1
|
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
|
2
|
Niu N, Ye J, Hu Z, Zhang J, Wang Y. Regulative Roles of Metabolic Plasticity Caused by Mitochondrial Oxidative Phosphorylation and Glycolysis on the Initiation and Progression of Tumorigenesis. Int J Mol Sci 2023; 24:ijms24087076. [PMID: 37108242 PMCID: PMC10139088 DOI: 10.3390/ijms24087076] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/23/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
One important feature of tumour development is the regulatory role of metabolic plasticity in maintaining the balance of mitochondrial oxidative phosphorylation and glycolysis in cancer cells. In recent years, the transition and/or function of metabolic phenotypes between mitochondrial oxidative phosphorylation and glycolysis in tumour cells have been extensively studied. In this review, we aimed to elucidate the characteristics of metabolic plasticity (emphasizing their effects, such as immune escape, angiogenesis migration, invasiveness, heterogeneity, adhesion, and phenotypic properties of cancers, among others) on tumour progression, including the initiation and progression phases. Thus, this article provides an overall understanding of the influence of abnormal metabolic remodeling on malignant proliferation and pathophysiological changes in carcinoma.
Collapse
Affiliation(s)
- Nan Niu
- Shenzhen Engineering Labortaory for Marine Algal Biotechnology, Longhua Innovation Institute for Biotechnology, College of Life Sciences and Oceanography, Lihu Campus of Shenzhen University, Shenzhen 518055, China
- College of Physics and Optoelectronic Engineering, Canghai Campus of Shenzhen University, Shenzhen 518060, China
| | - Jinfeng Ye
- Shenzhen Engineering Labortaory for Marine Algal Biotechnology, Longhua Innovation Institute for Biotechnology, College of Life Sciences and Oceanography, Lihu Campus of Shenzhen University, Shenzhen 518055, China
| | - Zhangli Hu
- Shenzhen Engineering Labortaory for Marine Algal Biotechnology, Longhua Innovation Institute for Biotechnology, College of Life Sciences and Oceanography, Lihu Campus of Shenzhen University, Shenzhen 518055, China
| | - Junbin Zhang
- Shenzhen Engineering Labortaory for Marine Algal Biotechnology, Longhua Innovation Institute for Biotechnology, College of Life Sciences and Oceanography, Lihu Campus of Shenzhen University, Shenzhen 518055, China
| | - Yun Wang
- Shenzhen Engineering Labortaory for Marine Algal Biotechnology, Longhua Innovation Institute for Biotechnology, College of Life Sciences and Oceanography, Lihu Campus of Shenzhen University, Shenzhen 518055, China
| |
Collapse
|
3
|
Eid RA, Alaa Edeen M, Shedid EM, Kamal ASS, Warda MM, Mamdouh F, Khedr SA, Soltan MA, Jeon HW, Zaki MSA, Kim B. Targeting Cancer Stem Cells as the Key Driver of Carcinogenesis and Therapeutic Resistance. Int J Mol Sci 2023; 24:ijms24021786. [PMID: 36675306 PMCID: PMC9861138 DOI: 10.3390/ijms24021786] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 01/18/2023] Open
Abstract
The emerging concept of cancer stem cells (CSCs) as the key driver behind carcinogenesis, progression, and diversity has displaced the prior model of a tumor composed of cells with similar subsequently acquired mutations and an equivalent capacity for renewal, invasion, and metastasis. This significant change has shifted the research focus toward targeting CSCs to eradicate cancer. CSCs may be characterized using cell surface markers. They are defined by their capacity to self-renew and differentiate, resist conventional therapies, and generate new tumors following repeated transplantation in xenografted mice. CSCs' functional capabilities are governed by various intracellular and extracellular variables such as pluripotency-related transcription factors, internal signaling pathways, and external stimuli. Numerous natural compounds and synthetic chemicals have been investigated for their ability to disrupt these regulatory components and inhibit stemness and terminal differentiation in CSCs, hence achieving clinical implications. However, no cancer treatment focuses on the biological consequences of these drugs on CSCs, and their functions have been established. This article provides a biomedical discussion of cancer at the time along with an overview of CSCs and their origin, features, characterization, isolation techniques, signaling pathways, and novel targeted therapeutic approaches. Additionally, we highlighted the factors endorsed as controlling or helping to promote stemness in CSCs. Our objective was to encourage future studies on these prospective treatments to develop a framework for their application as single or combined therapeutics to eradicate various forms of cancer.
Collapse
Affiliation(s)
- Refaat A. Eid
- Pathology Department, College of Medicine, King Khalid University, Abha P.O. Box 62529, Saudi Arabia
| | - Muhammad Alaa Edeen
- Cell Biology, Histology & Genetics Division, Biology Department, Faculty of Science, Zagazig University, Zagazig 44519, Egypt
- Correspondence: (M.A.E.); (B.K.)
| | - Eslam M. Shedid
- Biotechnology Division, Zoology Department, Faculty of Science, Benha University, Al Qalyubia Governorate, Banha 13511, Egypt
| | - Al Shaimaa S. Kamal
- Biotechnology Department, Faculty of Agriculture, Benha University, Al Qalyubia Governorate, Banha 13511, Egypt
| | - Mona M. Warda
- Biotechnology Division, Zoology Department, Faculty of Science, Benha University, Al Qalyubia Governorate, Banha 13511, Egypt
| | - Farag Mamdouh
- Biotechnology Division, Zoology Department, Faculty of Science, Benha University, Al Qalyubia Governorate, Banha 13511, Egypt
| | - Sohila A. Khedr
- Industrial Biotechnology Department, Faculty of Science, Tanta University, Tanta 31733, Egypt
| | - Mohamed A. Soltan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Sinai University, Ismailia 41611, Egypt
| | - Hee Won Jeon
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Mohamed Samir A. Zaki
- Anatomy Department, College of Medicine, King Khalid University, Abha P.O. Box 62529, Saudi Arabia
- Department of Histology and Cell Biology, College of Medicine, Zagazig University, Zagazig 31527, Egypt
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Correspondence: (M.A.E.); (B.K.)
| |
Collapse
|
4
|
Mohamed HT, Kamel G, El-Husseiny N, El-Sharkawy AA, El-Sherif AA, El-Shinawi M, Mohamed MM. Synchrotron Fourier-Transform Infrared Microspectroscopy: Characterization of in vitro polarized tumor-associated macrophages stimulated by the secretome of inflammatory and non-inflammatory breast cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119367. [PMID: 36202317 DOI: 10.1016/j.bbamcr.2022.119367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/13/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022]
Abstract
Studies suggested that the pathogenesis of inflammatory breast cancer (IBC) is related to inflammatory manifestations accompanied by specific cellular and molecular mechanisms in the IBC tumor microenvironment (TME). IBC is characterized by significantly higher infiltration of tumor-associated macrophages (TAMs) that contribute to its metastatic process via secreting many cytokines such as TNF, IL-6, IL-8, and IL-10 that enhance invasion and angiogenesis. Thus, there is a need to first understand how IBC-TME modulates the polarization of TAMs to better understand the role of TAMs in IBC. Herein, we used gene expression signature and Synchrotron Fourier-Transform Infrared Microspectroscopy (SR-μFTIR) to study the molecular and biochemical changes, respectively of in vitro polarized TAMs stimulated by the secretome of IBC and non-IBC cells. The gene expression signature showed significant differences in the macrophage's polarization-related genes between stimulated TAMs. FTIR spectra showed absorption bands in the region of 1700-1500 cm-1 attributed to the amide I ν(C=O), & νAS (CN), δ (NH), and amide II ν(CN), δ (NH) proteins bands. Moreover, three peaks of different intensities and areas were detected in the lipid region of the νCH2 and νCH3 stretching modes positioned within the 3000-2800 cm-1 range. The PCA analysis for the second derivative spectra of the amide regions discriminates between stimulated IBC and non-IBC TAMs. This study showed that IBC and non-IBC TMEs differentially modulate the polarization of TAMs and SR-μFTIR can determine these biochemical changes which will help to better understand the potential role of TAMs in IBC.
Collapse
Affiliation(s)
- Hossam Taha Mohamed
- Zoology Department, Faculty of Science, Cairo University, Giza 12613, Egypt; Faculty of Biotechnology, October University for Modern Sciences and Arts, Giza 12451, Egypt.
| | - Gihan Kamel
- Synchrotron-light for Experimental Science and Applications in the Middle East (SESAME), Allan, Jordan; Department of Physics, Faculty of Science, Helwan University, Cairo 11795, Egypt
| | - Noura El-Husseiny
- Zoology Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| | | | - Ahmed A El-Sherif
- Chemistry Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Mohamed El-Shinawi
- Department of General Surgery, Faculty of Medicine, Ain Shams University, Cairo, 11566, Egypt; Faculty of Medicine, Galala University, Suez 43511, Egypt
| | - Mona Mostafa Mohamed
- Zoology Department, Faculty of Science, Cairo University, Giza 12613, Egypt; Faculty of Science, Galala University, Suez 43511, Egypt
| |
Collapse
|
5
|
Characteristic of Ultrastructure of Mice B16 Melanoma Cells under the Influence of Different Lighting Regimes. Clocks Sleep 2022; 4:745-760. [PMID: 36547107 PMCID: PMC9777458 DOI: 10.3390/clockssleep4040056] [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: 10/26/2022] [Revised: 11/15/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Circadian rhythms of physiological processes, constantly being in a state of dynamic equilibrium and plastically associated with changes in environmental conditions, are the basis of homeostasis of an organism of human and other mammals. Violation of circadian rhythms due to significant disturbances in parameters of main environmental effectors (desynchronosis) leads to the development of pathological conditions and a more severe course of preexisting pathologies. We conducted the study of the ultrastructure of cells of mice transplantable malignant melanoma B16 under the condition of normal (fixed) lighting regime and under the influence of constant lighting. Results of the study show that melanoma B16 under fixed light regime represents a characteristic picture of this tumor-predominantly intact tissue with safe junctions of large, functionally active cells with highly irregular nuclei, developed organelles and a relatively low content of melanin. The picture of the B16 melanoma tissue structure and the ultrastructure of its cells under the action of constant lighting stand in marked contrast to the group with fixed light: under these conditions the tumor exhibits accelerated growth, a significant number of cells in the state of apoptosis and necrosis, ultrastructural signs of degradation of the structure and functions, and signs of embryonization of cells with the background of adaptation to oxygen deficiency.
Collapse
|
6
|
Mohamed HT, El-Sharkawy AA, El-Shinawi M, Schneider RJ, Mohamed MM. Inflammatory Breast Cancer: The Secretome of HCMV+ Tumor-Associated Macrophages Enhances Proliferation, Invasion, Colony Formation, and Expression of Cancer Stem Cell Markers. Front Oncol 2022; 12:899622. [PMID: 35847899 PMCID: PMC9281473 DOI: 10.3389/fonc.2022.899622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 05/25/2022] [Indexed: 11/13/2022] Open
Abstract
Inflammatory breast cancer (IBC) is a highly aggressive phenotype of breast cancer that is characterized by a high incidence early metastasis. We previously reported a significant association of human cytomegalovirus (HCMV) DNA in the carcinoma tissues of IBC patients but not in the adjacent normal tissues. HCMV-infected macrophages serve as “mobile vectors” for spreading and disseminating virus to different organs, and IBC cancer tissues are highly infiltrated by tumor-associated macrophages (TAMs) that enhance IBC progression and promote breast cancer stem cell (BCSC)-like properties. Therefore, there is a need to understand the role of HCMV-infected TAMs in IBC progression. The present study aimed to test the effect of the secretome (cytokines and secreted factors) of TAMs derived from HCMV+ monocytes isolated from IBC specimens on the proliferation, invasion, and BCSC abundance when tested on the IBC cell line SUM149. HCMV+ monocytes were isolated from IBC patients during modified radical mastectomy surgery and tested in vitro for polarization into TAMs using the secretome of SUM149 cells. MTT, clonogenic, invasion, real-time PCR arrays, PathScan Intracellular Signaling array, and cytokine arrays were used to characterize the secretome of HCMV+ TAMs for their effect on the progression of SUM149 cells. The results showed that the secretome of HCMV+ TAMs expressed high levels of IL-6, IL-8, and MCP-1 cytokines compared to HCMV- TAMs. In addition, the secretome of HCMV+ TAMs induced the proliferation, invasion, colony formation, and expression of BCSC-related genes in SUM149 cells compared to mock untreated cells. In addition, the secretome of HCMV+ TAMs activated the phosphorylation of intracellular signaling molecules p-STAT3, p-AMPKα, p-PRAS40, and p-SAPK/JNK in SUM149 cells. In conclusion, this study shows that the secretome of HCMV+ TAMs enhances the proliferation, invasion, colony formation, and BCSC properties by activating the phosphorylation of p-STAT3, p-AMPKα, p-PRAS40, and p-SAPK/JNK intracellular signaling molecules in IBC cells.
Collapse
Affiliation(s)
- Hossam Taha Mohamed
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt
- Faculty of Biotechnology, October University for Modern Sciences and Arts, Giza, Egypt
- *Correspondence: Hossam Taha Mohamed,
| | | | - Mohamed El-Shinawi
- Department of General Surgery, Faculty of Medicine, Ain Shams University, Cairo, Egypt
- Galala University, Suez, Egypt
| | - Robert J. Schneider
- Department of Microbiology, School of Medicine, New York University, New York, NY, United States
| | - Mona Mostafa Mohamed
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt
- Sector of International Cooperation, Galala University, Suez, Egypt
| |
Collapse
|
7
|
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
|
8
|
Ali S, Borin TF, Piranlioglu R, Ara R, Lebedyeva I, Angara K, Achyut BR, Arbab AS, Rashid MH. Changes in the tumor microenvironment and outcome for TME-targeting therapy in glioblastoma: A pilot study. PLoS One 2021; 16:e0246646. [PMID: 33544755 PMCID: PMC7864405 DOI: 10.1371/journal.pone.0246646] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) is a hypervascular and aggressive primary malignant tumor of the central nervous system. Recent investigations showed that traditional therapies along with antiangiogenic therapies failed due to the development of post-therapy resistance and recurrence. Previous investigations showed that there were changes in the cellular and metabolic compositions in the tumor microenvironment (TME). It can be said that tumor cell-directed therapies are ineffective and rethinking is needed how to treat GBM. It is hypothesized that the composition of TME-associated cells will be different based on the therapy and therapeutic agents, and TME-targeting therapy will be better to decrease recurrence and improve survival. Therefore, the purpose of this study is to determine the changes in the TME in respect of T-cell population, M1 and M2 macrophage polarization status, and MDSC population following different treatments in a syngeneic model of GBM. In addition to these parameters, tumor growth and survival were also studied following different treatments. The results showed that changes in the TME-associated cells were dependent on the therapeutic agents, and the TME-targeting therapy improved the survival of the GBM bearing animals. The current GBM therapies should be revisited to add agents to prevent the accumulation of bone marrow-derived cells in the TME or to prevent the effect of immune-suppressive myeloid cells in causing alternative neovascularization, the revival of glioma stem cells, and recurrence. Instead of concurrent therapy, a sequential strategy would be better to target TME-associated cells.
Collapse
Affiliation(s)
- Sehar Ali
- Laboratory of Tumor Angiogenesis Initiative, Georgia Cancer Center, Augusta University, Augusta, Georgia, United States of America
| | - Thaiz F. Borin
- Laboratory of Tumor Angiogenesis Initiative, Georgia Cancer Center, Augusta University, Augusta, Georgia, United States of America
| | - Raziye Piranlioglu
- Laboratory of Tumor Angiogenesis Initiative, Georgia Cancer Center, Augusta University, Augusta, Georgia, United States of America
| | - Roxan Ara
- Laboratory of Tumor Angiogenesis Initiative, Georgia Cancer Center, Augusta University, Augusta, Georgia, United States of America
| | - Iryna Lebedyeva
- Department of Chemistry and Physics, Augusta University, Augusta, Georgia, United States of America
| | - Kartik Angara
- Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, Michigan, United States of America
| | - Bhagelu R. Achyut
- Winship Cancer Institute, Emory University, Atlanta, Georgia, United States of America
| | - Ali Syed Arbab
- Laboratory of Tumor Angiogenesis Initiative, Georgia Cancer Center, Augusta University, Augusta, Georgia, United States of America
- * E-mail: (ASA); (MHR)
| | - Mohammad H. Rashid
- Laboratory of Tumor Angiogenesis Initiative, Georgia Cancer Center, Augusta University, Augusta, Georgia, United States of America
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- * E-mail: (ASA); (MHR)
| |
Collapse
|
9
|
Abstract
ATP is required for mammalian cells to remain viable and to perform genetically programmed functions. Maintenance of the ΔG′ATP hydrolysis of −56 kJ/mole is the endpoint of both genetic and metabolic processes required for life. Various anomalies in mitochondrial structure and function prevent maximal ATP synthesis through OxPhos in cancer cells. Little ATP synthesis would occur through glycolysis in cancer cells that express the dimeric form of pyruvate kinase M2. Mitochondrial substrate level phosphorylation (mSLP) in the glutamine-driven glutaminolysis pathway, substantiated by the succinate-CoA ligase reaction in the TCA cycle, can partially compensate for reduced ATP synthesis through both OxPhos and glycolysis. A protracted insufficiency of OxPhos coupled with elevated glycolysis and an auxiliary, fully operational mSLP, would cause a cell to enter its default state of unbridled proliferation with consequent dedifferentiation and apoptotic resistance, i.e., cancer. The simultaneous restriction of glucose and glutamine offers a therapeutic strategy for managing cancer.
Collapse
Affiliation(s)
- Thomas N Seyfried
- Biology Department, Boston College, 140 Commonwealth Avenue, Chestnut Hill, MA 02467, USA
| | - Gabriel Arismendi-Morillo
- Electron Microscopy Laboratory, Biological Researches Institute, Faculty of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Purna Mukherjee
- Biology Department, Boston College, 140 Commonwealth Avenue, Chestnut Hill, MA 02467, USA
| | - Christos Chinopoulos
- Department of Medical Biochemistry, Semmelweis University, Budapest, 1094, Hungary
| |
Collapse
|
10
|
Zhang LN, Zhang DD, Yang L, Gu YX, Zuo QP, Wang HY, Xu J, Liu DX. Roles of cell fusion between mesenchymal stromal/stem cells and malignant cells in tumor growth and metastasis. FEBS J 2020; 288:1447-1456. [PMID: 33070450 DOI: 10.1111/febs.15483] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/21/2020] [Accepted: 07/08/2020] [Indexed: 01/02/2023]
Abstract
Invasion and metastasis are the basic characteristics and important markers of malignant tumors, which are also the main cause of death in cancer patients. Epithelial-mesenchymal transition (EMT) is recognized as the first step of tumor invasion and metastasis. Many studies have demonstrated that cell fusion is a common phenomenon and plays a critical role in cancer development and progression. At present, cancer stem cell fusion has been considered as a new mechanism of cancer metastasis. Mesenchymal stromal/stem cell (MSC) is a kind of adult stem cells with high self-renewal ability and multidifferentiation potential, which is used as a very promising fusogenic candidate in the tumor microenvironment and has a crucial role in cancer progression. Many research results have shown that MSCs are involved in the regulation of tumor growth and metastasis through cell fusion. However, the role of cell fusion between MSCs and malignant cells in tumor growth and metastasis is still controversial. Several studies have demonstrated that MSCs can enhance malignant characteristics, promoting tumor growth and metastasis by fusing with malignant cells, while other conflicting reports believe that MSCs can reduce tumorigenicity upon fusion with malignant cells. In this review, we summarize the recent research on cell fusion events between MSCs and malignant cells in tumor growth and metastasis. The elucidation of the molecular mechanisms between MSC fusion and tumor metastasis may provide an effective strategy for tumor biotherapy.
Collapse
Affiliation(s)
- Li-Na Zhang
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China
| | - Di-Di Zhang
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China
| | - Lei Yang
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China
| | - Yu-Xuan Gu
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China
| | - Qiu-Ping Zuo
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China
| | - Hao-Yi Wang
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China
| | - Jia Xu
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China
| | - Dian-Xin Liu
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China
| |
Collapse
|
11
|
Seyfried TN, Mukherjee P, Iyikesici MS, Slocum A, Kalamian M, Spinosa JP, Chinopoulos C. Consideration of Ketogenic Metabolic Therapy as a Complementary or Alternative Approach for Managing Breast Cancer. Front Nutr 2020; 7:21. [PMID: 32219096 PMCID: PMC7078107 DOI: 10.3389/fnut.2020.00021] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 02/21/2020] [Indexed: 12/14/2022] Open
Abstract
Breast cancer remains as a significant cause of morbidity and mortality in women. Ultrastructural and biochemical evidence from breast biopsy tissue and cancer cells shows mitochondrial abnormalities that are incompatible with energy production through oxidative phosphorylation (OxPhos). Consequently, breast cancer, like most cancers, will become more reliant on substrate level phosphorylation (fermentation) than on oxidative phosphorylation (OxPhos) for growth consistent with the mitochondrial metabolic theory of cancer. Glucose and glutamine are the prime fermentable fuels that underlie therapy resistance and drive breast cancer growth through substrate level phosphorylation (SLP) in both the cytoplasm (Warburg effect) and the mitochondria (Q-effect), respectively. Emerging evidence indicates that ketogenic metabolic therapy (KMT) can reduce glucose availability to tumor cells while simultaneously elevating ketone bodies, a non-fermentable metabolic fuel. It is suggested that KMT would be most effective when used together with glutamine targeting. Information is reviewed for suggesting how KMT could reduce systemic inflammation and target tumor cells without causing damage to normal cells. Implementation of KMT in the clinic could improve progression free and overall survival for patients with breast cancer.
Collapse
Affiliation(s)
| | - Purna Mukherjee
- Biology Department, Boston College, Chestnut Hill, MA, United States
| | - Mehmet S. Iyikesici
- Medical Oncology, Kemerburgaz University Bahcelievler Medical Park Hospital, Istanbul, Turkey
| | - Abdul Slocum
- Medical Oncology, Chemo Thermia Oncology Center, Istanbul, Turkey
| | | | | | | |
Collapse
|
12
|
Manjunath Y, Porciani D, Mitchem JB, Suvilesh KN, Avella DM, Kimchi ET, Staveley-O’Carroll KF, Burke DH, Li G, Kaifi JT. Tumor-Cell-Macrophage Fusion Cells as Liquid Biomarkers and Tumor Enhancers in Cancer. Int J Mol Sci 2020; 21:E1872. [PMID: 32182935 PMCID: PMC7084898 DOI: 10.3390/ijms21051872] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 03/06/2020] [Accepted: 03/07/2020] [Indexed: 02/06/2023] Open
Abstract
Although molecular mechanisms driving tumor progression have been extensively studied, the biological nature of the various populations of circulating tumor cells (CTCs) within the blood is still not well understood. Tumor cell fusion with immune cells is a longstanding hypothesis that has caught more attention in recent times. Specifically, fusion of tumor cells with macrophages might lead to the development of metastasis by acquiring features such as genetic and epigenetic heterogeneity, chemotherapeutic resistance, and immune tolerance. In addition to the traditional FDA-approved definition of a CTC (CD45-, EpCAM+, cytokeratins 8+, 18+ or 19+, with a DAPI+ nucleus), an additional circulating cell population has been identified as being potential fusions cells, characterized by distinct, large, polymorphonuclear cancer-associated cells with a dual epithelial and macrophage/myeloid phenotype. Artificial fusion of tumor cells with macrophages leads to migratory, invasive, and metastatic phenotypes. Further studies might investigate whether these have a potential impact on the immune response towards the cancer. In this review, the background, evidence, and potential relevance of tumor cell fusions with macrophages is discussed, along with the potential role of intercellular connections in their formation. Such fusion cells could be a key component in cancer metastasis, and therefore, evolve as a diagnostic and therapeutic target in cancer precision medicine.
Collapse
Affiliation(s)
- Yariswamy Manjunath
- Department of Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA; (Y.M.); (J.B.M.); (K.N.S.); (D.M.A.); (E.T.K.); (K.F.S.-O.); (G.L.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
| | - David Porciani
- Department of Molecular Microbiology & Immunology, University of Missouri, Columbia, MO 65212, USA; (D.P.); (D.H.B.)
- Bond Life Sciences Center, University of Missouri, Columbia, MO 65212, USA
| | - Jonathan B. Mitchem
- Department of Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA; (Y.M.); (J.B.M.); (K.N.S.); (D.M.A.); (E.T.K.); (K.F.S.-O.); (G.L.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
| | - Kanve N. Suvilesh
- Department of Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA; (Y.M.); (J.B.M.); (K.N.S.); (D.M.A.); (E.T.K.); (K.F.S.-O.); (G.L.)
| | - Diego M. Avella
- Department of Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA; (Y.M.); (J.B.M.); (K.N.S.); (D.M.A.); (E.T.K.); (K.F.S.-O.); (G.L.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
| | - Eric T. Kimchi
- Department of Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA; (Y.M.); (J.B.M.); (K.N.S.); (D.M.A.); (E.T.K.); (K.F.S.-O.); (G.L.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
| | - Kevin F. Staveley-O’Carroll
- Department of Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA; (Y.M.); (J.B.M.); (K.N.S.); (D.M.A.); (E.T.K.); (K.F.S.-O.); (G.L.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
| | - Donald H. Burke
- Department of Molecular Microbiology & Immunology, University of Missouri, Columbia, MO 65212, USA; (D.P.); (D.H.B.)
- Bond Life Sciences Center, University of Missouri, Columbia, MO 65212, USA
- Department of Biochemistry, University of Missouri, Columbia, MO 65212, USA
| | - Guangfu Li
- Department of Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA; (Y.M.); (J.B.M.); (K.N.S.); (D.M.A.); (E.T.K.); (K.F.S.-O.); (G.L.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
- Department of Molecular Microbiology & Immunology, University of Missouri, Columbia, MO 65212, USA; (D.P.); (D.H.B.)
| | - Jussuf T. Kaifi
- Department of Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA; (Y.M.); (J.B.M.); (K.N.S.); (D.M.A.); (E.T.K.); (K.F.S.-O.); (G.L.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
| |
Collapse
|
13
|
Fusion of macrophages promotes breast cancer cell proliferation, migration and invasion through activating epithelial-mesenchymal transition and Wnt/β-catenin signaling pathway. Arch Biochem Biophys 2019; 676:108137. [PMID: 31605677 DOI: 10.1016/j.abb.2019.108137] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 09/02/2019] [Accepted: 10/08/2019] [Indexed: 12/13/2022]
Abstract
Cell fusion is a highly regulated process involved in cancer development, tissue regeneration and other physiological and pathological events. Many studies have shown that cancer cells can fuse with different types of cells such as mesenchymal stem cells (MSCs) and macrophages, which are behaved as two important fusogenic candidates in the tumor microenvironment. However, the underlying mechanisms of cell fusion between macrophages and malignant cells in cancer progression has not been fully clarified. The aim of the present study was to investigate the effects and mechanisms of cell fusion between macrophages and breast cancer cells on tumorigenesis and metastasis. Our results indicated that the hybrids exhibited enhanced proliferation, colony formation, migration and invasion capabilities, as well as suppressed apoptosis compared with parental breast cancer cells. Moreover, the hybrid cells displayed EMT with a significant downregulation of E-cadherin and upregulation of N-cadherin, Vimentin and Snail, as well as an obviously increased expression of MMP-2, MMP-9, uPA and S100A4. Mechanistically, we found that the TCF/LEF transcription factor activity of Wnt/β-catenin pathway and the expression of its downstream target genes including cyclin D1 and c-Myc were increased in the hybrid cells. Furthermore, our data confirmed that the promoting effects of fusion of macrophages on breast cancer cell proliferation, migration and invasion could be blocked by treatment with XAV-939, a Wnt/β-catenin signaling pathway inhibitor. In conclusion, our findings demonstrate that fusion of macrophages promotes proliferation, migration and invasion of breast cancer cells through activating EMT and Wnt/β-catenin signaling pathway. Our current study will further contribute to elucidate the mechanism of cell fusion in tumorigenesis and metastasis, and to develop a new therapeutic strategy for breast cancer treatment.
Collapse
|
14
|
Cancer cell fusion: a potential target to tackle drug-resistant and metastatic cancer cells. Drug Discov Today 2019; 24:1836-1844. [PMID: 31163272 DOI: 10.1016/j.drudis.2019.05.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/22/2019] [Accepted: 05/28/2019] [Indexed: 12/27/2022]
Abstract
Cell fusion is an integral, established phenomenon underlying various physiological processes in the cell cycle. Although research in cancer metastasis has hypothesised numerous molecular mechanisms and signalling pathways responsible for invasion and metastasis, the origin and progression of metastatic cells within primary tumours remains unclear. Recently, the role of cancer cell fusion in cancer metastasis and development of multidrug resistance (MDR) in tumours has gained prominence. However, evidence remains lacking to justify the role of cell fusion in cancer metastasis and drug resistance. Here, we highlight plausible mechanisms governing cell fusion with different cell types in the tumour microenvironment (TME), the clinical relevance of cancer cell fusion, its potential as a target for overcoming MDR and inhibiting metastasis, and putative modes of treatment.
Collapse
|
15
|
Li M, Li X, Zhao L, Zhou J, Cheng Y, Xu B, Wang J, Wei L. Spontaneous formation of tumorigenic hybrids between human omental adipose-derived stromal cells and endometrial cancer cells increased motility and heterogeneity of cancer cells. Cell Cycle 2019; 18:320-332. [PMID: 30636489 DOI: 10.1080/15384101.2019.1568743] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Recent reports indicate that mesenchymal stem cells (MSCs) can fuse with cancer cells to promote cancer progression. Omental adipose-derived stromal cells (O-ASCs) are similar to MSCs, which could be recruited to the stroma in endometrial cancer. The aim of our study was to investigate whether O-ASCs can fuse with endometrial cancer cells to influence cancer cells biological characteristics. We isolated O-ASCs from patients with endometrial cancer. O-ASCs and endometrial cancer cells were labeled with different fluorescent tags and directly co-cultured in an Opera high-throughput spinning-disk confocal microscopy system to observe the processes involved in the fusion, division and migration of hybrid cells. Immunofluorescence and high-throughput imaging analyzes were performed to evaluate proteins related to epithelial-mesenchymal transition (EMT).We found O-ASCs could spontaneously fuse with endometrial cancer cells, including cytomembrane and nuclear fusion. After fusion, endometrial cancer cells assume an elongated and fibroblast-like appearance that exhibit mesenchymal phenotypes. The hybrid cells proliferated through bipolar and multipolar divisions and exhibited more rapid migratory speeds than were observed in the parental cells (P < 0.01), potentially because of their EMT-associated changes, including the down-regulation of E-cadherin and up-regulation of Vimentin. Our results collectively suggest that tumorigenic hybrids spontaneously formed between human O-ASCs and endometrial cancer cells, and that the resulting cells enhanced cancer mobility and heterogeneity by accelerated migration and undergoing multipolar divisions. These data provide a new avenue for investigating the roles of O-ASCs in endometrial cancer.
Collapse
Affiliation(s)
- Mingxia Li
- a Department of Gynecology and Obstetrics , Peking University People's Hospital , Beijing , China.,b Department of Gynecology and Obstetrics, People's Liberation Army (PLA) Medical School , Chinese PLA General Hospital , Beijing , China
| | - Xiaoping Li
- a Department of Gynecology and Obstetrics , Peking University People's Hospital , Beijing , China
| | - Lijun Zhao
- a Department of Gynecology and Obstetrics , Peking University People's Hospital , Beijing , China
| | - Jingyi Zhou
- a Department of Gynecology and Obstetrics , Peking University People's Hospital , Beijing , China
| | - Yuan Cheng
- a Department of Gynecology and Obstetrics , Peking University People's Hospital , Beijing , China
| | - Bo Xu
- c State Key Laboratory of Natural and Biomietic Drugs, School of Pharmaceutical Sciences , Peking University , Beijing , China
| | - Jianliu Wang
- a Department of Gynecology and Obstetrics , Peking University People's Hospital , Beijing , China
| | - Lihui Wei
- a Department of Gynecology and Obstetrics , Peking University People's Hospital , Beijing , China
| |
Collapse
|
16
|
Zhang LN, Kong CF, Zhao D, Cong XL, Wang SS, Ma L, Huang YH. Fusion with mesenchymal stem cells differentially affects tumorigenic and metastatic abilities of lung cancer cells. J Cell Physiol 2018; 234:3570-3582. [PMID: 30417342 DOI: 10.1002/jcp.27011] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 06/25/2018] [Indexed: 12/13/2022]
Abstract
Cell fusion plays a crucial role in cancer progression and leads to massive aberrant changes in chromosome and gene expression involved in tumor metastasis. Cancer cells can fuse with many cell types, including stromal cells, epithelial cells, macrophages, and endothelial cells. Mesenchymal stem cells (MSCs) have been reported to migrate and incorporate into tumor sites during cancer progression. However, the underlying mechanism of stem cell fusion in tumor metastasis has not been fully deciphered. In this research, we established a cell fusion model between lung cancer cells and MSCs in vitro. We found that the hybrid cells showed enhanced metastatic capacity with increased expression of MMP-2 and MMP-9, whereas the proliferation ability was inhibited and cell cycle was blocked in the G0 /G1 phase with elevated expression of p21, p27, and p53. Moreover, the hybrid cells lost epithelial morphology and exhibited an epithelial-mesenchymal transition (EMT) change with downregulation of E-cadherin and upregulation of N-cadherin, Vimentin, α-SMA and Fibronectin1. Meanwhile, the expressions of EMT transcription factors, including Snail1, Slug, Twist1, Zeb1, and Zeb2, were also increased in hybrid cells. More important, the fusion hybrids acquired stem cell-like properties, which exhibited increased expression stem cell transcription factors Oct4, Sox2, Nanog, Kif4 as well as Bmi1. Taken together, our results suggested that cell fusion between lung cancer cells and MSCs offered enhanced metastatic capacity and characteristics of cancer stem cell by undergoing EMT. This study will contribute to explaning the origin of lung cancer stem cells and to elucidate the role of cell fusion in cancer metastasis.
Collapse
Affiliation(s)
- Li-Na Zhang
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China
| | - Chen-Fei Kong
- China-Japan Union Hospital, Jilin University, Changchun, China
| | - Dan Zhao
- China-Japan Union Hospital, Jilin University, Changchun, China
| | - Xian-Ling Cong
- China-Japan Union Hospital, Jilin University, Changchun, China
| | - Shen-Sen Wang
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China
| | - Ling Ma
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China
| | - Ying-Hui Huang
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing, China
| |
Collapse
|
17
|
Noubissi FK, Ogle BM. Cancer Cell Fusion: Mechanisms Slowly Unravel. Int J Mol Sci 2016; 17:ijms17091587. [PMID: 27657058 PMCID: PMC5037852 DOI: 10.3390/ijms17091587] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 08/26/2016] [Accepted: 09/12/2016] [Indexed: 01/15/2023] Open
Abstract
Although molecular mechanisms and signaling pathways driving invasion and metastasis have been studied for many years, the origin of the population of metastatic cells within the primary tumor is still not well understood. About a century ago, Aichel proposed that cancer cell fusion was a mechanism of cancer metastasis. This hypothesis gained some support over the years, and recently became the focus of many studies that revealed increasing evidence pointing to the possibility that cancer cell fusion probably gives rise to the metastatic phenotype by generating widespread genetic and epigenetic diversity, leading to the emergence of critical populations needed to evolve resistance to the treatment and development of metastasis. In this review, we will discuss the clinical relevance of cancer cell fusion, describe emerging mechanisms of cancer cell fusion, address why inhibiting cancer cell fusion could represent a critical line of attack to limit drug resistance and to prevent metastasis, and suggest one new modality for doing so.
Collapse
Affiliation(s)
| | - Brenda M Ogle
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA.
- Stem Cell Institute, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA.
- Masonic Cancer Center, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA.
- Lillehei Heart Institute, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA.
- Institute for Engineering and Medicine, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA.
| |
Collapse
|
18
|
Tumorigenic hybrids between mesenchymal stem cells and gastric cancer cells enhanced cancer proliferation, migration and stemness. BMC Cancer 2015; 15:793. [PMID: 26498753 PMCID: PMC4620013 DOI: 10.1186/s12885-015-1780-1] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 10/12/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Emerging evidence indicates that inappropriate cell-cell fusion might contribute to cancer progression. Similarly, mesenchymal stem cells (MSCs) can also fuse with other cells spontaneously and capable of adopting the phenotype of other cells. The aim of our study was to investigate the role of MSCs participated cell fusion in the tumorigenesis of gastric cancer. METHODS We fused human umbilical cord mesenchymal stem cells (hucMSCs) with gastric cancer cells in vitro by polyethylene glycol (PEG), the hybrid cells were sorted by flow cytometer. The growth and migration of hybrids were assessed by cell counting, cell colony formation and transwell assays. The proteins and genes related to epithelial- mesenchymal transition and stemness were tested by western blot, immunocytochemistry and real-time RT-PCR. The expression of CD44 and CD133 was examined by immunocytochemistry and flow cytometry. The xenograft assay was used to evaluation the tumorigenesis of the hybrids. RESULTS The obtained hybrids exhibited epithelial- mesenchymal transition (EMT) change with down-regulation of E-cadherin and up-regulation of Vimentin, N-cadherin, α-smooth muscle actin (α-SMA), and fibroblast activation protein (FAP). The hybrids also increased expression of stemness factors Oct4, Nanog, Sox2 and Lin28. The expression of CD44 and CD133 on hybrid cells was stronger than parental gastric cancer cells. Moreover, the migration and proliferation of heterotypic hybrids were enhanced. In addition, the heterotypic hybrids promoted the growth abilities of gastric xenograft tumor in vivo. CONCLUSIONS Taken together, our results suggest that cell fusion between hucMSCs and gastric cancer cells could contribute to tumorigenic hybrids with EMT and stem cell-like properties, which may provide a flexible tool for investigating the roles of MSCs in gastric cancer.
Collapse
|
19
|
Parris GE. Cell-Cell Fusion, Chemotaxis and Metastasis. INTERCELLULAR COMMUNICATION IN CANCER 2015:227-254. [DOI: 10.1007/978-94-017-7380-5_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
20
|
Song K, Song Y, Zhao XP, Shen H, Wang M, Yan TL, Liu K, Shang ZJ. Oral cancer/endothelial cell fusion experiences nuclear fusion and acquisition of enhanced survival potential. Exp Cell Res 2014; 328:156-163. [PMID: 25016285 DOI: 10.1016/j.yexcr.2014.07.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 06/19/2014] [Accepted: 07/01/2014] [Indexed: 12/14/2022]
Abstract
Most previous studies have linked cancer-macrophage fusion with tumor progression and metastasis. However, the characteristics of hybrid cells derived from oral cancer and endothelial cells and their involvement in cancer remained unknown. Double-immunofluorescent staining and fluorescent in situ hybridization (FISH) were performed to confirm spontaneous cell fusion between eGFP-labeled human umbilical vein endothelial cells (HUVECs) and RFP-labeled SCC9, and to detect the expression of vementin and cytokeratin 18 in the hybrids. The property of chemo-resistance of such hybrids was examined by TUNEL assay. The hybrid cells in xenografted tumor were identified by FISH and GFP/RFP dual-immunofluoresence staining. We showed that SCC9 cells spontaneously fused with cocultured endothelial cells, and the resultant hybrid cells maintained the division and proliferation activity after re-plating and thawing. Such hybrids expressed markers of both parental cells and became more resistant to chemotherapeutic drug cisplatin as compared to the parental SCC9 cells. Our in vivo data indicated that the hybrid cells contributed to tumor composition by using of immunostaining and FISH analysis, even though the hybrid cells and SCC9 cells were mixed with 1:10,000, according to the FACS data. Our study suggested that the fusion events between oral cancer and endothelial cells undergo nuclear fusion and acquire a new property of drug resistance and consequently enhanced survival potential. These experimental findings provide further supportive evidence for the theory that cell fusion is involved in cancer progression.
Collapse
Affiliation(s)
- Kai Song
- Department of Oral & Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Shandong Province, China; The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory for Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
| | - Yong Song
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory for Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China; Department of Stomatology, Liu Zhou People׳s Hospital, Guangxi, China
| | - Xiao-Ping Zhao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory for Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
| | - Hui Shen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory for Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
| | - Meng Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory for Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
| | - Ting-Lin Yan
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory for Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
| | - Ke Liu
- Department of Oral and Maxillofacial-Head and Neck oncology, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan 430079, China; The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory for Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China.
| | - Zheng-Jun Shang
- Department of Oral and Maxillofacial-Head and Neck oncology, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan 430079, China; The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory for Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China.
| |
Collapse
|
21
|
Shabo I, Olsson H, Elkarim R, Sun XF, Svanvik J. Macrophage Infiltration in Tumor Stroma is Related to Tumor Cell Expression of CD163 in Colorectal Cancer. CANCER MICROENVIRONMENT 2014; 7:61-9. [PMID: 24771466 DOI: 10.1007/s12307-014-0145-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 04/07/2014] [Indexed: 12/11/2022]
Abstract
The scavenger receptor, CD163, is a macrophage-specific marker. Recent studies have shown that CD163 expression in breast and rectal cancer cells is associated with poor prognosis. This study was conducted to evaluate the relationship between CD163 expression as a macrophage trait in cancer cells, and macrophage infiltration and its clinical significance in colorectal cancer. Immunostaining of CD163 and macrophage infiltration were evaluated in paraffin-embedded specimens, earlier analyzed for CD31, D2-40 and S-phase fraction, from primary tumors and normal colorectal mucosa of 75 patients with colorectal carcinoma. The outcomes were analyzed in relation to clinical-pathological data. CD163 expression was positive in cancer cells in 20 % of colorectal cancer patients and was related to advanced tumor stages (P = 0.008) and unfavorable prognosis (p = 0.001). High macrophage infiltration was related to shorter survival and positive CD163 expression in tumor cells. The prognostic impact of macrophage infiltration was independent of tumor stage and CD163 expression in cancer cells (p = 0.034). The expression of macrophage phenotype in colorectal cancer cells is associated with macrophage density in tumor stroma and lower survival rates. Macrophage infiltration has an independent prognostic impact on mortality in colorectal cancer. In accordance with previous experimental studies, these findings provide new insights into the role of macrophages in colorectal cancer.
Collapse
Affiliation(s)
- Ivan Shabo
- Department of surgery, County Council of Östergötland, Linköping, Sweden,
| | | | | | | | | |
Collapse
|
22
|
Breast cancer expression of DAP12 is associated with skeletal and liver metastases and poor survival. Clin Breast Cancer 2013; 13:371-7. [PMID: 23810293 DOI: 10.1016/j.clbc.2013.05.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 04/30/2013] [Accepted: 05/03/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND The transmembrane adapter protein, DAP12, transduces activation signals for several arrays of receptors, including human signal-regulatory protein, DAP12-associating lectin-1, triggering receptor expressed on myeloid cells-1, -2, and -3, in natural killer cells, granulocytes, monocytes/macrophages, and dendritic cells. The macrophage-specific antigen, Cluster of Differentiation 163 (CD163), is expressed in breast and colorectal cancers and is associated with early cancer recurrence and poor prognosis. It was recently shown that fusion between intestinal tumor cells and macrophages results in nuclear reprogramming with hybrid transcripts from both cells of origin. The role of DAP12 in the fusion process is not known. This study investigates the expression of DAP12 in BRC cells, and its relation to other macrophage traits and to the clinical progression of disease. MATERIALS AND METHODS Immunostaining of DAP12 and CD163 was performed and evaluated in paraffin-embedded specimens from 132 patients with BRC. The outcomes were analyzed in relation to clinicopathological data. RESULTS DAP12 expression in cancer cells was positive in 66 percent of the cancers and was associated with high tumor grade (P = .015), and with liver (P = .047) and skeletal (P = .067), but not with lung metastases (P = 1.00). Patients with BRC expressing DAP12 had poor prognosis, with higher recurrence rates of skeletal (P = .018) and liver metastases (P = .047), and shorter survival time (P = .0060). CONCLUSION We suggest that macrophage traits in BRC cells facilitate the metastatic process and that DAP12 expression might promote metastatic homing to bone and liver tissues.
Collapse
|
23
|
Man YG, Stojadinovic A, Mason J, Avital I, Bilchik A, Bruecher B, Protic M, Nissan A, Izadjoo M, Zhang X, Jewett A. Tumor-infiltrating immune cells promoting tumor invasion and metastasis: existing theories. J Cancer 2013; 4:84-95. [PMID: 23386907 PMCID: PMC3564249 DOI: 10.7150/jca.5482] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Accepted: 12/20/2012] [Indexed: 12/12/2022] Open
Abstract
It is a commonly held belief that infiltration of immune cells into tumor tissues and direct physical contact between tumor cells and infiltrated immune cells is associated with physical destructions of the tumor cells, reduction of the tumor burden, and improved clinical prognosis. An increasing number of studies, however, have suggested that aberrant infiltration of immune cells into tumor or normal tissues may promote tumor progression, invasion, and metastasis. Neither the primary reason for these contradictory observations, nor the mechanism for the reported diverse impact of tumor-infiltrating immune cells has been elucidated, making it difficult to judge the clinical implications of infiltration of immune cells within tumor tissues. This mini-review presents several existing hypotheses and models that favor the promoting impact of tumor-infiltrating immune cells on tumor invasion and metastasis, and also analyzes their strength and weakness.
Collapse
Affiliation(s)
- Yan-gao Man
- 1. Diagnostic and Translational Research Center, Henry Jackson Foundation, Gaithersburg, MD, USA
- 2. College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Alexander Stojadinovic
- 3. Surgical Oncology, Walter Reed National Military Medical Center, and Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Jeffrey Mason
- 4. Veterans Affair Medical Center, Washington, DC, USA
| | - Itzhak Avital
- 5. Bon Secours National Cancer Institute (BSNCI), Richmond VA, USA
| | - Anton Bilchik
- 6. John Wayne Cancer Institute; California Oncology Research Institute; and, David Geffen School of Medicine, University of California, Los Angeles, USA
| | | | - Mladjan Protic
- 8. Clinic of Abdominal, Endocrine, and Transplantation Surgery, Clinical Center of Vojvodina, University of Novi Sad - Medical Faculty, Novi Sad, Serbia
| | - Aviram Nissan
- 9. The Surgical Oncology Laboratory, Department of Surgery, Hadassah-Hebrew University Medical Center, Mount Scopus, Jerusalem, Israel
| | - Mina Izadjoo
- 1. Diagnostic and Translational Research Center, Henry Jackson Foundation, Gaithersburg, MD, USA
| | - Xichen Zhang
- 2. College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Anahid Jewett
- 10. Division of Oral Biology and Medicine, Jonsson Comprehensive Cancer Center, UCLA School of Dentistry, Los Angeles, CA, USA
| |
Collapse
|
24
|
Abstract
Metastasis involves the spread of cancer cells from the primary tumor to surrounding tissues and to distant organs and is the primary cause of cancer morbidity and mortality. In order to complete the metastatic cascade, cancer cells must detach from the primary tumor, intravasate into the circulatory and lymphatic systems, evade immune attack, extravasate at distant capillary beds, and invade and proliferate in distant organs. Currently, several hypotheses have been advanced to explain the origin of cancer metastasis. These involve an epithelial mesenchymal transition, an accumulation of mutations in stem cells, a macrophage facilitation process, and a macrophage origin involving either transformation or fusion hybridization with neoplastic cells. Many of the properties of metastatic cancer cells are also seen in normal macrophages. A macrophage origin of metastasis can also explain the long-standing "seed and soil" hypothesis and the absence of metastasis in plant cancers. The view of metastasis as a macrophage metabolic disease can provide novel insight for therapeutic management.
Collapse
Affiliation(s)
- Thomas N Seyfried
- Department of Biology, Boston College, Chestnut Hill, Massachusetts 02467, USA.
| | | |
Collapse
|
25
|
Ding J, Jin W, Chen C, Shao Z, Wu J. Tumor associated macrophage × cancer cell hybrids may acquire cancer stem cell properties in breast cancer. PLoS One 2012; 7:e41942. [PMID: 22848668 PMCID: PMC3405038 DOI: 10.1371/journal.pone.0041942] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 06/28/2012] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is one of the most frequently diagnosed cancers among women, and metastasis makes it lethal. Tumor-associated macrophages (TAMs) that acquire an alternatively activated macrophage (M2) phenotype may promote metastasis. However, the underlying mechanisms are still elusive. Here, we examined how TAMs interact with breast cancer cells to promote metastasis. Immunohistochemistry was used to examine the expression of the M2-specific antigen CD163 in paraffin-embedded mammary carcinoma blocks to explore fusion events in breast cancer patients. U937 cells were used as a substitute for human monocytes, and these cells differentiated into M2 macrophages following phorbol 12-myristate 13-acetate (PMA) and M-CSF stimulation. M2 macrophages and the breast cancer cell lines MCF-7 and MDA-MB-231 fused in the presence of 50% polyethylene glycol. Hybrids were isolated by fluorescence-activated cell sorting, and the relevant cell biological properties were compared with their parental counterparts. Breast cancer stem cell (BCSC)-related markers were quantified by immunofluorescence staining, RT-PCR, quantitative RT-PCR and/or western blotting. The tumor-initiating and metastatic capacities of the hybrids and their parental counterparts were assessed in NOD/SCID mice. We found that the CD163 expression rate in breast cancer tissues varied significantly and correlated with estrogen receptor status (p<0.05). The fusion efficiency of either breast cancer cell line with M2 macrophages ranged from 1.81 to 6.47% in the presence of PEG, and no significant difference was observed between the breast cancer cell lines used (p>0.05). Characterization of the fusion hybrids revealed a more aggressive phenotype, including increased migration, invasion and tumorigenicity, but reduced proliferative ability, compared with the parental lines. The hybrids also gained a CD44(+)CD24(-/low) phenotype and over-expressed epithelial-mesenchymal transition-associated genes. These results indicate that TAMs may promote breast cancer metastasis through cell fusion, and the hybrids may gain a BCSC phenotype.
Collapse
Affiliation(s)
- Jingxian Ding
- Department of Breast Surgery, Breast Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Fudan University Cancer Institute Research Center, Shanghai, China
| | - Wei Jin
- Department of Breast Surgery, Breast Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Canming Chen
- Department of Breast Surgery, Breast Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhiming Shao
- Department of Breast Surgery, Breast Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiong Wu
- Department of Breast Surgery, Breast Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- * E-mail:
| |
Collapse
|
26
|
Checinska A, Soengas MS. The gluttonous side of malignant melanoma: basic and clinical implications of macroautophagy. Pigment Cell Melanoma Res 2012; 24:1116-32. [PMID: 21995431 DOI: 10.1111/j.1755-148x.2011.00927.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
True to their inherent aggressive behavior, melanomas keep impressing the melanoma community with their ability to bypass tumor suppressor mechanisms. Name a pathway with the potential to control cell survival and melanoma cells will likely have it potentiated by multiple genetic or epigenetic alterations. In the context of progression and chemoresistance, large efforts have been dedicated to the identification of protective mechanisms associated with or linked to apoptotic death programs. These studies have guided the design of targeted anticancer strategies. Still, the promise for pro-apoptotic inducers as lead compounds for drug development has yet to come to fruition. It was then a question of time to identify alternative modulators of cell viability. An ideal candidate that is raising great expectations in the oncology field is autophagy, a catabolic process with multiple roles in cell homeostasis. Here we review the incipient literature on autophagy markers in melanocytic lesions. Intriguingly, histopathological studies are unveiling an intrinsic inter- and intratumor variability in the expression of autophagy modulators. Nonetheless, functional studies support a key role of autopaphagy programs in the response to a variety of stress factors. These include adaptive responses to nutrient deprivation, hypoxia and many anticancer agents, among other stimuli. Strategies are being also developed to mobilize the endocytic machinery and shift autolysosomes into death effectors. The opportunities that lie ahead in this field are exciting. Various authophagy mediators are potentially druggable. Moreover, animal models and the development of sophisticated screening methods offer a platform for multilevel academic-industrial collaborations. These efforts are expected to open avenues of research and, hopefully, lead to a more rational approach to melanoma treatment.
Collapse
Affiliation(s)
- Agnieszka Checinska
- Melanoma Laboratory, Molecular Pathology Programme, Centro Nacional de Investigaciones Oncológicas (Spanish National Cancer Research Centre), Madrid, Spain
| | | |
Collapse
|
27
|
Song K, Zhu F, Zhang HZ, Shang ZJ. Tumor necrosis factor-α enhanced fusions between oral squamous cell carcinoma cells and endothelial cells via VCAM-1/VLA-4 pathway. Exp Cell Res 2012; 318:1707-15. [PMID: 22664325 DOI: 10.1016/j.yexcr.2012.05.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 05/01/2012] [Accepted: 05/24/2012] [Indexed: 01/08/2023]
Abstract
Fusion between cancer cells and host cells, including endothelial cells, may strongly modulate the biological behavior of tumors. However, no one is sure about the driving factors and underlying mechanism involved in such fusion. We hypothesized in this study that inflammation, one of the main characteristics in tumor microenvironment, serves as a prominent catalyst for fusion events. Our results showed that oral cancer cells can fuse spontaneously with endothelial cells in co-culture and inflammatory cytokine tumor necrosis factor-α (TNF-α) increased fusion of human umbilical vein endothelium cells and oral cancer cells by up to 3-fold in vitro. Additionally, human oral squamous cell carcinoma cell lines and 35 out of 50 (70%) oral squamous carcinoma specimens express VLA-4, an integrin, previously implicated in fusions between human peripheral blood CD34-positive cells and murine cardiomyocytes. Expression of VCAM-1, a ligand for VLA-4, was evident on vascular endothelium of oral squamous cell carcinoma. Moreover, immunocytochemistry and flow cytometry analysis revealed that expression of VCAM-1 increased obviously in TNF-α-stimulated endothelial cells. Anti-VLA-4 or anti-VCAM-1 treatment can decrease significantly cancer-endothelial adhesion and block such fusion. Collectively, our results suggested that TNF-α could enhance cancer-endothelial cell adhesion and fusion through VCAM-1/VLA-4 pathway. This study provides insights into regulatory mechanism of cancer-endothelial cell fusion, and has important implications for the development of novel therapeutic strategies for prevention of metastasis.
Collapse
Affiliation(s)
- Kai Song
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST), Wuhan University, Wuhan, China
| | | | | | | |
Collapse
|
28
|
Mukhtar RA, Moore AP, Tandon VJ, Nseyo O, Twomey P, Adisa CA, Eleweke N, Au A, Baehner FL, Moore DH, McGrath MS, Olopade OI, Gray JW, Campbell MJ, Esserman LJ. Elevated levels of proliferating and recently migrated tumor-associated macrophages confer increased aggressiveness and worse outcomes in breast cancer. Ann Surg Oncol 2012; 19:3979-86. [PMID: 22622474 DOI: 10.1245/s10434-012-2415-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2011] [Indexed: 12/22/2022]
Abstract
PURPOSE Macrophages play a major role in inflammatory processes and have been associated with poor prognosis in a variety of cancers, including breast cancer. Previously, we investigated the relationship of a subset of tumor-associated macrophages (PCNA(+) TAMs) with clinicopathologic characteristics of breast cancer. We reported that high PCNA(+) TAM counts were associated with hormone receptor (HR)-negative, high-grade tumors and early recurrence. To further understand the significance of elevated PCNA(+) TAMs and the functionality of TAMs, we examined the expression of S100A8/S100A9 with the antibody Mac387. The heterodimeric S100A8/S100A9 complex plays a role in inflammation and is increased in several cancer types. METHODS We performed immunohistochemistry using the Mac387 antibody on 367 invasive human breast cancer cases. Results were compared to previous PCNA(+) TAM counts and were correlated with patient outcomes adjusting for HR status and histologic grade. RESULTS Like PCNA(+) TAMs, high Mac387 counts were associated with HR negativity, high tumor grade, younger age, and decreased recurrence-free survival. Mac387, however, appears to identify both a subset of macrophages and a subset of tumor cells. The concordance between Mac387 and PCNA(+) TAM counts was low and cases that had both high Mac387 and high PCNA(+) TAMs counts had a stronger association with early recurrence. CONCLUSIONS The presence of high numbers of PCNA(+) TAMs and Mac387-positive cells in breast cancers with poor outcomes may implicate a subset of TAMs in breast cancer pathogenesis, and may ultimately serve to develop potential cellular targets for therapeutic interventions.
Collapse
Affiliation(s)
- Rita A Mukhtar
- Department of Surgery, University of California, San Francisco, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Król M, Pawłowski KM, Majchrzak K, Gajewska M, Majewska A, Motyl T. Global gene expression profiles of canine macrophages and canine mammary cancer cells grown as a co-culture in vitro. BMC Vet Res 2012; 8:16. [PMID: 22353646 PMCID: PMC3315417 DOI: 10.1186/1746-6148-8-16] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Accepted: 02/21/2012] [Indexed: 12/24/2022] Open
Abstract
Background Solid tumours comprise various cells, including cancer cells, resident stromal cells, migratory haemopoietic cells and other. These cells regulate tumour growth and metastasis. Macrophages constitute probably the most important element of all interactions within the tumour microenvironment. However, the molecular mechanism, that guides tumour environment, still remains unknown. Exploring the underlying molecular mechanisms that orchestrate these phenomena has been the aim of our study. A co-culture of canine mammary cancer cells and macrophages was established and maintained for 72 hrs. Having sorted the cells, gene expression in cancer cells and macrophages, using DNA microarrays, was examined. The results were confirmed using real-time qPCR and confocal microscopy. Moreover, their ability for migration and invasion has been assessed. Results Microarray analysis showed that the up-regulated genes in the cancer cell lines are involved in 15 highly over-manifested pathways. The pathways that drew our diligent attention included: the inflammation pathway mediated by chemokine and cytokine, the Toll receptor signalling pathway and the B cell activation. The up-regulated genes in the macrophages were involved in only 18 significantly over-manifested pathways: the angiogenesis, the p53 pathway feedback loops2 and the Wnt signalling pathway. The microarray analysis revealed that co-culturing of cancer cells with macrophages initiated the myeloid-specific antigen expression in cancer cells, as well as cytokine/chemokine genes expression. This finding was confirmed at mRNA and protein level. Moreover, we showed that macrophages increase cancer migration and invasion. Conclusions The presence of macrophages in the cancer environment induces acquisition of the macrophage phenotype (specific antigens and chemokines/cytokines expression) in cancer cells. We presumed that cancer cells also acquire other myeloid features, such as: capabilities of cell rolling, spreading, migration and matrix invasion (what has also been confirmed by our results). It may, perhaps, be the result of myeloid-cancer cell hybrid formation, or cancer cells mimicking macrophages phenotype, owing to various proteins secreted by macrophages.
Collapse
Affiliation(s)
- Magdalena Król
- Department of Physiological Sciences, Warsaw University of Life Sciences - WULS, Warsaw, Poland.
| | | | | | | | | | | |
Collapse
|
30
|
Kraljevic Pavelic S, Sedic M, Bosnjak H, Spaventi S, Pavelic K. Metastasis: new perspectives on an old problem. Mol Cancer 2011; 10:22. [PMID: 21342498 PMCID: PMC3052211 DOI: 10.1186/1476-4598-10-22] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Accepted: 02/22/2011] [Indexed: 12/23/2022] Open
Abstract
Many hypotheses have been postulated to explain the intricate nature of the metastatic process, but none of them completely accounted for the actual biological and clinical observations. Consequently, metastasis still remains an open issue with only few metastasis-inducing proteins experimentally validated so far. Recently proposed novel metastatic model, where serial and parallel metastatic processes are adequately integrated, might help to bridge the current gap between experimental results and clinical observations. In addition, the identification, isolation and molecular characterization of cancer stem cells, a population of the cells within the tumour mass able to proliferate, self-renew and induce tumorigenesis, will shed new light on the complex molecular events mediating metastasis, invasion and resistance to therapy. Understanding the molecular basis of these tumour characteristics will usher in a new age of individualized cancer therapy. In this review article, we will provide a current overview of molecular mechanisms underpinning metastasis, and discuss recent findings in this field obtained by global molecular profiling strategies such as proteomics.
Collapse
|
31
|
Abstract
Emerging evidence suggests that many metastatic cancers arise from cells of the myeloid/macrophage lineage regardless of the primary tissue of origin. A myeloid origin of metastatic cancer stands apart from origins involving clonal evolution or epithelial-mesenchymal transitions. Evidence is reviewed demonstrating that numerous human cancers express multiple properties of macrophages including phagocytosis, fusogenicity, and gene/protein expression. It is unlikely that the macrophage properties expressed in metastatic cancers arise from sporadic random mutations in epithelial cells, but rather from damage to an already existing mesenchymal cell, e.g., a myeloid/macrophage-type cell. Such cells would naturally embody the capacity to express the multiple behaviors of metastatic cells. The view of metastasis as a myeloid/macrophage disease will impact future cancer research and anti-metastatic therapies.
Collapse
Affiliation(s)
- Leanne C. Huysentruyt
- Department of Medicine, Hematology and Oncology, University of California, San Francisco, San Francisco, CA USA
| | | |
Collapse
|
32
|
Seyfried TN, Shelton LM. Cancer as a metabolic disease. Nutr Metab (Lond) 2010; 7:7. [PMID: 20181022 PMCID: PMC2845135 DOI: 10.1186/1743-7075-7-7] [Citation(s) in RCA: 382] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2009] [Accepted: 01/27/2010] [Indexed: 01/08/2023] Open
Abstract
Emerging evidence indicates that impaired cellular energy metabolism is the defining characteristic of nearly all cancers regardless of cellular or tissue origin. In contrast to normal cells, which derive most of their usable energy from oxidative phosphorylation, most cancer cells become heavily dependent on substrate level phosphorylation to meet energy demands. Evidence is reviewed supporting a general hypothesis that genomic instability and essentially all hallmarks of cancer, including aerobic glycolysis (Warburg effect), can be linked to impaired mitochondrial function and energy metabolism. A view of cancer as primarily a metabolic disease will impact approaches to cancer management and prevention.
Collapse
|
33
|
Shabo I, Olsson H, Sun XF, Svanvik J. Expression of the macrophage antigen CD163 in rectal cancer cells is associated with early local recurrence and reduced survival time. Int J Cancer 2009; 125:1826-31. [PMID: 19582880 DOI: 10.1002/ijc.24506] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Expression of the macrophage antigen CD163 in breast cancer cells is recently shown to be related to early distant recurrence and shortened survival. In this study, 163 patients with rectal cancer, included in the Swedish rectal cancer trial and followed up for a median of 71 months, were examined for the expression of CD163 in the primary tumors. The cancer cells expressed CD163 in the primary tumors in 23% (n = 32) of the patients. In pretreatment biopsies from 101 patients, 10 had CD163-positive cancers and these patients had earlier local recurrence (p < 0.044) and reduced survival time (p < 0.045) compared with those with CD163-negative tumors. When studying surgical specimens from 61 patients randomized to preoperative irradiation (5 x 5 Gy delivered in 1 week), it was found that 31% were CD163 positive whereas the corresponding figure was only 17% for 78 patients who were nonirradiated (p < 0.044), which tentatively may be consistent with X-rays inducing fusion. In CD163-positive tumors there was a reduced apoptotic activity as measured with the Termina deoxynucleotidyl Transferase Biotin-dUTP Nick End Labeling (TUNEL) technique (p = 0.018). There tended also to be an increased proliferation activity measured as an expression of Ki-67 non significant (NS). It is concluded that primary rectal cancers may express CD-163, and this phenotypic macrophage trait is related to early local recurrence, shorter survival time and reduced apoptosis. Furthermore, the expression of CD163 is more common after irradiation.
Collapse
Affiliation(s)
- Ivan Shabo
- Division of Surgery, Institution of Clinical and Experimental Medicine, University of Linköping, Sweden
| | | | | | | |
Collapse
|
34
|
Abstract
We show that malignant melanoma cells display high levels of autophagy, a cytoplasmic process of protein and organelle digestion that provides an energy source in times of nutrient deprivation. In a panel of 12 cases of cutaneous malignant melanoma of the superficial spreading type, cells in florid melanoma in situ (MIS) and invasive cells in the dermis appeared to be undergoing autophagy. Autophagosomes were detected through immunohistochemistry using the marker LC3B (microtubule-associated light chain 3B), and by electron microscopy. Some autophagosomes contained melanized melanosomes, accounting for the phenomenon of 'coarse melanin' in malignant melanoma. Autophagosomes also contained the Golgi 58k protein, a structural component of the Golgi apparatus, and beta1,6-branched oligosaccharides, indicating that at least some of the autophagosomal proteins were glycosylated with these structures. The findings suggest that autophagy could be a constitutive metabolic state for invasive and metastatic melanoma cells. Interestingly, a similar phenotype was also expressed by tumor-associated melanophages. The findings are consistent with previous reports that endoplasmic reticulum (ER) stress drives melanoma progression, since ER stress is known to trigger autophagy. The results suggest that therapies inhibiting autophagy may be effective for the treatment of malignant melanoma by depriving cells of an important energy source.
Collapse
Affiliation(s)
- Rossitza Lazova
- Department of Dermatology, Yale University School of Medicine, New Haven, CT 06520-8059, USA
| | | | | |
Collapse
|