1
|
Ahmed F, Riza YM. A Systems Bioinformatics Analysis Indicates that Disruption of the lncRNA SFTA1P Network is Consistent with Impairing Surfactant Homeostasis and Respiratory Function Observed in Lung Adenocarcinoma. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2025. [PMID: 40353598 DOI: 10.1089/omi.2024.0209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Lung adenocarcinoma (LUAD) is one of the leading global health challenges wherein novel therapeutic targets are much needed. In this systems bioinformatics study, we report that disruption of the long noncoding RNA (lncRNA) SFTA1P-centered network, respiratory gaseous exchange and surfactant-associated Biological Network (rgsBNet), is consistent with impairing surfactant homeostasis and respiratory function, and thus warrants attention for future drug discovery and development. We analyzed data from The Cancer Genome Atlas LUAD cohort to identify differentially expressed mRNAs, lncRNAs, and microRNAs (miRNAs), followed by correlational analysis to examine the coexpression network of lncRNA SFTA1P and its potential role in LUAD pathogenesis. We observed the downregulation of lncRNA SFTA1P and its coexpressed network in LUAD. Intriguingly, this network appears to be associated with disrupting surfactant homeostasis and perturbing respiratory function, suggesting a potential role in LUAD progression. Additionally, we identified key transcription factors that correlate with the expression of genes crucial for respiratory gaseous exchange and surfactant homeostasis. The attendant regulatory mechanisms suggested that SFTA1P may act as a "sponge" for certain miRNAs, sequestering them away from their mRNA targets. In conclusion, this work uncovers novel insights into the molecular mechanisms governing surfactant homeostasis in LUAD and offers a possible avenue for therapeutic interventions aimed at ameliorating lung function and improving disease management. The downregulation of lncRNA SFTA1P and its coexpressed network highlights their potential as regulators of lung function and opens doors for further investigation into their role in LUAD progression and as potential therapeutic targets.
Collapse
Affiliation(s)
- Firoz Ahmed
- Department of Biological Sciences, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Yasir Mohamed Riza
- Faculty of Science, Department of Biochemistry, King Abdulaziz University, Jeddah, Saudi Arabia
- Centre of Artificial Intelligence for Precision Medicines, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
2
|
Mu X, Zhou Y, Yu Y, Zhang M, Liu J. The roles of cancer stem cells and therapeutic implications in melanoma. Front Immunol 2024; 15:1486680. [PMID: 39611156 PMCID: PMC11602477 DOI: 10.3389/fimmu.2024.1486680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 10/28/2024] [Indexed: 11/30/2024] Open
Abstract
Melanoma is a highly malignant skin tumor characterized by high metastasis and poor prognosis. Recent studies have highlighted the pivotal role of melanoma stem cells (MSCs)-a subpopulation of cancer stem cells (CSCs)-in driving tumor growth, metastasis, therapeutic resistance, and recurrence. Similar to CSCs in other cancers, MSCs possess unique characteristics, including specific surface markers, dysregulated signaling pathways, and the ability to thrive within complex tumor microenvironment (TME). This review explored the current landscape of MSC research, discussing the identification of MSC-specific surface markers, the role of key signaling pathways such as Wnt/β-catenin, Notch, and Hedgehog (Hh), and how interactions within the TME, including hypoxia and immune cells, contribute to MSC-mediated drug resistance and metastatic behavior. Furthermore, we also investigated the latest therapeutic strategies targeting MSCs, such as small-molecule inhibitors, immune-based approaches, and novel vaccine developments, with an emphasis on their potential to overcome melanoma progression and improve clinical outcomes. This review aims to provide valuable insights into the complex roles of MSCs in melanoma biology and offers perspectives for future research and therapeutic advances against this challenging disease.
Collapse
Affiliation(s)
- Xiaoli Mu
- The Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yixin Zhou
- The Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yongxin Yu
- The Department of Plastic and Reconstructive Surgery, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Mingyi Zhang
- The Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiyan Liu
- The Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
3
|
Ma G, Wang J, Fu J, Chen R, Liang M, Li M, Xia T, Liu X, Wang S. Heterogeneous circulating tumor cells correlate with responses to neoadjuvant chemotherapy and prognosis in patients with locally advanced breast cancer. Breast Cancer Res Treat 2023:10.1007/s10549-023-06942-y. [PMID: 37311933 DOI: 10.1007/s10549-023-06942-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 04/05/2023] [Indexed: 06/15/2023]
Abstract
Neoadjuvant chemotherapy (NCT) is the standard treatment for patients with locally advanced breast cancer (LABC). The predictive value of heterogeneous circulating tumor cells (CTCs) in NCT response has not been determined. All patients were staged as LABC, and blood samples were collected at the time of biopsy, and after the first and eighth NCT courses. Patients were divided into High responders (High-R) and Low responders (Low-R) according to Miller-Payne system and changes in Ki-67 levels after NCT treatment. A novel SE-i·FISH strategy was applied to detect CTCs. Heterogeneities were successfully analyzed in patients undergoing NCT. Total CTCs increased continuously and were higher in Low-R group, while in High-R group, CTCs increased slightly during NCT before returning to baseline levels. Triploid and tetraploid chromosome 8 increased in Low-R but not High-R group. The number of small CTCs in Low-R group increased significantly until the last sample, however, remained constant in High-R group. The patients with more CTCs had shorter PFS and OS than those with less CTCs after the eighth course of NCT. Total CTCs following NCT could predict patients' responses. More detailed characterizations of CTC blood profiles may improve predictive capacity and treatments of LABC.
Collapse
Affiliation(s)
- Ge Ma
- Department of Breast Surgery, The First Affiliated Hospital With Nanjing Medical University, 300 Guang-Zhou Road, Nanjing, 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Jingyi Wang
- Department of Breast Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, 29 Xinglong Lane, Changzhou, 213003, China
| | - Jingyue Fu
- Department of Breast Surgery, The First Affiliated Hospital With Nanjing Medical University, 300 Guang-Zhou Road, Nanjing, 210029, China
| | - Rui Chen
- Department of Breast Surgery, The First Affiliated Hospital With Nanjing Medical University, 300 Guang-Zhou Road, Nanjing, 210029, China
| | - Mengdi Liang
- Department of Breast Surgery, The First Affiliated Hospital With Nanjing Medical University, 300 Guang-Zhou Road, Nanjing, 210029, China
| | - Minghui Li
- Department of Breast Surgery, The First Affiliated Hospital With Nanjing Medical University, 300 Guang-Zhou Road, Nanjing, 210029, China
| | - Tiansong Xia
- Department of Breast Surgery, The First Affiliated Hospital With Nanjing Medical University, 300 Guang-Zhou Road, Nanjing, 210029, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Xiaoan Liu
- Department of Breast Surgery, The First Affiliated Hospital With Nanjing Medical University, 300 Guang-Zhou Road, Nanjing, 210029, China.
| | - Shui Wang
- Department of Breast Surgery, The First Affiliated Hospital With Nanjing Medical University, 300 Guang-Zhou Road, Nanjing, 210029, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
4
|
Ash S, Askenasy N. Immunotherapy for neuroblastoma by hematopoietic cell transplantation and post-transplant immunomodulation. Crit Rev Oncol Hematol 2023; 185:103956. [PMID: 36893946 DOI: 10.1016/j.critrevonc.2023.103956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 12/14/2022] [Accepted: 03/04/2023] [Indexed: 03/09/2023] Open
Abstract
Neuroblastoma represents a relatively common childhood tumor that imposes therapeutic difficulties. High risk neuroblastoma patients have poor prognosis, display limited response to radiochemotherapy and may be treated by hematopoietic cell transplantation. Allogeneic and haploidentical transplants have the distinct advantage of reinstitution of immune surveillance, reinforced by antigenic barriers. The key factors favorable to ignition of potent anti-tumor reactions are transition to adaptive immunity, recovery from lymphopenia and removal of inhibitory signals that inactivate immune cells at the local and systemic levels. Post-transplant immunomodulation may further foster anti-tumor reactivity, with positive but transient impact of infusions of lymphocytes and natural killer cells both from the donor, the recipient or third party. The most promising approaches include introduction of antigen-presenting cells in early post-transplant stages and neutralization of inhibitory signals. Further studies will likely shed light on the nature and actions of suppressor factors within tumor stroma and at the systemic level.
Collapse
Affiliation(s)
- Shifra Ash
- Department of Pediatric Hematology-Oncology, Rambam Medical Center, Haifa, Israel; Frankel Laboratory of Bone Marrow Transplantation, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.
| | - Nadir Askenasy
- Frankel Laboratory of Bone Marrow Transplantation, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| |
Collapse
|
5
|
Oh JH, Choi W, Ko E, Kang M, Tannenbaum A, Deasy JO. PathCNN: interpretable convolutional neural networks for survival prediction and pathway analysis applied to glioblastoma. Bioinformatics 2021; 37:i443-i450. [PMID: 34252964 PMCID: PMC8336441 DOI: 10.1093/bioinformatics/btab285] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
MOTIVATION Convolutional neural networks (CNNs) have achieved great success in the areas of image processing and computer vision, handling grid-structured inputs and efficiently capturing local dependencies through multiple levels of abstraction. However, a lack of interpretability remains a key barrier to the adoption of deep neural networks, particularly in predictive modeling of disease outcomes. Moreover, because biological array data are generally represented in a non-grid structured format, CNNs cannot be applied directly. RESULTS To address these issues, we propose a novel method, called PathCNN, that constructs an interpretable CNN model on integrated multi-omics data using a newly defined pathway image. PathCNN showed promising predictive performance in differentiating between long-term survival (LTS) and non-LTS when applied to glioblastoma multiforme (GBM). The adoption of a visualization tool coupled with statistical analysis enabled the identification of plausible pathways associated with survival in GBM. In summary, PathCNN demonstrates that CNNs can be effectively applied to multi-omics data in an interpretable manner, resulting in promising predictive power while identifying key biological correlates of disease. AVAILABILITY AND IMPLEMENTATION The source code is freely available at: https://github.com/mskspi/PathCNN.
Collapse
Affiliation(s)
- Jung Hun Oh
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Wookjin Choi
- Department of Computer Science, Virginia State University, Petersburg, VA 23806, USA
| | - Euiseong Ko
- Department of Computer Science, University of Nevada, Las Vegas, NV 89154, USA
| | - Mingon Kang
- Department of Computer Science, University of Nevada, Las Vegas, NV 89154, USA
| | - Allen Tannenbaum
- Departments of Computer Science and Applied Mathematics & Statistics, Stony Brook University, New York, NY 11794, USA
| | - Joseph O Deasy
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
6
|
Somasundaram R, Connelly T, Choi R, Choi H, Samarkina A, Li L, Gregorio E, Chen Y, Thakur R, Abdel-Mohsen M, Beqiri M, Kiernan M, Perego M, Wang F, Xiao M, Brafford P, Yang X, Xu X, Secreto A, Danet-Desnoyers G, Traum D, Kaestner KH, Huang AC, Hristova D, Wang J, Fukunaga-Kalabis M, Krepler C, Ping-Chen F, Zhou X, Gutierrez A, Rebecca VW, Vonteddu P, Dotiwala F, Bala S, Majumdar S, Dweep H, Wickramasinghe J, Kossenkov AV, Reyes-Arbujas J, Santiago K, Nguyen T, Griss J, Keeney F, Hayden J, Gavin BJ, Weiner D, Montaner LJ, Liu Q, Peiffer L, Becker J, Burton EM, Davies MA, Tetzlaff MT, Muthumani K, Wargo JA, Gabrilovich D, Herlyn M. Tumor-infiltrating mast cells are associated with resistance to anti-PD-1 therapy. Nat Commun 2021; 12:346. [PMID: 33436641 PMCID: PMC7804257 DOI: 10.1038/s41467-020-20600-7] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 12/10/2020] [Indexed: 12/11/2022] Open
Abstract
Anti-PD-1 therapy is used as a front-line treatment for many cancers, but mechanistic insight into this therapy resistance is still lacking. Here we generate a humanized (Hu)-mouse melanoma model by injecting fetal liver-derived CD34+ cells and implanting autologous thymus in immune-deficient NOD-scid IL2Rγnull (NSG) mice. Reconstituted Hu-mice are challenged with HLA-matched melanomas and treated with anti-PD-1, which results in restricted tumor growth but not complete regression. Tumor RNA-seq, multiplexed imaging and immunohistology staining show high expression of chemokines, as well as recruitment of FOXP3+ Treg and mast cells, in selective tumor regions. Reduced HLA-class I expression and CD8+/Granz B+ T cells homeostasis are observed in tumor regions where FOXP3+ Treg and mast cells co-localize, with such features associated with resistance to anti-PD-1 treatment. Combining anti-PD-1 with sunitinib or imatinib results in the depletion of mast cells and complete regression of tumors. Our results thus implicate mast cell depletion for improving the efficacy of anti-PD-1 therapy. Immune checkpoint therapies (ICT) are promising for treating various cancers, but response rates vary. Here the authors show, in mouse models, that tumor-infiltrating mast cells colocalize with regulatory T cells, coincide with local reduction of MHC-I and CD8 T cells, and is associated with resistance to ICT, which can be reversed by c-kit inhibitor treatment.
Collapse
Affiliation(s)
| | | | - Robin Choi
- The Wistar Institute, Philadelphia, PA, USA
| | | | | | - Ling Li
- The Wistar Institute, Philadelphia, PA, USA
| | | | | | - Rohit Thakur
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | | | | | | | | | - Fang Wang
- The Wistar Institute, Philadelphia, PA, USA
| | - Min Xiao
- The Wistar Institute, Philadelphia, PA, USA
| | | | - Xue Yang
- The Wistar Institute, Philadelphia, PA, USA
| | - Xiaowei Xu
- Department of Pathology and Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anthony Secreto
- Department of Medicine, Stem Cell and Xenograft Core, University of Pennsylvania, Philadelphia, PA, USA
| | - Gwenn Danet-Desnoyers
- Department of Medicine, Stem Cell and Xenograft Core, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel Traum
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexander C Huang
- Department of Pathology and Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Johannes Griss
- Division of Immunology, Allergy and Infectious Diseases (DIAID), Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | - Qin Liu
- The Wistar Institute, Philadelphia, PA, USA
| | | | | | - Elizabeth M Burton
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Michael A Davies
- Department of Melanoma Medical Oncology, University of California, San Francisco, CA, USA
| | - Michael T Tetzlaff
- Department of Pathology and Dermatology, University of California, San Francisco, CA, USA
| | - Kar Muthumani
- The Wistar Institute, Philadelphia, PA, USA.,GeneOne Life Science Inc., Fort Washington, PA, USA
| | - Jennifer A Wargo
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | | | | |
Collapse
|
7
|
Biological evaluation of pyrazolyl-urea and dihydro-imidazo-pyrazolyl-urea derivatives as potential anti-angiogenetic agents in the treatment of neuroblastoma. Oncotarget 2020; 11:3459-3472. [PMID: 32973970 PMCID: PMC7500105 DOI: 10.18632/oncotarget.27733] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 08/11/2020] [Indexed: 12/05/2022] Open
Abstract
Pyrazolyl-urea and dihydro-imidazo-pyrazolyl-urea compounds (STIRUR 13, STIRUR 41 and BUR 12) have been demonstrated to exert a strong inhibitory effect on interleukin 8 or N-formyl-methionyl-leucyl-phenylalanine-induced chemotaxis of human neutrophils. Since the migration of cancer cells is comparable to that of neutrophils, the purpose of this study is to evaluate the biological effect of STIRUR 13, STIRUR 41 and BUR 12 on ACN and HTLA-230, two neuroblastoma (NB) cell lines with different degree of malignancy. HTLA-230 cells, stage-IV NB cells, have high plasticity and can serve as progenitors of endothelial cells. The results herein reported show that the three tested compounds were not cytotoxic for both NB cells and did not alter their clonogenic potential. However, all compounds were able to inhibit the ability of HTLA-230 to form vascular-like structures. On the basis of these findings, pyrazolyl-urea and dihydro-imidazo-pyrazolyl-urea derivatives could be proposed as agents potentially effective in counteracting NB malignancy by inhibiting cell migration and tumor angiogenesis which represent important hallmarks responsible for cancer survival and progression.
Collapse
|
8
|
Hee E, Wong MK, Tan SH, Choo Z, Kuick CH, Ling S, Yong MH, Jain S, Lian DWQ, Ng EHQ, Yong YFL, Ren MH, Syed Sulaiman N, Low SYY, Chua YW, Syed MF, Lim TKH, Soh SY, Iyer P, Seng MSF, Lam JCM, Tan EEK, Chan MY, Tan AM, Chen Y, Chen Z, Chang KTE, Loh AHP. Neuroblastoma patient-derived cultures are enriched for a mesenchymal gene signature and reflect individual drug response. Cancer Sci 2020; 111:3780-3792. [PMID: 32777141 PMCID: PMC7540996 DOI: 10.1111/cas.14610] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/03/2020] [Accepted: 08/05/2020] [Indexed: 02/07/2023] Open
Abstract
Ex vivo evaluation of personalized models can facilitate individualized treatment selection for patients, and advance the discovery of novel therapeutic options. However, for embryonal malignancies, representative primary cultures have been difficult to establish. We developed patient‐derived cell cultures (PDCs) from chemo‐naïve and post–treatment neuroblastoma tumors in a consistent and efficient manner, and characterized their in vitro growth dynamics, histomorphology, gene expression, and functional chemo‐response. From 34 neuroblastoma tumors, 22 engrafted in vitro to generate 31 individual PDC lines, with higher engraftment seen with metastatic tumors. PDCs displayed characteristic immunohistochemical staining patterns of PHOX2B, TH, and GD2 synthase. Concordance of MYCN amplification, 1p and 11q deletion between PDCs and patient tumors was 83.3%, 72.7%, and 80.0% respectively. PDCs displayed a predominantly mesenchymal‐type gene expression signature and showed upregulation of pro‐angiogenic factors that were similarly enriched in culture medium and paired patient serum samples. When tested with standard‐of‐care cytotoxics at human Cmax‐equivalent concentrations, MYCN‐amplified and non‐MYCN‐amplified PDCs showed a differential response to cyclophosphamide and topotecan, which mirrored the corresponding patients’ responses, and correlated with gene signatures of chemosensitivity. In this translational proof‐of‐concept study, early‐phase neuroblastoma PDCs enriched for the mesenchymal cell subpopulation recapitulated the individual molecular and phenotypic profile of patient tumors, and highlighted their potential as a platform for individualized ex vivo drug‐response testing.
Collapse
Affiliation(s)
- Esther Hee
- VIVA-KKH Paediatric Brain and Solid Tumour Programme, KK Women's and Children's Hospital, Singapore, Singapore
| | - Meng Kang Wong
- VIVA-KKH Paediatric Brain and Solid Tumour Programme, KK Women's and Children's Hospital, Singapore, Singapore
| | - Sheng Hui Tan
- VIVA-KKH Paediatric Brain and Solid Tumour Programme, KK Women's and Children's Hospital, Singapore, Singapore
| | - Zhang'E Choo
- Neurodevelopment and Cancer Laboratory, Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chik Hong Kuick
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Sharon Ling
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Min Hwee Yong
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Sudhanshi Jain
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Derrick W Q Lian
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Eileen H Q Ng
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Yvonne F L Yong
- KK Research Centre, KK Women's and Children's Hospital, Singapore, Singapore
| | - Mee Hiong Ren
- KK Research Centre, KK Women's and Children's Hospital, Singapore, Singapore
| | - Nurfarhanah Syed Sulaiman
- VIVA-KKH Paediatric Brain and Solid Tumour Programme, KK Women's and Children's Hospital, Singapore, Singapore.,Department of Neurosurgery, National Neuroscience Institute, Singapore, Singapore
| | - Sharon Y Y Low
- VIVA-KKH Paediatric Brain and Solid Tumour Programme, KK Women's and Children's Hospital, Singapore, Singapore.,Neurosurgical Service, KK Women's and Children's Hospital, Singapore, Singapore.,SingHealth Duke-NUS Neuroscience Academic Clinical Program, Singapore, Singapore
| | - Yong Wei Chua
- Department of Anatomic Pathology, Singapore General Hospital, Singapore, Singapore
| | - Muhammad Fahmy Syed
- Department of Anatomic Pathology, Singapore General Hospital, Singapore, Singapore
| | - Tony K H Lim
- Department of Anatomic Pathology, Singapore General Hospital, Singapore, Singapore
| | - Shui Yen Soh
- Department of Paediatric Subspecialties Haematology Oncology Service, KK Women's and Children's Hospital, Singapore, Singapore
| | - Prasad Iyer
- Department of Paediatric Subspecialties Haematology Oncology Service, KK Women's and Children's Hospital, Singapore, Singapore
| | - Michaela S F Seng
- Department of Paediatric Subspecialties Haematology Oncology Service, KK Women's and Children's Hospital, Singapore, Singapore
| | - Joyce C M Lam
- Department of Paediatric Subspecialties Haematology Oncology Service, KK Women's and Children's Hospital, Singapore, Singapore
| | - Enrica E K Tan
- Department of Paediatric Subspecialties Haematology Oncology Service, KK Women's and Children's Hospital, Singapore, Singapore
| | - Mei Yoke Chan
- Department of Paediatric Subspecialties Haematology Oncology Service, KK Women's and Children's Hospital, Singapore, Singapore
| | - Ah Moy Tan
- Department of Paediatric Subspecialties Haematology Oncology Service, KK Women's and Children's Hospital, Singapore, Singapore
| | - Yong Chen
- Department of Paediatric Surgery, KK Women's and Children's Hospital, Singapore, Singapore
| | - Zhixiong Chen
- Neurodevelopment and Cancer Laboratory, Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kenneth T E Chang
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Amos Hong Pheng Loh
- VIVA-KKH Paediatric Brain and Solid Tumour Programme, KK Women's and Children's Hospital, Singapore, Singapore.,Department of Paediatric Surgery, KK Women's and Children's Hospital, Singapore, Singapore
| |
Collapse
|
9
|
Ma G, Jiang Y, Liang M, Li J, Wang J, Mao X, Veeramootoo JS, Xia T, Liu X, Wang S. Dynamic monitoring of CD45-/CD31+/DAPI+ circulating endothelial cells aneuploid for chromosome 8 during neoadjuvant chemotherapy in locally advanced breast cancer. Ther Adv Med Oncol 2020; 12:1758835920918470. [PMID: 32489429 PMCID: PMC7238307 DOI: 10.1177/1758835920918470] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 03/19/2020] [Indexed: 12/15/2022] Open
Abstract
Background: Neoadjuvant chemotherapy (NCT) is the standard treatment for patients with
locally advanced breast cancer (LABC). The aim of this study was to verify
this relationship, and to estimate the clinical value of aneuploid
circulating endothelial cells (CECs) in LABC patients with different NCT
responses. Methods: Breast cancer patients received an EC4-T4 NCT regimen. Peripheral blood
mononuclear cells were obtained before NCT, and after the first and last NCT
courses. A novel subtraction enrichment and immunostaining fluorescence
in situ hybridization (SE-iFISH) strategy was applied
for detection of circulating rare cells (CRCs). CECs (CD45–/CD31+/DAPI+) and
circulating tumor cells (CTCs) with different cytogenetic abnormalities
related to chromosome 8 aneuploidy were analyzed in LABC patients subjected
to NCT. Results: A total of 41 patients were enrolled. Firstly, CD31+/EpCAM+ aneuploid
endothelial-epithelial fusion cells were observed in LABC patients. Further,
aneuploid CECs in the peripheral blood showed a biphasic response during
NCT, as they initially increased and then decreased, whereas a strong
positive correlation was observed between aneuploid CECs and CTC
numbers. Conclusion: We determined that aneuploid CEC dynamics vary in patients with different
response to chemotherapy. Elucidating the potential cross-talk between CTCs
and aneuploid CECs may help characterize the process associated with the
development of chemotherapy resistance and metastasis.
Collapse
Affiliation(s)
- Ge Ma
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Yi Jiang
- The First Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Mengdi Liang
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - JiaYing Li
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Jingyi Wang
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Xinrui Mao
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | | | - Tiansong Xia
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Xiaoan Liu
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Shui Wang
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| |
Collapse
|
10
|
Lee YS, Krishnan A, Oughtred R, Rust J, Chang CS, Ryu J, Kristensen VN, Dolinski K, Theesfeld CL, Troyanskaya OG. A Computational Framework for Genome-wide Characterization of the Human Disease Landscape. Cell Syst 2019; 8:152-162.e6. [PMID: 30685436 PMCID: PMC7374759 DOI: 10.1016/j.cels.2018.12.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 10/16/2018] [Accepted: 12/20/2018] [Indexed: 01/21/2023]
Abstract
A key challenge for the diagnosis and treatment of complex human diseases is identifying their molecular basis. Here, we developed a unified computational framework, URSAHD (Unveiling RNA Sample Annotation for Human Diseases), that leverages machine learning and the hierarchy of anatomical relationships present among diseases to integrate thousands of clinical gene expression profiles and identify molecular characteristics specific to each of the hundreds of complex diseases. URSAHD can distinguish between closely related diseases more accurately than literature-validated genes or traditional differential-expression-based computational approaches and is applicable to any disease, including rare and understudied ones. We demonstrate the utility of URSAHD in classifying related nervous system cancers and experimentally verifying novel neuroblastoma-associated genes identified by URSAHD. We highlight the applications for potential targeted drug-repurposing and for quantitatively assessing the molecular response to clinical therapies. URSAHD is freely available for public use, including the use of underlying models, at ursahd.princeton.edu.
Collapse
Affiliation(s)
- Young-Suk Lee
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA; Department of Computer Science, Princeton University, Princeton, NJ, USA; School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Arjun Krishnan
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA; Departments of Computational Mathematics, Science, and Engineering and Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Rose Oughtred
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Jennifer Rust
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Christie S Chang
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Joseph Ryu
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Vessela N Kristensen
- Department of Genetics, Institute of Cancer Research, Oslo University Hospital, Radiumhospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Clinical Molecular Biology (EpiGen), Division of Medicine, Akershus University Hospital, Lørenskog, Norway
| | - Kara Dolinski
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Chandra L Theesfeld
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA.
| | - Olga G Troyanskaya
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA; Department of Computer Science, Princeton University, Princeton, NJ, USA; Flatiron Institute, Simons Foundation, New York, NY, USA.
| |
Collapse
|
11
|
Pieniazek M, Matkowski R, Donizy P. Macrophages in skin melanoma-the key element in melanomagenesis. Oncol Lett 2018; 15:5399-5404. [PMID: 29552183 PMCID: PMC5840697 DOI: 10.3892/ol.2018.8021] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 11/16/2017] [Indexed: 01/08/2023] Open
Abstract
Cutaneous melanoma is an aggressive cancer and its onset and growth are associated, through direct and indirect interactions, with the cancer microenvironment. The microenvironment comprises a dynamic complex of numerous types of cells (due to histogenesis) that constantly interact with each other through multiple cytokines and signaling proteins. Macrophages are one of the most thoroughly studied pleiotropic cells of the immune system. One of their major cytophysiological functions is their involvement in phagocytosis. Previous studies examining the microenvironment of melanomas and tumor-associated macrophages have revealed that they are involved in all stages of melanomagenesis. In the case of cancer initiation, they form an inflammatory microenvironment and then suppress the anticancer activity of the immune system, stimulate angiogenesis, enhance migration and invasion of the cancer cells, and ultimately contribute to the metastatic process. The present review provides a detailed overview on the function of macrophages in the melanoma microenvironment.
Collapse
Affiliation(s)
- Malgorzata Pieniazek
- Department of Clinical Oncology, Tadeusz Koszarowski Regional Oncology Centre, Opole 45-061, Poland
| | - Rafal Matkowski
- Department of Oncology and Division of Surgical Oncology, Wroclaw Medical University, Wroclaw 50-367, Poland
- Department of Surgical Oncology, Lower Silesian Oncology Centre, Wroclaw 53-413, Poland
| | - Piotr Donizy
- Department of Pathomorphology and Oncological Cytology, Wroclaw Medical University, Wroclaw 50-556, Poland
| |
Collapse
|
12
|
Jin L, Li ZF, Wang DK, Sun M, Qi W, Ma Q, Zhang L, Chu C, Chan EY, Lee SS, Wise H, To KF, Shi Y, Zhou N, Cheung WT. Molecular and functional characterization of tumor-induced factor (TIF): Hamster homolog of CXCL3 (GROγ) displays tumor suppressive activity. Cytokine 2018; 102:62-75. [DOI: 10.1016/j.cyto.2017.12.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 12/11/2017] [Accepted: 12/15/2017] [Indexed: 12/20/2022]
|
13
|
Duran CL, Howell DW, Dave JM, Smith RL, Torrie ME, Essner JJ, Bayless KJ. Molecular Regulation of Sprouting Angiogenesis. Compr Physiol 2017; 8:153-235. [PMID: 29357127 DOI: 10.1002/cphy.c160048] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The term angiogenesis arose in the 18th century. Several studies over the next 100 years laid the groundwork for initial studies performed by the Folkman laboratory, which were at first met with some opposition. Once overcome, the angiogenesis field has flourished due to studies on tumor angiogenesis and various developmental models that can be genetically manipulated, including mice and zebrafish. In addition, new discoveries have been aided by the ability to isolate primary endothelial cells, which has allowed dissection of various steps within angiogenesis. This review will summarize the molecular events that control angiogenesis downstream of biochemical factors such as growth factors, cytokines, chemokines, hypoxia-inducible factors (HIFs), and lipids. These and other stimuli have been linked to regulation of junctional molecules and cell surface receptors. In addition, the contribution of cytoskeletal elements and regulatory proteins has revealed an intricate role for mobilization of actin, microtubules, and intermediate filaments in response to cues that activate the endothelium. Activating stimuli also affect various focal adhesion proteins, scaffold proteins, intracellular kinases, and second messengers. Finally, metalloproteinases, which facilitate matrix degradation and the formation of new blood vessels, are discussed, along with our knowledge of crosstalk between the various subclasses of these molecules throughout the text. Compr Physiol 8:153-235, 2018.
Collapse
Affiliation(s)
- Camille L Duran
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - David W Howell
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Jui M Dave
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Rebecca L Smith
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Melanie E Torrie
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Jeffrey J Essner
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| |
Collapse
|
14
|
Tuccitto A, Tazzari M, Beretta V, Rini F, Miranda C, Greco A, Santinami M, Patuzzo R, Vergani B, Villa A, Manenti G, Cleris L, Giardiello D, Alison M, Rivoltini L, Castelli C, Perego M. Immunomodulatory Factors Control the Fate of Melanoma Tumor Initiating Cells. Stem Cells 2016; 34:2449-2460. [PMID: 27301067 DOI: 10.1002/stem.2413] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 04/29/2016] [Indexed: 12/19/2022]
Abstract
Melanoma is a highly heterogeneous tumor for which recent evidence supports a model of dynamic stemness. Melanoma cells might temporally acquire tumor-initiating properties or switch from a status of tumor-initiating cells (TICs) to a more differentiated one depending on the tumor context. However, factors driving these functional changes are still unknown. We focused on the role of cyto/chemokines in shaping TICs isolated directly from tumor specimens of two melanoma patients, namely Me14346S and Me15888S. We analyzed the secretion profile of TICs and of their corresponding melanoma differentiated cells and we tested the ability of cyto/chemokines to influence TIC self-renewal and differentiation. We found that TICs, grown in vitro as melanospheres, had a complex secretory profile as compared to their differentiated counterparts. Some factors, such as CCL-2 and IL-8, also produced by adherent melanoma cells and melanocytes did not influence TIC properties. Conversely, IL-6, released by differentiated cells, reduced TIC self-renewal and induced TIC differentiation while IL-10, produced by Me15888S, strongly promoted TIC self-renewal through paracrine/autocrine actions. Complete neutralization of IL-10 activity by gene silencing and antibody-mediated blocking of the IL-10Rα was required to sensitize Me15888S to IL-6-induced differentiation. For the first time these results show that functional heterogeneity of melanoma could be directly influenced by inflammatory and suppressive soluble factors, with IL-6 favoring TIC differentiation, and IL-10 supporting TIC self-renewal. Thus, understanding the tumor microenvironment (TME) role in modulating melanoma TIC phenotype is fundamental to identifying novel therapeutic targets to achieve long-lasting regression of metastatic melanoma. Stem Cells 2016;34:2449-2460.
Collapse
Affiliation(s)
- Alessandra Tuccitto
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.,Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Marcella Tazzari
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.,Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Valeria Beretta
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.,Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Francesca Rini
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.,Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Claudia Miranda
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.,Molecular Mechanism Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Angela Greco
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.,Molecular Mechanism Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Mario Santinami
- Melanoma and Sarcoma Unit, Department of Surgery, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Roberto Patuzzo
- Melanoma and Sarcoma Unit, Department of Surgery, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Barbara Vergani
- Consorzio MIA (Microscopy and Image Analysis), University of Milano-Bicocca, Milano, Italy
| | - Antonello Villa
- Consorzio MIA (Microscopy and Image Analysis), University of Milano-Bicocca, Milano, Italy
| | - Giacomo Manenti
- Department of Predictive and Preventive Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Loredana Cleris
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Daniele Giardiello
- Unit of Clinical Epidemiology and Trial Organization, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Malcolm Alison
- Centre for Tumour Biology, Barts Cancer Institute, Charterhouse Square, London, EC1M 6BQ, United Kingdom
| | - Licia Rivoltini
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.,Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Chiara Castelli
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy. .,Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.
| | - Michela Perego
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.,Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| |
Collapse
|
15
|
Somasundaram R, Herlyn M, Wagner SN. The role of tumor microenvironment in melanoma therapy resistance. Melanoma Manag 2016; 3:23-32. [PMID: 30190870 PMCID: PMC6094607 DOI: 10.2217/mmt.15.37] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 10/06/2015] [Indexed: 12/16/2022] Open
Abstract
Melanoma patients develop resistance to both chemotherapy and targeted-therapy drugs. Promising preclinical and clinical results with immune checkpoint inhibitors using antibodies directed against cytotoxic T-lymphocyte-associated protein 4 and programmed cell death protein 1 have re-energized the field of immune-based therapies in melanoma. However, similar to chemotherapy or targeted therapies, immune checkpoint blockade responds in only subsets of melanoma patients. A number of factors, including gene mutations, altered cell-signaling pathways and tumor heterogeneity can contribute to therapy resistance. Recent studies have highlighted the role of inflammatory tumor microenvironment on therapy resistance of cancer cells. Cancer cells either alone or in conjunction with the tumor stroma can contribute to an inflammatory microenvironment. Multimodal approaches of targeting the tumor microenvironment, in addition to malignant cells, may be necessary for better therapy responses.
Collapse
Affiliation(s)
| | - Meenhard Herlyn
- The Wistar Institute, 3601 Spruce St, Philadelphia, PA 19104, USA
| | - Stephan N Wagner
- Division of Immunology, Allergy & Infectious Diseases (DIAID), Department of Dermatology, Medical University of Vienna, 1090 Wien, Austria
| |
Collapse
|
16
|
Influence of carrier cells on the clinical outcome of children with neuroblastoma treated with high dose of oncolytic adenovirus delivered in mesenchymal stem cells. Cancer Lett 2016; 371:161-70. [DOI: 10.1016/j.canlet.2015.11.036] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 11/24/2015] [Accepted: 11/27/2015] [Indexed: 12/22/2022]
|
17
|
Resveratrol Protects PC12 Cell against 6-OHDA Damage via CXCR4 Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:730121. [PMID: 26681969 PMCID: PMC4670657 DOI: 10.1155/2015/730121] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 10/21/2015] [Accepted: 10/25/2015] [Indexed: 12/20/2022]
Abstract
Resveratrol, herbal nonflavonoid polyphenolic compound naturally derived from grapes, has long been acknowledged to possess extensive biological and pharmacological properties including antioxidant and anti-inflammatory ones and may exert a neuroprotective effect on neuronal damage in neurodegenerative diseases. However, the underlying molecular mechanisms remain undefined. In the present study, we intended to investigate the neuroprotective effects of resveratrol against 6-OHDA-induced neurotoxicity of PC12 cells and further explore the possible mechanisms involved. For this purpose, PC12 cells were exposed to 6-OHDA in the presence of resveratrol (0, 12.5, 25, and 50 μM). The results showed that resveratrol increased cell viability, alleviated the MMP reduction, and reduced the number of apoptotic cells as measured by MTT assay, JC-1 staining, and Hoechst/PI double staining (all p < 0.01). Immunofluorescent staining and Western blotting revealed that resveratrol averts 6-OHDA induced CXCR4 upregulation (p < 0.01). Our results demonstrated that resveratrol could effectively protect PC12 cells from 6-OHDA-induced oxidative stress and apoptosis via CXCR4 signaling pathway.
Collapse
|
18
|
Shankar V, Hori H, Kihira K, Lei Q, Toyoda H, Iwamoto S, Komada Y. Mesenchymal stromal cell secretome up-regulates 47 kDa CXCR4 expression, and induce invasiveness in neuroblastoma cell lines. PLoS One 2015; 10:e0120069. [PMID: 25774696 PMCID: PMC4361348 DOI: 10.1371/journal.pone.0120069] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 01/31/2015] [Indexed: 12/26/2022] Open
Abstract
Neuroblastoma accounts for 15% of childhood cancer deaths and presents with metastatic disease of the bone and the bone marrow at diagnosis in 70% of the cases. Previous studies have shown that the Mesenchymal Stromal Cell (MSC) secretome, triggers metastases in several cancer types such as breast and prostate cancer, but the specific role of the MSC factors in neuroblastoma metastasis is unclear. To better understand the effect of MSC secretome on chemokine receptors in neuroblastoma, and its role in metastasis, we studied a panel of 20 neuroblastoma cell lines, and compared their invasive potential towards MSC-conditioned-RPMI (mRPMI) and their cytokine receptor expression profiles. Western blot analysis revealed the expression of multiple CXCR4 isoforms in neuroblastoma cells. Among the five major isoforms, the expression of the 47 kDa isoform showed significant correlation with high invasiveness. Pretreatment with mRPMI up-regulated the expression of the 47 kDa CXCR4 isoform and also increased MMP-9 secretion, expression of integrin α3 and integrin β1, and the invasive potential of the cell; while blocking CXCR4 either with AMD 3100, a CXCR4 antagonist, or with an anti-47 kDa CXCR4 neutralizing antibody decreased the secretion of MMP-9, the expression of integrin α3 and integrin β1, and the invasive potential of the cell. Pretreatment with mRPMI also protected the 47 kDa CXCR4 isoform from ubiquitination and subsequent degradation. Our data suggest a modulatory role of the MSC secretome on the expression of the 47 kDa CXCR4 isoform and invasion potential of the neuroblastoma cells to the bone marrow.
Collapse
Affiliation(s)
- Vipin Shankar
- Department of Pediatrics, Mie University Graduate School of Medicine, 2-174, Edobashi, Tsu, Mie, Japan
| | - Hiroki Hori
- Department of Pediatrics, Mie University Graduate School of Medicine, 2-174, Edobashi, Tsu, Mie, Japan
- * E-mail:
| | - Kentaro Kihira
- Department of Pediatrics, Mie University Graduate School of Medicine, 2-174, Edobashi, Tsu, Mie, Japan
| | - Qi Lei
- Department of Pediatrics, Mie University Graduate School of Medicine, 2-174, Edobashi, Tsu, Mie, Japan
| | - Hidemi Toyoda
- Department of Pediatrics, Mie University Graduate School of Medicine, 2-174, Edobashi, Tsu, Mie, Japan
| | - Shotaro Iwamoto
- Department of Pediatrics, Mie University Graduate School of Medicine, 2-174, Edobashi, Tsu, Mie, Japan
| | - Yoshihiro Komada
- Department of Pediatrics, Mie University Graduate School of Medicine, 2-174, Edobashi, Tsu, Mie, Japan
| |
Collapse
|
19
|
Hetzler KL, Hardee JP, Puppa MJ, Narsale AA, Sato S, Davis JM, Carson JA. Sex differences in the relationship of IL-6 signaling to cancer cachexia progression. Biochim Biophys Acta Mol Basis Dis 2014; 1852:816-25. [PMID: 25555992 DOI: 10.1016/j.bbadis.2014.12.015] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 12/04/2014] [Accepted: 12/22/2014] [Indexed: 02/07/2023]
Abstract
A devastating aspect of cancer cachexia is severe loss of muscle and fat mass. Though cachexia occurs in both sexes, it is not well-defined in the female. The Apc(Min/+) mouse is genetically predisposed to develop intestinal tumors; circulating IL-6 is a critical regulator of cancer cachexia in the male Apc(Min/+) mouse. The purpose of this study was to examine the relationship between IL-6 signaling and cachexia progression in the female Apc(Min/+) mouse. Male and female Apc(Min/+) mice were examined during the initiation and progression of cachexia. Another group of females had IL-6 overexpressed between 12 and 14 weeks or 15-18 weeks of age to determine whether IL-6 could induce cachexia. Cachectic female Apc(Min/+) mice lost body weight, muscle mass, and fat mass; increased muscle IL-6 mRNA expression was associated with these changes, but circulating IL-6 levels were not. Circulating IL-6 levels did not correlate with downstream signaling in muscle in the female. Muscle IL-6r mRNA expression and SOCS3 mRNA expression as well as muscle IL-6r protein and STAT3 phosphorylation increased with severe cachexia in both sexes. Muscle SOCS3 protein increased in cachectic females but decreased in cachectic males. IL-6 overexpression did not affect cachexia progression in female Apc(Min/+) mice. Our results indicate that female Apc(Min/+) mice undergo cachexia progression that is at least initially IL-6-independent. Future studies in the female will need to determine mechanisms underlying regulation of IL-6 response and cachexia induction.
Collapse
Affiliation(s)
- Kimbell L Hetzler
- Integrative Muscle Biology Laboratory, Department of Exercise Science, University of South Carolina, 921 Assembly St., Columbia, SC 29208 USA
| | - Justin P Hardee
- Integrative Muscle Biology Laboratory, Department of Exercise Science, University of South Carolina, 921 Assembly St., Columbia, SC 29208 USA
| | - Melissa J Puppa
- Integrative Muscle Biology Laboratory, Department of Exercise Science, University of South Carolina, 921 Assembly St., Columbia, SC 29208 USA
| | - Aditi A Narsale
- Integrative Muscle Biology Laboratory, Department of Exercise Science, University of South Carolina, 921 Assembly St., Columbia, SC 29208 USA
| | - Shuichi Sato
- Integrative Muscle Biology Laboratory, Department of Exercise Science, University of South Carolina, 921 Assembly St., Columbia, SC 29208 USA
| | - J Mark Davis
- Integrative Muscle Biology Laboratory, Department of Exercise Science, University of South Carolina, 921 Assembly St., Columbia, SC 29208 USA
| | - James A Carson
- Integrative Muscle Biology Laboratory, Department of Exercise Science, University of South Carolina, 921 Assembly St., Columbia, SC 29208 USA.
| |
Collapse
|
20
|
Al-Zhoughbi W, Huang J, Paramasivan GS, Till H, Pichler M, Guertl-Lackner B, Hoefler G, Hoefler G. Tumor macroenvironment and metabolism. Semin Oncol 2014; 41:281-95. [PMID: 24787299 PMCID: PMC4012137 DOI: 10.1053/j.seminoncol.2014.02.005] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In this review we introduce the concept of the tumor macroenvironment and explore it in the context of metabolism. Tumor cells interact with the tumor microenvironment including immune cells. Blood and lymph vessels are the critical components that deliver nutrients to the tumor and also connect the tumor to the macroenvironment. Several factors are then released from the tumor itself but potentially also from the tumor microenvironment, influencing the metabolism of distant tissues and organs. Amino acids, and distinct lipid and lipoprotein species can be essential for further tumor growth. The role of glucose in tumor metabolism has been studied extensively. Cancer-associated cachexia is the most important tumor-associated systemic syndrome and not only affects the quality of life of patients with various malignancies but is estimated to be the cause of death in 15%-20% of all cancer patients. On the other hand, systemic metabolic diseases such as obesity and diabetes are known to influence tumor development. Furthermore, the clinical implications of the tumor macroenvironment are explored in the context of the patient's outcome with special consideration for pediatric tumors. Finally, ways to target the tumor macroenvironment that will provide new approaches for therapeutic concepts are described.
Collapse
Affiliation(s)
- Wael Al-Zhoughbi
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Jianfeng Huang
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | | | - Holger Till
- Department of Paediatric and Adolescent Surgery, Medical University of Graz, Graz, Austria
| | - Martin Pichler
- Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | | | - Gerald Hoefler
- Institute of Pathology, Medical University of Graz, Graz, Austria,Address correspondence to Gerald Hoefler, MD, Institute of Pathology, Medical University of Graz, Auenbruggerplatz 25, 8036 Graz, Austria
| | - Gerald Hoefler
- Institute of Pathology, Medical University of Graz, Graz, Austria.
| |
Collapse
|
21
|
Joseph JM, Farron AM, Renella R, Gapany C. Can Smaller-Scale Comprehensive Cancer Centers Provide Outstanding Care in Abdominal and Thoracic Pediatric Solid Tumor Surgery? Results of a 14-Year Retrospective Single-Center Analysis. Ann Surg Oncol 2014; 21:1726-31. [DOI: 10.1245/s10434-013-3455-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Indexed: 01/16/2023]
|
22
|
Abstract
During metastasis, cancer cells disseminate to other parts of the body by entering the bloodstream in a process that is called intravasation. They then extravasate at metastatic sites by attaching to endothelial cells that line blood vessels and crossing the vessel walls of tissues or organs. This Review describes how cancer cells cross the endothelial barrier during extravasation and how different receptors, signalling pathways and circulating cells such as leukocytes and platelets contribute to this process. Identification of the mechanisms that underlie cancer cell extravasation could lead to the development of new therapies to reduce metastasis.
Collapse
Affiliation(s)
- Nicolas Reymond
- 1] Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK. [2] Centre de Recherche de Biochimie Macromoléculaire, Centre National de la Recherche Scientifique (CNRS) - UMR5237, 1919 Route de Mende, 34293 Montpellier, Cedex 5, France. [3]
| | | | | |
Collapse
|
23
|
Tjomsland V, Bojmar L, Sandström P, Bratthäll C, Messmer D, Spångeus A, Larsson M. IL-1α expression in pancreatic ductal adenocarcinoma affects the tumor cell migration and is regulated by the p38MAPK signaling pathway. PLoS One 2013; 8:e70874. [PMID: 23951028 PMCID: PMC3741369 DOI: 10.1371/journal.pone.0070874] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 06/25/2013] [Indexed: 01/09/2023] Open
Abstract
The interplay between the tumor cells and the surrounding stroma creates inflammation, which promotes tumor growth and spread. The inflammation is a hallmark for pancreatic adenocarcinoma (PDAC) and is to high extent driven by IL-1α. IL-1α is expressed and secreted by the tumor cells and exerting its effect on the stroma, i.e. cancer associated fibroblasts (CAF), which in turn produce massive amount of inflammatory and immune regulatory factors. IL-1 induces activation of transcription factors such as nuclear factor-κβ (NF-κβ), but also activator protein 1 (AP-1) via the small G-protein Ras. Dysregulation of Ras pathways are common in cancer as this oncogene is the most frequently mutated in many cancers. In contrast, the signaling events leading up to the expression of IL-1α by tumor cells are not well elucidated. Our aim was to examine the signaling cascade involved in the induction of IL-1α expression in PDAC. We found p38MAPK, activated by the K-Ras signaling pathway, to be involved in the expression of IL-1α by PDAC as blocking this pathway decreased both the gene and protein expression of IL-1α. Blockage of the P38MAPK signaling in PDAC also dampened the ability of the tumor cell to induce inflammation in CAFs. In addition, the IL-1α autocrine signaling regulated the migratory capacity of PDAC cells. Taken together, the blockage of signaling pathways leading to IL-1α expression and/or neutralization of IL-1α in the PDAC microenvironment should be taken into consideration as possible treatment or complement to existing treatment of this cancer.
Collapse
Affiliation(s)
- Vegard Tjomsland
- Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Linda Bojmar
- Division of Surgery, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Per Sandström
- Division of Surgery, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | | | - Davorka Messmer
- Moores Cancer Center, University of California San Diego, La Jolla, California, United States of America
| | - Anna Spångeus
- Division of Internal Medicine and Department of Endocrinology, Department of Medical and Health Science, Linköping University, Linköping, Sweden
| | - Marie Larsson
- Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
- * E-mail:
| |
Collapse
|
24
|
IRF-8 controls melanoma progression by regulating the cross talk between cancer and immune cells within the tumor microenvironment. Neoplasia 2013; 14:1223-35. [PMID: 23308054 DOI: 10.1593/neo.121444] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 10/16/2012] [Accepted: 10/19/2012] [Indexed: 12/31/2022] Open
Abstract
The transcription factor interferon regulatory factor-8 (IRF-8) is crucial for myeloid cell development and immune response and also acts as a tumor suppressor gene. Here, we analyzed the role of IRF-8 in the cross talk between melanoma cells and tumor-infiltrating leukocytes. B16-F10 melanoma cells transplanted into IRF-8-deficient (IRF-8(-/-)) mice grow more rapidly, leading to higher numbers of lung metastasis, with respect to control animals. These events correlated with reduced dendritic cell and T cell infiltration, accumulation of myeloid-derived suppressor cells and a chemokine/chemokine receptor expression profile within the tumor microenvironment supporting tumor growth, angiogenesis, and metastasis. Noticeably, primary tumors developing in IRF-8(-/-) mice displayed a clear-cut inhibition of IRF-8 expression in melanoma cells. Injection of the demethylating agent 5-aza-2'-deoxycytidine into melanoma-bearing IRF-8(-/-) animals induced intratumoral IRF-8 expression and resulted in the re-establishment of a chemokine/ chemokine receptor pattern favoring leukocyte infiltration and melanoma growth arrest. Importantly, intrinsic IRF-8 expression was progressively down-modulated during melanoma growth in mice and in human metastatic melanoma cells with respect to primary tumors. Lastly, IRF-8 expression in melanoma cells was directly modulated by soluble factors, among which interleukin-27 (IL-27), released by immune cells from tumor-bearing mice. Collectively, these results underscore a key role of IRF-8 in the cross talk between melanoma and immune cells, thus revealing its critical function within the tumor microenvironment in regulating melanoma progression and invasiveness.
Collapse
|
25
|
Yang X, Hou J, Han Z, Wang Y, Hao C, Wei L, Shi Y. One cell, multiple roles: contribution of mesenchymal stem cells to tumor development in tumor microenvironment. Cell Biosci 2013; 3:5. [PMID: 23336752 PMCID: PMC3693909 DOI: 10.1186/2045-3701-3-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 11/20/2012] [Indexed: 12/13/2022] Open
Abstract
The discovery of tissue reparative and immunosuppressive abilities of mesenchymal stem cells (MSCs) has drawn more attention to tumor microenvironment and its role in providing the soil for the tumor cell growth. MSCs are recruited to tumor which is referred as the never healing wound and altered by the inflammation environment, thereby helping to construct the tumor microenvironment. The environment orchestrated by MSCs and other factors can be associated with angiogenesis, immunosuppression, inhibition of apoptosis, epithelial-mesenchymal transition (EMT), survival of cancer stem cells, which all contribute to tumor growth and progression. In this review, we will discuss how MSCs are recruited to the tumor microenvironment and what effects they have on tumor progression.
Collapse
Affiliation(s)
- Xue Yang
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, the Second Military Medicial University, 225 Changhai Road, Shanghai 200438, China.
| | | | | | | | | | | | | |
Collapse
|
26
|
Involvement of the CXCR7/CXCR4/CXCL12 axis in the malignant progression of human neuroblastoma. PLoS One 2012; 7:e43665. [PMID: 22916293 PMCID: PMC3423387 DOI: 10.1371/journal.pone.0043665] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 07/23/2012] [Indexed: 12/30/2022] Open
Abstract
Neuroblastoma (NB) is a typical childhood and heterogeneous neoplasm for which efficient targeted therapies for high-risk tumors are not yet identified. The chemokine CXCL12, and its receptors CXCR4 and CXCR7 have been involved in tumor progression and dissemination. While CXCR4 expression is associated to undifferentiated tumors and poor prognosis, the role of CXCR7, the recently identified second CXCL12 receptor, has not yet been elucidated in NB. In this report, CXCR7 and CXCL12 expressions were evaluated using a tissue micro-array including 156 primary and 56 metastatic NB tissues. CXCL12 was found to be highly associated to NB vascular and stromal structures. In contrast to CXCR4, CXCR7 expression was low in undifferentiated tumors, while its expression was stronger in matured tissues and specifically associated to differentiated neural tumor cells. As determined by RT-PCR, CXCR7 expression was mainly detected in N-and S-type NB cell lines, and was slightly induced upon NB cell differentiation in vitro. The relative roles of the two CXCL12 receptors were further assessed by overexpressing CXCR7 or CXCR4 receptor alone, or in combination, in the IGR-NB8 and the SH-SY5Y NB cell lines. In vitro functional analyses indicated that, in response to their common ligand, both receptors induced activation of ERK1/2 cascade, but not Akt pathway. CXCR7 strongly reduced in vitro growth, in contrast to CXCR4, and impaired CXCR4/CXCL12-mediated chemotaxis. Subcutaneous implantation of CXCR7-expressing NB cells showed that CXCR7 also significantly reduced in vivo growth. Moreover, CXCR7 affected CXCR4-mediated orthotopic growth in a CXCL12-producing environment. In such model, CXCR7, in association with CXCR4, did not induce NB cell metastatic dissemination. In conclusion, the CXCR7 and CXCR4 receptors revealed specific expression patterns and distinct functional roles in NB. Our data suggest that CXCR7 elicits anti-tumorigenic functions, and may act as a regulator of CXCR4/CXCL12-mediated signaling in NB.
Collapse
|
27
|
Cha YR, Fujita M, Butler M, Isogai S, Kochhan E, Siekmann AF, Weinstein BM. Chemokine signaling directs trunk lymphatic network formation along the preexisting blood vasculature. Dev Cell 2012; 22:824-36. [PMID: 22516200 DOI: 10.1016/j.devcel.2012.01.011] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Revised: 11/07/2011] [Accepted: 01/17/2012] [Indexed: 01/19/2023]
Abstract
The lymphatic system is crucial for fluid homeostasis, immune responses, and numerous pathological processes. However, the molecular mechanisms responsible for establishing the anatomical form of the lymphatic vascular network remain largely unknown. Here, we show that chemokine signaling provides critical guidance cues directing early trunk lymphatic network assembly and patterning. The chemokine receptors Cxcr4a and Cxcr4b are expressed in lymphatic endothelium, whereas chemokine ligands Cxcl12a and Cxcl12b are expressed in adjacent tissues along which the developing lymphatics align. Loss- and gain-of-function studies in zebrafish demonstrate that chemokine signaling orchestrates the stepwise assembly of the trunk lymphatic network. In addition to providing evidence for a lymphatic vascular guidance mechanism, these results also suggest a molecular basis for the anatomical coalignment of lymphatic and blood vessels.
Collapse
Affiliation(s)
- Young Ryun Cha
- Laboratory of Molecular Genetics, National Institute of Child Health and Human Development, National Institutes of Health, 6B/309, 6 Center Drive, Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Tissue-Specific Homing of Immune Cells in Malignant Skin Tumors. Pathol Oncol Res 2012; 18:749-59. [DOI: 10.1007/s12253-012-9529-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Accepted: 03/27/2012] [Indexed: 01/09/2023]
|
29
|
Apoptotic cells contribute to melanoma progression and this effect is partially mediated by the platelet-activating factor receptor. Mediators Inflamm 2012; 2012:610371. [PMID: 22577252 PMCID: PMC3346991 DOI: 10.1155/2012/610371] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2011] [Accepted: 02/01/2012] [Indexed: 12/31/2022] Open
Abstract
There is evidence that the platelet-activating factor receptor (PAFR) is involved in the clearance of apoptotic cells by macrophages, and that this is associated with anti-inflammatory phenotype. Our group has previously shown that coinjection of a large number of apoptotic cells can promote tumor growth from a subtumorigenic dose of melanoma cells. Here, we studied the involvement of the PAFR in the tumor growth promoting effect of apoptotic cells. A sub-tumorigenic dose of melanoma cells (Tm1) was coinjected with apoptotic Tm1 cells, subcutaneously in the flank of C57Bl/6 mice, and the volume was monitored for 30 days. Animals received the PAFR antagonists, WEB2170 or PCA4248 (5 mg/kg body weight) or vehicle, by peritumoral daily injection for 5 days. Results showed that PAFR antagonists significantly inhibited the tumor growth induced by the coinjection of a sub-tumorigenic dose of melanoma cells together with apoptotic cells. This was accompanied by inhibition of early neutrophil and macrophage infiltration. Addition of (platelet-activating factor) to this system has no significant effect. PAFR antagonists did not affect the promoting effect of carrageenan. We suggest that the recognition of apoptotic cells by phagocytes leads to activation of PAFR pathways, resulting in a microenvironment response favorable to melanoma growth.
Collapse
|
30
|
Melanoma-induced immunosuppression and its neutralization. Semin Cancer Biol 2012; 22:319-26. [PMID: 22349515 DOI: 10.1016/j.semcancer.2012.02.003] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 02/06/2012] [Indexed: 02/08/2023]
Abstract
Malignant melanoma is characterized by a rapid progression, metastasis to distant organs, and resistance to chemo- and radiotherapy. Well-defined immunogenic capacities of melanoma cells should allow a successful application of different immunotherapeutic strategies. However, the overall results of immunotherapeutic clinical studies are not satisfactory. These paradoxical observations are supposed to be due to the profound immunosuppression mediated by different mechanisms dealing with alterations in tumor and surrounding stroma cells. Melanoma microenvironment has been characterized by a remarkable accumulation of highly immunosuppressive regulatory leucocytes, in particular, myeloid-derived suppressor cells (MDSCs). Their migration, retention and high activity in the tumor lesions have been demonstrated to be induced by chronic inflammatory conditions developing in the tumor microenvironment and characterized by the long-term secretion of various inflammatory mediators (cytokines, chemokines, growth factors, reactive oxygen and nitrogen species, prostaglandins etc.) leading to further cancer progression. Here, we discuss the role of chronic inflammation in the recruitment and activation of MDSCs in melanoma lesions as well as therapeutic approaches of MDSC targeting to overcome tumor immunosuppressive microenvironment induced by chronic inflammation and enhance the efficiency of melanoma immunotherapies.
Collapse
|
31
|
Interleukin 1α sustains the expression of inflammatory factors in human pancreatic cancer microenvironment by targeting cancer-associated fibroblasts. Neoplasia 2011; 13:664-75. [PMID: 21847358 DOI: 10.1593/neo.11332] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2011] [Revised: 05/17/2011] [Accepted: 05/20/2011] [Indexed: 02/06/2023] Open
Abstract
The tumor microenvironment in pancreatic ductal adenocarcinoma (PDAC) is dynamic, with an extensive interaction between the stroma and tumor cells. The aim of this study was to delineate the cross talk between PDAC and cancer-associated fibroblasts (CAFs), with a focus on the mechanism creating the chronic inflammatory tumor milieu. We assessed the effects of the cross talk between PDAC and CAF cell lines on the creation and sustenance of the inflammatory tumor microenvironment in pancreatic cancer. The coculture of PDAC and CAF cell lines enhanced the levels of inflammatory factors including IL-1α, IL-6, CXCL8, VEGF-A, CCL20, and COX-2. CAFs were superior to tumor cells regarding the production of most inflammatory factors, and tumor cell-associated IL-1α was established as the initiator of the enhanced production of inflammatory factors through the binding of IL-1α to IL-1 receptor 1 (IL-1R1) expressed predominantly by CAFs. Furthermore, we found a correlation between IL-1α and CXCL8 expression levels in PDAC tissues and correlation between IL-1α expression and the clinical outcome of the patients. This confirmed an important role for the IL-1 signaling cascade in the creation and sustenance of a tumor favorable microenvironment. Neutralization of the IL-1α signaling efficiently diminished the cross talk-induced production of inflammatory factors. These data suggest that the cross talk between PDAC cells and the main stroma cell type, i.e. CAFs, is one essential factor in the formation of the inflammatory tumor environment, and we propose that neutralization of the IL-1α signaling might be a potential therapy for this cancer.
Collapse
|
32
|
Nakasone Y, Fujimoto M, Matsushita T, Hamaguchi Y, Huu DL, Yanaba M, Sato S, Takehara K, Hasegawa M. Host-derived MCP-1 and MIP-1α regulate protective anti-tumor immunity to localized and metastatic B16 melanoma. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 180:365-74. [PMID: 22037251 DOI: 10.1016/j.ajpath.2011.09.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Revised: 08/30/2011] [Accepted: 09/08/2011] [Indexed: 12/23/2022]
Abstract
Leukocytic infiltration into malignant melanoma lesions is tightly regulated by chemokines. To assess the role of the CC chemokines monocyte chemotactic protein-1 (MCP-1/chemokine ligand 2) and macrophage inflammatory protein-1α (MIP-1α/chemokine ligand 3) in this process, s.c. primary and metastatic B16 F10 melanoma tumor growth levels were examined in mice lacking MCP-1 or MIP-1α. Primary s.c. B16 F10 melanoma growth was augmented by loss of MCP-1 or MIP-1α. Similarly, lung metastasis was enhanced by the deficiency of MCP-1 or MIP-1α. Enhanced tumor outgrowth was associated with decreased percentages of infiltrating CD4(+) T cells, CD8(+) T cells, and natural killer cells. In the absence of MCP-1 or MIP-1α, melanoma outgrowth was correlated with reduced local expression of interferon-γ, IL-6, tumor necrosis factor-α, and transforming growth factor-β. Among these cytokines, reduced expression levels of interferon-γ and tumor necrosis factor-α on leukocytes from the spleen were associated with the development of lung metastasis in chemokine-deficient mice. The local s.c. administration of these four cytokines significantly augmented another chemokine's expression and suppressed primary melanoma growth in mice deficient for MCP-1 or MIP-1α. The s.c. injection of MCP-1 or MIP-1α significantly inhibited the primary tumor growth in wild-type mice. These results indicate that host-derived MCP-1 and MIP-1α regulate protective anti-tumor immunity to B16 F10 melanoma by promoting lymphocyte infiltration into the tumor and subsequent cytokine production.
Collapse
Affiliation(s)
- Yuko Nakasone
- Department of Dermatology, Kanazawa University Graduate School of Medical Science, Kanazawa, Ishikawa, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Berencsi K, Rani P, Zhang T, Gross L, Mastrangelo M, Meropol NJ, Herlyn D, Somasundaram R. In vitro migration of cytotoxic T lymphocyte derived from a colon carcinoma patient is dependent on CCL2 and CCR2. J Transl Med 2011; 9:33. [PMID: 21450101 PMCID: PMC3076246 DOI: 10.1186/1479-5876-9-33] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Accepted: 03/30/2011] [Indexed: 12/15/2022] Open
Abstract
Background Infiltration of colorectal carcinomas (CRC) with T-cells has been associated with good prognosis. There are some indications that chemokines could be involved in T-cell infiltration of tumors. Selective modulation of chemokine activity at the tumor site could attract immune cells resulting in tumor growth inhibition. In mouse tumor model systems, gene therapy with chemokines or administration of antibody (Ab)-chemokine fusion proteins have provided potent immune mediated tumor rejection which was mediated by infiltrating T cells at the tumor site. To develop such immunotherapeutic strategies for cancer patients, one must identify chemokines and their receptors involved in T-cell migration toward tumor cells. Methods To identify chemokine and chemokine receptors involved in T-cell migration toward CRC cells, we have used our previously published three-dimensional organotypic CRC culture system. Organotypic culture was initiated with a layer of fetal fibroblast cells mixed with collagen matrix in a 24 well tissue culture plate. A layer of CRC cells was placed on top of the fibroblast-collagen layer which was followed by a separating layer of fibroblasts in collagen matrix. Anti-CRC specific cytotoxic T lymphocytes (CTLs) mixed with fibroblasts in collagen matrix were placed on top of the separating layer. Excess chemokine ligand (CCL) or Abs to chemokine or chemokine receptor (CCR) were used in migration inhibition assays to identify the chemokine and the receptor involved in CTL migration. Results Inclusion of excess CCL2 in T-cell layer or Ab to CCL2 in separating layer of collagen fibroblasts blocked the migration of CTLs toward tumor cells and in turn significantly inhibited tumor cell apoptosis. Also, Ab to CCR2 in the separating layer of collagen and fibroblasts blocked the migration of CTLs toward tumor cells and subsequently inhibited tumor cell apoptosis. Expression of CCR2 in four additional CRC patients' lymphocytes isolated from infiltrating tumor tissues suggests their role in migration in other CRC patients. Conclusions Our data suggest that CCL2 secreted by tumor cells and CCR2 receptors on CTLs are involved in migration of CTLs towards tumor. Gene therapy of tumor cells with CCL2 or CCL2/anti-tumor Ab fusion proteins may attract CTLs that potentially could inhibit tumor growth.
Collapse
|
34
|
Schwartzbaum JA, Xiao Y, Liu Y, Tsavachidis S, Berger MS, Bondy ML, Chang JS, Chang SM, Decker PA, Ding B, Hepworth SJ, Houlston RS, Hosking FJ, Jenkins RB, Kosel ML, McCoy LS, McKinney PA, Muir K, Patoka JS, Prados M, Rice T, Robertson LB, Schoemaker MJ, Shete S, Swerdlow AJ, Wiemels JL, Wiencke JK, Yang P, Wrensch MR. Inherited variation in immune genes and pathways and glioblastoma risk. Carcinogenesis 2010; 31:1770-7. [PMID: 20668009 PMCID: PMC2950934 DOI: 10.1093/carcin/bgq152] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Revised: 07/14/2010] [Accepted: 07/19/2010] [Indexed: 12/26/2022] Open
Abstract
To determine whether inherited variations in immune function single-nucleotide polymorphisms (SNPs), genes or pathways affect glioblastoma risk, we analyzed data from recent genome-wide association studies in conjunction with predefined immune function genes and pathways. Gene and pathway analyses were conducted on two independent data sets using 6629 SNPs in 911 genes on 17 immune pathways from 525 glioblastoma cases and 602 controls from the University of California, San Francisco (UCSF) and a subset of 6029 SNPs in 893 genes from 531 cases and 1782 controls from MD Anderson (MDA). To further assess consistency of SNP-level associations, we also compared data from the UK (266 cases and 2482 controls) and the Mayo Clinic (114 cases and 111 controls). Although three correlated epidermal growth factor receptor (EGFR) SNPs were consistently associated with glioblastoma in all four data sets (Mantel-Haenzel P values = 1 × 10⁻⁵ to 4 × 10⁻³), independent replication is required as genome-wide significance was not attained. In gene-level analyses, eight immune function genes were significantly (minP < 0.05) associated with glioblastoma; the IL-2RA (CD25) cytokine gene had the smallest minP values in both UCSF (minP = 0.01) and MDA (minP = 0.001) data sets. The IL-2RA receptor is found on the surface of regulatory T cells potentially contributing to immunosuppression characteristic of the glioblastoma microenvironment. In pathway correlation analyses, cytokine signaling and adhesion-extravasation-migration pathways showed similar associations with glioblastoma risk in both MDA and UCSF data sets. Our findings represent the first systematic description of immune genes and pathways that characterize glioblastoma risk.
Collapse
Affiliation(s)
- Judith A Schwartzbaum
- Division of Epidemiology, College of Public Health, Ohio State University, Columbus, OH 43210, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
CX3CR1 promotes recruitment of human glioma-infiltrating microglia/macrophages (GIMs). Exp Cell Res 2010; 316:1553-66. [DOI: 10.1016/j.yexcr.2010.02.018] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2009] [Revised: 02/03/2010] [Accepted: 02/16/2010] [Indexed: 11/22/2022]
|
36
|
Calonge E, Alonso-Lobo JM, Escandón C, González N, Bermejo M, Santiago B, Mestre L, Pablos JL, Caruz A, Alcamí J. c/EBPbeta is a major regulatory element driving transcriptional activation of the CXCL12 promoter. J Mol Biol 2009; 396:463-72. [PMID: 19962993 DOI: 10.1016/j.jmb.2009.11.064] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2009] [Revised: 10/30/2009] [Accepted: 11/26/2009] [Indexed: 10/20/2022]
Abstract
CXCL12 is considered a constitutively expressed chemokine with homeostatic functions. However, induction of CXCL12 expression and its potential role in several pathologic conditions have been reported, suggesting that CXCL12 gene expression can be induced by different stimuli. To elucidate the molecular mechanisms involved in the regulation of CXCL12 gene expression, we aim to define the molecular factors that operate at the transcriptional level. Basal, constitutive expression of CXCL12 was dependent on basic helix-loop-helix factors. Transcriptional up-regulation of the CXCL12 gene was induced by cellular confluence or inflammatory stimuli such as interleukin-1 and interleukin-6, in a CCAAT/enhancer binding protein beta (c/EBPbeta)-dependent manner. Chromatin immunoprecipitation assays confirmed c/EBPbeta binding to a specific response element located at -1171 of the promoter region of CXCL12. Our data show that c/EBPbeta is a major regulatory element driving transcription of the CXCL12 gene in response to cytokines and cell confluence.
Collapse
Affiliation(s)
- E Calonge
- AIDS Immunopathology Unit, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Ctra. Majadahonda-Pozuelo, Km 2, 28220 Majadahonda, Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Witz IP. The tumor microenvironment: the making of a paradigm. CANCER MICROENVIRONMENT 2009; 2 Suppl 1:9-17. [PMID: 19701697 PMCID: PMC2756342 DOI: 10.1007/s12307-009-0025-8] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2009] [Accepted: 08/06/2009] [Indexed: 12/17/2022]
Abstract
What has been will be again, what has been done will be done again; there is nothing new under the sun (Ecclesiastes 1:9) Stephen Paget was the conceptual father of the role played by the Tumor Microenvironment (TME) in tumor progression. The focus of this essay is the developmental phase of the post Paget TME research. Attempts will be made to highlight some of the pioneering work of scientists from the late sixties through the eighties of last century who laid the foundations for the contemporary scientific achievements of TME research but whose ground breaking studies are rarely cited. This review should serve as a small tribute to their great work.
Collapse
Affiliation(s)
- Isaac P Witz
- Faculty of Life Sciences, Department of Cell Research & Immunology, Tel Aviv University, Ramat Aviv, Tel Aviv, 69978, Israel,
| |
Collapse
|
38
|
Meier R, Gross N. Chemokines in neuroectodermal tumors: and now for something completely different. Semin Cancer Biol 2008; 19:67. [PMID: 19013528 DOI: 10.1016/j.semcancer.2008.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2008] [Accepted: 10/20/2008] [Indexed: 11/25/2022]
|
39
|
Raffaghello L, Cocco C, Corrias MV, Airoldi I, Pistoia V. Chemokines in neuroectodermal tumour progression and metastasis. Semin Cancer Biol 2008; 19:97-102. [PMID: 19013246 DOI: 10.1016/j.semcancer.2008.10.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2008] [Revised: 10/13/2008] [Accepted: 10/15/2008] [Indexed: 12/28/2022]
Abstract
Chemokines and their receptors have emerged as pivotal regulators of tumour growth, progression, and metastasis. Here we review the current knowledge on chemokines and receptors likely involved in the development of metastasis of neuroectodermal tumours, with emphasis on neuroblastoma. In this respect, we discuss the controversial role of the CXCR4/CXCL12 axis in bone marrow localization of neuroblastoma cells. In addition, we focus on the ability of neuroblastoma-derived chemokines such as CCL2 and CX3CL1 to attract lymphoid cells to the tumour site. Finally, chemokine receptor and function in other neuroectodermal tumours of adulthood (i.e. melanoma and small cell lung cancer) are discussed.
Collapse
Affiliation(s)
- Lizzia Raffaghello
- Laboratory of Oncology, G. Gaslini Institute, Largo Gerolamo Gaslini 5, 16147 Genova, Italy
| | | | | | | | | |
Collapse
|
40
|
Gross N, Meier R. Chemokines in neuroectodermal cancers: the crucial growth signal from the soil. Semin Cancer Biol 2008; 19:103-10. [PMID: 19015030 DOI: 10.1016/j.semcancer.2008.10.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2008] [Revised: 10/23/2008] [Accepted: 10/23/2008] [Indexed: 12/30/2022]
Abstract
Although chemokines and their receptors were initially identified as regulators of cell trafficking during inflammation and immune response, they have emerged as crucial players in all stages of tumor development, primary growth, migration, angiogenesis, and establishment as metastases in distant target organs. Neuroectodermal tumors regroup neoplasms originating from the embryonic neural crest cells, which display clinical and biological similarities. These tumors are highly malignant and rapidly progressing diseases that disseminate to similar target organs such as bone marrow, bone, liver and lungs. There is increasing evidence that interaction of several chemokine receptors with corresponding chemokine ligands are implicated in the growth and invasive characteristics of these tumors. In this review we summarize the current knowledge on the role of CXCL12 chemokine and its CXCR4 and CXCR7 receptors in the progression and survival of neuroectodermal tumors, with particular emphasis on neuroblastoma, the most typical and enigmatic neuroectodermal childhood tumor.
Collapse
Affiliation(s)
- Nicole Gross
- Pediatric Oncology Research, Department of Pediatrics, University Hospital, Lausanne, Switzerland.
| | | |
Collapse
|