1
|
Smith JT, Chai RC. Bone niches in the regulation of tumour cell dormancy. J Bone Oncol 2024; 47:100621. [PMID: 39157742 PMCID: PMC11326946 DOI: 10.1016/j.jbo.2024.100621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 05/19/2024] [Accepted: 07/02/2024] [Indexed: 08/20/2024] Open
Abstract
Secondary metastases, accounting for 90 % of cancer-related deaths, pose a formidable challenge in cancer treatment, with bone being a prevalent site. Importantly, tumours may relapse, often in the skeleton even after successful eradication of the primary tumour, indicating that tumour cells may lay dormant within bone for extended periods of time. This review summarises recent findings in the mechanisms underlying tumour cell dormancy and the role of bone cells in this process. Hematopoietic stem cell (HSC) niches in bone provide a model for understanding regulatory microenvironments. Dormant tumour cells have been shown to exploit similar niches, with evidence suggesting interactions with osteoblast-lineage cells and other stromal cells via CXCL12-CXCR4, integrins, and TAM receptor signalling, especially through GAS6-AXL, led to dormancy, with exit of dormancy potentially regulated by osteoclastic bone resorption and neuronal signalling. A comprehensive understanding of dormant tumour cell niches and their regulatory mechanisms is essential for developing targeted therapies, a critical step towards eradicating metastatic tumours and stopping disease relapse.
Collapse
Affiliation(s)
- James T. Smith
- Bone Biology Lab, Cancer Plasticity and Dormancy Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Ryan C. Chai
- Bone Biology Lab, Cancer Plasticity and Dormancy Program, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia
| |
Collapse
|
2
|
Xiong G, Xie N, Nie M, Ling R, Yun B, Xie J, Ren L, Huang Y, Wang W, Yi C, Zhang M, Xu X, Zhang C, Zou B, Zhang L, Liu X, Huang H, Chen D, Cao W, Wang C. Single-cell transcriptomics reveals cell atlas and identifies cycling tumor cells responsible for recurrence in ameloblastoma. Int J Oral Sci 2024; 16:21. [PMID: 38424060 PMCID: PMC10904398 DOI: 10.1038/s41368-024-00281-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 03/02/2024] Open
Abstract
Ameloblastoma is a benign tumor characterized by locally invasive phenotypes, leading to facial bone destruction and a high recurrence rate. However, the mechanisms governing tumor initiation and recurrence are poorly understood. Here, we uncovered cellular landscapes and mechanisms that underlie tumor recurrence in ameloblastoma at single-cell resolution. Our results revealed that ameloblastoma exhibits five tumor subpopulations varying with respect to immune response (IR), bone remodeling (BR), tooth development (TD), epithelial development (ED), and cell cycle (CC) signatures. Of note, we found that CC ameloblastoma cells were endowed with stemness and contributed to tumor recurrence, which was dominated by the EZH2-mediated program. Targeting EZH2 effectively eliminated CC ameloblastoma cells and inhibited tumor growth in ameloblastoma patient-derived organoids. These data described the tumor subpopulation and clarified the identity, function, and regulatory mechanism of CC ameloblastoma cells, providing a potential therapeutic target for ameloblastoma.
Collapse
Affiliation(s)
- Gan Xiong
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Nan Xie
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Min Nie
- Department of Periodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Rongsong Ling
- Institute for Advanced Study, Shenzhen University, Shenzhen, China
| | - Bokai Yun
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Jiaxiang Xie
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Linlin Ren
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yaqi Huang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Wenjin Wang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Chen Yi
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Ming Zhang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Xiuyun Xu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Caihua Zhang
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Bin Zou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Leitao Zhang
- Department of Oral and Maxillofacial Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiqiang Liu
- Department of Oral and Maxillofacial Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hongzhang Huang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Demeng Chen
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei Cao
- Department of Oral and Maxillofacial & Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- National Center for Stomatology, National Clinical Research Center for Oral diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China.
| | - Cheng Wang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
3
|
Tripura C, Gunda S, Vishwakarma SK, Thatipalli AR, Jose J, Jerald MK, Khan AA, Pande G. Long-term and non-invasive in vivo tracking of DiD dye-labeled human hepatic progenitors in chronic liver disease models. World J Hepatol 2022; 14:1884-1898. [PMID: 36340748 PMCID: PMC9627437 DOI: 10.4254/wjh.v14.i10.1884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/02/2022] [Accepted: 10/04/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Chronic liver diseases (CLD) are the major public health burden due to the continuous increasing rate of global morbidity and mortality. The inherent limitations of organ transplantation have led to the development of stem cell-based therapy as a supportive and promising therapeutic option. However, identifying the fate of transplanted cells in vivo represents a crucial obstacle.
AIM To evaluate the potential applicability of DiD dye as a cell labeling agent for long-term, and non-invasive in vivo tracking of transplanted cells in the liver.
METHODS Magnetically sorted, epithelial cell adhesion molecule positive (1 × 106 cells/mL) fetal hepatic progenitor cells were labeled with DiD dye and transplanted into the livers of CLD-severe combined immunodeficiency (SCID) mice. Near-infrared (NIR) imaging was performed for in vivo tracking of the DiD-labeled transplanted cells along with colocalization of hepatic markers for up to 80 d. The existence of human cells within mouse livers was identified using Alu polymerase chain reaction and sequencing.
RESULTS NIR fluorescence imaging of CLD-SCID mice showed a positive fluorescence signal of DiD at days 7, 15, 30, 45, 60, and 80 post-transplantation. Furthermore, positive staining of cytokeratin, c-Met, and albumin colocalizing with DiD fluorescence clearly demonstrated that the fluorescent signal of hepatic markers emerged from the DiD-labeled transplanted cells. Recovery of liver function was also observed with serum levels of glutamic-oxaloacetic transaminase, glutamate-pyruvate transaminase, and bilirubin. The detection of human-specific Alu sequence from the transplanted mouse livers provided evidence for the survival of transplanted cells at day 80.
CONCLUSION DiD-labeling is promising for long-term and non-invasive in vivo cell tracking, and understanding the regenerative mechanisms incurred by the transplanted cells.
Collapse
Affiliation(s)
- Chaturvedula Tripura
- Cell and Stem Cell Biology, CSIR-Centre for Cellular and Molecular Biology, Hyderabad 500007, Telangana, India
| | - Srinivas Gunda
- Cell and Stem Cell Biology, CSIR-Centre for Cellular and Molecular Biology, Hyderabad 500007, Telangana, India
| | - Sandeep Kumar Vishwakarma
- Central Laboratory for Stem Cell Research and Translational Medicine, Centre for Liver Research and Diagnostics, Deccan College of Medical Sciences, Hyderabad 500058, Telangana, India
| | - Avinash Raj Thatipalli
- Cell and Stem Cell Biology, CSIR-Centre for Cellular and Molecular Biology, Hyderabad 500007, Telangana, India
| | - Jedy Jose
- Cell and Stem Cell Biology, CSIR-Centre for Cellular and Molecular Biology, Hyderabad 500007, Telangana, India
| | - Mahesh Kumar Jerald
- Cell and Stem Cell Biology, CSIR-Centre for Cellular and Molecular Biology, Hyderabad 500007, Telangana, India
| | - Aleem Ahmed Khan
- Central Laboratory for Stem Cell Research and Translational Medicine, Centre for Liver Research and Diagnostics, Deccan College of Medical Sciences, Hyderabad 500058, Telangana, India
| | - Gopal Pande
- Cell and Stem Cell Biology, CSIR-Centre for Cellular and Molecular Biology, Hyderabad 500007, Telangana, India
| |
Collapse
|
4
|
Li G, Fan M, Zheng Z, Zhang Y, Zhang Z, Huang Z, Luo W, Zhao W, Lai X, Chen H, Zeng F, Deng F. Osteoblastic protein kinase D1 contributes to the prostate cancer cells dormancy via GAS6-circadian clock signaling. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119296. [PMID: 35595103 DOI: 10.1016/j.bbamcr.2022.119296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 05/05/2022] [Accepted: 05/07/2022] [Indexed: 06/15/2023]
Abstract
Disseminated prostate cancer (PCa) is known to have a strong propensity for bone marrow. These disseminated tumor cells (DTCs) can survive in bone marrow for years without obvious proliferation, while maintaining the ability to develop into metastatic lesions. However, how DTCs kept dormant and recur is still uncertain. Here, we focus on the role of osteoblastic protein kinase D1 (PKD1) in PCa (PC-3 and DU145) dormancy using co-culture experiments. Using flow cytometry, western blotting, and immunofluorescence, we observed that in co-cultures osteoblasts could induce a dormant state in PCa cells, which is manifested by a fewer cell divisions, a decrease Ki-67-positive populations and a lower ERK/p38 ratio. In contrast, silencing of PKD1 gene in osteoblasts impedes co-cultured prostate cancer cell's dormancy ability. Mechanismly, protein kinase D1 (PKD1) in osteoblasts induces PCa dormancy via activating CREB1, which promoting the expression and secretion of growth arrest specific 6 (GAS6). Furthermore, GAS6-induced dormancy signaling significantly increased the expression of core circadian clock molecules in PCa cells, and a negative correlation of circadian clock proteins (BMAL1, CLOCK and DEC2) with recurrence-free survival is observed in metastatic prostate cancer patients. Interestingly, the expression of cell cycle factors (p21, p27, CDK1 and PCNA) which regulated by circadian clock also upregulated in response to GAS6 stimulation. Taken together, we provide evidence that osteoblastic PKD1/CREB1/GAS6 signaling regulates cellular dormancy of PCa cells, and highlights the importance of circadian clock in PCa cells dormancy.
Collapse
Affiliation(s)
- Guihuan Li
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Mingming Fan
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zenan Zheng
- Department of Oral & Maxillofacial Surgery, NanFang Hospital, Southern Medical University, Guangzhou 510900, China
| | - Yihe Zhang
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhishuai Zhang
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhibin Huang
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Wenyang Luo
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Wanlu Zhao
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaoju Lai
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Hua Chen
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Fangyin Zeng
- Department of Clinical Laboratory, Fifth Affiliated Hospital, Southern Medical University, Guangzhou 510900, China.
| | - Fan Deng
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
5
|
Zhang L, Feng X, Shen Y, Wang Y, Liu Z, Ma Y, Gu Y, Guo G, Duan L, Lu L, Liang Y, Lawrence T, Huang R. A novel
ZsGreen
knock‐in melanoma cell line reveals the function of
CD11b
in tumor phagocytosis. Immunol Cell Biol 2022; 100:691-704. [DOI: 10.1111/imcb.12575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 06/26/2022] [Accepted: 07/17/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Lichen Zhang
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine Xinxiang Medical University Xinxiang China
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine Xinxiang Medical University Xinxiang China
| | - Xinyu Feng
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine Xinxiang Medical University Xinxiang China
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine Xinxiang Medical University Xinxiang China
| | - Yingzhuo Shen
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine Xinxiang Medical University Xinxiang China
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine Xinxiang Medical University Xinxiang China
| | - Yingbin Wang
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine Xinxiang Medical University Xinxiang China
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine Xinxiang Medical University Xinxiang China
| | - Zhuangzhuang Liu
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine Xinxiang Medical University Xinxiang China
- Institute of Psychiatry and Neuroscience Xinxiang Medical University Xinxiang China
| | - Yuang Ma
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine Xinxiang Medical University Xinxiang China
| | - Yanrong Gu
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine Xinxiang Medical University Xinxiang China
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine Xinxiang Medical University Xinxiang China
| | - Guo Guo
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine Xinxiang Medical University Xinxiang China
| | - Liangwei Duan
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine Xinxiang Medical University Xinxiang China
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine Xinxiang Medical University Xinxiang China
| | - Liaoxun Lu
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine Xinxiang Medical University Xinxiang China
- Institute of Psychiatry and Neuroscience Xinxiang Medical University Xinxiang China
| | - Yinming Liang
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine Xinxiang Medical University Xinxiang China
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine Xinxiang Medical University Xinxiang China
- Institute of Psychiatry and Neuroscience Xinxiang Medical University Xinxiang China
| | - Toby Lawrence
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine Xinxiang Medical University Xinxiang China
- Centre for Inflammation Biology and Cancer Immunology, Cancer Research UK King's Health Partners Centre, School of Immunology and Microbial Sciences King's College London London UK
| | - Rong Huang
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine Xinxiang Medical University Xinxiang China
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine Xinxiang Medical University Xinxiang China
| |
Collapse
|
6
|
Abstract
Cell cycle involves a series of changes that lead to cell growth and division. Cell cycle analysis is crucial to understand cellular responses to changing environmental conditions. Since its inception, flow cytometry has been particularly useful for cell cycle analysis at single cell level due to its speed and precision. Previously, flow cytometric cell cycle analysis relied solely on the measurement of cellular DNA content. Later, methods were developed for multiparametric analysis. This review explains the journey of flow cytometry to understand different molecular and cellular events underlying cell cycle using various protocols. Recent advances in the field that overcome the shortcomings of traditional flow cytometry and expand its scope for cell cycle studies are also discussed.
Collapse
|
7
|
Quayle LA, Spicer A, Ottewell PD, Holen I. Transcriptomic Profiling Reveals Novel Candidate Genes and Signalling Programs in Breast Cancer Quiescence and Dormancy. Cancers (Basel) 2021; 13:cancers13163922. [PMID: 34439077 PMCID: PMC8392441 DOI: 10.3390/cancers13163922] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 07/30/2021] [Indexed: 01/11/2023] Open
Abstract
Metastatic recurrence, the major cause of breast cancer mortality, is driven by reactivation of dormant disseminated tumour cells that are defined by mitotic quiescence and chemoresistance. The molecular mechanisms underpinning mitotic quiescence in cancer are poorly understood, severely limiting the development of novel therapies for removal of residual, metastasis-initiating tumour cells. Here, we present a molecular portrait of the quiescent breast cancer cell transcriptome across the four main breast cancer sub-types (luminal, HER2-enriched, basal-like and claudin-low) and identify a novel quiescence-associated 22-gene signature using an established lipophilic-dye (Vybrant® DiD) retention model and whole-transcriptomic profiling (mRNA-Seq). Using functional association network analysis, we elucidate the molecular interactors of these signature genes. We then go on to demonstrate that our novel 22-gene signature strongly correlates with low tumoural proliferative activity, and with dormant disease and late metastatic recurrence (≥5 years after primary tumour diagnosis) in metastatic breast cancer in multiple clinical cohorts. These genes may govern the formation and persistence of disseminated tumour cell populations responsible for breast cancer recurrence, and therefore represent prospective novel candidates to inform future development of therapeutic strategies to target disseminated tumour cells in breast cancer, eliminate minimal residual disease and prevent metastatic recurrence.
Collapse
Affiliation(s)
- Lewis A. Quayle
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; (A.S.); (P.D.O.); (I.H.)
- Correspondence: ; Tel.: +44-114-215-9209
| | - Amy Spicer
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; (A.S.); (P.D.O.); (I.H.)
- The Francis Crick Institute, Midland Road, London NW1 1AT, UK
| | - Penelope D. Ottewell
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; (A.S.); (P.D.O.); (I.H.)
| | - Ingunn Holen
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; (A.S.); (P.D.O.); (I.H.)
| |
Collapse
|
8
|
Tuy K, Rickenbacker L, Hjelmeland AB. Reactive oxygen species produced by altered tumor metabolism impacts cancer stem cell maintenance. Redox Biol 2021; 44:101953. [PMID: 34052208 PMCID: PMC8212140 DOI: 10.1016/j.redox.2021.101953] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/11/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023] Open
Abstract
Controlling reactive oxygen species (ROS) at sustainable levels can drive multiple facets of tumor biology, including within the cancer stem cell (CSC) population. Tight regulation of ROS is one key component in CSCs that drives disease recurrence, cell signaling, and therapeutic resistance. While ROS are well-appreciated to need oxygen and are a product of oxidative phosphorylation, there are also important roles for ROS under hypoxia. As hypoxia promotes and sustains major stemness pathways, further consideration of ROS impacts on CSCs in the tumor microenvironment is important. Furthermore, glycolytic shifts that occur in cancer and may be promoted by hypoxia are associated with multiple mechanisms to mitigate oxidative stress. This altered metabolism provides survival advantages that sustain malignant features, such as proliferation and self-renewal, while producing the necessary antioxidants that reduce damage from oxidative stress. Finally, disease recurrence is believed to be attributed to therapy resistant CSCs which can be quiescent and have changes in redox status. Effective DNA damage response pathways and/or a slow-cycling state can protect CSCs from the genomic catastrophe induced by irradiation and genotoxic agents. This review will explore the delicate, yet complex, relationship between ROS and its pleiotropic role in modulating the CSC.
Collapse
Affiliation(s)
- Kaysaw Tuy
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lucas Rickenbacker
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anita B Hjelmeland
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
9
|
Nik Nabil WN, Xi Z, Song Z, Jin L, Zhang XD, Zhou H, De Souza P, Dong Q, Xu H. Towards a Framework for Better Understanding of Quiescent Cancer Cells. Cells 2021; 10:cells10030562. [PMID: 33807533 PMCID: PMC7999675 DOI: 10.3390/cells10030562] [Citation(s) in RCA: 21] [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: 01/13/2021] [Revised: 02/22/2021] [Accepted: 03/02/2021] [Indexed: 12/15/2022] Open
Abstract
Quiescent cancer cells (QCCs) are cancer cells that are reversibly suspended in G0 phase with the ability to re-enter the cell cycle and initiate tumor growth, and, ultimately, cancer recurrence and metastasis. QCCs are also therapeutically challenging due to their resistance to most conventional cancer treatments that selectively act on proliferating cells. Considering the significant impact of QCCs on cancer progression and treatment, better understanding of appropriate experimental models, and the evaluation of QCCs are key questions in the field that have direct influence on potential pharmacological interventions. Here, this review focuses on existing and emerging preclinical models and detection methods for QCCs and discusses their respective features and scope for application. By providing a framework for selecting appropriate experimental models and investigative methods, the identification of the key players that regulate the survival and activation of QCCs and the development of more effective QCC-targeting therapeutic agents may mitigate the consequences of QCCs.
Collapse
Affiliation(s)
- Wan Najbah Nik Nabil
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (W.N.N.N.); (Z.X.); (Z.S.)
- Pharmaceutical Services Programme, Ministry of Health, Petaling Jaya 46200, Malaysia
| | - Zhichao Xi
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (W.N.N.N.); (Z.X.); (Z.S.)
| | - Zejia Song
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (W.N.N.N.); (Z.X.); (Z.S.)
| | - Lei Jin
- School of Medicine and Public Health, The University of Newcastle, Newcastle, NSW 2308, Australia;
| | - Xu Dong Zhang
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, NSW 2308, Australia;
| | - Hua Zhou
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China;
| | - Paul De Souza
- School of Medicine, Western Sydney University, Sydney, NSW 2751, Australia;
| | - Qihan Dong
- Chinese Medicine Anti-Cancer Evaluation Program, Greg Brown Laboratory, Central Clinical School and Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
- Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
- Correspondence: (Q.D.); (H.X.)
| | - Hongxi Xu
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China;
- Correspondence: (Q.D.); (H.X.)
| |
Collapse
|
10
|
Cseresnyes Z, Hassan MIA, Dahse HM, Voigt K, Figge MT. Quantitative Impact of Cell Membrane Fluorescence Labeling on Phagocytosis Measurements in Confrontation Assays. Front Microbiol 2020; 11:1193. [PMID: 32582113 PMCID: PMC7289966 DOI: 10.3389/fmicb.2020.01193] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 05/11/2020] [Indexed: 12/18/2022] Open
Abstract
Phagocytosis is series of steps where the pathogens and the immune cells interact during an invasion. This starts with the adhesion process between the host and pathogen cells, and is followed by the engulfment of the pathogens. Many analytical methods that are applied to characterize phagocytosis based on imaging the host-pathogen confrontation assays rely on the fluorescence labeling of cells. However, the potential effect of the membrane labeling on the quantitative results of the confrontation assays has not been studied in detail. In this study, we determine whether the fluorescence labeling processes themselves influence the results of the phagocytosis measurements. Here, alveolar macrophages, which form one of the most important compartments of the innate immune system, were used as an example of host cells, whereas Aspergillus fumigatus and Lichtheimia corymbifera that cause aspergillosis and mucormycosis, respectively, were studied as examples for pathogens. At first, our study investigated the importance of the sequence of steps of the fixation process when preparing the confrontation assay sample for microscopy studies. Here we showed that applying the fixation agent before the counter-staining causes miscalculations during the determination of the phagocytic measures. Furthermore, we also found that staining the macrophages with various concentrations of DID, as a typical membrane label, in most cases altered the capability of macrophages to phagocytose FITC-stained A. fumigatus and L. corymbifera spores in comparison with unlabeled macrophages. This effect of the DID staining showed a differential character dependent upon the labeling status and the specific type of pathogen. Moreover, labeling the spores of A. fumigatus and L. corymbifera with FITC increased the phagocytic measures during confrontation with unlabeled macrophages when compared to label-free spores. Overall, our study confirms that the staining process itself may significantly manipulate the quantitative outcome of the confrontation assay. As a result of our study, we also developed a user-friendly image analysis tool that analyses confrontation assays both with and without fluorescence labeling of the host cells and of the pathogens. Our image analysis algorithm saves experimental work effort and time, provides more precise results when calculating the phagocytic measures, and delivers a convenient analysis tool for the biologists to monitor host-pathogen interactions as they happen without the artifacts that fluorescence labeling imposes on biological interactions.
Collapse
Affiliation(s)
- Zoltan Cseresnyes
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
| | - Mohamed I. Abdelwahab Hassan
- Jena Microbial Resource Collection, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
- Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University Jena, Jena, Germany
- Department of Pests and Plant Protection, National Research Centre, Giza, Egypt
| | - Hans-Martin Dahse
- Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
| | - Kerstin Voigt
- Jena Microbial Resource Collection, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
- Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University Jena, Jena, Germany
| | - Marc Thilo Figge
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Jena, Germany
- Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University Jena, Jena, Germany
| |
Collapse
|
11
|
Voronin DV, Kozlova AA, Verkhovskii RA, Ermakov AV, Makarkin MA, Inozemtseva OA, Bratashov DN. Detection of Rare Objects by Flow Cytometry: Imaging, Cell Sorting, and Deep Learning Approaches. Int J Mol Sci 2020; 21:E2323. [PMID: 32230871 PMCID: PMC7177904 DOI: 10.3390/ijms21072323] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/25/2020] [Accepted: 03/25/2020] [Indexed: 12/14/2022] Open
Abstract
Flow cytometry nowadays is among the main working instruments in modern biology paving the way for clinics to provide early, quick, and reliable diagnostics of many blood-related diseases. The major problem for clinical applications is the detection of rare pathogenic objects in patient blood. These objects can be circulating tumor cells, very rare during the early stages of cancer development, various microorganisms and parasites in the blood during acute blood infections. All of these rare diagnostic objects can be detected and identified very rapidly to save a patient's life. This review outlines the main techniques of visualization of rare objects in the blood flow, methods for extraction of such objects from the blood flow for further investigations and new approaches to identify the objects automatically with the modern deep learning methods.
Collapse
Affiliation(s)
- Denis V. Voronin
- Laboratory of Biomedical Photoacoustics, Saratov State University, 410012 Saratov, Russia
- Department of Physical and Colloid Chemistry, National University of Oil and Gas (Gubkin University), 119991 Moscow, Russia
| | - Anastasiia A. Kozlova
- Laboratory of Biomedical Photoacoustics, Saratov State University, 410012 Saratov, Russia
| | - Roman A. Verkhovskii
- Laboratory of Biomedical Photoacoustics, Saratov State University, 410012 Saratov, Russia
- School of Urbanistics, Civil Engineering and Architecture, Yuri Gagarin State Technical University of Saratov, 410054 Saratov, Russia
| | - Alexey V. Ermakov
- Laboratory of Biomedical Photoacoustics, Saratov State University, 410012 Saratov, Russia
- Department of Biomedical Engineering, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Mikhail A. Makarkin
- Laboratory of Biomedical Photoacoustics, Saratov State University, 410012 Saratov, Russia
| | - Olga A. Inozemtseva
- Laboratory of Biomedical Photoacoustics, Saratov State University, 410012 Saratov, Russia
| | - Daniil N. Bratashov
- Laboratory of Biomedical Photoacoustics, Saratov State University, 410012 Saratov, Russia
| |
Collapse
|
12
|
Liang S, Louchami K, Holvoet B, Verbeke R, Deroose CM, Manshian B, Soenen SJ, Lentacker I, Himmelreich U. Tri-modal In vivo Imaging of Pancreatic Islets Transplanted Subcutaneously in Mice. Mol Imaging Biol 2019; 20:940-951. [PMID: 29671177 DOI: 10.1007/s11307-018-1192-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE Transplantation of pancreatic islets (PIs) is a promising therapeutic approach for type 1 diabetes. The main obstacle for this strategy is that the outcome of islet engraftment depends on the engraftment site. It was our aim to develop a strategy for using non-invasive imaging techniques to assess the location and fate of transplanted PIs longitudinally in vivo. PROCEDURES In order to overcome the limitations of individual imaging techniques and cross-validate findings by different modalities, we have combined fluorine magnetic resonance imaging (F-19 MRI), fluorescence imaging (FLI), and bioluminescent imaging (BLI) for studying subcutaneously transplanted PIs and beta cell-like cells (INS-1E cell line) in vivo. We optimized the transduction (using lentiviral vectors) and labeling procedures (using perfluoro crown ether nanoparticles with a fluorescence dye) for PIs and INS-1E cell imaging. RESULTS The feasibility of using the proposed imaging methods for PI assessment was demonstrated both in vitro and in vivo. Our data suggested that F-19 MRI is suitable for high-resolution localization of transplanted cells and PIs; FLI is essential for confirmation of contrast localization by histology; and BLI is a reliable method to assess cell viability and survival after transplantation. No significant side effects on cell viability and function have been observed. CONCLUSIONS The proposed tri-modal imaging platform is a valuable approach for the assessment of engrafted PIs in vivo. It is potentially suitable for comparing different transplantation sites and evaluating novel strategies for improving PI transplantation technique in the future.
Collapse
Affiliation(s)
- Sayuan Liang
- Biomedical MRI, Department of Imaging & Pathology, University of Leuven, Leuven, Belgium.,Bio-Imaging Lab, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp, Belgium.,Philips Research China, Shanghai, China
| | - Karim Louchami
- Biomedical MRI, Department of Imaging & Pathology, University of Leuven, Leuven, Belgium.,Laboratory of Experimental Hormonology, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Bryan Holvoet
- Nuclear Medicine & Molecular Imaging, Department of Imaging & Pathology, University of Leuven, Leuven, Belgium
| | - Rein Verbeke
- Laboratory for General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium
| | - Christophe M Deroose
- Nuclear Medicine & Molecular Imaging, Department of Imaging & Pathology, University of Leuven, Leuven, Belgium
| | - Bella Manshian
- Biomedical MRI, Department of Imaging & Pathology, University of Leuven, Leuven, Belgium
| | - Stefaan J Soenen
- Biomedical MRI, Department of Imaging & Pathology, University of Leuven, Leuven, Belgium
| | - Ine Lentacker
- Laboratory for General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium
| | - Uwe Himmelreich
- Biomedical MRI, Department of Imaging & Pathology, University of Leuven, Leuven, Belgium.
| |
Collapse
|
13
|
Quayle LA, Ottewell PD, Holen I. Chemotherapy resistance and stemness in mitotically quiescent human breast cancer cells identified by fluorescent dye retention. Clin Exp Metastasis 2018; 35:831-846. [PMID: 30377878 PMCID: PMC6267670 DOI: 10.1007/s10585-018-9946-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 10/26/2018] [Indexed: 12/12/2022]
Abstract
Metastatic recurrence in breast cancer is a major cause of mortality and often occurs many years after removal of the primary tumour. This process is driven by the reactivation of disseminated tumour cells that are characterised by mitotic quiescence and chemotherapeutic resistance. The ability to reliably isolate and characterise this cancer cell population is critical to enable development of novel therapeutic strategies for prevention of breast cancer recurrence. Here we describe the identification and characterisation of a sub-population of slow-cycling tumour cells in the MCF-7 and MDA-MB-231 human breast cancer cell lines based on their ability to retain the lipophilic fluorescent dye Vybrant® DiD for up to six passages in culture. Vybrant® DiD-retaining (DiD+) cells displayed significantly increased aldehyde dehydrogenase activity and exhibited significantly reduced sensitivity to chemotherapeutic agents compared to their rapidly dividing, Vybrant® DiD-negative (DiD−) counterparts. In addition, DiD+ cells were exclusively capable of initiating population re-growth following withdrawal of chemotherapy. The DiD+ population displayed only partial overlap with the CD44+CD24−/low cell surface protein marker signature widely used to identify breast cancer stem cells, but was enriched for CD44+CD24+ cells. Real-time qPCR profiling revealed differential expression of epithelial-to-mesenchymal transition and stemness genes between DiD+ and DiD− populations. This is the first demonstration that both MCF-7 and MDA-MB-231 human breast cancer lines contain a latent therapy-resistant population of slow-cycling cells capable of initiating population regrowth post-chemotherapy. Our data support that label-retaining cells can serve as a model for identification of molecular mechanisms driving tumour cell quiescence and de novo chemoresistance and that further characterisation of this prospective tumour-reinitiating population could yield novel therapeutic targets for elimination of the cells responsible for breast cancer recurrence.
Collapse
Affiliation(s)
- Lewis A Quayle
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK.
| | - Penelope D Ottewell
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Ingunn Holen
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| |
Collapse
|
14
|
Widner DB, Park SH, Eber MR, Shiozawa Y. Interactions Between Disseminated Tumor Cells and Bone Marrow Stromal Cells Regulate Tumor Dormancy. Curr Osteoporos Rep 2018; 16:596-602. [PMID: 30128835 PMCID: PMC6156930 DOI: 10.1007/s11914-018-0471-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
PURPOSE OF REVIEW To succinctly summarize recent findings concerning dormancy regulating interactions between bone marrow stromal cells and disseminated tumor cells. RECENT FINDINGS Recent studies have highlighted roles of the bone marrow microenviroment, including osteoblasts, mesenchymal stem cells (MSCs), and endothelial cells, in inducing or maintaining cancer cell dormancy. Key pathways of interest include: osteoblast-induced transforming growth factor (TGF)-β2 signaling, transfer of MSC-derived exosomes containing dormancy inducing microRNA, cancer cell cannibalism of MSCs, and endothelial cell secretion of thrombospondin 1 (TSP1). The bone marrow is a common site of metastatic disease recurrence following a period of cancer cell dormancy. Understanding why disseminated tumor cells enter into dormancy and later resume cell proliferation and growth is vital to developing effective therapeutics against these cells. The bone marrow stroma and the various pathways through which it participates in crosstalk with cancer cells are essential to furthering understanding of how dormancy is regulated.
Collapse
Affiliation(s)
- D Brooke Widner
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157-1082, USA
| | - Sun H Park
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157-1082, USA
| | - Matthew R Eber
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157-1082, USA
| | - Yusuke Shiozawa
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157-1082, USA.
| |
Collapse
|
15
|
Bilousova T, Elias C, Miyoshi E, Alam MP, Zhu C, Campagna J, Vadivel K, Jagodzinska B, Gylys KH, John V. Suppression of tau propagation using an inhibitor that targets the DK-switch of nSMase2. Biochem Biophys Res Commun 2018; 499:751-757. [PMID: 29604274 DOI: 10.1016/j.bbrc.2018.03.209] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 03/27/2018] [Indexed: 11/19/2022]
Abstract
Targeting of molecular pathways involved in the cell-to-cell propagation of pathological tau species is a novel approach for development of disease-modifying therapies that could block tau pathology and attenuate cognitive decline in patients with Alzheimer's disease and other tauopathies. We discovered cambinol through a screening effort and show that it is an inhibitor of cell-to-cell tau propagation. Our in vitro data demonstrate that cambinol inhibits neutral sphingomyelinase 2 (nSMase2) enzyme activity in dose response fashion, and suppresses extracellular vesicle (EV) production while reducing tau seed propagation. Our in vivo testing with cambinol shows that it can reduce the nSMase2 activity in the brain after oral administration. Our molecular docking and simulation analysis reveals that cambinol can target the DK-switch in the nSMase2 active site.
Collapse
Affiliation(s)
- Tina Bilousova
- Drug Discovery Lab, Department of Neurology, University of California, Los Angeles, CA, USA; Gylys Lab, School of Nursing, University of California, Los Angeles, CA, USA
| | - Chris Elias
- Drug Discovery Lab, Department of Neurology, University of California, Los Angeles, CA, USA
| | - Emily Miyoshi
- Gylys Lab, School of Nursing, University of California, Los Angeles, CA, USA
| | - Mohammad Parvez Alam
- Drug Discovery Lab, Department of Neurology, University of California, Los Angeles, CA, USA
| | - Chunni Zhu
- Drug Discovery Lab, Department of Neurology, University of California, Los Angeles, CA, USA
| | - Jesus Campagna
- Drug Discovery Lab, Department of Neurology, University of California, Los Angeles, CA, USA
| | - Kanagasabai Vadivel
- Drug Discovery Lab, Department of Neurology, University of California, Los Angeles, CA, USA
| | - Barbara Jagodzinska
- Drug Discovery Lab, Department of Neurology, University of California, Los Angeles, CA, USA
| | - Karen Hoppens Gylys
- Gylys Lab, School of Nursing, University of California, Los Angeles, CA, USA
| | - Varghese John
- Drug Discovery Lab, Department of Neurology, University of California, Los Angeles, CA, USA.
| |
Collapse
|
16
|
Pascual G, Avgustinova A, Mejetta S, Martín M, Castellanos A, Attolini CSO, Berenguer A, Prats N, Toll A, Hueto JA, Bescós C, Di Croce L, Benitah SA. Targeting metastasis-initiating cells through the fatty acid receptor CD36. Nature 2016; 541:41-45. [PMID: 27974793 DOI: 10.1038/nature20791] [Citation(s) in RCA: 917] [Impact Index Per Article: 114.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 11/16/2016] [Indexed: 12/14/2022]
Abstract
The fact that the identity of the cells that initiate metastasis in most human cancers is unknown hampers the development of antimetastatic therapies. Here we describe a subpopulation of CD44bright cells in human oral carcinomas that do not overexpress mesenchymal genes, are slow-cycling, express high levels of the fatty acid receptor CD36 and lipid metabolism genes, and are unique in their ability to initiate metastasis. Palmitic acid or a high-fat diet specifically boosts the metastatic potential of CD36+ metastasis-initiating cells in a CD36-dependent manner. The use of neutralizing antibodies to block CD36 causes almost complete inhibition of metastasis in immunodeficient or immunocompetent orthotopic mouse models of human oral cancer, with no side effects. Clinically, the presence of CD36+ metastasis-initiating cells correlates with a poor prognosis for numerous types of carcinomas, and inhibition of CD36 also impairs metastasis, at least in human melanoma- and breast cancer-derived tumours. Together, our results indicate that metastasis-initiating cells particularly rely on dietary lipids to promote metastasis.
Collapse
Affiliation(s)
- Gloria Pascual
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Alexandra Avgustinova
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Stefania Mejetta
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Mercè Martín
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Andrés Castellanos
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Camille Stephan-Otto Attolini
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Antoni Berenguer
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Neus Prats
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Agustí Toll
- IMIM, Department of Dermatology, Hospital del Mar, 08003 Barcelona
| | - Juan Antonio Hueto
- Vall D´Hebron Hospital, Barcelona, Department of Oral and Maxillofacial Surgery, Universitat Autònoma de Barcelona, Barcelona 08035 Spain
| | - Coro Bescós
- Vall D´Hebron Hospital, Barcelona, Department of Oral and Maxillofacial Surgery, Universitat Autònoma de Barcelona, Barcelona 08035 Spain
| | - Luciano Di Croce
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Dr. Aiguader 88, 08003 Barcelona, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona 08002, Spain
| | - Salvador Aznar Benitah
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
| |
Collapse
|
17
|
Cai X, Zhang CJ, Ting Wei Lim F, Chan SJ, Bandla A, Chuan CK, Hu F, Xu S, Thakor NV, Liao LD, Liu B. Organic Nanoparticles with Aggregation-Induced Emission for Bone Marrow Stromal Cell Tracking in a Rat PTI Model. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:6576-6585. [PMID: 27592863 DOI: 10.1002/smll.201601630] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 06/24/2016] [Indexed: 05/13/2023]
Abstract
Stem-cell based therapy is an emerging therapeutic approach for ischemic stroke treatment. Bone marrow stromal cells (BMSCs) are in common use as a cell source for stem cell therapy and show promising therapeutic outcomes for stroke treatment. One challenge is to develop a reliable tracking strategy to monitor the fate of BMSCs and assess their therapeutic effects in order to improve the success rate of such treatment. Herein, TPEEP, a fluorogen with aggregation-induced emission characteristics and near-infrared emission are designed and synthesized and further fabricated into organic nanoparticles (NPs). The obtained NPs show high fluorescence quantum yield, low cytotoxicity with good physical and photostability, which display excellent tracking performance of BMSCs in vitro and in vivo. Using a rat photothrombotic ischemia model as an example, the NP-labeled BMSCs are able to migrate to the stroke lesion site to yield bright red fluorescence. Immunofluorescence staining shows that the NP labeling does not affect the normal function of BMSCs, proving their good biocompatibility in vivo. These merits make TPEEP NP a potential cell tracker to evaluate the fate of BMSCs in cell therapy.
Collapse
Affiliation(s)
- Xiaolei Cai
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, 28 Medical Drive, #05-01, Singapore, 117456
| | - Chong-Jing Zhang
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585
| | - Frances Ting Wei Lim
- Singapore Institute for Neurotechnology (SINAPSE), National University of Singapore, 28 Medical Drive, Singapore, 117456
| | - Su Jing Chan
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 2129, USA
| | - Aishwarya Bandla
- Singapore Institute for Neurotechnology (SINAPSE), National University of Singapore, 28 Medical Drive, Singapore, 117456
| | - Chan Kim Chuan
- Singapore Institute for Neurotechnology (SINAPSE), National University of Singapore, 28 Medical Drive, Singapore, 117456
| | - Fang Hu
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585
| | - Shidang Xu
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585
| | - Nitish V Thakor
- Singapore Institute for Neurotechnology (SINAPSE), National University of Singapore, 28 Medical Drive, Singapore, 117456
| | - Lun-De Liao
- Singapore Institute for Neurotechnology (SINAPSE), National University of Singapore, 28 Medical Drive, Singapore, 117456
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, 35 Keyan Road, Zhunan Town, Miaoli County, 35053, Taiwan
| | - Bin Liu
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585
| |
Collapse
|
18
|
Axl is required for TGF-β2-induced dormancy of prostate cancer cells in the bone marrow. Sci Rep 2016; 6:36520. [PMID: 27819283 PMCID: PMC5098246 DOI: 10.1038/srep36520] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 10/14/2016] [Indexed: 01/02/2023] Open
Abstract
Disseminated prostate cancer (PCa) cells in the marrow survive for years without evidence of proliferation, while maintaining the capacity to develop into metastatic lesions. These dormant disseminated tumor cells (DTCs) may reside in close proximity to osteoblasts, while expressing high levels of Axl, one of the tyrosine kinase receptors for growth arrest specific 6 (Gas6). Yet how Axl regulates DTC proliferation in marrow remains undefined. Here, we explored the impact of the loss of Axl in PCa cells (PC3 and DU145) on the induction of their dormancy when they are co-cultured with a pre-osteoblastic cell line, MC3T3-E1. MC3T3-E1 cells dramatically decrease the proliferation of PCa cells, however this suppressive effect of osteoblasts is significantly reduced by the reduction of Axl expression in PCa cells. Interestingly, expression of both TGF-β and its receptors were regulated by Axl expression in PCa cells, while specific blockade of TGF-β signaling limited the ability of the osteoblasts to induce dormancy of PCa cells. Finally, we found that both Gas6 and Axl are required for TGF-β2-mediated cell growth suppression. Taken together, these data suggest that a loop between the Gas6/Axl axis and TGF-β2 signaling plays a significant role in the induction of PCa cell dormancy.
Collapse
|
19
|
Lehmann TP, Juzwa W, Filipiak K, Sujka-Kordowska P, Zabel M, Głowacki J, Głowacki M, Jagodziński PP. Quantification of the asymmetric migration of the lipophilic dyes, DiO and DiD, in homotypic co-cultures of chondrosarcoma SW-1353 cells. Mol Med Rep 2016; 14:4529-4536. [PMID: 27748852 PMCID: PMC5101988 DOI: 10.3892/mmr.2016.5793] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 08/15/2016] [Indexed: 12/15/2022] Open
Abstract
DiO and DiD are lipophilic cell labelling dyes used in the staining of cells in vivo and in vitro. The aim of the present study was to quantify the asymmetrical distribution of dyes in co-cultured cells and to measure the intercellular transfer of DiO and DiD. DiO and DiD were applied separately to stain two identical populations of SW-1353 human chondrosarcoma cells that were subsequently co-cultured (homotypic co-culture). The intercellular migration of dyes in the co-cultured cells was measured by flow cytometry and recorded under a fluorescent microscope. DiD and DiO caused no effect on the proliferation of cells, the degradation rate of the two dyes was comparable and crossover effects between dyes were negligible. The results of the present study suggested that asymmetrical intercellular migration of DiD and DiO was responsible for the asymmetrical distribution of these dyes in co-cultured cells. To take advantage of the lipophilic dyes migration in the double-stained co-cultured cells we suggest to apply mixed-dyes controls prior to the flow cytometric analysis. These controls are performed by staining cells with a 1:1 mix of the two dyes and would enable the estimation of the intensity of intercellular contact in co-culture systems. A 1:1 premix of DiO and DiD was applied to estimate cellular effect on intercellular exchange of lipid dyes in co-cultures incubated with cycloheximide and cytochalasin B. The cellular effect contributed 6–7% of intercellular migration of the lipophilic dyes, DiO and DiD. The majority of the observed intercellular transfer of these dyes was due to non-cellular, passive transfer.
Collapse
Affiliation(s)
- Tomasz P Lehmann
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 60‑781 Poznan, Poland
| | - Wojciech Juzwa
- Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, 60‑627 Poznan, Poland
| | - Krystyna Filipiak
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60‑781 Poznan, Poland
| | - Patrycja Sujka-Kordowska
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60‑781 Poznan, Poland
| | - Maciej Zabel
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60‑781 Poznan, Poland
| | | | - Maciej Głowacki
- Department of Paediatric Orthopaedics and Traumatology, Poznan University of Medical Sciences, 61‑545 Poznan, Poland
| | - Paweł P Jagodziński
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 60‑781 Poznan, Poland
| |
Collapse
|
20
|
Wang N, Reeves KJ, Brown HK, Fowles ACM, Docherty FE, Ottewell PD, Croucher PI, Holen I, Eaton CL. The frequency of osteolytic bone metastasis is determined by conditions of the soil, not the number of seeds; evidence from in vivo models of breast and prostate cancer. J Exp Clin Cancer Res 2015; 34:124. [PMID: 26480944 PMCID: PMC4615337 DOI: 10.1186/s13046-015-0240-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 10/12/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND While both preclinical and clinical studies suggest that the frequency of growing skeletal metastases is elevated in individuals with higher bone turnover, it is unclear whether this is a result of increased numbers of tumour cells arriving in active sites or of higher numbers of tumour cells being induced to divide by the bone micro-environment. Here we have investigated how the differences in bone turnover affect seeding of tumour cells and/or development of overt osteolytic bone metastasis using in vivo models of hormone-independent breast and prostate cancer. METHODS Cohorts of 6 (young) and 16 (mature)-week old BALB/c nude mice were culled 1, 7 and 21 days after received intracardiac injection of luciferase expressing human prostate (PC3) or breast cancer (MDA-MB-231) cell lines labelled with a fluorescent cell membrane dye (Vybrant DiD). The presence of growing bone metastases was determined by bioluminescence using an in vivo imaging system (IVIS) and followed by anatomical confirmation of tumour metastatic sites post mortem, while the presence of individual fluorescently labelled tumour cells was evaluated using two-photon microscopy ex vivo. The bone remodelling activities were compared between young and mature naïve mice (both male and female) using micro-CT analysis, ELISA and bone histomorphometry. RESULTS Both prostate and breast cancer cells generated higher numbers of overt skeletal lesions in young mice (~80%) than in mature mice (~20%). Although mature mice presented with fewer overt bone metastases, the number of tumour cells arriving/colonizing in the tibias was comparable between young and mature animals. Young naïve mice had lower bone volume but higher bone formation and resorption activities compared to mature animals. CONCLUSIONS Our studies suggest that higher frequencies of growing osteolytic skeletal metastases in these models are linked to increased bone turnover and not to the initial number of tumour cells entering the bone microenvironment.
Collapse
Affiliation(s)
- Ning Wang
- The Mellanby Centre for Bone Research, Department of Human Metabolism, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK.
| | - Kimberley J Reeves
- Break Through Breast Cancer Research Unit, Paterson Institute for Cancer Research Manchester, Manchester, UK.
| | - Hannah K Brown
- The Mellanby Centre for Bone Research, Department of Human Metabolism, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK.
| | - Anne C M Fowles
- The Mellanby Centre for Bone Research, Department of Human Metabolism, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK.
| | - Freyja E Docherty
- The Mellanby Centre for Bone Research, Department of Human Metabolism, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK.
| | - Penelope D Ottewell
- Department of Oncology, Medical School, University of Sheffield, Sheffield, UK.
| | - Peter I Croucher
- Bone Biology Division, Garvan Institute of Medical Research, Sydney, Australia.
| | - Ingunn Holen
- Department of Oncology, Medical School, University of Sheffield, Sheffield, UK.
| | - Colby L Eaton
- The Mellanby Centre for Bone Research, Department of Human Metabolism, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK.
| |
Collapse
|
21
|
Wang N, Docherty F, Brown HK, Reeves K, Fowles A, Lawson M, Ottewell PD, Holen I, Croucher PI, Eaton CL. Mitotic quiescence, but not unique "stemness," marks the phenotype of bone metastasis-initiating cells in prostate cancer. FASEB J 2015; 29:3141-50. [PMID: 25888599 DOI: 10.1096/fj.14-266379] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 03/31/2015] [Indexed: 01/09/2023]
Abstract
This study aimed to identify subpopulations of prostate cancer cells that are responsible for the initiation of bone metastases. Using rapidly dividing human prostate cancer cell lines, we identified mitotically quiescent subpopulations (<1%), which we compared with the rapidly dividing populations for patterns of gene expression and for their ability to migrate to the skeletons of athymic mice. The study used 2-photon microscopy to map the presence/distribution of fluorescently labeled, quiescent cells and luciferase expression to determine the presence of growing bone metastases. We showed that the mitotically quiescent cells were very significantly more tumorigenic in forming bone metastases than fast-growing cells (55 vs. 15%) and had a unique gene expression profile. The quiescent cells were not uniquely stem cell like, with no expression of CD133 but had the same level expression of other putative prostate stem cell markers (CD44 and integrins α2/β1), when compared to the rapidly proliferating population. In addition, mitotic quiescence was associated with very high levels of C-X-C chemokine receptor type 4 (CXCR4) production. Inhibition of CXCR4 activity altered the homing of quiescent tumor cells to bone. Our studies suggest that mitotic dormancy is a unique phenotype that facilitates tumor cell colonization of the skeleton in prostate cancer.
Collapse
Affiliation(s)
- Ning Wang
- *Department of Human Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom; Breakthrough Breast Cancer Research Unit, Paterson Institute for Cancer Research, Manchester, United Kingdom; Department of Oncology, Medical School, University of Sheffield, Sheffield, United Kingdom; and Bone Biology Division, Garvan Institute of Medical Research, Sydney, Australia
| | - Freyja Docherty
- *Department of Human Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom; Breakthrough Breast Cancer Research Unit, Paterson Institute for Cancer Research, Manchester, United Kingdom; Department of Oncology, Medical School, University of Sheffield, Sheffield, United Kingdom; and Bone Biology Division, Garvan Institute of Medical Research, Sydney, Australia
| | - Hannah K Brown
- *Department of Human Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom; Breakthrough Breast Cancer Research Unit, Paterson Institute for Cancer Research, Manchester, United Kingdom; Department of Oncology, Medical School, University of Sheffield, Sheffield, United Kingdom; and Bone Biology Division, Garvan Institute of Medical Research, Sydney, Australia
| | - Kim Reeves
- *Department of Human Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom; Breakthrough Breast Cancer Research Unit, Paterson Institute for Cancer Research, Manchester, United Kingdom; Department of Oncology, Medical School, University of Sheffield, Sheffield, United Kingdom; and Bone Biology Division, Garvan Institute of Medical Research, Sydney, Australia
| | - Anne Fowles
- *Department of Human Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom; Breakthrough Breast Cancer Research Unit, Paterson Institute for Cancer Research, Manchester, United Kingdom; Department of Oncology, Medical School, University of Sheffield, Sheffield, United Kingdom; and Bone Biology Division, Garvan Institute of Medical Research, Sydney, Australia
| | - Michelle Lawson
- *Department of Human Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom; Breakthrough Breast Cancer Research Unit, Paterson Institute for Cancer Research, Manchester, United Kingdom; Department of Oncology, Medical School, University of Sheffield, Sheffield, United Kingdom; and Bone Biology Division, Garvan Institute of Medical Research, Sydney, Australia
| | - Penelope D Ottewell
- *Department of Human Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom; Breakthrough Breast Cancer Research Unit, Paterson Institute for Cancer Research, Manchester, United Kingdom; Department of Oncology, Medical School, University of Sheffield, Sheffield, United Kingdom; and Bone Biology Division, Garvan Institute of Medical Research, Sydney, Australia
| | - Ingunn Holen
- *Department of Human Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom; Breakthrough Breast Cancer Research Unit, Paterson Institute for Cancer Research, Manchester, United Kingdom; Department of Oncology, Medical School, University of Sheffield, Sheffield, United Kingdom; and Bone Biology Division, Garvan Institute of Medical Research, Sydney, Australia
| | - Peter I Croucher
- *Department of Human Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom; Breakthrough Breast Cancer Research Unit, Paterson Institute for Cancer Research, Manchester, United Kingdom; Department of Oncology, Medical School, University of Sheffield, Sheffield, United Kingdom; and Bone Biology Division, Garvan Institute of Medical Research, Sydney, Australia
| | - Colby L Eaton
- *Department of Human Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom; Breakthrough Breast Cancer Research Unit, Paterson Institute for Cancer Research, Manchester, United Kingdom; Department of Oncology, Medical School, University of Sheffield, Sheffield, United Kingdom; and Bone Biology Division, Garvan Institute of Medical Research, Sydney, Australia
| |
Collapse
|
22
|
Wang N, Docherty FE, Brown HK, Reeves KJ, Fowles ACM, Ottewell PD, Dear TN, Holen I, Croucher PI, Eaton CL. Prostate cancer cells preferentially home to osteoblast-rich areas in the early stages of bone metastasis: evidence from in vivo models. J Bone Miner Res 2014; 29:2688-96. [PMID: 24956445 DOI: 10.1002/jbmr.2300] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 06/06/2014] [Accepted: 06/13/2014] [Indexed: 12/31/2022]
Abstract
It has been suggested that metastasis-initiating cells gain a foothold in bone by homing to a metastastatic microenvironment (or "niche"). Whereas the precise nature of this niche remains to be established, it is likely to contain bone cell populations including osteoblasts and osteoclasts. In the mouse tibia, the distribution of osteoblasts on endocortical bone surfaces is non-uniform, and we hypothesize that studying co-localization of individual tumor cells with resident cell populations will reveal the identity of critical cellular components of the niche. In this study, we have mapped the distribution of three human prostate cancer cell lines (PC3-NW1, LN-CaP, and C4 2B4) colonizing the tibiae of athymic mice following intracardiac injection and evaluated their interaction with potential metastatic niches. Prostate cancer cells labeled with the fluorescent cell membrane dye (Vybrant DiD) were found by two-photon microscopy to be engrafted in the tibiae in close proximity (∼40 µm) to bone surfaces and 70% more cancer cells were detected in the lateral compared to the medial endocortical bone regions. This was associated with a 5-fold higher number of osteoblasts and 7-fold higher bone formation rate on the lateral endocortical bone surface compared to the medial side. By disrupting cellular interactions mediated by the chemokine (C-X-C motif) receptor 4 (CXCR4)/chemokine ligand 12 (CXCL12) axis with the CXCR4 inhibitor AMD3100, the preferential homing pattern of prostate cancer cells to osteoblast-rich bone surfaces was disrupted. In this study, we map the location of prostate cancer cells that home to endocortical regions in bone and our data demonstrate that homing of prostate cancer cells is associated with the presence and activity of osteoblast lineage cells, and suggest that therapies targeting osteoblast niches should be considered to prevent development of incurable prostate cancer bone metastases.
Collapse
Affiliation(s)
- Ning Wang
- The Mellanby Centre for Bone Research, Department of Human Metabolism, The University of Sheffield, Sheffield, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|