highly plastic and heterogenous triple-negative breast cancer (TNBC), in which a significant representation of malignant cancer stem cells (CSCs) is displayed. Herein, we selectively isolated and characterized EVs from CSC or differentiated cancer cells (DCC; EVsCSC and EVsDCC , respectively) from the MDA-MB-231 TNBC cell line. Our results showed that EVsCSC and EVsDCC contain distinct bioactive cargos and therefore elicit a differential effect on stromal cells in the TME. Specifically, EVsDCC activated secretory cancer associated fibroblasts (CAFs), triggering IL-6/IL-8 signaling and sustaining CSC phenotype maintenance. Complementarily, EVsCSC promoted the activation of α-SMA+ myofibroblastic CAFs subpopulations and increased the endothelial remodeling, enhancing the invasive potential of TNBC cells in vitro and in vivo. In addition, solely the EVsCSC mediated signaling prompted the transformation of healthy lungs into receptive niches able to support metastatic growth of breast cancer cells.
Collapse
Affiliation(s)
- Patricia González-Callejo
- Drug Delivery & Targeting, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Petra Gener
- Drug Delivery & Targeting, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Zamira V Díaz-Riascos
- Drug Delivery & Targeting, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain.,Functional Validation & Preclinical Research (FVPR), Vall d‧Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Passeig de la Vall d'Hebron, Barcelona, Spain
| | - Sefora Conti
- Integrative Cell and Tissue Dynamics Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain
| | - Patricia Cámara-Sánchez
- Drug Delivery & Targeting, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain.,Functional Validation & Preclinical Research (FVPR), Vall d‧Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Passeig de la Vall d'Hebron, Barcelona, Spain
| | - Roger Riera
- Nanoscopy for Nanomedicine Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain.,Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| | - Sandra Mancilla
- Drug Delivery & Targeting, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain.,Functional Validation & Preclinical Research (FVPR), Vall d‧Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Passeig de la Vall d'Hebron, Barcelona, Spain
| | - Miguel García-Gabilondo
- Neurovascular Research Laboratory, Vall d‧Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Passeig Vall d'Hebron, Barcelona, Spain
| | - Vicente Peg
- Department of Pathology, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Diego Arango
- Department of Molecular Oncology, Biomedical Research Institute of Lleida, Lleida, Spain.,Biomedical Research in Digestive Tract Tumors, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Passeig de la Vall d'Hebron, Barcelona, Spain
| | - Anna Rosell
- Neurovascular Research Laboratory, Vall d‧Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Passeig Vall d'Hebron, Barcelona, Spain
| | - Anna Labernadie
- Integrative Cell and Tissue Dynamics Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain
| | - Xavier Trepat
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain.,Integrative Cell and Tissue Dynamics Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), Passeig Lluís Companys, Barcelona, Spain
| | - Lorenzo Albertazzi
- Nanoscopy for Nanomedicine Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain.,Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| | - Simó Schwartz
- Drug Delivery & Targeting, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Joaquin Seras-Franzoso
- Drug Delivery & Targeting, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain.,Department of Genetics and Microbiology, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Ibane Abasolo
- Drug Delivery & Targeting, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain.,Functional Validation & Preclinical Research (FVPR), Vall d‧Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Passeig de la Vall d'Hebron, Barcelona, Spain
| |
Collapse
|
28
|
Ma Y, Zhu Y, Shang L, Qiu Y, Shen N, Wang J, Adam T, Wei W, Song Q, Li J, Wicha MS, Luo M. LncRNA XIST regulates breast cancer stem cells by activating proinflammatory IL-6/STAT3 signaling. Oncogene 2023; 42:1419-1437. [PMID: 36922677 PMCID: PMC10154203 DOI: 10.1038/s41388-023-02652-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 02/19/2023] [Accepted: 02/24/2023] [Indexed: 03/17/2023]
Abstract
Aberrant expression of XIST, a long noncoding RNA (lncRNA) initiating X chromosome inactivation (XCI) in early embryogenesis, is a common feature of breast cancer (BC). However, the roles of post-XCI XIST in breast carcinogenesis remain elusive. Here we identify XIST as a key regulator of breast cancer stem cells (CSCs), which exhibit aldehyde dehydrogenase positive (ALDH+) epithelial- (E) and CD24loCD44hi mesenchymal-like (M) phenotypes. XIST is variably expressed across the spectrum of BC subtypes, and doxycycline (DOX)-inducible knockdown (KD) of XIST markedly inhibits spheroid/colony forming capacity, tumor growth and tumor-initiating potential. This phenotype is attributed to impaired E-CSC in luminal and E- and M-CSC activities in triple-negative (TN) BC. Gene expression profiling unveils that XIST KD most significantly affects cytokine-cytokine receptor interactions, leading to markedly suppressed expression of proinflammatory cytokines IL-6 and IL-8 in ALDH- bulk BC cells. Exogenous IL-6, but not IL-8, rescues the reduced sphere-forming capacity and proportion of ALDH+ E-CSCs in luminal and TN BC upon XIST KD. XIST functions as a nuclear sponge for microRNA let-7a-2-3p to activate IL-6 production from ALDH- bulk BC cells, which acts in a paracrine fashion on ALDH+ E-CSCs that display elevated cell surface IL-6 receptor (IL6R) expression. This promotes CSC self-renewal via STAT3 activation and expression of key CSC factors including c-MYC, KLF4 and SOX9. Together, this study supports a novel role of XIST by derepressing let-7 controlled paracrine IL-6 proinflammatory signaling to promote CSC self-renewal.
Collapse
Affiliation(s)
- Yuxi Ma
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Department of Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yongyou Zhu
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Li Shang
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- University of Michigan Rogel Cancer Center, Ann Arbor, MI, 48109, USA
| | - Yan Qiu
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Na Shen
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jonathan Wang
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Tiffany Adam
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Wei Wei
- Department of Breast and Thyroid Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Qingxuan Song
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Jun Li
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Max S Wicha
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.
- University of Michigan Rogel Cancer Center, Ann Arbor, MI, 48109, USA.
| | - Ming Luo
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.
- University of Michigan Rogel Cancer Center, Ann Arbor, MI, 48109, USA.
- Department of Breast and Thyroid Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, China.
| |
Collapse
|
29
|
Verstappe J, Berx G. A role for partial epithelial-to-mesenchymal transition in enabling stemness in homeostasis and cancer. Semin Cancer Biol 2023; 90:15-28. [PMID: 36773819 DOI: 10.1016/j.semcancer.2023.02.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/19/2023] [Accepted: 02/02/2023] [Indexed: 02/12/2023]
Abstract
Stem cells have self-renewal capacities and the ability to give rise to differentiated cells thereby sustaining tissues during homeostasis and injury. This structural hierarchy extends to tumours which harbor stem-like cells deemed cancer stem cells that propagate the tumour and drive metastasis and relapse. The process of epithelial-to-mesenchymal transition (EMT), which plays an important role in development and cancer cell migration, was shown to be correlated with stemness in both homeostasis and cancer indicating that stemness can be acquired and is not necessarily an intrinsic trait. Nowadays it is experimentally proven that the activation of an EMT program does not necessarily drive cells towards a fully mesenchymal phenotype but rather to hybrid E/M states. This review offers the latest advances in connecting the EMT status and stem-cell state of both non-transformed and cancer cells. Recent literature clearly shows that hybrid EMT states have a higher probability of acquiring stem cell traits. The position of a cell along the EMT-axis which coincides with a stem cell-like state is known as the stemness window. We show how the original EMT-state of a cell dictates the EMT/MET inducing programmes required to reach stemness. Lastly we present the mechanism of stemness regulation and the regulatory feedback loops which position cells at a certain EMT state along the EMT axis.
Collapse
Affiliation(s)
- Jeroen Verstappe
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Geert Berx
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| |
Collapse
|
30
|
Jang YS, Kim TW, Ryu JS, Kong HJ, Jang SH, Nam GH, Kim JH, Jeon S. Upregulation of programmed death ligand-1 in tumor-associated macrophages affects chemotherapeutic response in ovarian cancer cells. PLoS One 2023; 18:e0277285. [PMID: 36757936 PMCID: PMC9910707 DOI: 10.1371/journal.pone.0277285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/24/2022] [Indexed: 02/10/2023] Open
Abstract
To better understand the mechanism of chemoresistance in ovarian cancer cells, we aimed to investigate the influence of macrophages on the tumor cell response to carboplatin and identify the genes associated with chemoresistance. We mimicked the tumor microenvironment (TME) using a co-culture technique and compared the proliferation of ovarian cells with and without macrophages. We also examined M1 and M2 marker expression and the expression of key TME genes. Post the co-culture, we treated ovarian cancer cells with carboplatin and elucidated the function of programmed death-ligand 1 (PD-L1) in carboplatin chemoresistance. We investigated CD68 and PD-L1 expression in normal and cancerous ovarian tissues using immunohistochemistry (IHC). Finally, we analyzed the association between CD68 or PD-L1 expression and survival outcomes. Inducible nitric oxide synthase (iNOS) was downregulated, while the gene expression of M2 macrophage markers was increased in ovarian cancer cells. PD-L1, vascular endothelial growth factor (VEGF), Interleukin (IL)-6, IL-10, IL-12, signal transducer and activator of transcription 3 (STAT3), B-cell lymphoma 2 (BCL2), multidrug resistance 1 (MDR1), and colony stimulating factor 1 (CSF-1) were upregulated. Notably, PD-L1 was upregulated in both the ovarian cancer cells and macrophages. Ovarian cancer cells co-cultured with macrophages exhibited statistically significant carboplatin resistance compared to single-cultured ovarian cancer cells. PD-L1 silencing induced chemosensitivity in both types of co-cultured ovarian cancer cells. However, IHC results revealed no correlation between PD-L1 expression and patient survival or cancer stage. CD68 expression was significantly increased in cancer cells compared to normal or benign ovarian tumor cells, but it was not associated with the survival outcomes of ovarian cancer patients. Our study demonstrated that ovarian cancer cells interact with macrophages to induce the M2 phenotype. We also established that PD-L1 upregulation in both ovarian cancer cells and macrophages is a key factor for carboplatin chemoresistance.
Collapse
Affiliation(s)
- Yong Soo Jang
- Department of Obstetrics and Gynecology, College of Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Tae Wan Kim
- Soonchunhyang Innovative Convergence Research Center, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
- Department of Medical Life Science, Soonchunhyang University, Asan, Korea
| | - Jae Sung Ryu
- Soonchunhyang Innovative Convergence Research Center, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
- Department of Medical Life Science, Soonchunhyang University, Asan, Korea
| | - Hye Jeong Kong
- Soonchunhyang Innovative Convergence Research Center, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
- Department of Medical Life Science, Soonchunhyang University, Asan, Korea
| | - Si Hyeong Jang
- Department of Pathology, College of Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Gye Hyun Nam
- Department of Obstetrics and Gynecology, College of Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Jae Hoon Kim
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Seob Jeon
- Department of Obstetrics and Gynecology, College of Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
- * E-mail:
| |
Collapse
|
31
|
Faria L, Canato S, Jesus TT, Gonçalves M, Guerreiro PS, Lopes CS, Meireles I, Morais-de-Sá E, Paredes J, Janody F. Activation of an actin signaling pathway in pre-malignant mammary epithelial cells by P-cadherin is essential for transformation. Dis Model Mech 2023; 16:dmm049652. [PMID: 36808468 PMCID: PMC9983776 DOI: 10.1242/dmm.049652] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 01/19/2023] [Indexed: 02/23/2023] Open
Abstract
Alterations in the expression or function of cell adhesion molecules have been implicated in all steps of tumor progression. Among those, P-cadherin is highly enriched in basal-like breast carcinomas, playing a central role in cancer cell self-renewal, collective cell migration and invasion. To establish a clinically relevant platform for functional exploration of P-cadherin effectors in vivo, we generated a humanized P-cadherin Drosophila model. We report that actin nucleators, Mrtf and Srf, are main P-cadherin effectors in fly. We validated these findings in a human mammary epithelial cell line with conditional activation of the SRC oncogene. We show that, prior to promoting malignant phenotypes, SRC induces a transient increase in P-cadherin expression, which correlates with MRTF-A accumulation, its nuclear translocation and the upregulation of SRF target genes. Moreover, knocking down P-cadherin, or preventing F-actin polymerization, impairs SRF transcriptional activity. Furthermore, blocking MRTF-A nuclear translocation hampers proliferation, self-renewal and invasion. Thus, in addition to sustaining malignant phenotypes, P-cadherin can also play a major role in the early stages of breast carcinogenesis by promoting a transient boost of MRTF-A-SRF signaling through actin regulation.
Collapse
Affiliation(s)
- Lídia Faria
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
- Master Programme in Oncology, School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
| | - Sara Canato
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
- Physiology and Cancer Program, Champalimaud Foundation, Avenida de Brasília, 1400-038 Lisboa, Portugal
| | - Tito T. Jesus
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
| | - Margarida Gonçalves
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Patrícia S. Guerreiro
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
- Vector B2B - Drug Developing - Associação Para Investigação em Biotecnologia, Av. Prof. Egas Moniz, Edifício Egas Moniz, 1649-028 Lisboa, Portugal
| | - Carla S. Lopes
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Isabel Meireles
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
| | - Eurico Morais-de-Sá
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Joana Paredes
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
- FMUP, Medical Faculty of University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Florence Janody
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, P-2780-156 Oeiras, Portugal
| |
Collapse
|
32
|
Zhang L, Chen W, Liu S, Chen C. Targeting Breast Cancer Stem Cells. Int J Biol Sci 2023; 19:552-570. [PMID: 36632469 PMCID: PMC9830502 DOI: 10.7150/ijbs.76187] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 12/09/2022] [Indexed: 01/04/2023] Open
Abstract
The potential roles of breast cancer stem cells (BCSCs) in tumor initiation and recurrence have been recognized for many decades. Due to their strong capacity for self-renewal and differentiation, BCSCs are the major reasons for poor clinical outcomes and low therapeutic response. Several hypotheses on the origin of cancer stem cells have been proposed, including critical gene mutations in stem cells, dedifferentiation of somatic cells, and cell plasticity remodeling by epithelial-mesenchymal transition (EMT) and the tumor microenvironment. Moreover, the tumor microenvironment, including cellular components and cytokines, modulates the self-renewal and therapeutic resistance of BCSCs. Small molecules, antibodies, and chimeric antigen receptor (CAR)-T cells targeting BCSCs have been developed, and their applications in combination with conventional therapies are undergoing clinical trials. In this review, we focus on the features of BCSCs, emphasize the major factors and tumor environment that regulate the stemness of BCSCs, and discuss potential BCSC-targeting therapies.
Collapse
Affiliation(s)
- Lu Zhang
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; State Key Laboratory of Genetic Engineering; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; The Shanghai paracrine Key Laboratory of Medical Epigenetics; Shanghai Key Laboratory of Radiation Oncology; The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology; Shanghai Medical College; Fudan University, Shanghai 200032, China
| | - Wenmin Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming 650201, China.,Kunming College of Life Sciences, the University of the Chinese Academy of Sciences, Kunming 650201, China
| | - Suling Liu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; State Key Laboratory of Genetic Engineering; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; The Shanghai paracrine Key Laboratory of Medical Epigenetics; Shanghai Key Laboratory of Radiation Oncology; The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology; Shanghai Medical College; Fudan University, Shanghai 200032, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing 211166, China.,✉ Corresponding authors: Ceshi Chen, E-mail: or Suling Liu, E-mail:
| | - Ceshi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming 650201, China.,Academy of Biomedical Engineering, Kunming Medical University, Kunming 650500, China.,The Third Affiliated Hospital, Kunming Medical University, Kunming 650118, China.,✉ Corresponding authors: Ceshi Chen, E-mail: or Suling Liu, E-mail:
| |
Collapse
|
33
|
Abou Shousha S, Baheeg S, Ghoneim H, Zoheir M, Hemida M, Shahine Y. The effect of Fas/FasL pathway blocking on apoptosis and stemness within breast cancer tumor microenvironment (preclinical study). Breast Dis 2023; 42:163-176. [PMID: 37334575 DOI: 10.3233/bd-220077] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Evasion of the immune system is the tumor's key strategy for its maintenance and progression. Thus, targeting the tumor microenvironment (TME) is considered one of the most promising approaches for fighting cancer, where immune cells within the TME play a vital role in immune surveillance and cancer elimination.FasL is one of the most important death ligands expressed by tumor-infiltrating lymphocytes (TILs) and plays a vital role in eliminating Fas-expressing cancer cells via Fas/FasL pathway-induced apoptosis. However, tumor cells can express elevated levels of FasL inducing apoptosis to TILs. Fas/FasL expression is linked to the maintenance of cancer stem cells (CSCs) within the TME, contributing to tumor aggressiveness, metastasis, recurrence, and chemoresistance.This study is considered the first study designed to block the overexpressed FasL on the tumor cells within TME mimicking tissue culture system using rFas molecules and supplementing the Fas enriched tissue culture system with blocked Fas - peripheral blood mononuclear cells PBMCs (using anti-Fas mAb) to protect them from tumor counterattack and augment their ability to induce tumor cell apoptosis and stemness inhibition.A significantly increased level of apoptosis and decreased expression of CD 44 (CSCs marker) was observed within the east tumor tissue culture system enriched with Fas molecules and anti-Fas treated PBMCs and the one enriched with Fas molecules only compared to the breast tumor tissues cultured alone (p < 0.001). Accordingly, we can consider the current study as a promising proposed immunotherapeutic strategy for breast cancer.
Collapse
Affiliation(s)
- Seham Abou Shousha
- Medical Research Institute, Immunology and Allergy Department, Alexandria University, Alexandria, Egypt
| | - Suzan Baheeg
- National Institute of Oceanography and Fisheries, Central Laboratories Unit, Alexandria, Egypt
| | - Hossam Ghoneim
- Medical Research Institute, Immunology and Allergy Department, Alexandria University, Alexandria, Egypt
| | - Malak Zoheir
- Medical Research Institute, Pathology Department, Alexandria University, Alexandria, Egypt
| | - Mahmoud Hemida
- Medical Research Institute, Department of Surgery, Alexandria University, Alexandria, Egypt
| | - Yasmine Shahine
- Faculty of Pharmacy, Department of Microbiology & Immunology, Pharos University in Alexandria, Alexandria, Egypt
| |
Collapse
|
34
|
Xu L, Chen Z, Wang Y, Li Y, Wang Z, Li F, Xi X. Polyphyllin VII as a Potential Drug for Targeting Stemness in Hepatocellular Cancer via STAT3 Signaling. Curr Cancer Drug Targets 2023; 23:325-331. [PMID: 36284387 DOI: 10.2174/1568009623666221024103834] [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: 05/12/2022] [Revised: 09/07/2022] [Accepted: 09/13/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND At present, the treatment of hepatocellular carcinoma (HCC) is disturbed by the treatment failure and recurrence caused by the residual liver cancer stem cells (CSCs). Therefore, drugs targeting HCC CSCs should be able to effectively eliminate HCC and prevent its recurrence. In this study, we demonstrated the effect of Polyphyllin VII (PP7) on HCC CSCs and explored their potential mechanism. METHODS HepG2 and Huh7 cells were used to analyze the antitumor activity of PP7 by quantifying cell growth and metastasis as well as to study the effect on stemness. RESULTS Our results demonstrated that PP7 promoted apoptosis and significantly inhibited proliferation and migration of both HepG2 and Huh7 cells. PP7 also inhibited tumor spheroid formation and induced significant changes in the expression of stemness markers (CD133 and OCT-4). These effects of PP7 were mediated by STAT3 signaling. CONCLUSION PP7 can effectively suppress tumor initiation, growth, and metastasis and inhibit stemness through regulation of STAT3 signaling pathway in liver cancer cells. Our data would add more evidence to further clarify the therapeutic effect of PP7 against HCC.
Collapse
Affiliation(s)
- Liuhang Xu
- Department of Immunology, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, No. 30 Renmin Nanlu, Shiyan City, Hubei Province 442000, P.R. China
| | - Ziqi Chen
- Department of Immunology, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, No. 30 Renmin Nanlu, Shiyan City, Hubei Province 442000, P.R. China
| | - Yangbin Wang
- Department of Immunology, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, No. 30 Renmin Nanlu, Shiyan City, Hubei Province 442000, P.R. China
| | - Yulin Li
- Department of Immunology, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, No. 30 Renmin Nanlu, Shiyan City, Hubei Province 442000, P.R. China
| | - Zhongyu Wang
- Department of Immunology, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, No. 30 Renmin Nanlu, Shiyan City, Hubei Province 442000, P.R. China
| | - Fangzhou Li
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Shiyan, No. 30 Renmin Nanlu, Shiyan City, Hubei Province 442000, P.R. China
| | - Xueyan Xi
- Department of Immunology, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, No. 30 Renmin Nanlu, Shiyan City, Hubei Province 442000, P.R. China.,Renmin Hospital, Hubei University of Medicine, Shiyan, No. 30 Renmin Nanlu, Shiyan City, Hubei Province 442000, P.R. China.,Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, No. 30 Renmin Nanlu, Shiyan City, Hubei Province 442000, P.R. China
| |
Collapse
|
35
|
Liu Q, Guan C, Liu C, Li H, Wu J, Sun C. Targeting hypoxia-inducible factor-1alpha: A new strategy for triple-negative breast cancer therapy. Biomed Pharmacother 2022; 156:113861. [DOI: 10.1016/j.biopha.2022.113861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/30/2022] [Accepted: 10/08/2022] [Indexed: 11/02/2022] Open
|
36
|
F. V, V. D. P, C. M, M. LI, C. D, G. P, D. C, A. T, M. G, S. DF, M. T, V. V, G. S. Targeting epigenetic alterations in cancer stem cells. FRONTIERS IN MOLECULAR MEDICINE 2022; 2:1011882. [PMID: 39086963 PMCID: PMC11285701 DOI: 10.3389/fmmed.2022.1011882] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/08/2022] [Indexed: 08/02/2024]
Abstract
Oncogenes or tumor suppressor genes are rarely mutated in several pediatric tumors and some early stage adult cancers. This suggests that an aberrant epigenetic reprogramming may crucially affect the tumorigenesis of these tumors. Compelling evidence support the hypothesis that cancer stem cells (CSCs), a cell subpopulation within the tumor bulk characterized by self-renewal capacity, metastatic potential and chemo-resistance, may derive from normal stem cells (NSCs) upon an epigenetic deregulation. Thus, a better understanding of the specific epigenetic alterations driving the transformation from NSCs into CSCs may help to identify efficacious treatments to target this aggressive subpopulation. Moreover, deepening the knowledge about these alterations may represent the framework to design novel therapeutic approaches also in the field of regenerative medicine in which bioengineering of NSCs has been evaluated. Here, we provide a broad overview about: 1) the role of aberrant epigenetic modifications contributing to CSC initiation, formation and maintenance, 2) the epigenetic inhibitors in clinical trial able to specifically target the CSC subpopulation, and 3) epigenetic drugs and stem cells used in regenerative medicine for cancer and diseases.
Collapse
Affiliation(s)
- Verona F.
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Pantina V. D.
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| | - Modica C.
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| | - Lo Iacono M.
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - D’Accardo C.
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Porcelli G.
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Cricchio D.
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| | - Turdo A.
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Gaggianesi M.
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| | - Di Franco S.
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| | - Todaro M.
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Veschi V.
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| | - Stassi G.
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| |
Collapse
|
37
|
Oct4 cooperates with c-Myc to improve mesenchymal-to-endothelial transition and myocardial repair of cardiac-resident mesenchymal stem cells. Stem Cell Res Ther 2022; 13:445. [PMID: 36056383 PMCID: PMC9438134 DOI: 10.1186/s13287-022-03120-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 08/04/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cardiac-resident mesenchymal stem cells (cMSCs) can exhibit fibrotic, proinflammatory, and proangiogenic phenotype in response to myocardial ischemia (Isch). How their phenotypic fate decisions are determined remains poorly understood. Here, we demonstrate that the cooperation of Oct4 and c-Myc in cMSCs creates a preferable mesenchymal-to-endothelial transition (MEndoT) to promote angiogenesis and consequent myocardial repair. METHODS We collected MSCs from cardiac and peripheral blood of rat with left ventricular Isch (LV Isch) 30 days after myocardial infarction (MI) or sham operation. After a comparison of characterization between cMSCs and peripheral blood MSCs (pbMSCs), we conducted transcriptome analysis and RNA sequencing of cMSCs. Using loss/gain-of-function approaches to understand the cooperation of c-Myc and Oct4 on MEndoT of cMSCs under hypoxic condition, we explored the mechanisms through transcriptome and functional experiment, and chromatin immunoprecipitation. Next, we transplanted male cMSCs with overexpression or inhibition of c-Myc/Oct4 into the infarcted myocardium of female rats and evaluated infarct size, cell retention, inflammation, remodeling, and function after 30 days. RESULTS LV Isch switched cMSCs toward both inflammatory and proangiogenic phenotypes, with increased secretion of inflammatory cytokines as well as decreased expression of proangiogenic factors. The effect of LV Isch on pbMSCs was less remarkable. Gene expression heatmap showed imbalance in expression of Oct4 and c-Myc regulating genes associated with remodeling of cMSCs. We provided evidence that cMSCs-specific c-Myc- versus Oct4-overexpression showed divergent genomic signatures, and their corresponding target genes play an important role in regulating cMSCs phenotypic changes. In particular, Oct4 accelerated angiogenesis induced by c-Myc overexpression in cMSCs and inhibited their phenotypic transition into inflammatory cells and fibroblast. Mechanistically, exogenous Oct4 caused c-Myc to translocate from the nucleus to the cytoplasm and activated some of its target signalings including VEGF signaling. Although transplantation of cMSCs alone did not improve LV remodeling and function, cMSCs co-transfected with c-Myc and Oct4 promoted a more positive effect in their survival and reparative properties, increased animal survival, reduced infarct size, decreased scar thickness, inhibited LV remodeling, and improved heart function 30 days after MI. Significantly, Oct4 promoted MEndoT ("Rescue me" signal) of cMSCs after both c-Myc stimulation in vitro and transplantation into the infarcted heart. CONCLUSIONS Myocardial Isch drives resident cMSCs toward multiple phenotypes. Oct4 interacts with c-Myc to promote MEndoT capacity of cMSCs and improve their survival and reparative effects through upregulation of angiogenesis-related signaling pathways. These findings may identify novel targets for stem cell therapy.
Collapse
|
38
|
Aguilar-Cazares D, Chavez-Dominguez R, Marroquin-Muciño M, Perez-Medina M, Benito-Lopez JJ, Camarena A, Rumbo-Nava U, Lopez-Gonzalez JS. The systemic-level repercussions of cancer-associated inflammation mediators produced in the tumor microenvironment. Front Endocrinol (Lausanne) 2022; 13:929572. [PMID: 36072935 PMCID: PMC9441602 DOI: 10.3389/fendo.2022.929572] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/01/2022] [Indexed: 12/15/2022] Open
Abstract
The tumor microenvironment is a dynamic, complex, and redundant network of interactions between tumor, immune, and stromal cells. In this intricate environment, cells communicate through membrane-membrane, ligand-receptor, exosome, soluble factors, and transporter interactions that govern cell fate. These interactions activate the diverse and superfluous signaling pathways involved in tumor promotion and progression and induce subtle changes in the functional activity of infiltrating immune cells. The immune response participates as a selective pressure in tumor development. In the early stages of tumor development, the immune response exerts anti-tumor activity, whereas during the advanced stages, the tumor establishes mechanisms to evade the immune response, eliciting a chronic inflammation process that shows a pro-tumor effect. The deregulated inflammatory state, in addition to acting locally, also triggers systemic inflammation that has repercussions in various organs and tissues that are distant from the tumor site, causing the emergence of various symptoms designated as paraneoplastic syndromes, which compromise the response to treatment, quality of life, and survival of cancer patients. Considering the tumor-host relationship as an integral and dynamic biological system, the chronic inflammation generated by the tumor is a communication mechanism among tissues and organs that is primarily orchestrated through different signals, such as cytokines, chemokines, growth factors, and exosomes, to provide the tumor with energetic components that allow it to continue proliferating. In this review, we aim to provide a succinct overview of the involvement of cancer-related inflammation at the local and systemic level throughout tumor development and the emergence of some paraneoplastic syndromes and their main clinical manifestations. In addition, the involvement of these signals throughout tumor development will be discussed based on the physiological/biological activities of innate and adaptive immune cells. These cellular interactions require a metabolic reprogramming program for the full activation of the various cells; thus, these requirements and the by-products released into the microenvironment will be considered. In addition, the systemic impact of cancer-related proinflammatory cytokines on the liver-as a critical organ that produces the leading inflammatory markers described to date-will be summarized. Finally, the contribution of cancer-related inflammation to the development of two paraneoplastic syndromes, myelopoiesis and cachexia, will be discussed.
Collapse
Affiliation(s)
- Dolores Aguilar-Cazares
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
| | - Rodolfo Chavez-Dominguez
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
- Posgrado en Ciencias Biologicas, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| | - Mario Marroquin-Muciño
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
- Laboratorio de Quimioterapia Experimental, Departamento de Bioquimica, Escuela Nacional de Ciencias Biologicas, Instituto Politecnico Nacional, Mexico City, Mexico
| | - Mario Perez-Medina
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
- Laboratorio de Quimioterapia Experimental, Departamento de Bioquimica, Escuela Nacional de Ciencias Biologicas, Instituto Politecnico Nacional, Mexico City, Mexico
| | - Jesus J. Benito-Lopez
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
- Posgrado en Ciencias Biologicas, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| | - Angel Camarena
- Laboratorio de Human Leukocyte Antigen (HLA), Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
| | - Uriel Rumbo-Nava
- Clinica de Neumo-Oncologia, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
| | - Jose S. Lopez-Gonzalez
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
| |
Collapse
|
39
|
Macrophage induced ERK-TGF-β1 signaling in MCF7 breast cancer cells result in reversible cancer stem cell plasticity and epithelial mesenchymal transition. Biochim Biophys Acta Gen Subj 2022; 1866:130215. [PMID: 35905921 DOI: 10.1016/j.bbagen.2022.130215] [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: 03/10/2022] [Revised: 06/22/2022] [Accepted: 07/21/2022] [Indexed: 10/31/2022]
Abstract
BACKGROUND Breast cancer is a heterogenous disease composed of multiple clonal populations and the mechanism by which the tumor microenvironment induces cancer stem cell plasticity is not fully understood. METHODS MCF7 breast cancer cells were treated with macrophage conditioned medium (MɸCM). PD98059 and SB431542 were used for ERK and TGF-βR inhibition respectively. Epithelial-mesenchymal transition (EMT) and cancer stem cell markers (CSC) were studied using qRT-PCR and flowcytometry. SCID mice were used for animal experiments. RESULTS MɸCM- induced ERK/TGF-β1 signaling led to enrichment of CSC and EMT in MCF7 cells and mammospheres. These effects were abrogated by both MEK inhibitor PD98059 (TGF-β1 synthesis) and SB431542 (TGF-β1 signaling). The increase in CSC was both hybrid (ALDH1+) and mesenchymal (CD44+ CD24- cells). Increase in hybrid E/M state was at a single cell level as confirmed by the increase in both claudin-1 (E) and vimentin (M). This did not have any growth advantage in SCID mice and monitoring of CSC and EMT markers before and after growth in SCID mice indicated reversal of these markers in tumor cells recovered from mice. Removal of MɸCM and neutralization of TNF-α, IL-6 and IL-1β in MɸCM abrogated ERK phosphorylation, TGF-β and CSC enrichment indicating the requirement of continuous signaling for maintenance. CONCLUSIONS ERK signaling plays an important role in MɸCM- induced EMT and CSC plasticity which is completely reversible upon withdrawal of signals. GENERAL SIGNIFICANCE Our experimental observations support the semi-independent nature of EMT-stemness connection which is very dynamic and reversible depending on the microenvironment.
Collapse
|
40
|
Zhou H, He Q, Li C, Alsharafi BLM, Deng L, Long Z, Gan Y. Focus on the tumor microenvironment: A seedbed for neuroendocrine prostate cancer. Front Cell Dev Biol 2022; 10:955669. [PMID: 35938167 PMCID: PMC9355504 DOI: 10.3389/fcell.2022.955669] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
The tumor microenvironment (TME) is a microecology consisting of tumor and mesenchymal cells and extracellular matrices. The TME plays important regulatory roles in tumor proliferation, invasion, metastasis, and differentiation. Neuroendocrine differentiation (NED) is a mechanism by which castration resistance develops in advanced prostate cancer (PCa). NED is induced after androgen deprivation therapy and neuroendocrine prostate cancer (NEPC) is established finally. NEPC has poor prognosis and short overall survival and is a major cause of death in patients with PCa. Both the cellular and non-cellular components of the TME regulate and induce NEPC formation through various pathways. Insights into the roles of the TME in NEPC evolution, growth, and progression have increased over the past few years. These novel insights will help refine the NEPC formation model and lay the foundation for the discovery of new NEPC therapies targeting the TME.
Collapse
Affiliation(s)
- Hengfeng Zhou
- Andrology Center, Department of Urology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Qiangrong He
- Andrology Center, Department of Urology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Chao Li
- Andrology Center, Department of Urology, the Third Xiangya Hospital, Central South University, Changsha, China
| | | | - Liang Deng
- Andrology Center, Department of Urology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhi Long
- Andrology Center, Department of Urology, the Third Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Zhi Long, ; Yu Gan,
| | - Yu Gan
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Zhi Long, ; Yu Gan,
| |
Collapse
|
41
|
Wang L, Jin Z, Master RP, Maharjan CK, Carelock ME, Reccoppa TBA, Kim MC, Kolb R, Zhang W. Breast Cancer Stem Cells: Signaling Pathways, Cellular Interactions, and Therapeutic Implications. Cancers (Basel) 2022; 14:3287. [PMID: 35805056 PMCID: PMC9265870 DOI: 10.3390/cancers14133287] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/02/2022] [Accepted: 07/02/2022] [Indexed: 02/01/2023] Open
Abstract
Breast cancer stem cells (BCSCs) constitute a small population of cells within breast cancer and are characterized by their ability to self-renew, differentiate, and recapitulate the heterogeneity of the tumor. Clinically, BCSCs have been correlated with cancer progression, metastasis, relapse, and drug resistance. The tumorigenic roles of BCSCs have been extensively reviewed and will not be the major focus of the current review. Here, we aim to highlight how the crucial intrinsic signaling pathways regulate the fate of BCSCs, including the Wnt, Notch, Hedgehog, and NF-κB signaling pathways, as well as how different cell populations crosstalk with BCSCs within the TME, including adipocytes, endothelial cells, fibroblasts, and immune cells. Based on the molecular and cellular activities of BCSCs, we will also summarize the targeting strategies for BCSCs and related clinical trials. This review will highlight that BCSC development in breast cancer is impacted by both BCSC endogenous signaling and external factors in the TME, which provides an insight into how to establish a comprehensively therapeutic strategy to target BCSCs for breast cancer treatments.
Collapse
Affiliation(s)
- Lei Wang
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (L.W.); (Z.J.); (R.P.M.); (C.K.M.); (M.E.C.); (T.B.A.R.); (M.-C.K.); (R.K.)
- Immunology Concentration, Biomedical Graduate Program, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Zeng Jin
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (L.W.); (Z.J.); (R.P.M.); (C.K.M.); (M.E.C.); (T.B.A.R.); (M.-C.K.); (R.K.)
- Cancer Biology Concentration, Biomedical Graduate Program, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Rohan P. Master
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (L.W.); (Z.J.); (R.P.M.); (C.K.M.); (M.E.C.); (T.B.A.R.); (M.-C.K.); (R.K.)
| | - Chandra K. Maharjan
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (L.W.); (Z.J.); (R.P.M.); (C.K.M.); (M.E.C.); (T.B.A.R.); (M.-C.K.); (R.K.)
| | - Madison E. Carelock
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (L.W.); (Z.J.); (R.P.M.); (C.K.M.); (M.E.C.); (T.B.A.R.); (M.-C.K.); (R.K.)
- Cancer Biology Concentration, Biomedical Graduate Program, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Tiffany B. A. Reccoppa
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (L.W.); (Z.J.); (R.P.M.); (C.K.M.); (M.E.C.); (T.B.A.R.); (M.-C.K.); (R.K.)
- Department of Biology, College of Liberal Arts & Sciences, University of Florida, Gainesville, FL 32610, USA
| | - Myung-Chul Kim
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (L.W.); (Z.J.); (R.P.M.); (C.K.M.); (M.E.C.); (T.B.A.R.); (M.-C.K.); (R.K.)
| | - Ryan Kolb
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (L.W.); (Z.J.); (R.P.M.); (C.K.M.); (M.E.C.); (T.B.A.R.); (M.-C.K.); (R.K.)
- UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
| | - Weizhou Zhang
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (L.W.); (Z.J.); (R.P.M.); (C.K.M.); (M.E.C.); (T.B.A.R.); (M.-C.K.); (R.K.)
- UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
42
|
Breast Cancer Metastasis: Mechanisms and Therapeutic Implications. Int J Mol Sci 2022; 23:ijms23126806. [PMID: 35743249 PMCID: PMC9224686 DOI: 10.3390/ijms23126806] [Citation(s) in RCA: 162] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/15/2022] [Accepted: 06/17/2022] [Indexed: 02/05/2023] Open
Abstract
Breast cancer is the most common malignancy in women worldwide. Metastasis is the leading cause of high mortality in most cancers. Although predicting the early stage of breast cancer before metastasis can increase the survival rate, breast cancer is often discovered or diagnosed after metastasis has occurred. In general, breast cancer has a poor prognosis because it starts as a local disease and can spread to lymph nodes or distant organs, contributing to a significant impediment in breast cancer treatment. Metastatic breast cancer cells acquire aggressive characteristics from the tumor microenvironment (TME) through several mechanisms including epithelial–mesenchymal transition (EMT) and epigenetic regulation. Therefore, understanding the nature and mechanism of breast cancer metastasis can facilitate the development of targeted therapeutics focused on metastasis. This review discusses the mechanisms leading to metastasis and the current therapies to improve the early diagnosis and prognosis in patients with metastatic breast cancer.
Collapse
|
43
|
Gao F, Zhang G, Liu Y, He Y, Sheng Y, Sun X, Du Y, Yang C. Activation of CD44 signaling in leader cells induced by tumor-associated macrophages drives collective detachment in luminal breast carcinomas. Cell Death Dis 2022; 13:540. [PMID: 35680853 PMCID: PMC9184589 DOI: 10.1038/s41419-022-04986-4] [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: 02/04/2022] [Revised: 05/17/2022] [Accepted: 05/30/2022] [Indexed: 01/21/2023]
Abstract
Collective detachment of cancer cells at the invading front could generate efficient metastatic spread. However, how cancer cell clusters shed from the leading front remains unknown. We previously reported that the dynamic expression of CD44 in breast cancers (BrCas) at collectively invading edges was associated with tumor-associated macrophages (TAMs). In this study, we first observed that the highly expressed CD44 (CD44high) cancer cell clusters were located in the BrCa circulating vessels, accompanied by CD206+ TAMs. Next, we identified that the cancer cell clusters can be converted to an invasive CD44high state which was induced by TAMs, thus giving rise to CD44-associated signaling mediated cohesive detachment. Then, we showed that disrupting CD44-signaling inhibited the TAMs triggered cohesive detaching using 3D organotypic culture and mouse models. Furthermore, our mechanistic study showed that the acquisition of CD44high state was mediated by the MDM2/p53 pathway activation which was induced by CCL8 released from TAMs. Blocking of CCL8 could inhibit the signaling cascade which decreased the CD44-mediated cohesive detachment and spread. Our findings uncover a novel mechanism underlying collective metastasis in BrCas that may be helpful to seek for potential targets.
Collapse
Affiliation(s)
- Feng Gao
- grid.412528.80000 0004 1798 5117Department of Clinical Laboratory, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, 200233 China ,grid.412528.80000 0004 1798 5117Department of Molecular Biology Laboratory, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, 200233 China
| | - Guoliang Zhang
- grid.412528.80000 0004 1798 5117Department of Molecular Biology Laboratory, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, 200233 China
| | - Yiwen Liu
- grid.412528.80000 0004 1798 5117Department of Molecular Biology Laboratory, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, 200233 China
| | - Yiqing He
- grid.412528.80000 0004 1798 5117Department of Molecular Biology Laboratory, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, 200233 China
| | - Yumeng Sheng
- grid.412528.80000 0004 1798 5117Department of Clinical Laboratory, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, 200233 China
| | - Xiaodan Sun
- grid.412528.80000 0004 1798 5117Department of Clinical Laboratory, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, 200233 China
| | - Yan Du
- grid.412528.80000 0004 1798 5117Department of Molecular Biology Laboratory, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, 200233 China
| | - Cuixia Yang
- grid.412528.80000 0004 1798 5117Department of Clinical Laboratory, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, 200233 China ,grid.412528.80000 0004 1798 5117Department of Molecular Biology Laboratory, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, 200233 China
| |
Collapse
|
44
|
Tabibzadeh S. Repair, regeneration and rejuvenation require un-entangling pluripotency from senescence. Ageing Res Rev 2022; 80:101663. [PMID: 35690382 DOI: 10.1016/j.arr.2022.101663] [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: 03/07/2022] [Revised: 05/27/2022] [Accepted: 06/04/2022] [Indexed: 11/16/2022]
Abstract
There is a notion that pluripotency and senescence, represent two extremes of life of cells. Pluripotent cells display epigenetic youth, unlimited proliferative capacity and pluripotent differentiating potential whereas cells that reach the Hayflick limit, transit to senescence, undergo permanent inhibition of cell replication and create an aging tissue landscape. However, pluripotency and senescence appear to be intimately linked and are jointly generated in many different contexts such as during embryogenesis or formation of tissue spheroids, in stem cell niches, cancer, or by induction of a pluripotent state (induced pluripotency). Tissue damage and senescence provide signals that are critical to generation of a pluripotent state and, in turn, pluripotency, induces senescence. Thus, it follows, that precisely timed control of senescence is required for harnessing the full benefits of both senescence and its associated pluripotency during tissue regeneration or rejuvenation.
Collapse
Affiliation(s)
- Siamak Tabibzadeh
- Frontiers in Bioscience Research Institute in Aging and Cancer, 16471 Scientific Way, Irvine, CA 92618.
| |
Collapse
|
45
|
Sun M, Chen X, Yang Z. Single cell mass spectrometry studies reveal metabolomic features and potential mechanisms of drug-resistant cancer cell lines. Anal Chim Acta 2022; 1206:339761. [PMID: 35473873 PMCID: PMC9046687 DOI: 10.1016/j.aca.2022.339761] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 03/18/2022] [Accepted: 03/22/2022] [Indexed: 12/28/2022]
Abstract
Irinotecan (Iri) is a key drug to treat metastatic colorectal cancer, but its clinical activity is often limited by de novo and acquired drug resistance. Studying the underlying mechanisms of drug resistance is necessary for developing novel therapeutic strategies. In this study, we used both regular and irinotecan-resistant (Iri-resistant) colorectal cell lines as models, and performed single cell mass spectrometry (SCMS) metabolomics studies combined with analyses from cytotoxicity assay, western blot, flow cytometry, quantitative real-time polymerase chain reaction (qPCR), and reactive oxygen species (ROS). Our SCMS results indicate that Iri-resistant cancer cells possess higher levels of unsaturated lipids compared with the regular cancer cells. In addition, multiple protein biomarkers and their corresponding mRNAs of colon cancer stem cells are overexpressed in Iri-resistance cells. Particularly, stearoyl-CoA desaturase 1 (SCD1) is upregulated with the development of drug resistance in Iri-resistant cells, whereas inhibiting the activity of SCD1 efficiently increase their sensitivity to Iri treatment. In addition, we demonstrated that SCD1 directly regulates the expression of ALDH1A1, which contributes to the cancer stemness and ROS level in Iri-resistant cell lines.
Collapse
|
46
|
Wu M, Zhang X, Zhang W, Chiou YS, Qian W, Liu X, Zhang M, Yan H, Li S, Li T, Han X, Qian P, Liu S, Pan Y, Lobie PE, Zhu T. Cancer stem cell regulated phenotypic plasticity protects metastasized cancer cells from ferroptosis. Nat Commun 2022; 13:1371. [PMID: 35296660 PMCID: PMC8927306 DOI: 10.1038/s41467-022-29018-9] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 02/22/2022] [Indexed: 02/07/2023] Open
Abstract
Cancer cells display phenotypic equilibrium between the stem-like and differentiated states during neoplastic homeostasis. The functional and mechanistic implications of this subpopulation plasticity remain largely unknown. Herein, it is demonstrated that the breast cancer stem cell (BCSC) secretome autonomously compresses the stem cell population. Co-implantation with BCSCs decreases the tumor-initiating capacity yet increases metastasis of accompanying cancer cells, wherein DKK1 is identified as a pivotal factor secreted by BCSCs for such functions. DKK1-promotes differentiation is indispensable for disseminated tumor cell metastatic outgrowth. In contrast, DKK1 inhibitors substantially relieve the metastatic burden by restraining metastatic cells in the dormant state. DKK1 increases the expression of SLC7A11 to protect metastasizing cancer cells from lipid peroxidation and ferroptosis. Combined treatment with a ferroptosis inducer and a DKK1 inhibitor exhibits synergistic effects in diminishing metastasis. Hence, this study deciphers the contribution of CSC-regulated phenotypic plasticity in metastatic colonization and provides therapeutic approaches to limit metastatic outgrowth. The contribution of breast cancer stem cells (BCSCs) to metastasis needs further elucidation. Here, the authors show that BCSCs secrete DKK1 to protect metastasizing cancer cells from ferroptosis via upregulation of SLC7A11, and further show that the combination of a ferroptosis inducer with a DKK1 inhibitor reduces metastasis.
Collapse
Affiliation(s)
- Mingming Wu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.,The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiao Zhang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.,The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Weijie Zhang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.,The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yi Shiou Chiou
- Tsinghua-Berkeley Shenzhen Institute and Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Shenzhen, China.,Master Degree Program in Toxicology, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan.,Shenzhen Bay Laboratory, Shenzhen, China
| | - Wenchang Qian
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.,The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiangtian Liu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.,The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Min Zhang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.,The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Hong Yan
- Department of Pathology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Shilan Li
- Department of Pathology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Tao Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xinghua Han
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Pengxu Qian
- Center for Stem Cell and Regenerative Medicine, Department of Basic Medical Sciences and Institute of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Suling Liu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, Shanghai Medical College, Key Laboratory of Breast Cancer in Shanghai, Innovation Center for Cell Signaling Network, Cancer Institute, Fudan University, Shanghai, China
| | - Yueyin Pan
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Peter E Lobie
- Tsinghua-Berkeley Shenzhen Institute and Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Shenzhen, China. .,Shenzhen Bay Laboratory, Shenzhen, China.
| | - Tao Zhu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China. .,The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
47
|
Xu XL, Huang ZY, Yu K, Li J, Fu XW, Deng SL. Estrogen Biosynthesis and Signal Transduction in Ovarian Disease. Front Endocrinol (Lausanne) 2022; 13:827032. [PMID: 35299973 PMCID: PMC8921451 DOI: 10.3389/fendo.2022.827032] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/03/2022] [Indexed: 12/01/2022] Open
Abstract
Estrogen mainly binds to estrogen receptors (ERs) to regulate menstrual cycles and reproduction. The expression of ERalpha (ERα), ERbeta (ERβ), and G-protein-coupled estrogen receptor (GPER) mRNA could be detected in ovary, suggesting that they play an important role in estrogen signal transduction in ovary. And many studies have revealed that abnormal expression of estrogen and its receptors is closely related to ovarian disease or malignant tumors. With the continuous development and research of animal models, tissue-specific roles of both ERα and ERβ have been demonstrated in animals, which enable people to have a deeper understanding of the potential role of ER in regulating female reproductive diseases. Nevertheless, our current understanding of ERs expression and function in ovarian disease is, however, incomplete. To elucidate the biological mechanism behind ERs in the ovary, this review will focus on the role of ERα and ERβ in polycystic ovary syndrome (PCOS), ovarian cancer and premature ovarian failure (POF) and discuss the major challenges of existing therapies to provide a reference for the treatment of estrogen target tissue ovarian diseases.
Collapse
Affiliation(s)
- Xue-Ling Xu
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zheng-Yuan Huang
- Department of Metabolism, Digestion and Reproduction, Chelsea and Westminster Hospital, Imperial College London, London, United Kingdom
| | - Kun Yu
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jun Li
- Department of Reproductive Medicine, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiang-Wei Fu
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shou-Long Deng
- National Health Commission of China (NHC) Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| |
Collapse
|
48
|
Jing Y, Liang W, Zhang L, Tang J, Huang Z. The Role of Mesenchymal Stem Cells in the Induction of Cancer-Stem Cell Phenotype. Front Oncol 2022; 12:817971. [PMID: 35251985 PMCID: PMC8891610 DOI: 10.3389/fonc.2022.817971] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/19/2022] [Indexed: 11/13/2022] Open
Abstract
Cancer stem cells (CSCs) modify and form their microenvironment by recruiting and activating specific cell types such as mesenchymal stem cells (MSCs). Tumor-infiltrating MSCs help to establish a suitable tumor microenvironment for the restoration of CSCs and tumor progression. In addition, crosstalk between cancer cells and MSCs in the microenvironment induces a CSC phenotype in cancer cells. Many mechanisms are involved in crosstalk between CSCs/cancer cells and MSCs including cell-cell interaction, secretion of exosomes, and paracrine secretion of several molecules including inflammatory mediators, cytokines, and growth factors. Since this crosstalk may contribute to drug resistance, metastasis, and tumor growth, it is suggested that blockade of the crosstalk between MSCs and CSCs/cancer cells can provide a new avenue to improving the cancer therapeutic tools. In this review, we will discuss the role of MSCs in the induction of cancer stem cell phenotype and the restoration of CSCs. We also discuss targeting the crosstalk between MSCs and CSCs/cancer cells as a therapeutic strategy.
Collapse
Affiliation(s)
- Yuanming Jing
- Department of Gastrointestinal Surgery, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Wenqing Liang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Lin Zhang
- Department of Pharmacy, Shaoxing People’s Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, China
| | - Junjun Tang
- Department of Radiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Zongliang Huang, ; Junjun Tang ,
| | - Zongliang Huang
- Department of Radiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Zongliang Huang, ; Junjun Tang ,
| |
Collapse
|
49
|
Aramini B, Masciale V, Grisendi G, Bertolini F, Maur M, Guaitoli G, Chrystel I, Morandi U, Stella F, Dominici M, Haider KH. Dissecting Tumor Growth: The Role of Cancer Stem Cells in Drug Resistance and Recurrence. Cancers (Basel) 2022; 14:cancers14040976. [PMID: 35205721 PMCID: PMC8869911 DOI: 10.3390/cancers14040976] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 02/12/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary Cancer is one of the most debated problems all over the world. Cancer stem cells are considered responsible of tumor initiation, metastasis, drug resistance, and recurrence. This subpopulation of cells has been found into the tumor bulk and showed the capacity to self-renew, differentiate, up to generate a new tumor. In the last decades, several studies have been set on the molecular mechanisms behind their specific characteristics as the Wnt/β-catenin signaling, Notch signaling, Hedgehog signaling, transcription factors, etc. The most powerful part of CSCs is represented by the niches as “promoter” of their self-renewal and “protector” from the common oncological treatment as chemotherapy and radiotherapy. In our review article we highlighted the primary mechanisms involved in CSC tumorigenesis for the setting of further targets to control the metastatic process. Abstract Emerging evidence suggests that a small subpopulation of cancer stem cells (CSCs) is responsible for initiation, progression, and metastasis cascade in tumors. CSCs share characteristics with normal stem cells, i.e., self-renewal and differentiation potential, suggesting that they can drive cancer progression. Consequently, targeting CSCs to prevent tumor growth or regrowth might offer a chance to lead the fight against cancer. CSCs create their niche, a specific area within tissue with a unique microenvironment that sustains their vital functions. Interactions between CSCs and their niches play a critical role in regulating CSCs’ self-renewal and tumorigenesis. Differences observed in the frequency of CSCs, due to the phenotypic plasticity of many cancer cells, remain a challenge in cancer therapeutics, since CSCs can modulate their transcriptional activities into a more stem-like state to protect themselves from destruction. This plasticity represents an essential step for future therapeutic approaches. Regarding self-renewal, CSCs are modulated by the same molecular pathways found in normal stem cells, such as Wnt/β-catenin signaling, Notch signaling, and Hedgehog signaling. Another key characteristic of CSCs is their resistance to standard chemotherapy and radiotherapy treatments, due to their capacity to rest in a quiescent state. This review will analyze the primary mechanisms involved in CSC tumorigenesis, with particular attention to the roles of CSCs in tumor progression in benign and malignant diseases; and will examine future perspectives on the identification of new markers to better control tumorigenesis, as well as dissecting the metastasis process.
Collapse
Affiliation(s)
- Beatrice Aramini
- Division of Thoracic Surgery, Department of Experimental Diagnostic and Specialty Medicine–DIMES of the Alma Mater Studiorum, University of Bologna, G.B. Morgagni-L. Pierantoni Hospital, 47121 Forlì, Italy;
- Thoracic Surgery Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy; (V.M.); (U.M.)
- Correspondence:
| | - Valentina Masciale
- Thoracic Surgery Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy; (V.M.); (U.M.)
| | - Giulia Grisendi
- Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy; (G.G.); (F.B.); (M.M.); (G.G.); (I.C.); (M.D.)
| | - Federica Bertolini
- Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy; (G.G.); (F.B.); (M.M.); (G.G.); (I.C.); (M.D.)
| | - Michela Maur
- Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy; (G.G.); (F.B.); (M.M.); (G.G.); (I.C.); (M.D.)
| | - Giorgia Guaitoli
- Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy; (G.G.); (F.B.); (M.M.); (G.G.); (I.C.); (M.D.)
| | - Isca Chrystel
- Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy; (G.G.); (F.B.); (M.M.); (G.G.); (I.C.); (M.D.)
| | - Uliano Morandi
- Thoracic Surgery Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy; (V.M.); (U.M.)
| | - Franco Stella
- Division of Thoracic Surgery, Department of Experimental Diagnostic and Specialty Medicine–DIMES of the Alma Mater Studiorum, University of Bologna, G.B. Morgagni-L. Pierantoni Hospital, 47121 Forlì, Italy;
| | - Massimo Dominici
- Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy; (G.G.); (F.B.); (M.M.); (G.G.); (I.C.); (M.D.)
| | | |
Collapse
|
50
|
Felcher CM, Bogni ES, Kordon EC. IL-6 Cytokine Family: A Putative Target for Breast Cancer Prevention and Treatment. Int J Mol Sci 2022; 23:ijms23031809. [PMID: 35163731 PMCID: PMC8836921 DOI: 10.3390/ijms23031809] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 12/11/2022] Open
Abstract
The IL-6 cytokine family is a group of signaling molecules with wide expression and function across vertebrates. Each member of the family signals by binding to its specific receptor and at least one molecule of gp130, which is the common transmembrane receptor subunit for the whole group. Signal transduction upon stimulation of the receptor complex results in the activation of multiple downstream cascades, among which, in mammary cells, the JAK-STAT3 pathway plays a central role. In this review, we summarize the role of the IL-6 cytokine family—specifically IL-6 itself, LIF, OSM, and IL-11—as relevant players during breast cancer progression. We have compiled evidence indicating that this group of soluble factors may be used for early and more precise breast cancer diagnosis and to design targeted therapy to treat or even prevent metastasis development, particularly to the bone. Expression profiles and possible therapeutic use of their specific receptors in the different breast cancer subtypes are also described. In addition, participation of these cytokines in pathologies of the breast linked to lactation and involution of the gland, as post-partum breast cancer and mastitis, is discussed.
Collapse
Affiliation(s)
- Carla M. Felcher
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Universidad de Buenos Aires—Consejo Nacional de Investigaciones Científicas y Técnicas (IFIBYNE-UBA-CONICET), Ciudad Autónoma de Buenos Aires (CABA) 1428, Argentina; (C.M.F.); (E.S.B.)
| | - Emilia S. Bogni
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Universidad de Buenos Aires—Consejo Nacional de Investigaciones Científicas y Técnicas (IFIBYNE-UBA-CONICET), Ciudad Autónoma de Buenos Aires (CABA) 1428, Argentina; (C.M.F.); (E.S.B.)
| | - Edith C. Kordon
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Universidad de Buenos Aires—Consejo Nacional de Investigaciones Científicas y Técnicas (IFIBYNE-UBA-CONICET), Ciudad Autónoma de Buenos Aires (CABA) 1428, Argentina; (C.M.F.); (E.S.B.)
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires (CABA) 1428, Argentina
- Correspondence:
| |
Collapse
|