1
|
Schnee M, Sieler M, Dörnen J, Dittmar T. Effects of polystyrene nano- and microplastics on human breast epithelial cells and human breast cancer cells. Heliyon 2024; 10:e38686. [PMID: 39449700 PMCID: PMC11497447 DOI: 10.1016/j.heliyon.2024.e38686] [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: 05/22/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024] Open
Abstract
The continuous littering of the environment with plastic and the resulting nano- and microplastics produced from various processes are ever-increasing problems. These materials also affect humans, as the absorption and accumulation of nano- and microplastics and their effects on health have thus far been rarely researched, which also applies to cancer. In the present study, the absorption of different sizes of polystyrene (PS) nano- and microplastics (PS particles) into human breast epithelial cells and human breast cancer cells was investigated. Subsequently, how the proliferation, colony and mammosphere formation abilities, cell fusion and migration of the cells were influenced by the PS particles were investigated. Our data revealed granularity-, dose- and cell line-dependent absorption of the PS particles, with the highest absorption observed in the MDA-MB-231-DSP1-7 cells and the lowest in the M13SV1_Syn1-DSP8-11 cells. Neither the colony-forming ability nor the cell fusion activity increased with the addition of PS particles. In contrast, slight, partially significant stimulatory effects on both proliferation and cell migration were observed, although these effects depended on the particle quantity and size and the cell line used. In summary, PS particles are absorbed by human breast epithelial and human breast cancer cells and influence cells that may be associated with cancer progression.
Collapse
Affiliation(s)
- Maximilian Schnee
- Institute of Immunology, Center for Biomedical Research and Education (ZBAF), Witten/Herdecke University, Witten, Germany
| | - Mareike Sieler
- Institute of Immunology, Center for Biomedical Research and Education (ZBAF), Witten/Herdecke University, Witten, Germany
| | - Jessica Dörnen
- Institute of Immunology, Center for Biomedical Research and Education (ZBAF), Witten/Herdecke University, Witten, Germany
- Faculty of Medicine, Ruhr University Bochum, Bochum, Germany
| | - Thomas Dittmar
- Institute of Immunology, Center for Biomedical Research and Education (ZBAF), Witten/Herdecke University, Witten, Germany
| |
Collapse
|
2
|
Shultes PV, Weaver DT, Tadele DS, Barker-Clarke RJ, Scott JG. Cell-cell fusion in cancer: The next cancer hallmark? Int J Biochem Cell Biol 2024; 175:106649. [PMID: 39186970 DOI: 10.1016/j.biocel.2024.106649] [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: 06/04/2024] [Revised: 08/13/2024] [Accepted: 08/21/2024] [Indexed: 08/28/2024]
Abstract
In this review, we consider the role of cell-cell fusion in cancer development and progression through an evolutionary lens. We begin by summarizing the origins of fusion proteins (fusogens), of which there are many distinct classes that have evolved through convergent evolution. We then use an evolutionary framework to highlight how the persistence of fusion over generations and across different organisms can be attributed to traits that increase fitness secondary to fusion; these traits map well to the expanded hallmarks of cancer. By studying the tumor microenvironment, we can begin to identify the key selective pressures that may favor higher rates of fusion compared to healthy tissues. The paper concludes by discussing the increasing number of research questions surrounding fusion, recommendations for how to answer them, and the need for a greater interest in exploring cell fusion and evolutionary principles in oncology moving forward.
Collapse
Affiliation(s)
- Paulameena V Shultes
- Translational Hematology and Oncology (THOR), Cleveland Clinic, Cleveland, OH 44120, USA; School of Medicine, Case Western Reserve University, Cleveland, OH 44120, USA
| | - Davis T Weaver
- Translational Hematology and Oncology (THOR), Cleveland Clinic, Cleveland, OH 44120, USA; School of Medicine, Case Western Reserve University, Cleveland, OH 44120, USA
| | - Dagim S Tadele
- Translational Hematology and Oncology (THOR), Cleveland Clinic, Cleveland, OH 44120, USA; Oslo University Hospital, Ullevål, Department of Medical Genetics, Oslo, Norway
| | - Rowan J Barker-Clarke
- Translational Hematology and Oncology (THOR), Cleveland Clinic, Cleveland, OH 44120, USA
| | - Jacob G Scott
- Translational Hematology and Oncology (THOR), Cleveland Clinic, Cleveland, OH 44120, USA; School of Medicine, Case Western Reserve University, Cleveland, OH 44120, USA; Physics Department, Case Western Reserve University, Cleveland, OH 44120, USA.
| |
Collapse
|
3
|
Marcotte E, Goyeneche A, Abdouh M, Burnier JV, Burnier MN. The Phenotypical Characterization of Dual-Nature Hybrid Cells in Uveal Melanoma. Cancers (Basel) 2024; 16:3231. [PMID: 39335202 PMCID: PMC11429545 DOI: 10.3390/cancers16183231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/20/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Metastasis, occurring years after primary diagnosis, represents a poor prognosis in uveal melanoma (UM)-affected individuals. The nature of cells involved in this process is under debate. Circulating hybrid cells that have combined tumor and immune cell features found in blood were predictive of metastasis and may correspond to dual-nature cells (DNC) in the primary tumor. Herein, we sought to determine the presence of DNCs in primary UM tumors, the cell types involved in their genesis, and their ability to be formed in vitro. METHODS UM lesions (n = 38) were immunolabeled with HMB45 in combination with immune-cell-specific antibodies. In parallel, we co-cultured UM cells and peripheral blood mononuclear cells (PBMCs) to analyze DNC formation. RESULTS HMB45+/CD45+ DNCs were present in 90% (26/29) of the tumors, HMB45+/CD8+ DNCs were present in 93% (26/28), and HMB45+/CD68+ DNCs were present in 71% (17/24). DNCs formed with CD8+ and CD68+ cells were positively correlated to the infiltration of their respective immune cells. Notably, UM cells were prone to hybridize with PBMCs in vitro. CONCLUSIONS This phenotypical characterization of DNCs in UM demonstrates that CD8+ T-cells and macrophages are capable of DNC formation, and they are important for better understanding metastatic dissemination, thus paving the path towards novel therapeutic avenues.
Collapse
Affiliation(s)
- Emily Marcotte
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- The MUHC-McGill University Ocular Pathology & Translational Research Laboratory, McGill University, Montreal, QC H4A 3J1, Canada
| | - Alicia Goyeneche
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- The MUHC-McGill University Ocular Pathology & Translational Research Laboratory, McGill University, Montreal, QC H4A 3J1, Canada
| | - Mohamed Abdouh
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- The MUHC-McGill University Ocular Pathology & Translational Research Laboratory, McGill University, Montreal, QC H4A 3J1, Canada
| | - Julia Valdemarin Burnier
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- Experimental Pathology Unit, Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC H3A 3T2, Canada
| | - Miguel Noel Burnier
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- The MUHC-McGill University Ocular Pathology & Translational Research Laboratory, McGill University, Montreal, QC H4A 3J1, Canada
| |
Collapse
|
4
|
Marabitti V, Vulpis E, Nazio F, Campello S. Mitochondrial Transfer as a Strategy for Enhancing Cancer Cell Fitness:Current Insights and Future Directions. Pharmacol Res 2024; 208:107382. [PMID: 39218420 DOI: 10.1016/j.phrs.2024.107382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/08/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
It is now recognized that tumors are not merely masses of transformed cells but are intricately interconnected with healthy cells in the tumor microenvironment (TME), forming complex and heterogeneous structures. Recent studies discovered that cancer cells can steal mitochondria from healthy cells to empower themselves, while reducing the functions of their target organ. Mitochondrial transfer, i.e. the intercellular movement of mitochondria, is recently emerging as a novel process in cancer biology, contributing to tumor growth, metastasis, and resistance to therapy by shaping the metabolic landscape of the tumor microenvironment. This review highlights the influence of transferred mitochondria on cancer bioenergetics, redox balance and apoptotic resistance, which collectively foster aggressive cancer phenotype. Furthermore, the therapeutic implications of mitochondrial transfer are discussed, emphasizing the potential of targeting these pathways to overcome drug resistance and improve treatment efficacy.
Collapse
Affiliation(s)
- Veronica Marabitti
- Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy
| | - Elisabetta Vulpis
- Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy
| | - Francesca Nazio
- Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy
| | - Silvia Campello
- Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy.
| |
Collapse
|
5
|
Anderson AN, Conley P, Klocke CD, Sengupta SK, Pang A, Farley HC, Gillingham AR, Dawson AD, Fan Y, Jones JA, Gibbs SL, Skalet AH, Wu G, Wong MH. Detection of neoplastic-immune hybrid cells with metastatic properties in uveal melanoma. Biomark Res 2024; 12:67. [PMID: 39030653 PMCID: PMC11264923 DOI: 10.1186/s40364-024-00609-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/18/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND Uveal melanoma is the most common non-cutaneous melanoma and is an intraocular malignancy affecting nearly 7,000 individuals per year worldwide. Of these, approximately 50% will progress to metastatic disease for which there are currently no effective curative therapies. Despite advances in molecular profiling and metastatic stratification of uveal melanoma tumors, little is known regarding their underlying biology of metastasis. Our group has identified a disseminated neoplastic cell population characterized by co-expression of immune and melanoma proteins, circulating hybrid cells (hybrids), in patients with uveal melanoma. Compared to circulating tumor cells, which lack expression of immune proteins, hybrids are detected at an increased prevalence in peripheral blood and can be used as a non-invasive biomarker to predict metastatic progression. METHODS To ascertain mechanisms underlying enhanced hybrid cell dissemination we identified hybrid cells within primary uveal melanoma tumors using single cell RNA sequencing (n = 8) and evaluated their gene expression and predicted ligand-receptor interactions in relation to other melanoma and immune cells within the primary tumor. We then verified expression of upregulated hybrid pathways within patient-matched tumor and peripheral blood hybrids (n = 4) using cyclic immunofluorescence and quantified their protein expression relative to other non-hybrid tumor and disseminated tumor cells. RESULTS Among the top upregulated genes and pathways in hybrid cells were those involved in enhanced cell motility and cytoskeletal rearrangement, immune evasion, and altered cellular metabolism. In patient-matched tumor and peripheral blood, we verified gene expression by examining concordant protein expression for each pathway category: TMSB10 (cell motility), CD74 (immune evasion) and GPX1 (metabolism). Both TMSB10 and GPX1 were expressed on significantly higher numbers of disseminated hybrid cells compared to circulating tumor cells, and CD74 and GPX1 were expressed on more disseminated hybrids than tumor-resident hybrids. Lastly, we identified that hybrid cells express ligand-receptor signaling pathways implicated in promoting metastasis including GAS6-AXL, CXCL12-CXCR4, LGALS9-P4HB and IGF1-IGFR1. CONCLUSION These findings highlight the importance of TMSB10, GPX1 and CD74 for successful hybrid cell dissemination and survival in circulation. Our results contribute to the understanding of uveal melanoma tumor progression and interactions between tumor cells and immune cells in the tumor microenvironment that may promote metastasis.
Collapse
Affiliation(s)
- Ashley N Anderson
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Patrick Conley
- Department of Medical Informatics and Clinical Epidemiology, OHSU, Portland, OR, USA
| | - Christopher D Klocke
- Department of Medical Informatics and Clinical Epidemiology, OHSU, Portland, OR, USA
| | - Sidharth K Sengupta
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Amara Pang
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Hannah C Farley
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA
- Department of Biomedical Engineering, OHSU, Portland, OR, USA
| | - Abigail R Gillingham
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Aubrey D Dawson
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Yichen Fan
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Jocelyn A Jones
- Department of Biomedical Engineering, OHSU, Portland, OR, USA
| | - Summer L Gibbs
- Department of Biomedical Engineering, OHSU, Portland, OR, USA
- Knight Cancer Institute, OHSU, Portland, OR, USA
| | - Alison H Skalet
- Casey Eye Institute, OHSU, Portland, OR, USA
- Knight Cancer Institute, OHSU, Portland, OR, USA
| | - Guanming Wu
- Department of Medical Informatics and Clinical Epidemiology, OHSU, Portland, OR, USA
- Knight Cancer Institute, OHSU, Portland, OR, USA
| | - Melissa H Wong
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA.
- Knight Cancer Institute, OHSU, Portland, OR, USA.
| |
Collapse
|
6
|
Lu Y, Chen D, Wang B, Chai W, Yan M, Chen Y, Zhan Y, Yang R, Zhou E, Dai S, Li Y, Dong R, Zheng B. Single-cell landscape of undifferentiated pleomorphic sarcoma. Oncogene 2024; 43:1353-1368. [PMID: 38459120 DOI: 10.1038/s41388-024-03001-8] [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: 08/24/2023] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/10/2024]
Abstract
Undifferentiated pleomorphic sarcoma (UPS) is a highly aggressive malignant soft tissue tumor with a poor prognosis; however, the identity and heterogeneity of tumor populations remain elusive. Here, eight major cell clusters were identified through the RNA sequencing of 79,569 individual cells of UPS. UPS originates from mesenchymal stem cells (MSCs) and features undifferentiated subclusters. UPS subclusters were predicted to exist in two bulk RNA datasets, and had a prognostic value in The Cancer Genome Atlas (TCGA) dataset. The functional heterogeneity of malignant UPS cells and the immune microenvironment were characterized. Additionally, the fused cells were innovatively detected by expressing both monocyte/macrophage markers and other subcluster-associated genes. Based on the ligand-receptor interaction analysis, cellular interactions with epidermal growth factor receptor (EGFR) and vascular endothelial growth factor receptor (VEGFR) were abundant. Furthermore, 73% of patients with UPS (48/66) showed positive EGFR expression, which was associated with a poor prognosis. EGFR blockade with cetuximab inhibited tumor growth in a patient-derived xenograft model. Our transcriptomic studies delineate the landscape of UPS intratumor heterogeneity and serve as a foundational resource for further discovery and therapeutic exploration.
Collapse
Affiliation(s)
- Yifei Lu
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, 201102, China
| | - Deqian Chen
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, 201102, China
| | - Bingnan Wang
- Department of Musculoskeletal Oncology, Shanghai Cancer Center, and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Wenjun Chai
- Department of Animal Experimental Center, Fudan University Shanghai Cancer Center, Shanghai, 201102, China
| | - Mingxia Yan
- Department of Animal Experimental Center, Fudan University Shanghai Cancer Center, Shanghai, 201102, China
| | - Yong Chen
- Department of Musculoskeletal Oncology, Shanghai Cancer Center, and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yong Zhan
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, 201102, China
| | - Ran Yang
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, 201102, China
| | - Enqing Zhou
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, 201102, China
| | - Shuyang Dai
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, 201102, China
| | - Yi Li
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, 201102, China
| | - Rui Dong
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Birth Defects, Shanghai, 201102, China.
| | - Biqiang Zheng
- Department of Musculoskeletal Oncology, Shanghai Cancer Center, and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
7
|
Ali AM, Raza A. scRNAseq and High-Throughput Spatial Analysis of Tumor and Normal Microenvironment in Solid Tumors Reveal a Possible Origin of Circulating Tumor Hybrid Cells. Cancers (Basel) 2024; 16:1444. [PMID: 38611120 PMCID: PMC11010995 DOI: 10.3390/cancers16071444] [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: 03/01/2024] [Revised: 03/31/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
Metastatic cancer is a leading cause of death in cancer patients worldwide. While circulating hybrid cells (CHCs) are implicated in metastatic spread, studies documenting their tissue origin remain sparse, with limited candidate approaches using one-two markers. Utilizing high-throughput single-cell and spatial transcriptomics, we identified tumor hybrid cells (THCs) co-expressing epithelial and macrophage markers and expressing a distinct transcriptome. Rarely, normal tissue showed these cells (NHCs), but their transcriptome was easily distinguishable from THCs. THCs with unique transcriptomes were observed in breast and colon cancers, suggesting this to be a generalizable phenomenon across cancer types. This study establishes a framework for HC identification in large datasets, providing compelling evidence for their tissue residence and offering comprehensive transcriptomic characterization. Furthermore, it sheds light on their differential function and identifies pathways that could explain their newly acquired invasive capabilities. THCs should be considered as potential therapeutic targets.
Collapse
Affiliation(s)
- Abdullah Mahmood Ali
- Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Edward P Evans MDS Center, Herbert Irving Comprehensive Cancer Center, New York, NY 10032, USA
| | - Azra Raza
- Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Edward P Evans MDS Center, Herbert Irving Comprehensive Cancer Center, New York, NY 10032, USA
| |
Collapse
|
8
|
Wang R, Hu P, Wang F, Lyu J, Ou Y, Edderkaoui M, Zhang Y, Lewis MS, Pandol SJ, Zhau HE, Chung LWK. Spontaneous Fusion with Transformed Mesenchymal Stromal Cells Results in Complete Heterogeneity in Prostate Cancer Cells. Cancers (Basel) 2024; 16:951. [PMID: 38473313 PMCID: PMC10931070 DOI: 10.3390/cancers16050951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 02/16/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Tumor cells gain advantages in growth and survival by acquiring genotypic and phenotypic heterogeneity. Interactions with bystander cells in the tumor microenvironment contribute to the progression of heterogeneity. We have shown that fusion between tumor and bystander cells is one form of interaction, and that tumor-bystander cell fusion has contrasting effects. By trapping fusion hybrids in the heterokaryon or synkaryon state, tumor-bystander cell fusion prevents the progression of heterogeneity. However, if trapping fails, fusion hybrids will resume replication to form derivative clones with diverse genomic makeups and behavioral phenotypes. To determine the characteristics of bystander cells that influence the fate of fusion hybrids, we co-cultured prostate mesenchymal stromal cell lines and their spontaneously transformed sublines with LNCaP as well as HPE-15 prostate cancer cells. Subclones derived from cancer-stromal fusion hybrids were examined for genotypic and phenotypic diversifications. Both stromal cell lines were capable of fusing with cancer cells, but only fusion hybrids with the transformed stromal subline generated large numbers of derivative subclones. Each subclone had distinct cell morphologies and growth behaviors and was detected with complete genomic hybridization. The health conditions of the bystander cell compartment play a crucial role in the progression of tumor cell heterogeneity.
Collapse
Affiliation(s)
- Ruoxiang Wang
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (P.H.); (F.W.); (J.L.); (M.E.); (S.J.P.); (H.E.Z.); (L.W.K.C.)
| | - Peizhen Hu
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (P.H.); (F.W.); (J.L.); (M.E.); (S.J.P.); (H.E.Z.); (L.W.K.C.)
| | - Fubo Wang
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (P.H.); (F.W.); (J.L.); (M.E.); (S.J.P.); (H.E.Z.); (L.W.K.C.)
| | - Ji Lyu
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (P.H.); (F.W.); (J.L.); (M.E.); (S.J.P.); (H.E.Z.); (L.W.K.C.)
| | - Yan Ou
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (Y.O.); (Y.Z.)
| | - Mouad Edderkaoui
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (P.H.); (F.W.); (J.L.); (M.E.); (S.J.P.); (H.E.Z.); (L.W.K.C.)
| | - Yi Zhang
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (Y.O.); (Y.Z.)
| | - Michael S. Lewis
- Department of Medicine and Pathology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
| | - Stephen J. Pandol
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (P.H.); (F.W.); (J.L.); (M.E.); (S.J.P.); (H.E.Z.); (L.W.K.C.)
| | - Haiyen E. Zhau
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (P.H.); (F.W.); (J.L.); (M.E.); (S.J.P.); (H.E.Z.); (L.W.K.C.)
| | - Leland W. K. Chung
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (P.H.); (F.W.); (J.L.); (M.E.); (S.J.P.); (H.E.Z.); (L.W.K.C.)
| |
Collapse
|
9
|
Han D, He X, Huang Y, Gao M, Guo T, Ren X, Liao X, Chen X, Pang X, Cheng S. A Multifunctional Delivery System for Remodulating Cell Behaviors of Circulating Malignant Cells to Prevent Cell Fusion. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303309. [PMID: 37590231 PMCID: PMC10582411 DOI: 10.1002/advs.202303309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/30/2023] [Indexed: 08/19/2023]
Abstract
Cell fusion plays a critical role in cancer progression and metastasis. However, effective modulation of the cell fusion behavior and timely evaluation on the cell fusion to provide accurate information for personalized therapy are facing challenges. Here, it demonstrates that the cancer cell fusion behavior can be efficiently modulated and precisely detected through employing a multifunctional delivery vector to realize cancer targeting delivery of a genome editing plasmid and a molecular beacon-based AND logic gate. The multifunctional delivery vector decorated by AS1411 conjugated hyaluronic acid and NLS-GE11 peptide conjugated hyaluronic acid can specifically target circulating malignant cells (CMCs) of cancer patients to deliver the genome editing plasmid for epidermal growth factor receptor (EGFR) knockout. The cell fusion between CMCs and endothelial cells can be detected by the AND logic gate delivered by the multifunctional vector. After EGFR knockout, the edited CMCs exhibit dramatically inhibited cell fusion capability, while unedited CMCs can easily fuse with human umbilical vein endothelial cells (HUVEC) to form hybrid cells. This study provides a new therapeutic strategy for preventing cancer progression and a reliable tool for evaluating cancer cell fusion for precise personalized therapy.
Collapse
Affiliation(s)
- Di Han
- Key Laboratory of Biomedical Polymers of Ministry of EducationDepartment of ChemistryWuhan UniversityWuhanHubei430072China
| | - Xiao‐Yan He
- School of Life SciencesAnhui Medical UniversityHefeiAnhui230011China
| | - Yun Huang
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Anhui Medical UniversityAnhui Public Health Clinical CenterHefeiAnhui230011China
| | - Min Gao
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Anhui Medical UniversityAnhui Public Health Clinical CenterHefeiAnhui230011China
| | - Tao Guo
- Department of Thyroid and Breast SurgeryThe First Affiliated Hospital of Anhui Medical UniversityAnhui Public Health Clinical CenterHefeiAnhui230011China
| | - Xiao‐He Ren
- Key Laboratory of Biomedical Polymers of Ministry of EducationDepartment of ChemistryWuhan UniversityWuhanHubei430072China
| | - Xin‐Ru Liao
- Key Laboratory of Biomedical Polymers of Ministry of EducationDepartment of ChemistryWuhan UniversityWuhanHubei430072China
| | - Xue‐Si Chen
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchunJilin130022China
| | - Xuan Pang
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchunJilin130022China
| | - Si‐Xue Cheng
- Key Laboratory of Biomedical Polymers of Ministry of EducationDepartment of ChemistryWuhan UniversityWuhanHubei430072China
| |
Collapse
|
10
|
Dittmar T, Sieler M, Hass R. Why do certain cancer cells alter functionality and fuse? Biol Chem 2023; 404:951-960. [PMID: 37246410 DOI: 10.1515/hsz-2023-0162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/11/2023] [Indexed: 05/30/2023]
Abstract
Cancer cell fusion represents a rare event. However, the surviving cancer hybrid cells after a post-hybrid selection process (PHSP) can overgrow other cancer cells by exhibiting a proliferation advantage and/or expression of cancer stem-like properties. Addition of new tumor properties during hetero-fusion of cancer cells e.g. with mesenchymal stroma-/stem-like cells (MSC) contribute to enhanced tumor plasticity via acquisition of new/altered functionalities. This provides new avenues for tumor development and metastatic behavior. Consequently, the present review article will also address the question as to whether cancer cell fusion represents a general and possibly evolutionary-conserved program or rather a random process?
Collapse
Affiliation(s)
- Thomas Dittmar
- Institute of Immunology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, Stockumer Str. 10, D-58448 Witten, Germany
| | - Mareike Sieler
- Institute of Immunology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, Stockumer Str. 10, D-58448 Witten, Germany
| | - Ralf Hass
- Department of Obstetrics and Gynecology, Biochemistry and Tumor Biology Laboratory, Hannover Medical School, D-30625 Hannover, Germany
| |
Collapse
|
11
|
Cozzo AJ, Coleman MF, Hursting SD. You complete me: tumor cell-myeloid cell nuclear fusion as a facilitator of organ-specific metastasis. Front Oncol 2023; 13:1191332. [PMID: 37427108 PMCID: PMC10324515 DOI: 10.3389/fonc.2023.1191332] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 05/25/2023] [Indexed: 07/11/2023] Open
Abstract
Every cancer genome is unique, resulting in potentially near infinite cancer cell phenotypes and an inability to predict clinical outcomes in most cases. Despite this profound genomic heterogeneity, many cancer types and subtypes display a non-random distribution of metastasis to distant organs, a phenomenon known as organotropism. Proposed factors in metastatic organotropism include hematogenous versus lymphatic dissemination, the circulation pattern of the tissue of origin, tumor-intrinsic factors, compatibility with established organ-specific niches, long-range induction of premetastatic niche formation, and so-called "prometastatic niches" that facilitate successful colonization of the secondary site following extravasation. To successfully complete the steps required for distant metastasis, cancer cells must evade immunosurveillance and survive in multiple new and hostile environments. Despite substantial advances in our understanding of the biology underlying malignancy, many of the mechanisms used by cancer cells to survive the metastatic journey remain a mystery. This review synthesizes the rapidly growing body of literature demonstrating the relevance of an unusual cell type known as "fusion hybrid" cells to many of the hallmarks of cancer, including tumor heterogeneity, metastatic conversion, survival in circulation, and metastatic organotropism. Whereas the concept of fusion between tumor cells and blood cells was initially proposed over a century ago, only recently have technological advancements allowed for detection of cells containing components of both immune and neoplastic cells within primary and metastatic lesions as well as among circulating malignant cells. Specifically, heterotypic fusion of cancer cells with monocytes and macrophages results in a highly heterogeneous population of hybrid daughter cells with enhanced malignant potential. Proposed mechanisms behind these findings include rapid, massive genome rearrangement during nuclear fusion and/or acquisition of monocyte/macrophage features such as migratory and invasive capability, immune privilege, immune cell trafficking and homing, and others. Rapid acquisition of these cellular traits may increase the likelihood of both escape from the primary tumor site and extravasation of hybrid cells at a secondary location that is amenable to colonization by that particular hybrid phenotype, providing a partial explanation for the patterns observed in some cancers with regard to sites of distant metastases.
Collapse
Affiliation(s)
- Alyssa J. Cozzo
- Duke University School of Medicine, Durham, NC, United States
- Department of Pathology, Duke University Medical Center, Durham, NC, United States
| | - Michael F. Coleman
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Stephen D. Hursting
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, United States
| |
Collapse
|
12
|
Ye X, Huang X, Fu X, Zhang X, Lin R, Zhang W, Zhang J, Lu Y. Myeloid-like tumor hybrid cells in bone marrow promote progression of prostate cancer bone metastasis. J Hematol Oncol 2023; 16:46. [PMID: 37138326 PMCID: PMC10155318 DOI: 10.1186/s13045-023-01442-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 04/19/2023] [Indexed: 05/05/2023] Open
Abstract
BACKGROUND Bone metastasis is the leading cause of death in patients with prostate cancer (PCa) and currently has no effective treatment. Disseminated tumor cells in bone marrow often obtain new characteristics to cause therapy resistance and tumor recurrence. Thus, understanding the status of disseminated prostate cancer cells in bone marrow is crucial for developing a new treatment. METHODS We analyzed the transcriptome of disseminated tumor cells from a single cell RNA-sequencing data of PCa bone metastases. We built a bone metastasis model through caudal artery injection of tumor cells, and sorted the tumor hybrid cells by flow cytometry. We performed multi-omics analysis, including transcriptomic, proteomic and phosphoproteomic analysis, to compare the difference between the tumor hybrid cells and parental cells. In vivo experiments were performed to analyze the tumor growth rate, metastatic and tumorigenic potential, drug and radiation sensitivity in hybrid cells. Single cell RNA-sequencing and CyTOF were performed to analyze the impact of hybrid cells on tumor microenvironment. RESULTS Here, we identified a unique cluster of cancer cells in PCa bone metastases, which expressed myeloid cell markers and showed a significant change in pathways related to immune regulation and tumor progression. We found that cell fusion between disseminated tumor cells and bone marrow cells can be source of these myeloid-like tumor cells. Multi-omics showed the pathways related to cell adhesion and proliferation, such as focal adhesion, tight junction, DNA replication, and cell cycle, were most significantly changed in these hybrid cells. In vivo experiment showed hybrid cells had a significantly increased proliferative rate, and metastatic potential. Single cell RNA-sequencing and CyTOF showed tumor-associated neutrophils/monocytes/macrophages were highly enriched in hybrid cells-induced tumor microenvironment with a higher immunosuppressive capacity. Otherwise, the hybrid cells showed an enhanced EMT phenotype with higher tumorigenicity, and were resistant to docetaxel and ferroptosis, but sensitive to radiotherapy. CONCLUSION Taken together, our data demonstrate that spontaneous cell fusion in bone marrow can generate myeloid-like tumor hybrid cells that promote the progression of bone metastasis, and these unique population of disseminated tumor cells can provide a potential therapeutic target for PCa bone metastasis.
Collapse
Affiliation(s)
- Xinyu Ye
- School of Medicine, Southern University of Science and Technology, No. 1088 Xue Yuan Blvd, Shenzhen, 518055, Guangdong, China
| | - Xin Huang
- School of Medicine, Southern University of Science and Technology, No. 1088 Xue Yuan Blvd, Shenzhen, 518055, Guangdong, China
| | - Xing Fu
- School of Medicine, Southern University of Science and Technology, No. 1088 Xue Yuan Blvd, Shenzhen, 518055, Guangdong, China
| | - Xiao Zhang
- School of Medicine, Southern University of Science and Technology, No. 1088 Xue Yuan Blvd, Shenzhen, 518055, Guangdong, China
| | - Risheng Lin
- School of Medicine, Southern University of Science and Technology, No. 1088 Xue Yuan Blvd, Shenzhen, 518055, Guangdong, China
| | - Wen Zhang
- School of Medicine, Southern University of Science and Technology, No. 1088 Xue Yuan Blvd, Shenzhen, 518055, Guangdong, China
| | - Jian Zhang
- School of Medicine, Southern University of Science and Technology, No. 1088 Xue Yuan Blvd, Shenzhen, 518055, Guangdong, China.
| | - Yi Lu
- School of Medicine, Southern University of Science and Technology, No. 1088 Xue Yuan Blvd, Shenzhen, 518055, Guangdong, China.
| |
Collapse
|
13
|
Warrier NM, Kelkar N, Johnson CT, Govindarajan T, Prabhu V, Kumar P. Understanding cancer stem cells and plasticity: Towards better therapeutics. Eur J Cell Biol 2023; 102:151321. [PMID: 37137199 DOI: 10.1016/j.ejcb.2023.151321] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 05/05/2023] Open
Abstract
The ability of cancer cells to finally overcome various lines of treatment in due course has always baffled the scientific community. Even with the most promising therapies, relapse is ultimately seen, and this resilience has proved to be a major hurdle in the management of cancer. Accumulating evidence now attributes this resilience to plasticity. Plasticity is the ability of cells to change their properties and is substantial as it helps in normal tissue regeneration or post-injury repair processes. It also helps in the overall maintenance of homeostasis. Unfortunately, this critical ability of cells, when activated incorrectly, can lead to numerous diseases, including cancer. Therefore, in this review, we focus on the plasticity aspect with an emphasis on cancer stem cells (CSCs). We discuss the various forms of plasticity that provide survival advantages to CSCs. Moreover, we explore various factors that affect plasticity. Furthermore, we provide the therapeutic implications of plasticity. Finally, we provide an insight into the future targeted therapies involving plasticity for better clinical outcomes.
Collapse
Affiliation(s)
- Neerada Meenakshi Warrier
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Nachiket Kelkar
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Carol Tresa Johnson
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | | | - Vijendra Prabhu
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| | - Praveen Kumar
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| |
Collapse
|
14
|
Okeyo KO, Hiyaji R, Oana H. A single-cell surgery microfluidic device for transplanting tumor cytoplasm into dendritic cells without nuclei mixing. Biotechnol J 2023; 18:e2200135. [PMID: 36412930 DOI: 10.1002/biot.202200135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 11/12/2022] [Accepted: 11/14/2022] [Indexed: 11/23/2022]
Abstract
This study aimed to demonstrate the feasibility of generating tumor cell vaccine models by single-cell surgery in a microfluidic device that integrates one-to-one electrofusion, shear flow reseparation, and on-device culture. The device was microfabricated from polydimethylsiloxane (PDMS) and consisted of microorifices (aperture size: ∼3 μm) for one-to-one fusion, and microcages for on-device culture. Using the device, we could achieve one-to-one electrofusion of leukemic plasmacytoid dendritic cells (DC-like cells) and Jurkat cells with a fusion efficiency of ∼ 80%. Fusion via the narrow microorifices allowed DC-like cells to acquire cytoplasmic contents of the Jurkat cells while preventing nuclei mixing. After fusion, the DC-like cells were selectively reseparated from the Jurkat cells by shear flow application to generate tumor nuclei-free antigen-recipient DC-like (tarDC-like) cells. When cultured as single cells on the device, these cells could survive under gentle medium perfusion with a median survival time of 11.5 h, although a few cells could survive longer than 36 h. Overall, this study demonstrates single-cell surgery in a microfluidic device for potential generation of dendritic cell vaccines which are uncontaminated with tumor nucleic materials. We believe that this study will inspire the generation of safer tumor cell vaccines for cancer immunotherapy.
Collapse
Affiliation(s)
- Kennedy Omondi Okeyo
- Institute for Life and Medical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Ryuta Hiyaji
- Department of Mechanical Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hidehiro Oana
- Department of Mechanical Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
15
|
Dai X, Shao Y, Tian X, Cao X, Ye L, Gao P, Cheng H, Wang X. Fusion between Glioma Stem Cells and Mesenchymal Stem Cells Promotes Malignant Progression in 3D-Bioprinted Models. ACS APPLIED MATERIALS & INTERFACES 2022; 14:35344-35356. [PMID: 35881920 DOI: 10.1021/acsami.2c06658] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The interaction between glioma stem cells (GSCs) and mesenchymal stem cells (MSCs) in the glioma microenvironment is considered to be an important factor in promoting tumor progression, but the mechanism is still not fully elucidated. To further elucidate the interaction between GSCs and MSCs, two 3D-bioprinted tumor models (low-temperature molding and coaxial bioprinting) were used to simulate the tumor growth microenvironment. Cell fusion between GSCs and MSCs was found by the method of Cre-LoxP switch gene and RFP/GFP dual-color fluorescence tracing. The fused cells coexpressed biomarkers of GSCs and MSCs, showing stronger proliferation, cloning, and invasion abilities than GSCs and MSCs. In addition, the fused cells have stronger tumorigenic properties in nude mice, showing the pathological features of malignant tumors. In conclusion, GSCs and MSCs undergo cell fusion in 3D-bioprinted models, and the fused cells have a higher degree of malignancy than parental cells, which promotes the progression of glioma.
Collapse
Affiliation(s)
- Xingliang Dai
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, P. R. China
| | - Yuxuan Shao
- Department of Clinical Medicine, the First Clinical College of Anhui Medical University, Hefei 230032, P. R. China
| | - Xuefeng Tian
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
| | - Xiaoyan Cao
- Department of Clinical Medicine, the First Clinical College of Anhui Medical University, Hefei 230032, P. R. China
| | - Lei Ye
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
| | - Peng Gao
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
| | - Hongwei Cheng
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
| | - Xianwen Wang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, P. R. China
| |
Collapse
|
16
|
Zhao J, Ruan J, Lv G, Shan Q, Fan Z, Wang H, Du Y, Ling L. Cell membrane-based biomimetic nanosystems for advanced drug delivery in cancer therapy: A comprehensive review. Colloids Surf B Biointerfaces 2022; 215:112503. [PMID: 35429736 DOI: 10.1016/j.colsurfb.2022.112503] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/08/2022] [Accepted: 04/08/2022] [Indexed: 12/30/2022]
Abstract
Natural types of cells display distinct characteristics with homotypic targeting and extended circulation in the blood, which are worthy of being explored as promising drug delivery systems (DDSs) for cancer therapy. To enhance their delivery efficiency, these cells can be combined with therapeutic agents and artificial nanocarriers to construct the next generation of DDSs in the form of biomimetic nanomedicines. In this review, we present the recent advances in cell membrane-based DDSs (CDDSs) and their applications for efficient cancer therapy. Different sources of cell membranes are discussed, mainly including red blood cells (RBC), leukocytes, cancer cells, stem cells and hybrid cells. Moreover, the extraction methods used for obtaining such cells and the mechanism contributing to the functional action of these biomimetic CDDSs are explained. Finally, a future perspective is proposed to highlight the limitations of CDDSs and the possible resolutions toward clinical transformation of currently developed biomimetic chemotherapies.
Collapse
Affiliation(s)
- Jianing Zhao
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Ministry of Education of China), School of Pharmacy, Yantai University, Yantai 264005, China
| | - Jian Ruan
- Yantai Center for Food and Drug Control, Yantai 264005, China
| | - Guangyao Lv
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Ministry of Education of China), School of Pharmacy, Yantai University, Yantai 264005, China
| | - Qi Shan
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Ministry of Education of China), School of Pharmacy, Yantai University, Yantai 264005, China
| | - Zhiping Fan
- Institute of BioPharmaceutical Research, Liaocheng University, Liaocheng 252059, China
| | - Hongbo Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Ministry of Education of China), School of Pharmacy, Yantai University, Yantai 264005, China.
| | - Yuan Du
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Ministry of Education of China), School of Pharmacy, Yantai University, Yantai 264005, China.
| | - Longbing Ling
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Ministry of Education of China), School of Pharmacy, Yantai University, Yantai 264005, China.
| |
Collapse
|
17
|
Monocyte-Macrophage Lineage Cell Fusion. Int J Mol Sci 2022; 23:ijms23126553. [PMID: 35742997 PMCID: PMC9223484 DOI: 10.3390/ijms23126553] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/07/2022] [Accepted: 06/10/2022] [Indexed: 02/06/2023] Open
Abstract
Cell fusion (fusogenesis) occurs in natural and pathological conditions in prokaryotes and eukaryotes. Cells of monocyte–macrophage lineage are highly fusogenic. They create syncytial multinucleated giant cells (MGCs) such as osteoclasts (OCs), MGCs associated with the areas of infection/inflammation, and foreign body-induced giant cells (FBGCs). The fusion of monocytes/macrophages with tumor cells may promote cancer metastasis. We describe types and examples of monocyte–macrophage lineage cell fusion and the role of actin-based structures in cell fusion.
Collapse
|
18
|
Ibragimova M, Tsyganov M, Litviakov N. Tumour Stem Cells in Breast Cancer. Int J Mol Sci 2022; 23:ijms23095058. [PMID: 35563449 PMCID: PMC9099719 DOI: 10.3390/ijms23095058] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/27/2022] [Accepted: 04/30/2022] [Indexed: 12/12/2022] Open
Abstract
Tumour stem cells (CSCs) are a self-renewing population that plays important roles in tumour initiation, recurrence, and metastasis. Although the medical literature is extensive, problems with CSC identification and cancer therapy remain. This review provides the main mechanisms of CSC action in breast cancer (BC): CSC markers and signalling pathways, heterogeneity, plasticity, and ecological behaviour. The dynamic heterogeneity of CSCs and the dynamic transitions of CSC− non-CSCs and their significance for metastasis are considered.
Collapse
Affiliation(s)
- Marina Ibragimova
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 5, Kooperativny Street, 634050 Tomsk, Russia; (M.T.); (N.L.)
- Laboratory of Genetic Technologies, Siberian State Medical University, 2, Moscow Tract, 634050 Tomsk, Russia
- Biological Institute, National Research Tomsk State University, 36, Lenin, 634050 Tomsk, Russia
- Correspondence:
| | - Matvey Tsyganov
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 5, Kooperativny Street, 634050 Tomsk, Russia; (M.T.); (N.L.)
| | - Nikolai Litviakov
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 5, Kooperativny Street, 634050 Tomsk, Russia; (M.T.); (N.L.)
- Laboratory of Genetic Technologies, Siberian State Medical University, 2, Moscow Tract, 634050 Tomsk, Russia
- Biological Institute, National Research Tomsk State University, 36, Lenin, 634050 Tomsk, Russia
| |
Collapse
|
19
|
Generation of Cancer Stem/Initiating Cells by Cell-Cell Fusion. Int J Mol Sci 2022; 23:ijms23094514. [PMID: 35562905 PMCID: PMC9101717 DOI: 10.3390/ijms23094514] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/10/2022] [Accepted: 04/17/2022] [Indexed: 02/04/2023] Open
Abstract
CS/ICs have raised great expectations in cancer research and therapy, as eradication of this key cancer cell type is expected to lead to a complete cure. Unfortunately, the biology of CS/ICs is rather complex, since no common CS/IC marker has yet been identified. Certain surface markers or ALDH1 expression can be used for detection, but some studies indicated that cancer cells exhibit a certain plasticity, so CS/ICs can also arise from non-CS/ICs. Another problem is intratumoral heterogeneity, from which it can be inferred that different CS/IC subclones must be present in the tumor. Cell–cell fusion between cancer cells and normal cells, such as macrophages and stem cells, has been associated with the generation of tumor hybrids that can exhibit novel properties, such as an enhanced metastatic capacity and even CS/IC properties. Moreover, cell–cell fusion is a complex process in which parental chromosomes are mixed and randomly distributed among daughter cells, resulting in multiple, unique tumor hybrids. These, if they have CS/IC properties, may contribute to the heterogeneity of the CS/IC pool. In this review, we will discuss whether cell–cell fusion could also lead to the origin of different CS/ICs that may expand the overall CS/IC pool in a primary tumor.
Collapse
|
20
|
Kaigorodova EV, Kozik AV, Zavaruev IS, Grishchenko MY. Hybrid/Atypical Forms of Circulating Tumor Cells: Current State of the Art. BIOCHEMISTRY (MOSCOW) 2022; 87:380-390. [PMID: 35527376 PMCID: PMC8993035 DOI: 10.1134/s0006297922040071] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Cancer is one of the most common diseases worldwide, and its treatment is associated with many challenges such as drug and radioresistance and formation of metastases. These difficulties are due to tumor heterogeneity, which has many causes. One may be the cell fusion, a process that is relevant to both physiological (e.g., wound healing) and pathophysiological (cancer and viral infection) processes. This literature review aimed to summarize the existing data on the hybrid/atypical forms of circulating cancer cells and their role in tumor progression. For that, the bioinformatics search in universal databases, such as PubMed, NCBI, and Google Scholar was conducted by using the keywords “hybrid cancer cells”, “cancer cell fusion”, etc. In this review the latest information related to the hybrid tumor cells, theories of their genesis, characteristics of different variants with data from our own researches are presented. Many aspects of the hybrid cell research are still in their infancy. However, with the level of knowledge already accumulated, circulating hybrids such as CAML and CHC could be considered as promising biomarkers of cancerous tumors, and even more as a new approach to cancer treatment.
Collapse
Affiliation(s)
- Evgeniya V Kaigorodova
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634050, Russia.
- Siberian State Medical University, Tomsk, 634050, Russia
| | - Alexey V Kozik
- Siberian State Medical University, Tomsk, 634050, Russia
| | | | | |
Collapse
|
21
|
Tretyakova MS, Subbalakshmi AR, Menyailo ME, Jolly MK, Denisov EV. Tumor Hybrid Cells: Nature and Biological Significance. Front Cell Dev Biol 2022; 10:814714. [PMID: 35242760 PMCID: PMC8886020 DOI: 10.3389/fcell.2022.814714] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 01/25/2022] [Indexed: 11/13/2022] Open
Abstract
Metastasis is the leading cause of cancer death and can be realized through the phenomenon of tumor cell fusion. The fusion of tumor cells with other tumor or normal cells leads to the appearance of tumor hybrid cells (THCs) exhibiting novel properties such as increased proliferation and migration, drug resistance, decreased apoptosis rate, and avoiding immune surveillance. Experimental studies showed the association of THCs with a high frequency of cancer metastasis; however, the underlying mechanisms remain unclear. Many other questions also remain to be answered: the role of genetic alterations in tumor cell fusion, the molecular landscape of cells after fusion, the lifetime and fate of different THCs, and the specific markers of THCs, and their correlation with various cancers and clinicopathological parameters. In this review, we discuss the factors and potential mechanisms involved in the occurrence of THCs, the types of THCs, and their role in cancer drug resistance and metastasis, as well as potential therapeutic approaches for the prevention, and targeting of tumor cell fusion. In conclusion, we emphasize the current knowledge gaps in the biology of THCs that should be addressed to develop highly effective therapeutics and strategies for metastasis suppression.
Collapse
Affiliation(s)
- Maria S Tretyakova
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Ayalur R Subbalakshmi
- Cancer Systems Biology Laboratory, Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru, India
| | - Maxim E Menyailo
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Mohit Kumar Jolly
- Cancer Systems Biology Laboratory, Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru, India
| | - Evgeny V Denisov
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| |
Collapse
|
22
|
Lazebnik Y. Cell fusion as a link between the SARS-CoV-2 spike protein, COVID-19 complications, and vaccine side effects. Oncotarget 2021; 12:2476-2488. [PMID: 34917266 PMCID: PMC8664391 DOI: 10.18632/oncotarget.28088] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 09/24/2021] [Indexed: 12/23/2022] Open
Abstract
A distinctive feature of the SARS-CoV-2 spike protein is its ability to efficiently fuse cells, thus producing syncytia found in COVID-19 patients. This commentary proposes how this ability enables spike to cause COVID-19 complications as well as side effects of COVID-19 vaccines, and suggests how these effects can be prevented.
Collapse
|
23
|
Dittmar T, Weiler J, Luo T, Hass R. Cell-Cell Fusion Mediated by Viruses and HERV-Derived Fusogens in Cancer Initiation and Progression. Cancers (Basel) 2021; 13:5363. [PMID: 34771528 PMCID: PMC8582398 DOI: 10.3390/cancers13215363] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/22/2021] [Accepted: 10/24/2021] [Indexed: 12/13/2022] Open
Abstract
Cell fusion is a well-known, but still scarcely understood biological phenomenon, which might play a role in cancer initiation, progression and formation of metastases. Although the merging of two (cancer) cells appears simple, the entire process is highly complex, energy-dependent and tightly regulated. Among cell fusion-inducing and -regulating factors, so-called fusogens have been identified as a specific type of proteins that are indispensable for overcoming fusion-associated energetic barriers and final merging of plasma membranes. About 8% of the human genome is of retroviral origin and some well-known fusogens, such as syncytin-1, are expressed by human (cancer) cells. Likewise, enveloped viruses can enable and facilitate cell fusion due to evolutionarily optimized fusogens, and are also capable to induce bi- and multinucleation underlining their fusion capacity. Moreover, multinucleated giant cancer cells have been found in tumors derived from oncogenic viruses. Accordingly, a potential correlation between viruses and fusogens of human endogenous retroviral origin in cancer cell fusion will be summarized in this review.
Collapse
Affiliation(s)
- Thomas Dittmar
- Institute of Immunology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, 58448 Witten, Germany;
| | - Julian Weiler
- Institute of Immunology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, 58448 Witten, Germany;
| | - Tianjiao Luo
- Biochemistry and Tumor Biology Laboratory, Department of Obstetrics and Gynecology, Hannover Medical School, 30625 Hannover, Germany;
| | - Ralf Hass
- Biochemistry and Tumor Biology Laboratory, Department of Obstetrics and Gynecology, Hannover Medical School, 30625 Hannover, Germany;
| |
Collapse
|