1
|
Xu M, Zhang G, Cui T, Liu J, Wang Q, Shang D, Yu T, Guo B, Huang J, Li C. Cross-modal integration of bulk RNA-seq and single-cell RNA sequencing data to reveal T-cell exhaustion in colorectal cancer. J Cell Mol Med 2024; 28:e70101. [PMID: 39344205 PMCID: PMC11439987 DOI: 10.1111/jcmm.70101] [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: 05/22/2024] [Revised: 08/20/2024] [Accepted: 09/09/2024] [Indexed: 10/01/2024] Open
Abstract
Colorectal cancer (CRC) is a relatively common malignancy clinically and the second leading cause of cancer-related deaths. Recent studies have identified T-cell exhaustion as playing a crucial role in the pathogenesis of CRC. A long-standing challenge in the clinical management of CRC is to understand how T cells function during its progression and metastasis, and whether potential therapeutic targets for CRC treatment can be predicted through T cells. Here, we propose DeepTEX, a multi-omics deep learning approach that integrates cross-model data to investigate the heterogeneity of T-cell exhaustion in CRC. DeepTEX uses a domain adaptation model to align the data distributions from two different modalities and applies a cross-modal knowledge distillation model to predict the heterogeneity of T-cell exhaustion across diverse patients, identifying key functional pathways and genes. DeepTEX offers valuable insights into the application of deep learning in multi-omics, providing crucial data for exploring the stages of T-cell exhaustion associated with CRC and relevant therapeutic targets.
Collapse
Affiliation(s)
- Mingcong Xu
- School of Computer Science and TechnologyHarbin University of Science and TechnologyHarbinChina
- Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, The First Affiliated Hospital, University of South ChinaHengyangHunanChina
| | - Guorui Zhang
- Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, The First Affiliated Hospital, University of South ChinaHengyangHunanChina
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South ChinaHengyangHunanChina
| | - Ting Cui
- Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, The First Affiliated Hospital, University of South ChinaHengyangHunanChina
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South ChinaHengyangHunanChina
| | - Jiaqi Liu
- Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, The First Affiliated Hospital, University of South ChinaHengyangHunanChina
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South ChinaHengyangHunanChina
- Hunan Provincial Key Laboratory of Multi‐Omics and Artificial Intelligence of Cardiovascular DiseasesUniversity of South ChinaHengyangHunanChina
| | - Qiuyu Wang
- Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, The First Affiliated Hospital, University of South ChinaHengyangHunanChina
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South ChinaHengyangHunanChina
- Hunan Provincial Key Laboratory of Multi‐Omics and Artificial Intelligence of Cardiovascular DiseasesUniversity of South ChinaHengyangHunanChina
| | - Desi Shang
- Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, The First Affiliated Hospital, University of South ChinaHengyangHunanChina
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South ChinaHengyangHunanChina
- Hunan Provincial Key Laboratory of Multi‐Omics and Artificial Intelligence of Cardiovascular DiseasesUniversity of South ChinaHengyangHunanChina
| | - Tingting Yu
- Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, The First Affiliated Hospital, University of South ChinaHengyangHunanChina
| | - Bingzhou Guo
- College of Artificial Intelligence and Big Data for Medical Sciences, Shandong First Medical UniversityJinanShandongChina
| | - Jinjie Huang
- School of Computer Science and TechnologyHarbin University of Science and TechnologyHarbinChina
| | - Chunquan Li
- Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, The First Affiliated Hospital, University of South ChinaHengyangHunanChina
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South ChinaHengyangHunanChina
- Hunan Provincial Key Laboratory of Multi‐Omics and Artificial Intelligence of Cardiovascular DiseasesUniversity of South ChinaHengyangHunanChina
| |
Collapse
|
2
|
Sun J, Luo J, Jiang F, Zhao J, Zhou S, Wang L, Zhang D, Ding Y, Li X. Exploring the cross-cancer effect of circulating proteins and discovering potential intervention targets for 13 site-specific cancers. J Natl Cancer Inst 2024; 116:565-573. [PMID: 38039160 DOI: 10.1093/jnci/djad247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/23/2023] [Accepted: 11/20/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND The proteome is an important reservoir of potential therapeutic targets for cancer. This study aimed to examine the causal associations between plasma proteins and cancer risk and to identify proteins with cross-cancer effects. METHODS Genetic instruments for 3991 plasma proteins were extracted from a large-scale proteomic study. Summary-level data of 13 site-specific cancers were derived from publicly available datasets. Proteome-wide Mendelian randomization and colocalization analyses were used to investigate the causal effect of circulating proteins on cancers. Protein-protein interactions and druggability assessment were conducted to prioritize potential therapeutic targets. Finally, systematical Mendelian randomization analysis between healthy lifestyle factors and cancer-related proteins was conducted to identify which proteins could act as interventional targets by lifestyle changes. RESULTS Genetically determined circulating levels of 58 proteins were statistically significantly associated with 7 site-specific cancers. A total of 39 proteins were prioritized by colocalization, of them, 11 proteins (ADPGK, CD86, CLSTN3, CSF2RA, CXCL10, GZMM, IL6R, NCR3, SIGLEC5, SIGLEC14, and TAPBP) were observed to have cross-cancer effects. Notably, 5 of these identified proteins (CD86, CSF2RA, CXCL10, IL6R, and TAPBP) have been targeted for drug development in cancer therapy; 8 proteins (ADPGK, CD86, CXCL10, GZMM, IL6R, SIGLEC5, SIGLEC14, TAPBP) could be modulated by healthy lifestyles. CONCLUSION Our study identified 39 circulating protein biomarkers with convincing causal evidence for 7 site-specific cancers, with 11 proteins demonstrating cross-cancer effects, and prioritized the proteins as potential intervention targets by either drugs or lifestyle changes, which provided new insights into the etiology, prevention, and treatment of cancers.
Collapse
Affiliation(s)
- Jing Sun
- Department of Big Data in Health Science School of Public Health, and Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jia Luo
- Department of Epidemiology and Health Statistics, the School of Public Health of Qingdao University, Qingdao, Shandong Province, China
| | - Fangyuan Jiang
- Department of Big Data in Health Science School of Public Health, and Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jianhui Zhao
- Department of Big Data in Health Science School of Public Health, and Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Siyun Zhou
- Department of Big Data in Health Science School of Public Health, and Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lijuan Wang
- Centre for Global Health, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics, the School of Public Health of Qingdao University, Qingdao, Shandong Province, China
| | - Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xue Li
- Department of Big Data in Health Science School of Public Health, and Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
3
|
Mokhtari K, Peymani M, Rashidi M, Hushmandi K, Ghaedi K, Taheriazam A, Hashemi M. Colon cancer transcriptome. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 180-181:49-82. [PMID: 37059270 DOI: 10.1016/j.pbiomolbio.2023.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/31/2023] [Accepted: 04/06/2023] [Indexed: 04/16/2023]
Abstract
Over the last four decades, methodological innovations have continuously changed transcriptome profiling. It is now feasible to sequence and quantify the transcriptional outputs of individual cells or thousands of samples using RNA sequencing (RNA-seq). These transcriptomes serve as a connection between cellular behaviors and their underlying molecular mechanisms, such as mutations. This relationship, in the context of cancer, provides a chance to unravel tumor complexity and heterogeneity and uncover novel biomarkers or treatment options. Since colon cancer is one of the most frequent malignancies, its prognosis and diagnosis seem to be critical. The transcriptome technology is developing for an earlier and more accurate diagnosis of cancer which can provide better protectivity and prognostic utility to medical teams and patients. A transcriptome is a whole set of expressed coding and non-coding RNAs in an individual or cell population. The cancer transcriptome includes RNA-based changes. The combined genome and transcriptome of a patient may provide a comprehensive picture of their cancer, and this information is beginning to affect treatment decision-making in real-time. A full assessment of the transcriptome of colon (colorectal) cancer has been assessed in this review paper based on risk factors such as age, obesity, gender, alcohol use, race, and also different stages of cancer, as well as non-coding RNAs like circRNAs, miRNAs, lncRNAs, and siRNAs. Similarly, they have been examined independently in the transcriptome study of colon cancer.
Collapse
Affiliation(s)
- Khatere Mokhtari
- Department of Modern Biology, ACECR Institute of Higher Education (Isfahan Branch), Isfahan, Iran
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, 4815733971, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, 4815733971, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Kamran Ghaedi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
4
|
Yu W, He G, Zhang W, Ye Z, Zhong Z, Huang S. INHBB is a novel prognostic biomarker and correlated with immune infiltrates in gastric cancer. Front Genet 2022; 13:933862. [PMID: 36118865 PMCID: PMC9478859 DOI: 10.3389/fgene.2022.933862] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 08/12/2022] [Indexed: 12/24/2022] Open
Abstract
Inhibin subunit beta B (INHBB) is a potential prognostic biomarker for a variety of cancers. However, its role in gastric cancer (GC) remains elusive. The differential expression data of INHBB in tumor and normal tissues were extracted from several databases and genetic alterations of INHBB were assessed by cBioPortal. Kaplan-Meier analysis was used to evaluate the survival rate of patients with GC with INHBB and association with clinical features in GC. Cox regression analysis was used to explore the prognostic value of clinical indicators and INHBB in GC, and a nomogram prognostic model was established. In addition, the predictive validity of the nomogram model was assessed by time-depended receiver operating characteristic (ROC) and calibration curves. Functional enrichment analyses were conducted to functionally annotate INHBB. Notably, we found that the quantitative assessment of immune cell subpopulation infiltration correlated with INHBB expression. INHBB expression is upregulated in GC and is correlated with several clinical features including prognostic indicators and a histological type. Genetic alterations were observed in INHBB, its DNA methylation level was negatively correlated with INHBB expression. High INHBB expression is associated with a poor prognosis and is an independent risk factor for prognosis in GC, along with age and residual tumor. The nomogram model showed a good prediction ability and was validated by time-depended ROC and calibration curves. Functional enrichment analysis indicated that INHBB-associated genes were enriched in tumor microenvironment Gene Ontology (GO) terms and were correlated with tumor-associated pathways. INHBB has a regulatory function in immune cell infiltration, especially macrophage infiltration in GC. Specifically, patients with GC with high INHBB expression and high macrophage infiltration have a worse prognosis. INHBB expression was negatively correlated with the expression of chemokines/chemokine receptors and plays a regulatory role in immunoinhibitor/immunostimulator-involved pathways. INHBB is a potential prognostic biomarker for GC and may drive the abnormal activity of critical cancer-associated pathways, potentially contributing to immune cell infiltration to promote GC development.
Collapse
Affiliation(s)
- Weifeng Yu
- Gastroenterology Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Gastroenterology Department, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Guihua He
- Gastroenterology Department, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Wang Zhang
- Gastroenterology Department, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Zhenhao Ye
- Gastroenterology Department, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Zishao Zhong
- Gastroenterology Department, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- *Correspondence: Zishao Zhong, ; Suiping Huang,
| | - Suiping Huang
- Gastroenterology Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Gastroenterology Department, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Zishao Zhong, ; Suiping Huang,
| |
Collapse
|
5
|
Singh A, Chatterjee A, Rakshit S, Shanmugam G, Mohanty LM, Sarkar K. Neem Leaf Glycoprotein in immunoregulation of cancer. Hum Immunol 2022; 83:768-777. [DOI: 10.1016/j.humimm.2022.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/09/2022] [Accepted: 08/16/2022] [Indexed: 11/04/2022]
|
6
|
Braoudaki M, Ahmad MS, Mustafov D, Seriah S, Siddiqui MN, Siddiqui SS. Chemokines and chemokine receptors in colorectal cancer; multifarious roles and clinical impact. Semin Cancer Biol 2022; 86:436-449. [PMID: 35700938 DOI: 10.1016/j.semcancer.2022.06.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 11/19/2022]
Abstract
Colorectal cancer (CRC) is considered the second cause of cancer death worldwide. The early diagnosis plays a key role in patient prognosis and subsequently overall survival. Similar to several types of cancer, colorectal cancer is also characterised by drug resistance and heterogeneity that contribute to its complexity -especially at advanced stages. However, despite the extensive research related to the identification of biomarkers associated to early diagnosis, accurate prognosis and the management of CRC patients, little progress has been made thus far. Therefore, the mortality rates, especially at advanced stages, remain high. A large family of chemoattractant cytokines called chemokines are known for their significant role in inflammation and immunity. Chemokines released by the different tumorous cells play a key role in increasing the complexity of the tumour's microenvironment. The current review investigates the role of chemokines and chemokine receptors in colorectal cancer and their potential as clinical molecular signatures that could be effectively used as a personalised therapeutic approach. We discussed how chemokine and chemokine receptors regulate the microenvironment and lead to heterogeneity in CRC. An important aspect of chemokines is their role in drug resistance which has been extensively discussed. This review also provides an overview of the current advances in the search for chemokines and chemokine receptors in CRC.
Collapse
Affiliation(s)
- Maria Braoudaki
- Dept of Clinical, Pharmaceutical and Biological Sciences, School of Life and Medical Sciences, University of Hertfordshire, UK
| | - Mohammed Saqif Ahmad
- Dept of Clinical, Pharmaceutical and Biological Sciences, School of Life and Medical Sciences, University of Hertfordshire, UK
| | - Denis Mustafov
- Dept of Clinical, Pharmaceutical and Biological Sciences, School of Life and Medical Sciences, University of Hertfordshire, UK
| | - Sara Seriah
- Dept of Clinical, Pharmaceutical and Biological Sciences, School of Life and Medical Sciences, University of Hertfordshire, UK
| | - Mohammad Naseem Siddiqui
- Department of Biosciences, Faculty of Natural Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Shoib Sarwar Siddiqui
- Dept of Clinical, Pharmaceutical and Biological Sciences, School of Life and Medical Sciences, University of Hertfordshire, UK.
| |
Collapse
|
7
|
High Expression of Interferon Pathway Genes CXCL10 and STAT2 Is Associated with Activated T-Cell Signature and Better Outcome of Oral Cancer Patients. J Pers Med 2022; 12:jpm12020140. [PMID: 35207629 PMCID: PMC8877377 DOI: 10.3390/jpm12020140] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 02/05/2023] Open
Abstract
To improve the survival rate of cancer patients, biomarkers for both early diagnosis and patient stratification for appropriate therapeutics play crucial roles in precision oncology. Investigation of altered gene expression and the relevant molecular pathways in cancer cells are helpful for discovering such biomarkers. In this study, we explore the potential prognostic biomarkers for oral cancer patients through systematically analyzing five oral cancer transcriptomic data sets (TCGA, GSE23558, GSE30784, GSE37991, and GSE138206). Gene Set Enrichment Analysis (GSEA) was individually applied to each data set and the upregulated Hallmark molecular pathways of each data set were intersected to generate 13 common pathways including interferon-α/γ pathways. Among the 5 oral cancer data sets, 43 interferon pathway genes were commonly upregulated and 17 genes exhibited prognostic values in TCGA cohort. After validating in another oral cancer cohort (GSE65858), high expressions of C-X-C motif chemokine ligand 10 (CXCL10) and Signal transducer and activator of transcription 2 (STAT2) were confirmed to be good prognostic biomarkers. GSEA of oral cancers stratified by CXCL10/STAT2 expression showed that activation of T-cell pathways and increased tumor infiltration scores of Type 1 T helper (Th1) and CD8+ T cells were associated with high CXCL10/STAT2 expression. These results suggest that high CXCL10/STAT2 expression can predict a favorable outcome in oral cancer patients.
Collapse
|
8
|
Karin N. Chemokines in the Landscape of Cancer Immunotherapy: How They and Their Receptors Can Be Used to Turn Cold Tumors into Hot Ones? Cancers (Basel) 2021; 13:6317. [PMID: 34944943 PMCID: PMC8699256 DOI: 10.3390/cancers13246317] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 02/07/2023] Open
Abstract
Over the last decade, monoclonal antibodies to immune checkpoint inhibitors (ICI), also known as immune checkpoint blockers (ICB), have been the most successful approach for cancer therapy. Starting with mAb to cytotoxic T lymphocyte antigen 4 (CTLA-4) inhibitors in metastatic melanoma and continuing with blockers of the interactions between program cell death 1 (PD-1) and its ligand program cell death ligand 1 (PDL-1) or program cell death ligand 2 (PDL-2), that have been approved for about 20 different indications. Yet for many cancers, ICI shows limited success. Several lines of evidence imply that the limited success in cancer immunotherapy is associated with attempts to treat patients with "cold tumors" that either lack effector T cells, or in which these cells are markedly suppressed by regulatory T cells (Tregs). Chemokines are a well-defined group of proteins that were so named due to their chemotactic properties. The current review focuses on key chemokines that not only attract leukocytes but also shape their biological properties. CXCR3 is a chemokine receptor with 3 ligands. We suggest using Ig-based fusion proteins of two of them: CXL9 and CXCL10, to enhance anti-tumor immunity and perhaps transform cold tumors into hot tumors. Potential differences between CXCL9 and CXCL10 regarding ICI are discussed. We also discuss the possibility of targeting the function or deleting a key subset of Tregs that are CCR8+ by monoclonal antibodies to CCR8. These cells are preferentially abundant in several tumors and are likely to be the key drivers in suppressing anti-cancer immune reactivity.
Collapse
Affiliation(s)
- Nathan Karin
- Department of Immunology, Faculty of Medicine, Technion, P.O. Box 9697, Haifa 31096, Israel
| |
Collapse
|
9
|
Cxcl10 chemokine induces migration of ING4-deficient breast cancer cells via a novel crosstalk mechanism between the Cxcr3 and Egfr receptors. Mol Cell Biol 2021; 42:e0038221. [PMID: 34871062 DOI: 10.1128/mcb.00382-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The chemokine Cxcl10 has been associated with poor prognosis in breast cancer, but the mechanism is not well understood. Our previous study have shown that CXCL10 was repressed by the ING4 tumor suppressor, suggesting a potential inverse functional relationship. We thus investigated a role for Cxcl10 in the context of ING4 deficiencies in breast cancer. We first analyzed public gene expression datasets and found that patients with CXCL10-high/ING4-low expressing tumors had significantly reduced disease-free survival in breast cancer. In vitro, Cxcl10 induced migration of ING4-deleted breast cancer cells, but not of ING4-intact cells. Using inhibitors, we found that Cxcl10-induced migration of ING4-deleted cells required Cxcr3, Egfr, and the Gβγ subunits downstream of Cxcr3, but not Gαi. Immunofluorescent imaging showed that Cxcl10 induced early transient colocalization between Cxcr3 and Egfr in both ING4-intact and ING4-deleted cells, which recurred only in ING4-deleted cells. A peptide agent that binds to the internal juxtamembrane domain of Egfr inhibited Cxcr3/Egfr colocalization and cell migration. Taken together, these results presented a novel mechanism of Cxcl10 that elicits migration of ING4-deleted cells, in part by inducing a physical or proximal association between Cxcr3 and Egfr and signaling downstream via Gβγ. These results further indicated that ING4 plays a critical role in the regulation of Cxcl10 signaling that enables breast cancer progression.
Collapse
|
10
|
Contribution of CXCR3-mediated signaling in the metastatic cascade of solid malignancies. Biochim Biophys Acta Rev Cancer 2021; 1876:188628. [PMID: 34560199 DOI: 10.1016/j.bbcan.2021.188628] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/15/2021] [Accepted: 09/19/2021] [Indexed: 12/20/2022]
Abstract
Metastasis is a significant cause of the mortality resulting from solid malignancies. The process of metastasis is complex and is regulated by numerous cancer cell-intrinsic and -extrinsic factors. CXCR3 is a chemokine receptor that is frequently expressed by cancer cells, endothelial cells and immune cells. CXCR3A signaling in cancer cells tends to promote the invasive and migratory phenotype of cancer cells. Indirectly, CXCR3 modulates the anti-tumor immune response resulting in variable effects that can permit or inhibit metastatic progression. Finally, the activity of CXCR3B in endothelial cells is generally angiostatic, which limits the access of cancer cells to key conduits to secondary sites. However, the interaction of these activities within a tumor and the presence of opposing CXCR3 splice variants clouds the picture of the role of CXCR3 in metastasis. Consequently, thorough analysis of the contributions of CXCR3 to cancer metastasis is necessary. This review is an in-depth examination of the involvement of CXCR3 in the metastatic process of solid malignancies.
Collapse
|
11
|
Kanumuri R, Chelluboyina AK, Biswal J, Vignesh R, Pandian J, Venu A, Vaishnavi B, Leena DJ, Jeyaraman J, Ganesan K, Aradhyam GK, Venkatraman G, Rayala SK. Small peptide inhibitor from the sequence of RUNX3 disrupts PAK1-RUNX3 interaction and abrogates its phosphorylation-dependent oncogenic function. Oncogene 2021; 40:5327-5341. [PMID: 34253860 DOI: 10.1038/s41388-021-01927-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 06/09/2021] [Accepted: 06/24/2021] [Indexed: 02/06/2023]
Abstract
P21 Activated Kinase 1 (PAK1) is an oncogenic serine/threonine kinase known to play a significant role in the regulation of cytoskeleton and cell morphology. Runt-related transcription factor 3 (RUNX3) was initially known for its tumor suppressor function, but recent studies have reported the oncogenic role of RUNX3 in various cancers. Previous findings from our laboratory provided evidence that Threonine 209 phosphorylation of RUNX3 acts as a molecular switch in dictating the tissue-specific dualistic functions of RUNX3 for the first time. Based on these proofs and to explore the translational significance of these findings, we designed a small peptide (RMR) from the protein sequence of RUNX3 flanking the Threonine 209 phosphorylation site. The selection of this specific peptide from multiple possible peptides was based on their binding energies, hydrogen bonding, docking efficiency with the active site of PAK1 and their ability to displace PAK1-RUNX3 interaction in our prediction models. We found that this peptide is stable both in in vitro and in vivo conditions, not toxic to normal cells and inhibits the Threonine 209 phosphorylation in RUNX3 by PAK1. We also tested the efficacy of this peptide to block the RUNX3 Threonine 209 phosphorylation mediated tumorigenic functions in in vitro cell culture models, patient-derived explant (PDE) models and in in vivo tumor xenograft models. These results proved that this peptide has the potential to be developed as an efficient therapeutic molecule for targeting RUNX3 Threonine 209 phosphorylation-dependent tumor phenotypes.
Collapse
Affiliation(s)
- Rahul Kanumuri
- Department of Biotechnology, Indian Institute of technology Madras (IITM), Chennai, Tamilnadu, India
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education & Research (Deemed to be University), Porur, Chennai, Tamilnadu, India
| | - Aruna Kumar Chelluboyina
- Department of Biotechnology, Indian Institute of technology Madras (IITM), Chennai, Tamilnadu, India
- Division of General Medical Sciences - Oncology, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Jayashree Biswal
- Structural Biology and Bio-Computing Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi, India
| | - Ravichandran Vignesh
- Department of Biotechnology, Indian Institute of technology Madras (IITM), Chennai, Tamilnadu, India
| | - Jaishree Pandian
- Unit of Excellence in Cancer Genetics, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Akkanapally Venu
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education & Research (Deemed to be University), Porur, Chennai, Tamilnadu, India
| | - B Vaishnavi
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education & Research (Deemed to be University), Porur, Chennai, Tamilnadu, India
| | - D J Leena
- Department of Pathology, Sri Ramachandra Institute of Higher Education & Research (Deemed to be University), Porur, Chennai, Tamilnadu, India
| | - Jeyakanthan Jeyaraman
- Structural Biology and Bio-Computing Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi, India
| | - Kumaresan Ganesan
- Unit of Excellence in Cancer Genetics, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Gopala Krishna Aradhyam
- Department of Biotechnology, Indian Institute of technology Madras (IITM), Chennai, Tamilnadu, India
| | - Ganesh Venkatraman
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education & Research (Deemed to be University), Porur, Chennai, Tamilnadu, India.
| | - Suresh K Rayala
- Department of Biotechnology, Indian Institute of technology Madras (IITM), Chennai, Tamilnadu, India.
| |
Collapse
|
12
|
Circulating Biomarkers of Colorectal Cancer (CRC)-Their Utility in Diagnosis and Prognosis. J Clin Med 2021; 10:jcm10112391. [PMID: 34071492 PMCID: PMC8199026 DOI: 10.3390/jcm10112391] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/25/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023] Open
Abstract
The global burden of colorectal cancer (CRC) is expected to increase, with 2.2 million new cases and 1.1 million annual deaths by 2030. Therefore, the establishment of novel biomarkers useful in the early diagnosis of CRC is of utmost importance. A number of publications have documented the significance of the overexpression of several specific proteins, such as inflammatory mediators, in CRC progression. However, little is known about the potential utility of these proteins as circulating blood tumor biomarkers of CRC. Therefore, in the present review we report the results of our previous original studies as well as the findings of other authors who investigated whether inflammatory mediators might be used as novel biomarkers in the diagnosis and prognosis of CRC. Our study revealed that among all of the tested proteins, serum M-CSF, CXCL-8, IL-6 and TIMP-1 have the greatest value in the diagnosis and progression of CRC. Serum TIMP-1 is useful in differentiating between CRC and colorectal adenomas, whereas M-CSF and CRP are independent prognostic factors for the survival of patients with CRC. This review confirms the promising significance of these proteins as circulating biomarkers for CRC. However, due to their non-specific nature, further validation of their sensitivity and specificity is required.
Collapse
|
13
|
Mechanisms of Immune Escape and Resistance to Checkpoint Inhibitor Therapies in Mismatch Repair Deficient Metastatic Colorectal Cancers. Cancers (Basel) 2021; 13:cancers13112638. [PMID: 34072037 PMCID: PMC8199207 DOI: 10.3390/cancers13112638] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary A subset of colorectal cancers (CRCs) is characterized by a mismatch repair deficiency that is frequently associated with microsatellite instability (MSI). The compromised DNA repair machinery leads to the accumulation of tumor neoantigens affecting the sensitivity of MSI metastatic CRC to immune checkpoint inhibitors (CPIs), both upfront and in later lines of treatment. However, up to 30% of MSI CRCs exhibit primary resistance to frontline immune based therapy, and an additional subset develops acquired resistance. Here, we first discuss the clinical and molecular features of MSI CRCs and then we review how the loss of antigenicity, immunogenicity, and a hostile tumor microenvironment could influence primary and acquired resistance to CPIs. Finally, we describe strategies to improve the outcome of MSI CRC patients upon CPI treatment. Abstract Immune checkpoint inhibitors (CPIs) represent an effective therapeutic strategy for several different types of solid tumors and are remarkably effective in mismatch repair deficient (MMRd) tumors, including colorectal cancer (CRC). The prevalent view is that the elevated and dynamic neoantigen burden associated with the mutator phenotype of MMRd fosters enhanced immune surveillance of these cancers. In addition, recent findings suggest that MMRd tumors have increased cytosolic DNA, which triggers the cGAS STING pathway, leading to interferon-mediated immune response. Unfortunately, approximately 30% of MMRd CRC exhibit primary resistance to CPIs, while a substantial fraction of tumors acquires resistance after an initial benefit. Profiling of clinical samples and preclinical studies suggests that alterations in the Wnt and the JAK-STAT signaling pathways are associated with refractoriness to CPIs. Intriguingly, mutations in the antigen presentation machinery, such as loss of MHC or Beta-2 microglobulin (B2M), are implicated in initial immune evasion but do not impair response to CPIs. In this review, we outline how understanding the mechanistic basis of immune evasion and CPI resistance in MMRd CRC provides the rationale for innovative strategies to increase the subset of patients benefiting from CPIs.
Collapse
|
14
|
Long J, YaoYi Sui Z, Sui Y, Fang S. SphK1 Promotes Cancer Progression through Activating JAK/STAT Pathway and Up-Regulating S1PR1 Expression in Colon Cancer Cells. Anticancer Agents Med Chem 2021; 22:254-260. [PMID: 33797381 DOI: 10.2174/1871520621666210401105344] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 02/09/2021] [Accepted: 02/21/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND SphK1 is a conserved lipid kinase, which can catalyze formation of tumor promoting factor sphingosine phosphate-1 (S1P). OBJECTIVE To investigate effect of SphK1 on proliferation/migration of colon cancer cells and associated mechanisms. METHODS Transcription of SphK1 gene in colon cancer cells was detected. Gene transcription of SphK1 was inhibited by transfecting with si-SphK1 gene in colon cancer cells. Effects of SphK1 inhibition (si-SphK1) on cell migration/proliferation were detected using transwell system and MTS. Gene transcription of SIP, S1PR1, S1PR2, S1PR3, and activation of JAK/STAT3 pathway were examined using RT-PCR and western blot assay. S1PR1 over-expressing plasmid was constructed and transfected into cells. Effects of S1PR1 over-expression on migration/proliferation of si-SphK1 transfected colon cancer cells and activation of JAK/STAT3 pathway were determined using RT-PCR and western blotting. RESULTS Gene transcription of SphK1 in SW480 and HT-29 colon cancer cells was significantly inhibited by transfection of si-SphK1 gene. Transwell migration and MTS findings showed that si-SphK1 transfection (si-SphK1 group) could reduce migration quantity and cell viability of colon cancer cells compared to negative control (NC) (p<0.0001). SphK1 inhibition (si-SphK1 group) significantly down-regulated S1PR1 and S1PR3 gene transcription in SW480 and HT-29 cells (p<0.0001), and decreased activation level of JAKSTAT3 signaling pathway compared to NC group (p<0.05). Over-expression of S1PR1 reversed inhibitory effects of si-SphK1 on migration/proliferation of SW480 and activation of JAK/Stat3. CONCLUSION SphK1 promoted proliferation and migration of colon cancer cells through promoting JAK/STAT activation and up-regulating S1PR1 expression.
Collapse
Affiliation(s)
- Jianting Long
- Department of Medicinal Oncology, The First Affiliated Hospital, SUN Yat-Sen University, Guangzhou, 510080. China
| | - Zhijia YaoYi Sui
- Department of Medicinal Oncology, The First Affiliated Hospital, SUN Yat-Sen University, Guangzhou, 510080. China
| | - Yi Sui
- Department of Clinical Nutrition, The First Affiliated Hospital, SUN Yat-Sen University, Guangzhou, 510080. China
| | - Shi Fang
- Department of Clinical Nutrition, The First Affiliated Hospital, SUN Yat-Sen University, Guangzhou, 510080. China
| |
Collapse
|
15
|
Identification of Differential Intestinal Mucosa Transcriptomic Biomarkers for Ulcerative Colitis by Bioinformatics Analysis. DISEASE MARKERS 2020; 2020:8876565. [PMID: 33144895 PMCID: PMC7596466 DOI: 10.1155/2020/8876565] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/05/2020] [Accepted: 09/23/2020] [Indexed: 02/07/2023]
Abstract
Background Ulcerative colitis (UC) is a complicated disease caused by the interaction between genetic and environmental factors that affect mucosal homeostasis and triggers inappropriate immune response. The purpose of the study was to identify significant biomarkers with potential therapeutic targets and the underlying mechanisms. Methods The gene expression profiles of GSE48958, GSE73661, and GSE59071 are from the GEO database. Differentially expressed genes (DEGs) were screened by the GEO2R tool. Next, the Database for Annotation, Visualization and Integrated Discovery (DAVID) was applied to analyze gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway. Then, protein-protein interaction (PPI) was visualized by Cytoscape with Search Tool for the Retrieval of Interacting Genes (STRING). Results There were a total of 128 common DEGs genes, including 86 upregulated genes enriched in extracellular space, regulation of inflammatory response, chemokine-mediated signaling pathway, response to lipopolysaccharide, and cell proliferation, while 42 downregulated genes enriched in the integral component of the membrane, the integral component of the plasma membrane, apical plasma membrane, symporter activity, and chloride channel activity. The KEGG pathway analysis results demonstrated that DEGs were particularly enriched in cytokine-cytokine receptor interaction, TNF signaling pathway, chemokine signaling pathway, pertussis, and rheumatoid arthritis. 18 central modules of the PPI networks were selected with Cytotype MCODE. Furthermore, 18 genes were found to significantly enrich in the extracellular space, inflammatory response, chemokine-mediated signaling pathway, TNF signaling pathway, regulation of cell proliferation, and immune response via reanalysis of DAVID. Conclusion The study identified DEGs, key target genes, functional pathways, and pathway analysis of UC, which may provide potential molecular targets and diagnostic biomarkers for UC.
Collapse
|
16
|
Pączek S, Łukaszewicz-Zając M, Mroczko B. Chemokines-What Is Their Role in Colorectal Cancer? Cancer Control 2020; 27:1073274820903384. [PMID: 32103675 PMCID: PMC7066593 DOI: 10.1177/1073274820903384] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Colorectal cancer (CRC) is one of the leading causes of cancer-related death. It
is the second most frequently diagnosed malignancy in Europe and third
worldwide. Colorectal malignancies diagnosed at an early stage offer a promising
survival rate. However, advanced tumors often present distant metastases even
after the complete resection of a primary tumor. Therefore, novel biomarkers of
CRC are sorely needed in the diagnosis and prognosis of this common malignancy.
A family of chemokines are composed of small, secreted proteins. They are best
known for their ability to stimulate the migration of several cell types. Some
investigations have indicated that chemokines are involved in cancer
development, including CRC. This article presents current knowledge regarding
chemokines and their specific receptors in CRC progression. Moreover, the prime
aim of this review is to summarize the potential role of these proteins as
biomarkers in the diagnosis and prognosis of CRC.
Collapse
Affiliation(s)
- Sara Pączek
- Department of Biochemical Diagnostics, Medical University of Bialystok, Poland
| | | | - Barbara Mroczko
- Department of Biochemical Diagnostics, Medical University of Bialystok, Poland.,Department of Neurodegeneration Diagnostics, Medical University of Bialystok, Poland
| |
Collapse
|
17
|
Kim SI, Kang JW, Noh JK, Jung HR, Lee YC, Lee JW, Kong M, Eun YG. Gene signature for prediction of radiosensitivity in human papillomavirus-negative head and neck squamous cell carcinoma. Radiat Oncol J 2020; 38:99-108. [PMID: 33012153 PMCID: PMC7533413 DOI: 10.3857/roj.2020.00136] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 05/06/2020] [Indexed: 11/08/2022] Open
Abstract
Purpose The probability of recurrence of cancer after adjuvant or definitive radiotherapy in patients with human papillomavirus-negative (HPV(–)) head and neck squamous cell carcinoma (HNSCC) varies for each patient. This study aimed to identify and validate radiation sensitivity signature (RSS) of patients with HPV(–) HNSCC to predict the recurrence of cancer after radiotherapy. Materials and Methods Clonogenic survival assays were performed to assess radiosensitivity in 14 HNSCC cell lines. We identified genes closely correlated with radiosensitivity and validated them in The Cancer Genome Atlas (TCGA) cohort. The validated RSS were analyzed by ingenuity pathway analysis (IPA) to identify canonical pathways, upstream regulators, diseases and functions, and gene networks related to radiosensitive genes in HPV(–) HNSCC. Results The survival fraction of 14 HNSCC cell lines after exposure to 2 Gy of radiation ranged from 48% to 72%. Six genes were positively correlated and 35 genes were negatively correlated with radioresistance, respectively. RSS was validated in the HPV(–) TCGA HNSCC cohort (n = 203), and recurrence-free survival (RFS) rate was found to be significantly lower in the radioresistant group than in the radiosensitive group (p = 0.035). Cell death and survival, cell-to-cell signaling, and cellular movement were significantly enriched in RSS, and RSSs were highly correlated with each other. Conclusion We derived a HPV(–) HNSCC-specific RSS and validated it in an independent cohort. The outcome of adjuvant or definitive radiotherapy in HPV(–) patients with HNSCC can be predicted by analyzing their RSS, which might help in establishing a personalized therapeutic plan.
Collapse
Affiliation(s)
- Su Il Kim
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul, Korea
| | - Jeong Wook Kang
- Department of Otolaryngology-Head and Neck Surgery, Kyung Hee University Medical Center, Seoul, Korea
| | - Joo Kyung Noh
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul, Korea
| | - Hae Rim Jung
- Department of Otolaryngology-Head and Neck Surgery, Kyung Hee University Medical Center, Seoul, Korea
| | - Young Chan Lee
- Department of Otolaryngology-Head and Neck Surgery, Kyung Hee University Medical Center, Seoul, Korea
| | - Jung Woo Lee
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Kyung Hee University, Seoul, Korea
| | - Moonkyoo Kong
- Division of Lung & Head and Neck Oncology, Department of Radiation Oncology, Kyung Hee University Medical Center, Seoul, Korea
| | - Young-Gyu Eun
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul, Korea.,Department of Otolaryngology-Head and Neck Surgery, Kyung Hee University Medical Center, Seoul, Korea
| |
Collapse
|
18
|
Immune Cell Infiltration and Identifying Genes of Prognostic Value in the Papillary Renal Cell Carcinoma Microenvironment by Bioinformatics Analysis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5019746. [PMID: 32775427 PMCID: PMC7399742 DOI: 10.1155/2020/5019746] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 06/21/2020] [Accepted: 07/07/2020] [Indexed: 12/21/2022]
Abstract
Papillary renal cell carcinoma (PRCC) is one of the most common histological subtypes of renal cell carcinoma. Type 1 and type 2 PRCC are reported to be clinically and biologically distinct. However, little is known about immune infiltration and the expression patterns of immune-related genes in these two histologic subtypes, thereby limiting the development of immunotherapy for PRCC. Thus, we analyzed the expression of 22 immune cells in type 1 and type 2 PRCC tissues by combining The Cancer Genome Atlas (TCGA) database with the ESTIMATE and CIBERSORT algorithms. Subsequently, we extracted a list of differentially expressed genes associated with the immune microenvironment. Multichip mRNA microarray data sets for PRCC were downloaded from the Gene Expression Omnibus (GEO) to further validate our findings. We found that the immune scores and stromal scores were associated with overall survival in patients with type 2 PRCC rather than type 1 PRCC. Tumor-infiltrating M1 and M2 macrophages could predict the clinical outcome by reflecting the host's immune capacity against type 2 PRCC. Furthermore, CCL19/CCR7, CXCL12/CXCR4, and CCL20/CCR6 were shown to be potential new targets for tumor gene therapy in type 2 PRCC. Our findings provide valuable resources for improving immunotherapy for PRCC.
Collapse
|
19
|
Karin N. CXCR3 Ligands in Cancer and Autoimmunity, Chemoattraction of Effector T Cells, and Beyond. Front Immunol 2020; 11:976. [PMID: 32547545 PMCID: PMC7274023 DOI: 10.3389/fimmu.2020.00976] [Citation(s) in RCA: 171] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 04/24/2020] [Indexed: 12/15/2022] Open
Abstract
CXCR3 is a chemokine receptor with three ligands; CXCL9, CXCL10, and CXCL11. CXCL11 binds CXCR3 with a higher affinity than the other ligands leading to receptor internalization. Long ago we reported that one of these chemokines, CXCL10, not only attracts CXCR3+ CD4+ and CD8+ effector T cells to sites of inflammation, but also direct their polarization into highly potent effector T cells. Later we showed that CXCL11 directs the linage development of T-regulatory-1 cells (Tr1). We also observed that CXCL11 and CXCL10 induce different signaling cascades via CXCR3. Collectively this suggests that CXCR3 ligands differentially regulate the biological function of T cells via biased signaling. It is generally accepted that tumor cells evolved to express several chemokine receptors and secrete their ligands. Vast majority of these chemokines support tumor growth by different mechanisms that are discussed. We suggest that CXCL10 and possibly CXCL9 differ from other chemokines by their ability to restrain tumor growth and enhance anti-tumor immunity. Along with this an accumulating number of studies showed in various human cancers a clear association between poor prognosis and low expression of CXCL10 at tumor sites, and vice versa. Finally, we discuss the possibility that CXCL9 and CXCL10 may differ in their biological function via biased signaling and its possible relevance to cancer immunotherapy. The current mini review focuses on exploring the role of CXCR3 ligands in directing the biological properties of CD4+ and CD8+ T cells in the context of cancer and autoimmunity. We believe that the combined role of these chemokines in attracting T cells and also directing their biological properties makes them key drivers of immune function.
Collapse
Affiliation(s)
- Nathan Karin
- Department of Immunology, Faculty of Medicine, Technion- Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
20
|
Nozaki E, Kobayashi T, Ohnishi H, Ohtsuka K, Masaki T, Watanabe T, Sugiyama M. C-X-C motif receptor 3A enhances proliferation and invasiveness of colorectal cancer cells, and is mediated by C-X-C motif ligand 10. Oncol Lett 2020; 19:2495-2501. [PMID: 32194750 DOI: 10.3892/ol.2020.11326] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 11/11/2019] [Indexed: 12/28/2022] Open
Abstract
The chemokine CXCL10 and its receptor CXCR3 have been demonstrated to be implicated in cancer cell proliferation and metastasis. CXCR3 has three splice variants: CXCR3A, CXCR3B and CXCR3-alt. CXCR3A and B serve multiple roles in the growth and invasiveness of a number of cancer types. However, the roles of CXCR3 isoforms in colorectal cancer (CRC) cells remain unclear. In the current study, the effects of CXCL10 and CXCR3 isoforms on proliferation and invasion of CRC cells was examined. Proliferation and invasiveness of the CRC cell line HCT116, which were transfected with CXCR3A or CXCR3B in the presence of CXCL10, were evaluated in vitro using MTT, scratch wound healing and transwell assays. MTT assay indicated that regardless of the presence or absence of CXCL10, the proliferative ability of CXCR3A-transfected HCT116 cells was enhanced compared with blank and mock cells. Scratch wound healing and transwell assays indicated that invasiveness of CXCR3A-transfected cells was greater compared with blank and mock cells. However, HCT116 cells transfected with CXCR3B did not exhibit changes in their proliferative or invasive ability. mRNA expression of MMP9, which is associated with signaling downstream of the CXCL10/CXCR3A pathway, was increased 4-fold in CXCR3A-transfected HCT116 cells compared with control cells. The results of the present study indicated that CXCL10-enhanced proliferation and invasiveness of the CRC cell line HCT116 was likely mediated by CXCR3A, but not by CXCR3B.
Collapse
Affiliation(s)
- Eriko Nozaki
- Department of Laboratory Medicine, Kyorin University School of Medicine, Mitaka, Tokyo 181-8611, Japan
| | - Takaaki Kobayashi
- Department of Medical Oncology, Kyorin University School of Medicine, Mitaka, Tokyo 181-8611, Japan
| | - Hiroaki Ohnishi
- Department of Laboratory Medicine, Kyorin University School of Medicine, Mitaka, Tokyo 181-8611, Japan
| | - Kouki Ohtsuka
- Department of Laboratory Medicine, Kyorin University School of Medicine, Mitaka, Tokyo 181-8611, Japan
| | - Tadahiko Masaki
- Department of Surgery, Kyorin University School of Medicine, Mitaka, Tokyo 181-8611, Japan
| | - Takashi Watanabe
- Department of Laboratory Medicine, Kyorin University School of Medicine, Mitaka, Tokyo 181-8611, Japan
| | - Masanori Sugiyama
- Department of Surgery, Kyorin University School of Medicine, Mitaka, Tokyo 181-8611, Japan
| |
Collapse
|
21
|
Chen J, Chen QL, Wang WH, Chen XL, Hu XQ, Liang ZQ, Cao YB, Cao YM, Su SB. Prognostic and predictive values of CXCL10 in colorectal cancer. Clin Transl Oncol 2020; 22:1548-1564. [PMID: 32016676 DOI: 10.1007/s12094-020-02299-6] [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] [Received: 12/20/2019] [Accepted: 01/09/2020] [Indexed: 01/05/2023]
Abstract
BACKGROUND The role of CXCL10 in progression and prognosis of colorectal cancer (CRC) has been studied for years, yet results remain controversial. AIM This study aims to explore the relationship between CXCL10 and CRC progression and prognosis. METHODS We evaluated plasma CXCL10 in CRC patients using ELISA. We also performed a meta-analysis of the associations between CXCL10 and overall survival (OS), disease-free survival (DFS), disease-specific survival (DSS), relapse-free survival (RFS), and clinicopathological features. Finally, correlations between CXCL10 and methylation or immune infiltration were performed using TCGA data. RESULTS ELISA analysis showed that CXCL10 was associated with age, red blood cells, blood platelets, and blood urea nitrogen. A separate analysis of 3,763 patients from 24 studies revealed that there were significant associations between low CXCL10 expression and OS (HR 1.25, 95% CI 1.01-1.53), DFS (HR 1.65, 95% CI 1.17-2.34), and RFS (HR 1.43, 95% CI 1.20-1.71) in CRC. Additionally, downregulated CXCL10 expression was significantly correlated with age [odds ratio (OR) 1.31, 95% CI 1.13-1.52], metastasis (OR 1.34, 95% CI 1.11-1.63), recurrence (OR 1.46, 95% CI 1.16-1.83), tumor location (OR 1.88, 95% CI 1.58-2.24), differentiation (OR 0.57, 95% CI 0.35-0.93), microsatellite instability (OR 0.23, 95% CI 0.15-0.35), BRAF mutation (OR 1.62, 95% CI 1.25-2.08), p53 mutation (OR 0.28, 95% CI 0.16-0.47), and CIMP (OR 0.27, 95% CI 0.17-0.43). Furthermore, significant associations were observed between CXCL10 and methylation and immune infiltration. CONCLUSIONS The study suggests that CXCL10 might be a potential target for the treatment of CRC. TRIAL REGISTRATION NCT03189992. Registered 4 June 2017, https://www.clinicaltrials.gov/ct2/show/study/NCT03189992?term=NCT03189992&rank=1 .
Collapse
Affiliation(s)
- J Chen
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200082, China.,Research Center for Traditional Chinese Medicine Complexity System, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.,Shanghai TCM-Integrated Institute of Vascular Anomalies, Shanghai, 200082, China.,Institute of Vascular Anomalies, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200082, China
| | - Q-L Chen
- Research Center for Traditional Chinese Medicine Complexity System, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - W-H Wang
- Department of Medical Oncology, Shuguang Hospital Affiliated Baoshan Branch, Shanghai University of Traditional Chinese Medicine, Shanghai, 201901, China
| | - X-L Chen
- Research Center for Traditional Chinese Medicine Complexity System, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - X-Q Hu
- Research Center for Traditional Chinese Medicine Complexity System, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.,Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Z-Q Liang
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200082, China
| | - Y-B Cao
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200082, China.,Shanghai TCM-Integrated Institute of Vascular Anomalies, Shanghai, 200082, China.,Institute of Vascular Anomalies, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200082, China
| | - Y-M Cao
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200082, China.,Shanghai TCM-Integrated Institute of Vascular Anomalies, Shanghai, 200082, China.,Institute of Vascular Anomalies, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200082, China
| | - S-B Su
- Research Center for Traditional Chinese Medicine Complexity System, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
22
|
Tuomisto AE, Mäkinen MJ, Väyrynen JP. Systemic inflammation in colorectal cancer: Underlying factors, effects, and prognostic significance. World J Gastroenterol 2019; 25:4383-4404. [PMID: 31496619 PMCID: PMC6710177 DOI: 10.3748/wjg.v25.i31.4383] [Citation(s) in RCA: 186] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 06/07/2019] [Accepted: 07/19/2019] [Indexed: 02/06/2023] Open
Abstract
Systemic inflammation is a marker of poor prognosis preoperatively present in around 20%-40% of colorectal cancer patients. The hallmarks of systemic inflammation include an increased production of proinflammatory cytokines and acute phase proteins that enter the circulation. While the low-level systemic inflammation is often clinically silent, its consequences are many and may ultimately lead to chronic cancer-associated wasting, cachexia. In this review, we discuss the pathogenesis of cancer-related systemic inflammation, explore the role of systemic inflammation in promoting cancer growth, escaping antitumor defense, and shifting metabolic pathways, and how these changes are related to less favorable outcome.
Collapse
Affiliation(s)
- Anne E Tuomisto
- Cancer and Translational Medicine Research Unit, University of Oulu, Oulu 90220, Finland
- Department of Pathology, Oulu University Hospital and Medical Research Center Oulu, Oulu 90220, Finland
| | - Markus J Mäkinen
- Cancer and Translational Medicine Research Unit, University of Oulu, Oulu 90220, Finland
- Department of Pathology, Oulu University Hospital and Medical Research Center Oulu, Oulu 90220, Finland
| | - Juha P Väyrynen
- Cancer and Translational Medicine Research Unit, University of Oulu, Oulu 90220, Finland
- Department of Pathology, Oulu University Hospital and Medical Research Center Oulu, Oulu 90220, Finland
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115, United States
| |
Collapse
|
23
|
Zhang J, Shen L, Deng Y, Sun X, Wang Y, Yao Y, Zhang H, Zou W, Zhang Z, Wan J, Yang L, Zhu J, Zhang Z. A novel LARCassigner3 classification predicts outcomes in patients with locally advanced rectal cancer treated with neoadjuvant chemoradiotherapy: a retrospective training and validation analysis. Cancer Manag Res 2019; 11:4153-4170. [PMID: 31123421 PMCID: PMC6511254 DOI: 10.2147/cmar.s196662] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 03/19/2019] [Indexed: 12/14/2022] Open
Abstract
Purpose: To build and validate a predictive model of outcome for patients with locally advanced rectal cancer (LARC) treated with neoadjuvant chemoradiotherapy. Materials and methods: We developed a LARCassigner3 classifier based on tumor and paired normal tissues of patients treated with neoadjuvant chemoradiation and surgery from January 2007 to December 2012 in Fudan University Shanghai Cancer Center. Excluding 23 pairs of tissues failed in the RNA quality test, rested 197 patients were divided into discovery (n=98) and validation (n=99) cohorts randomly. Median follow-up time was 58 months. We used the Kaplan-Meier method to estimate disease-free survival (DFS), overall survival (OS), local recurrent, and distant metastatic rate We constructed a multivariate Cox model to identify the variables independently associated with progression-free and OS. Results: We identified three classifier genes related to relevant colorectal cancer features (CXCL9, SFRP2, and CD44) that formed the LARCassigner3 classifier assay. In the discovery set, the median DFS was 48.1 months (95% confidence interval (CI) 47.3-49.5) in the low-risk group and 23.4 months (95% CI 22.1-24.8) in the high-risk group (p=0.0134); the median OS was 39.2 months (95% CI 38.4-40.3) in the high-risk group and 19.1 months (95% CI 18.3-20.7) in the low-risk group (p=0.0134); 5-year distant metastasis was 13.9% (95% CI 9.0-21.3) in the low-risk group and 49.8% (95% CI 38.7-60.9) in the high-risk group (p=0.0072). Additionally, the different responses to neoadjuvant chemoradiotherapy and the LARCassigner3 low-risk and high-risk groups was statistically significant (p=0.004) in the discovery cohort. Similar results were obtained in the internal evaluation cohort. Conclusions: Patients with LARCassigner3 low-risk tumors were associated with a good prognosis. The clinical utility of using LARCassigner3 subtyping for the identification of patients for neoadjuvant chemoradiotherapy requires validation in dependent clinical trial cohorts.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Lijun Shen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Yun Deng
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Xiaoyang Sun
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Yaqi Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Ye Yao
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Hui Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Wei Zou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Zhiyuan Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Juefeng Wan
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Lifeng Yang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Ji Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Zhen Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| |
Collapse
|
24
|
Li X, Zhong Q, Luo D, Du Q, Liu W. The prognostic value of CXC subfamily ligands in stage I-III patients with colorectal cancer. PLoS One 2019; 14:e0214611. [PMID: 30973890 PMCID: PMC6459597 DOI: 10.1371/journal.pone.0214611] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 03/17/2019] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE To investigate the value of CXC subfamily ligands in stage I-III patients with colorectal cancer, in order to find a new predictor for CRC patients. METHODS We used Gene Expression Omnibus (GEO) database to collect the gene expression of CXC subfamily ligands and corresponding clinical data. The survival analysis was performed by "survival" package of Rsoftware. The CRC patients' DFS and the relationship between the expression levels of CXC subfamily ligands were evaluated by the univariate Cox regression analysis. RESULTS By using microarray data, there were 14 CXC subfamily ligands identified from dataset GSE39582. Seven CXC subfamily ligands were significantly correlated with DFS in CRC patients. (p<0.05),including CXCL1, CXCL3, CXCL9, CXCL10, CXCL11, CXCL13, and CXCL14. From multivariate Cox regression analyze, four CXC subfamily ligands (CXCL9, CXCL10, CXCL11, and CXCL13) were significantly associated with CRC patients' DFS (all p<0.05). Three CXC subfamily ligands (CXCL10, CXCL11, and CXCL13) were significantly associated with CRC patients' Overall survival (OS) (all p<0.05). Both CXCL11 and CXCL13 had the similar prediction values for DFS and OS. CONCLUSION There were seven CXC subfamily ligands were significantly correlated with DFS in CRC patients. Different expression level of four CXC subfamily ligands (CXCL9, CXCL10, CXCL11, and CXCL13) and Three CXC subfamily ligands (CXCL10, CXCL11, and CXCL13) were related to CRC patients' DFS and OS. There are still needs more experiments to confirm our conclusions. Next step we will make animal experiment about the genes in order to verified the predictive value of the CXC subfamily ligands.
Collapse
Affiliation(s)
- Xiangde Li
- Department of Radiation Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Qiulu Zhong
- Department of Radiation Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Danjing Luo
- Department of Radiation Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Qinghua Du
- Department of Radiation Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Wenqi Liu
- Department of Radiation Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
25
|
Xu G, Zhou Y, Zhou F. Development and validation of an immunity-related classifier of nine chemokines for predicting recurrence in stage I-III patients with colorectal cancer after operation. Cancer Manag Res 2018; 10:4051-4064. [PMID: 30323661 PMCID: PMC6173492 DOI: 10.2147/cmar.s174452] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Introduction Chemokines are closely related with tumor immunity, progression, and metastasis. We aimed to construct a multi-RNA classifier of chemokine family genes for predicting tumor recurrence in stage I-III patients with colorectal cancer (CRC) after operation. Patients and methods By analyzing microarray data, the Cox regression analysis was conducted to determine survival-related chemokine family genes and develop a multi-RNA classifier in the training set. The prognostic value of this multi-RNA classifier was further validated in the internal validation and external independent sets. Receiver operating characteristic curves were used to compare the prediction ability of the combined model of this multi-RNA classifier and stage, and this multi-RNA classifier and stage alone. Results Nine survival-related chemokines were identified in the training set. We identified a nine-chemokine classifier and classified the patients as high-risk or low-risk. Compared with CRC patients with high-risk scores, CRC patients with low-risk scores had longer disease-free survival in the training (HR=2.353, 95% CI=1.480-3.742, P<0.001), internal validation (HR=2.389, 95% CI=1.428-3.996, P<0.001), and external independent (HR=3.244, 95% CI=1.813-5.807, P<0.001) sets. This nine-chemokine classifier was an independent prognostic factor in these datasets (P<0.05). The combined model of this nine-chemokine classifier and tumor stage may tend to have higher accuracy than stage alone in the training (area under curve 0.727 vs 0.626, P<0.01), internal validation (0.668 vs 0.584, P=0.03), and external independent (0.704 vs 0.678, P>0.05) sets. This nine-chemokine classifier may only be applied in Marisa's C2, C5, and C6 subtypes patients. Conclusion Our nine-chemokine classifier is a reliable prognostic tool for some specific biological subtypes of CRC patients. It might contribute to guide the personalized treatment for high-risk patients.
Collapse
Affiliation(s)
- Guozeng Xu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China, .,Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan, China,
| | - Yuehan Zhou
- Department of Pharmacology, Guilin Medical University, Guilin 541004, Guangxi, China
| | - Fuxiang Zhou
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China, .,Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan, China,
| |
Collapse
|
26
|
Ahlén Bergman E, Hartana CA, Johansson M, Linton LB, Berglund S, Hyllienmark M, Lundgren C, Holmström B, Palmqvist K, Hansson J, Alamdari F, Huge Y, Aljabery F, Riklund K, Winerdal ME, Krantz D, Zirakzadeh AA, Marits P, Sjöholm LK, Sherif A, Winqvist O. Increased CD4 + T cell lineage commitment determined by CpG methylation correlates with better prognosis in urinary bladder cancer patients. Clin Epigenetics 2018; 10:102. [PMID: 30075815 PMCID: PMC6076404 DOI: 10.1186/s13148-018-0536-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 07/24/2018] [Indexed: 12/24/2022] Open
Abstract
Background Urinary bladder cancer is a common malignancy worldwide. Environmental factors and chronic inflammation are correlated with the disease risk. Diagnosis is performed by transurethral resection of the bladder, and patients with muscle invasive disease preferably proceed to radical cystectomy, with or without neoadjuvant chemotherapy. The anti-tumour immune responses, known to be initiated in the tumour and draining lymph nodes, may play a major role in future treatment strategies. Thus, increasing the knowledge of tumour-associated immunological processes is important. Activated CD4+ T cells differentiate into four main separate lineages: Th1, Th2, Th17 and Treg, and they are recognized by their effector molecules IFN-γ, IL-13, IL-17A, and the transcription factor Foxp3, respectively. We have previously demonstrated signature CpG sites predictive for lineage commitment of these four major CD4+ T cell lineages. Here, we investigate the lineage commitment specifically in tumour, lymph nodes and blood and relate them to the disease stage and response to neoadjuvant chemotherapy. Results Blood, tumour and regional lymph nodes were obtained from patients at time of transurethral resection of the bladder and at radical cystectomy. Tumour-infiltrating CD4+ lymphocytes were significantly hypomethylated in all four investigated lineage loci compared to CD4+ lymphocytes in lymph nodes and blood (lymph nodes vs tumour-infiltrating lymphocytes: IFNG -4229 bp p < 0.0001, IL13 -11 bp p < 0.05, IL17A -122 bp p < 0.01 and FOXP3 -77 bp p > 0.05). Examination of individual lymph nodes displayed different methylation signatures, suggesting possible correlation with future survival. More advanced post-cystectomy tumour stages correlated significantly with increased methylation at the IFNG -4229 bp locus. Patients with complete response to neoadjuvant chemotherapy displayed significant hypomethylation in CD4+ T cells for all four investigated loci, most prominently in IFNG p < 0.0001. Neoadjuvant chemotherapy seemed to result in a relocation of Th1-committed CD4+ T cells from blood, presumably to the tumour, indicated by shifts in the methylation patterns, whereas no such shifts were seen for lineages corresponding to IL13, IL17A and FOXP3. Conclusion Increased lineage commitment in CD4+ T cells, as determined by demethylation in predictive CpG sites, is associated with lower post-cystectomy tumour stage, complete response to neoadjuvant chemotherapy and overall better outcome, suggesting epigenetic profiling of CD4+ T cell lineages as a useful readout for clinical staging. Electronic supplementary material The online version of this article (10.1186/s13148-018-0536-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Emma Ahlén Bergman
- Unit of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| | - Ciputra Adijaya Hartana
- Unit of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Markus Johansson
- Department of Urology, Sundsvall Hospital, Sundsvall, Sweden.,Department of surgical and perioperative Sciences, Urology and Andrology, Umeå University, Umeå, Sweden
| | - Ludvig B Linton
- Unit of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Sofia Berglund
- Unit of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | | | - Christian Lundgren
- Unit of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Benny Holmström
- Department of Urology, Akademiska University Hospital, Uppsala, Sweden
| | - Karin Palmqvist
- Department of surgical and perioperative Sciences, Urology and Andrology, Umeå University, Umeå, Sweden.,Department of Surgery, Urology Section, Östersund County Hospital, Östersund, Sweden
| | - Johan Hansson
- Centre for Research and Development, Faculty of Medicine, Uppsala University, County Council of Gävleborg, Uppsala, Sweden
| | | | - Ylva Huge
- Department of Clinical and Experimental Medicine, Division of Urology, Linköping University, Linköping, Sweden
| | - Firas Aljabery
- Department of Clinical and Experimental Medicine, Division of Urology, Linköping University, Linköping, Sweden
| | - Katrine Riklund
- Department of Radiation Sciences, Diagnostic Radiology, Umeå University, Umeå, Sweden
| | - Malin E Winerdal
- Unit of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - David Krantz
- Unit of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - A Ali Zirakzadeh
- Unit of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.,Department of surgical and perioperative Sciences, Urology and Andrology, Umeå University, Umeå, Sweden
| | - Per Marits
- Unit of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Louise K Sjöholm
- Center for Molecular Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Amir Sherif
- Department of surgical and perioperative Sciences, Urology and Andrology, Umeå University, Umeå, Sweden.,Department of Radiation Sciences, Diagnostic Radiology, Umeå University, Umeå, Sweden
| | - Ola Winqvist
- Unit of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
27
|
Nesseler JP, Schaue D, McBride WH, Nickers P. [Inflammatory and immune biomarkers of radiation response]. Cancer Radiother 2018; 22:180-192. [PMID: 29650389 DOI: 10.1016/j.canrad.2017.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 09/08/2017] [Indexed: 02/07/2023]
Abstract
In radiotherapy, the treatment is adapted to each individual to protect healthy tissues but delivers most of time a standard dose according to the tumor histology and site. The only biomarkers studied to individualize the treatment are the HPV status with radiation dose de-escalation strategies, and tumor hypoxia with dose escalation to hypoxic subvolumes using FMISO- or FAZA-PET imaging. In the last decades, evidence has grown about the contribution of the immune system to radiation tumor response. Many preclinical studies have identified some of the mechanisms involved. In this context, we have realised a systematic review to highlight potential inflammatory and immune biomarkers of radiotherapy response. Some are inside the tumor microenvironment, as lymphocyte infiltration or PD-L1 expression, others are circulating biomarkers, including different types of hematological cells, cytokines and chemokines.
Collapse
Affiliation(s)
- J P Nesseler
- Department of radiation oncology, David Geffen school of medicine, university of California at Los Angeles, 10833 Le Conte avenue, 90095-1714 Los Angeles, CA, États-Unis.
| | - D Schaue
- Department of radiation oncology, David Geffen school of medicine, university of California at Los Angeles, 10833 Le Conte avenue, 90095-1714 Los Angeles, CA, États-Unis
| | - W H McBride
- Department of radiation oncology, David Geffen school of medicine, university of California at Los Angeles, 10833 Le Conte avenue, 90095-1714 Los Angeles, CA, États-Unis
| | - P Nickers
- Départment de radiothérapie, centre François-Baclesse, rue Émile-Mayrisch, 4240 Esch-sur-Alzette, Luxembourg
| |
Collapse
|
28
|
Kistner L, Doll D, Holtorf A, Nitsche U, Janssen KP. Interferon-inducible CXC-chemokines are crucial immune modulators and survival predictors in colorectal cancer. Oncotarget 2017; 8:89998-90012. [PMID: 29163806 PMCID: PMC5685727 DOI: 10.18632/oncotarget.21286] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 08/26/2017] [Indexed: 12/22/2022] Open
Abstract
Tumor-infiltrating T-cells are strongly associated with prognosis in colorectal cancer, but the mechanisms governing intratumoral lymphocyte recruitment are unclear. We investigated the clinical relevance and functional contribution of interferon-regulated CXC-chemokines CXCL9, CXCL10, and CXCL11, described as T-cells attractants. Their expression was significantly elevated in tumors as compared to normal colon in 163 patients with colon cancer, represented an independent positive predictor of post-operative survival, and was highly significantly correlated with the presence of tumor-infiltrating cytotoxic CD8+ T-cells and CD4+ TH1-effector cells. The regulation of chemokine expression was investigated in established cell lines and in tissue explants from resected tumor specimen (n=22). All colorectal cancer cell lines tested, as well as stroma or endothelial cells, produced CXC-chemokines in response to cytokine stimulation. Moreover, resected tumor explants could be stimulated to produce CXC-chemokines, even in cases with initially low CXC-levels. Lastly, a causative role of chemokine expression was evaluated with an orthotopic mouse model, based on isogenic rectal CT26 cancer cells, engineered to express CXCL10. The orthotopic model demonstrated a protective and anti-metastatic role of intratumoral CXCL10 expression, mediated mainly by adaptive immunity.
Collapse
Affiliation(s)
- Larissa Kistner
- Department of Surgery, Klinikum rechts der Isar, TUM, Munich, Germany
| | - Dietrich Doll
- Department of Surgery, Klinikum rechts der Isar, TUM, Munich, Germany.,Current/Present Address: St. Marienhospital Vechta, Vechta, Germany
| | - Anne Holtorf
- Department of Surgery, Klinikum rechts der Isar, TUM, Munich, Germany
| | - Ulrich Nitsche
- Department of Surgery, Klinikum rechts der Isar, TUM, Munich, Germany
| | | |
Collapse
|
29
|
Lin C, Yan H, Yang J, Li L, Tang M, Zhao X, Nie C, Luo N, Wei Y, Yuan Z. Combination of DESI2 and IP10 gene therapy significantly improves therapeutic efficacy against murine carcinoma. Oncotarget 2017; 8:56281-56295. [PMID: 28915590 PMCID: PMC5593561 DOI: 10.18632/oncotarget.17623] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 04/20/2017] [Indexed: 02/05/2023] Open
Abstract
DESI2 (also known as PNAS-4) is a novel pro-apoptotic gene activated during the early response to DNA damage. We previously reported that overexpression of DESI2 induces S phase arrest and apoptosis by activating checkpoint kinases. The present study was designed to test whether combination of DESI2 and IP10 could improve the therapy efficacy in vitro and in vivo. The recombinant plasmid co-expressing DESI2 and IP10 was encapsulated with DOTAP/Cholesterol nanoparticle. Immunocompetent mice bearing CT26 colon carcinoma and LL2 lung cancer were treated with the complex. We found that, in vitro, the combination of DESI2 and IP10 more efficiently inhibited proliferation of CT26, LL2, SKOV3 and A549 cancer cells via apoptosis. In vivo, the combined gene therapy more significantly inhibited tumor growth and efficiently prolonged the survival of tumor bearing mice. Mechanistically, the augmented antitumor activity in vivo was associated with induction of apoptosis and inhibition of angiogenesis. The anti-angiogenesis was further mimicked by inhibiting proliferation of immortalized HUVEC cells in vitro. Meanwhile, the infiltration of lymphocytes also contributed to the enhanced antitumor effects. Depletion of CD8+ T lymphocytes significantly abrogated the antitumor activity, whereas depletion of CD4+ T cells or NK cells showed partial abrogation. Our data suggest that the combined gene therapy of DESI2 and IP10 can significantly enhance the antitumor activity as apoptosis inducer, angiogenesis inhibitor and immune response initiator. The present study may provide a novel and effective method for treating cancer.
Collapse
Affiliation(s)
- Chao Lin
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Chengdu, Sichuan University, Chengdu, 610041, China
| | - HuaYing Yan
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Chengdu, Sichuan University, Chengdu, 610041, China
- Department of Functional Imaging, Sichuan Provincial Women's and Children's Hospital, Chengdu, 610031, China
| | - Jun Yang
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Chengdu, Sichuan University, Chengdu, 610041, China
| | - Lei Li
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Chengdu, Sichuan University, Chengdu, 610041, China
| | - Mei Tang
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Chengdu, Sichuan University, Chengdu, 610041, China
| | - Xinyu Zhao
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Chengdu, Sichuan University, Chengdu, 610041, China
| | - Chunlai Nie
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Chengdu, Sichuan University, Chengdu, 610041, China
| | - Na Luo
- Nankai University School of Medicine, Collaborative Innovation Center of Biotherapy, Tianjin, 300071, China
| | - Yuquan Wei
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Chengdu, Sichuan University, Chengdu, 610041, China
| | - Zhu Yuan
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Chengdu, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
30
|
Gnjatic S, Bronte V, Brunet LR, Butler MO, Disis ML, Galon J, Hakansson LG, Hanks BA, Karanikas V, Khleif SN, Kirkwood JM, Miller LD, Schendel DJ, Tanneau I, Wigginton JM, Butterfield LH. Identifying baseline immune-related biomarkers to predict clinical outcome of immunotherapy. J Immunother Cancer 2017; 5:44. [PMID: 28515944 PMCID: PMC5432988 DOI: 10.1186/s40425-017-0243-4] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 04/26/2017] [Indexed: 12/31/2022] Open
Abstract
As cancer strikes, individuals vary not only in terms of factors that contribute to its occurrence and development, but as importantly, in their capacity to respond to treatment. While exciting new therapeutic options that mobilize the immune system against cancer have led to breakthroughs for a variety of malignancies, success is limited to a subset of patients. Pre-existing immunological features of both the host and the tumor may contribute to how patients will eventually fare with immunotherapy. A broad understanding of baseline immunity, both in the periphery and in the tumor microenvironment, is needed in order to fully realize the potential of cancer immunotherapy. Such interrogation of the tumor, blood, and host immune parameters prior to treatment is expected to identify biomarkers predictive of clinical outcome as well as to elucidate why some patients fail to respond to immunotherapy. To approach these opportunities for progress, the Society for Immunotherapy of Cancer (SITC) reconvened the Immune Biomarkers Task Force. Comprised of an international multidisciplinary panel of experts, Working Group 4 sought to make recommendations that focus on the complexity of the tumor microenvironment, with its diversity of immune genes, proteins, cells, and pathways naturally present at baseline and in circulation, and novel tools to aid in such broad analyses.
Collapse
Affiliation(s)
- Sacha Gnjatic
- Department of Hematology/Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, S5-105, 1470 Madison Avenue, Box 1128, New York, NY 10029 USA
| | - Vincenzo Bronte
- Head of Immunology Section, University of Verona, Piazzale Le L. A. Scuro, 10, Verona, Italy
| | - Laura Rosa Brunet
- Immodulon Therapeutics Ltd, Stockley Park, 6-9 The Square, Uxbridge, UK
| | - Marcus O Butler
- Princess Margaret Hospital/Ontario Cancer Institute, RM 9-622, 610 University Ave, Toronto, ON Canada
| | - Mary L Disis
- University of Washington, Tumor Vaccine Group, 850 Mercer Street, Box 358050, Seattle, WA 98109 USA
| | - Jérôme Galon
- INSERM - Cordeliers Research Center, Integrative Cancer Immunology Laboratory, 15 rue de l'Ecole de Médecine, Paris, France
| | - Leif G Hakansson
- CanImGuide Therapeutics AB, Domkyrkovägen 23, Hoellviken, Sweden
| | - Brent A Hanks
- Duke University Medical Center, 308 Research Drive, LSRC, Room C203, Box 3819, Durham, NC 27708 USA
| | - Vaios Karanikas
- Roche Innovation Center Zurich, Wagistrasse 18, Schlieren, Switzerland
| | - Samir N Khleif
- Georgia Cancer Center, Augusta University, 1120 15th Street, CN-2101A, Augusta, GA 30912 USA
| | - John M Kirkwood
- University of Pittsburgh, Hillman Cancer Center-Research Pavilion, 5117 Centre Avenue, Suite 1.32, Pittsburg, PA 15213 USA
| | - Lance D Miller
- Wake Forest School of Medicine, 1 Medical Center Blvd, Winston Salem, NC 27157 USA
| | - Dolores J Schendel
- Medigene Immunotherapies GmbH, Lochhamer Strasse 11, Planegg-Martinsried, Germany
| | | | - Jon M Wigginton
- MacroGenics, Inc., 9704 Medical Center Drive, Rockville, MD 20850 USA
| | - Lisa H Butterfield
- Department of Medicine, Surgery and Immunology, University of Pittsburgh Cancer Institute, 5117 Centre Avenue, Pittsburgh, PA 15213 USA
| |
Collapse
|
31
|
You N, Li J, Huang X, Wu K, Tang Y, Wang L, Li H, Mi N, Zheng L. COMMD7 promotes hepatocellular carcinoma through regulating CXCL10. Biomed Pharmacother 2017; 88:653-657. [PMID: 28142122 DOI: 10.1016/j.biopha.2017.01.046] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 12/28/2016] [Accepted: 01/06/2017] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is still a heavy threat to public health. However, novel therapeutic and diagnostic method for HCC is still urgently needed thus far. Based on sequence analysis and homology comparison, we previously reported a novel gene termed COMMD7, which is mapped to 20q11.22 and promotes cell proliferation in HCC cells. But the molecular mechanisms underlying the pro-tumor property of COMMD7 are not fully addressed yet. In this study, we demonstrate that the conditional medium derived from COMMD7-overexpressed HCC cell promotes proliferation of naïve HCC cells. The over-expression of COMMD7 significantly induced the migratory and invasive in HCC cells. Mechanistic study found that over-expression of COMMD7 induces C-X-C motif chemokine 10 (CXCL10) expression. Blocking CXCL10 signal transduction by neutralizing antibody abolished COMMD7-mediated cell proliferation and migration. In conclusion, COMMD7 promotes hepatocellular carcinoma through regulating CXCL10. The present data suggests a potential role of CXCL10 in the oncogenic function of COMMD7, and will lead to a better understanding of the development of HCC.
Collapse
Affiliation(s)
- Nan You
- Department of Hepatobiliary Surgery, Xinqiao Hospital of Third Military Medical University, Chongqing 400037, China
| | - Jing Li
- Department of Hepatobiliary Surgery, Xinqiao Hospital of Third Military Medical University, Chongqing 400037, China
| | - Xiaobing Huang
- Department of Hepatobiliary Surgery, Xinqiao Hospital of Third Military Medical University, Chongqing 400037, China
| | - Ke Wu
- Department of Hepatobiliary Surgery, Xinqiao Hospital of Third Military Medical University, Chongqing 400037, China
| | - Yichen Tang
- Department of Hepatobiliary Surgery, Xinqiao Hospital of Third Military Medical University, Chongqing 400037, China
| | - Liang Wang
- Department of Hepatobiliary Surgery, Xinqiao Hospital of Third Military Medical University, Chongqing 400037, China
| | - Hongyan Li
- Department of Hepatobiliary Surgery, Xinqiao Hospital of Third Military Medical University, Chongqing 400037, China
| | - Na Mi
- Department of Hepatobiliary Surgery, Xinqiao Hospital of Third Military Medical University, Chongqing 400037, China
| | - Lu Zheng
- Department of Hepatobiliary Surgery, Xinqiao Hospital of Third Military Medical University, Chongqing 400037, China.
| |
Collapse
|
32
|
Wu X, Giobbie-Hurder A, Liao X, Lawrence D, McDermott D, Zhou J, Rodig S, Hodi FS. VEGF Neutralization Plus CTLA-4 Blockade Alters Soluble and Cellular Factors Associated with Enhancing Lymphocyte Infiltration and Humoral Recognition in Melanoma. Cancer Immunol Res 2016; 4:858-868. [PMID: 27549123 DOI: 10.1158/2326-6066.cir-16-0084] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 07/29/2016] [Indexed: 12/18/2022]
Abstract
Immune recognition of tumor targets by specific cytotoxic lymphocytes is essential for the effective rejection of tumors. A phase I clinical trial of ipilimumab (an antibody that blocks CTLA-4 function) in combination with bevacizumab (an antibody that inhibits angiogenesis) in patients with metastatic melanoma found favorable clinical outcomes were associated with increased tumor endothelial activation and lymphocyte infiltration. To better understand the underlying mechanisms, we sought features and factors that changed as a function of treatment in patients. Ipilimumab plus bevacizumab (Ipi-Bev) increased tumor vascular expression of ICAM1 and VCAM1. Treatment also altered concentrations of many circulating cytokines and chemokines, including increases of CXCL10, IL1α, TNFα, CXCL1, IFNα2, and IL8, with decreases in VEGF-A in most patients. IL1α and TNFα induced expression of E-selectin, CXCL1, and VCAM1 on melanoma tumor-associated endothelial cells (TEC) in vitro and promoted adhesion of activated T cells onto TEC. VEGFA inhibited TNFα-induced expression of ICAM1 and VCAM1 and T-cell adhesion, which was blocked by bevacizumab. CXCL10 promoted T-cell migration across TEC in vitro, was frequently expressed by melanoma cells, and was upregulated in a subset of tumors in treated patients. Robust upregulation of CXCL10 in tumors was accompanied by increased T-cell infiltration. Ipi-Bev also augmented humoral immune responses recognizing targets in melanoma, tumor endothelial, and tumor mesenchymal stem cells. Our findings suggest that Ipi-Bev therapy augments immune recognition in the tumor microenvironment through enhancing lymphocyte infiltration and antibody responses. IL1α, TNFα, and CXCL10, together with VEGF neutralization, contribute to Ipi-Bev-induced melanoma immune recognition. Cancer Immunol Res; 4(10); 858-68. ©2016 AACR.
Collapse
Affiliation(s)
- Xinqi Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Anita Giobbie-Hurder
- Center for Immuno-oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts. Department of Biostatistics, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Xiaoyun Liao
- Center for Immuno-oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts. Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Donald Lawrence
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | | | - Jun Zhou
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Scott Rodig
- Center for Immuno-oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts. Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - F Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts. Melanoma Disease Center, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
33
|
An integrative approach for the identification of prognostic and predictive biomarkers in rectal cancer. Oncotarget 2016; 6:32561-74. [PMID: 26359356 PMCID: PMC4741712 DOI: 10.18632/oncotarget.4935] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 08/20/2015] [Indexed: 12/22/2022] Open
Abstract
Introduction Colorectal cancer is the third most common cancer in the world, a small fraction of which is represented by locally advanced rectal cancer (LARC). If not medically contraindicated, preoperative chemoradiotherapy, represent the standard of care for LARC patients. Unfortunately, patients shows a wide range of response rates in which approximately 20% has a complete pathological response, whereas in 20 to 40% the response is poor or absent. Results The following specific gene signature, able to discriminate responders' patients from non-responders, were founded: AKR1C3, CXCL11, CXCL10, IDO1, CXCL9, MMP12 and HLA-DRA. These genes are mainly involved in immune system pathways and interact with drugs traditionally used in the adjuvant treatment of rectal cancer. Discussion The present study suggests that new ideas for therapy could be found not only limited to studying genes differentially expressed between the two groups of patients but deepening the mechanisms, associated to response, in which they are involved. Methods Gene expression studies performed by: Agostini et al., Rimkus et al. and Kim et al. have been merged through a meta-analysis of the raw data. Gene expression data-sets have been processed using A-MADMAN. Common differentially expressed gene (DEG) were identified through SAM analysis. To further characterize the identified DEG we deeply investigated its biological role using an integrative computational biology approach.
Collapse
|
34
|
Yoo JY, Jeong JW, Fazleabas AT, Tayade C, Young SL, Lessey BA. Protein Inhibitor of Activated STAT3 (PIAS3) Is Down-Regulated in Eutopic Endometrium of Women with Endometriosis. Biol Reprod 2016; 95:11. [PMID: 27226311 PMCID: PMC5029430 DOI: 10.1095/biolreprod.115.137158] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 05/09/2016] [Indexed: 01/07/2023] Open
Abstract
Endometriosis is a major cause of chronic pelvic pain and infertility. Activation of STAT3 appears central to the inflammatory phenotype of eutopic endometrium in women with endometriosis. However, the molecular mechanism by which this occurs remains unknown. Our objective is to determine how STAT3 activity is regulated in endometriosis. Protein inhibitor of activated STAT3 (PIAS3) is a negative regulator of STAT3 activity. We examined the levels of PIAS3 in endometrium from women with and without endometriosis using Western blot analysis and immunohistochemistry. Levels of PIAS3 are significantly lower, in contrast with phosphorylation of STAT3, in women with endometriosis compared to women without endometriosis. Furthermore, induction of endometriosis in the baboon showed a significant reduction of PIAS3 expression during the progression of the disease. Interferon-γ (INFγ) reduces PIAS3 protein levels and increases phospho-STAT3 levels through CXCL10 in endometrial cells, Ishikawa, and 12Z cells. These results suggest that attenuation of PIAS3 causes aberrant activation of STAT3 in endometriosis, leading to inflammatory changes that may impair fertility or cause pain.
Collapse
Affiliation(s)
- Jung-Yoon Yoo
- Department of Obstetrics, Gynecology & Reproductive Biology, Michigan State University, Grand Rapids, Michigan
| | - Jae-Wook Jeong
- Department of Obstetrics, Gynecology & Reproductive Biology, Michigan State University, Grand Rapids, Michigan
| | - Asgerally T Fazleabas
- Department of Obstetrics, Gynecology & Reproductive Biology, Michigan State University, Grand Rapids, Michigan
| | - Chandrakant Tayade
- Department of Obstetrics and Gynecology, Queens University, Kingston, Canada
| | - Steven L Young
- Department of Obstetrics and Gynecology, University of North Carolina, Chapel Hill, North Carolina
| | - Bruce A Lessey
- Department of Obstetrics and Gynecology, University of South Carolina School of Medicine, Greenville Health System, Greenville, South Carolina
| |
Collapse
|
35
|
The Role of Chemokines in Promoting Colorectal Cancer Invasion/Metastasis. Int J Mol Sci 2016; 17:ijms17050643. [PMID: 27136535 PMCID: PMC4881469 DOI: 10.3390/ijms17050643] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 04/18/2016] [Accepted: 04/25/2016] [Indexed: 12/18/2022] Open
Abstract
Colorectal cancer (CRC) is one of the leading causes of cancer-related death worldwide. Although most of the primary CRC can be removed by surgical resection, advanced tumors sometimes show recurrences in distant organs such as the liver, lung, lymph node, bone or peritoneum even after complete resection of the primary tumors. In these advanced and metastatic CRC, it is the tumor-stroma interaction in the tumor microenvironment that often promotes cancer invasion and/or metastasis through chemokine signaling. The tumor microenvironment contains numerous host cells that may suppress or promote cancer aggressiveness. Several types of host-derived myeloid cells reside in the tumor microenvironment, and the recruitment of them is under the control of chemokine signaling. In this review, we focus on the functions of chemokine signaling that may affect tumor immunity by recruiting several types of bone marrow-derived cells (BMDC) to the tumor microenvironment of CRC.
Collapse
|
36
|
Wu Z, Huang X, Han X, Li Z, Zhu Q, Yan J, Yu S, Jin Z, Wang Z, Zheng Q, Wang Y. The chemokine CXCL9 expression is associated with better prognosis for colorectal carcinoma patients. Biomed Pharmacother 2016; 78:8-13. [PMID: 26898419 DOI: 10.1016/j.biopha.2015.12.021] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Revised: 11/20/2015] [Accepted: 12/15/2015] [Indexed: 01/25/2023] Open
Abstract
The chemokine CXCL9 has been demonstrated to play an important role in the development of human malignancies. However, its prognostic significance in cancer patients remains unclear and less is known about its role in colonrectal carcinoma (CRC) patients. In this study, we found that the relative mRNA expression level of CXCL9 in primary colorectal tumor tissues was significantly higher than that in corresponding normal colon tissues. CXCL9 protein expression was also detected in 102 of 130 primary CRC patients by immunochemistry. Thus, CXCL9 might play a vital role in the progression of colorectal cancer. By analyzing the correlation between clinicopathological factors of patients and expression of CXCL9 protein, we showed that the expression of CXCL9 was significantly associated with tumor differentiation, tumor invasion, lymph node metastasis, distant metastasis, and vascular invasion, but not with other factors of CRC patients including age, gender, tumor location and tumor size. Furthermore, by performing Kaplan-Meier method as well as Cox's univariate and multivariate hazard regression model, we found that the higher the CXCL9 expression, the higher overall survival rate was observed, and CXCL9 expression was a significant independent prognostic factor for CRC patients. Therefore, CXCL9 is a useful predictor of better clinical outcome in CRC patients.
Collapse
Affiliation(s)
- Zhenqian Wu
- Department of Surgery, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University, 600 Yishan Road, 200233 Shanghai, China
| | - Xiuyan Huang
- Department of Surgery, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University, 600 Yishan Road, 200233 Shanghai, China
| | - Xiaodong Han
- Department of Surgery, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University, 600 Yishan Road, 200233 Shanghai, China
| | - Zhongnan Li
- Department of Surgery, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University, 600 Yishan Road, 200233 Shanghai, China
| | - Qinchao Zhu
- Department of Surgery, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University, 600 Yishan Road, 200233 Shanghai, China
| | - Jun Yan
- Department of Surgery, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University, 600 Yishan Road, 200233 Shanghai, China
| | - Song Yu
- Department of Surgery, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University, 600 Yishan Road, 200233 Shanghai, China
| | - Zhiming Jin
- Department of Surgery, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University, 600 Yishan Road, 200233 Shanghai, China
| | - Zhigang Wang
- Department of Surgery, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University, 600 Yishan Road, 200233 Shanghai, China
| | - Qi Zheng
- Department of Surgery, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University, 600 Yishan Road, 200233 Shanghai, China
| | - Yu Wang
- Department of Surgery, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University, 600 Yishan Road, 200233 Shanghai, China.
| |
Collapse
|
37
|
Bai M, Chen X, Ba YI. CXCL10/CXCR3 overexpression as a biomarker of poor prognosis in patients with stage II colorectal cancer. Mol Clin Oncol 2015; 4:23-30. [PMID: 26870351 PMCID: PMC4726926 DOI: 10.3892/mco.2015.665] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 07/28/2015] [Indexed: 12/21/2022] Open
Abstract
The CXCL10/CXCR3 axis of inflammatory mediators is one of the most important groups of chemokine axes, which has been proven to be a lymphocyte-associated metastasis mediator in several tumors. The term inflammatory adhesions refers to tumors found to be attached to the surrouding tissues during surgery, although no cancer cell infiltration is later identified on pathological examination. The aim of the present study was to investigate the clinical characteristics of stage II colorectal cancer (CRC) and determine the correlation between the CXCL10/CXCR3 axis, inflammatory adhesions and prognosis. Clinicohistopathological data were collected from 401 CRC patients who had undergone R0 resection. Statistical analysis was performed with SPSS 17.0 software. Immunohistochemistry (IHC) was applied to measure the expression of CXCL10 and CXCR3 in 71 recurrent CRC patients, 72 non-recurrent CRC patients and 10 samples from normal peritumoral tissues, all retrieved from the Tianjin Medical University Cancer Institute and Hospital, Tianjin, China. Inflammatory adhesions, tumor location and size and the number of high-risk factors for reccurrence were more significantly associated with overall survival (OS) rather than disease-free survival in all the patients as determined by the log-rank and Cox's regression hazard analysis. Further analysis demonstrated that only the presence of inflammatory adhesions (P=0.025) was associated with the OS of recurrent patients. Patients with recurrence exhibited higher CXCR3 (P<0.001) and CXCL10 (P<0.001) expression compared with non-recurrent patients, as determined by IHC. The correlation between clinicopathological variables, CXCL10/CXCR3 expression and survival was also analyzed: Inflammatory adhesions and general tumor type (ulcerated vs. elevated) exhibited a significant correlation with CXCR3; however, the expression of CXCL10 was not significantly correlated with tumor location, histological type, size, gender, or preoperative carcinoembryonic antigen and hemoglobin levels. Furthermore, patients exhibiting a high expression of CXCR3 presented with a higher risk of relapse; among those, patients with inflammatory adhesions always exhibited worse survival. However, no such association was identified for CXCL10 expression. In conclusion, CXCR3 expression may be used as a prognostic marker and may contribute to the prediction of clinical outcome in stage II CRC patients.
Collapse
Affiliation(s)
- Ming Bai
- Department of Gastrointestinal Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Xia Chen
- Department of Pediatric Hematology Center, Institute of hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300041, P.R. China
| | - Y I Ba
- Department of Gastrointestinal Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| |
Collapse
|
38
|
Sato Y, Motoyama S, Nanjo H, Wakita A, Yoshino K, Sasaki T, Nagaki Y, Liu J, Imai K, Saito H, Minamiya Y. CXCL10 Expression Status is Prognostic in Patients with Advanced Thoracic Esophageal Squamous Cell Carcinoma. Ann Surg Oncol 2015; 23:936-42. [PMID: 26464192 DOI: 10.1245/s10434-015-4909-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Indexed: 02/05/2023]
Abstract
BACKGROUND CXCL10, a member of the CXC chemokine family, is known to mediate chemotaxis, apoptosis, angiogenesis, and cell growth. It is also reportedly involved in tumor development and can affect prognosis in several cancers. However, the precise relationship between CXCL10 and the prognosis of patients with esophageal squamous cell carcinoma (ESCC) is not fully understood. METHODS We used ESCC tissue microarrays containing samples from 177 patients to test whether the CXCL10 expression status, determined using immunohistochemical analysis, is predictive of prognosis. We also tested whether CXCL10 expression status could serve as a clinically useful marker for evaluating the need for adjuvant chemotherapy after surgery. RESULTS We found that high CXCL10 expression in clinical samples was an independent prognostic factor and was predictive of a favorable 5-year overall survival and disease-specific survival (p = 0.0102 and 0.0332, respectively). Additionally, no significant difference was detected between patients in the CXCL10-high group treated with surgery alone and those treated with surgery followed by adjuvant chemotherapy. In the CXCL10-low group, on the other hand, patients treated with surgery followed by adjuvant chemotherapy had better 5-year overall survival than those treated with surgery alone. CONCLUSIONS High CXCL10 expression is an independent prognostic factor and has the potential to serve as a clinically useful marker of the need for adjuvant chemotherapy after surgery in patients with advanced thoracic ESCC.
Collapse
Affiliation(s)
- Yusuke Sato
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan.
| | - Satoru Motoyama
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Hiroshi Nanjo
- Department of Pathology, Akita University Graduate School of Medicine, Akita, Japan
| | - Akiyuki Wakita
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Kei Yoshino
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Tomohiko Sasaki
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Yushi Nagaki
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Jiajia Liu
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Kazuhiro Imai
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Hajime Saito
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Yoshihiro Minamiya
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| |
Collapse
|
39
|
Hu Z, Brooks SA, Dormoy V, Hsu CW, Hsu HY, Lin LT, Massfelder T, Rathmell WK, Xia M, Al-Mulla F, Al-Temaimi R, Amedei A, Brown DG, Prudhomme KR, Colacci A, Hamid RA, Mondello C, Raju J, Ryan EP, Woodrick J, Scovassi AI, Singh N, Vaccari M, Roy R, Forte S, Memeo L, Salem HK, Lowe L, Jensen L, Bisson WH, Kleinstreuer N. Assessing the carcinogenic potential of low-dose exposures to chemical mixtures in the environment: focus on the cancer hallmark of tumor angiogenesis. Carcinogenesis 2015; 36 Suppl 1:S184-S202. [PMID: 26106137 PMCID: PMC4492067 DOI: 10.1093/carcin/bgv036] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 12/12/2014] [Accepted: 12/15/2014] [Indexed: 01/09/2023] Open
Abstract
One of the important 'hallmarks' of cancer is angiogenesis, which is the process of formation of new blood vessels that are necessary for tumor expansion, invasion and metastasis. Under normal physiological conditions, angiogenesis is well balanced and controlled by endogenous proangiogenic factors and antiangiogenic factors. However, factors produced by cancer cells, cancer stem cells and other cell types in the tumor stroma can disrupt the balance so that the tumor microenvironment favors tumor angiogenesis. These factors include vascular endothelial growth factor, endothelial tissue factor and other membrane bound receptors that mediate multiple intracellular signaling pathways that contribute to tumor angiogenesis. Though environmental exposures to certain chemicals have been found to initiate and promote tumor development, the role of these exposures (particularly to low doses of multiple substances), is largely unknown in relation to tumor angiogenesis. This review summarizes the evidence of the role of environmental chemical bioactivity and exposure in tumor angiogenesis and carcinogenesis. We identify a number of ubiquitous (prototypical) chemicals with disruptive potential that may warrant further investigation given their selectivity for high-throughput screening assay targets associated with proangiogenic pathways. We also consider the cross-hallmark relationships of a number of important angiogenic pathway targets with other cancer hallmarks and we make recommendations for future research. Understanding of the role of low-dose exposure of chemicals with disruptive potential could help us refine our approach to cancer risk assessment, and may ultimately aid in preventing cancer by reducing or eliminating exposures to synergistic mixtures of chemicals with carcinogenic potential.
Collapse
Affiliation(s)
- Zhiwei Hu
- To whom correspondence should be addressed. Tel: +1 614 685 4606; Fax: +1-614-247-7205;
| | - Samira A. Brooks
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Valérian Dormoy
- INSERM U1113, team 3 “Cell Signalling and Communication in Kidney and Prostate Cancer”, University of Strasbourg, Facultée de Médecine, 67085 Strasbourg, France
- Department of Cell and Developmental Biology, University of California, Irvine, CA 92697, USA
| | - Chia-Wen Hsu
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892-3375, USA
| | - Hsue-Yin Hsu
- Department of Life Sciences, Tzu-Chi University, Taiwan, Republic of China
| | - Liang-Tzung Lin
- Department of Microbiology and Immunology, Taipei Medical University, Taiwan, Republic of China
| | - Thierry Massfelder
- INSERM U1113, team 3 “Cell Signalling and Communication in Kidney and Prostate Cancer”, University of Strasbourg, Facultée de Médecine, 67085 Strasbourg, France
| | - W. Kimryn Rathmell
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Menghang Xia
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892-3375, USA
| | - Fahd Al-Mulla
- Department of Life Sciences, Tzu-Chi University, Taiwan, Republic of China
| | | | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Firenze, Florence 50134, Italy
| | - Dustin G. Brown
- Department of Environmental and Radiological Health Sciences
, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523, USA
| | - Kalan R. Prudhomme
- Environmental and Molecular Toxicology, Environmental Health Science Center, Oregon State University, Corvallis, OR 97331, USA
| | - Annamaria Colacci
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna, Italy
| | - Roslida A. Hamid
- Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor, Malaysia
| | - Chiara Mondello
- Institute of Molecular Genetics, National Research Council, Pavia 27100, Italy
| | - Jayadev Raju
- Regulatory Toxicology Research Division, Bureau of Chemical Safety, Food Directorate
, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada
| | - Elizabeth P. Ryan
- Department of Environmental and Radiological Health Sciences
, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523, USA
| | - Jordan Woodrick
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, WashingtonDC 20057, USA
| | - A. Ivana Scovassi
- Institute of Molecular Genetics, National Research Council, Pavia 27100, Italy
| | - Neetu Singh
- Advanced Molecular Science Research Centre (Centre for Advance Research), King George’s Medical University, Lucknow, Uttar Pradesh 226003, India
| | - Monica Vaccari
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna, Italy
| | - Rabindra Roy
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, WashingtonDC 20057, USA
| | - Stefano Forte
- Mediterranean Institute of Oncology, Viagrande 95029, Italy
| | - Lorenzo Memeo
- Mediterranean Institute of Oncology, Viagrande 95029, Italy
| | - Hosni K. Salem
- Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt
| | - Leroy Lowe
- Getting to Know Cancer, Truro, Nova Scotia B2N 1X5, Canada
| | - Lasse Jensen
- Division of Cardiovascular Medicine, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden and
| | - William H. Bisson
- Environmental and Molecular Toxicology, Environmental Health Science Center, Oregon State University, Corvallis, OR 97331, USA
| | - Nicole Kleinstreuer
- Integrated Laboratory Systems, Inc., in support of the National Toxicology Program Interagency Center for the Evaluation of Alternative Toxicological Methods, NIEHS, MD K2-16, RTP, NC 27709, USA
| |
Collapse
|
40
|
Akeus P, Langenes V, Kristensen J, von Mentzer A, Sparwasser T, Raghavan S, Quiding-Järbrink M. Treg-cell depletion promotes chemokine production and accumulation of CXCR3(+) conventional T cells in intestinal tumors. Eur J Immunol 2015; 45:1654-66. [PMID: 25754875 DOI: 10.1002/eji.201445058] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 01/26/2015] [Accepted: 03/02/2015] [Indexed: 12/20/2022]
Abstract
Colorectal cancer (CRC) is one of the most prevalent tumor types worldwide and tumor-infiltrating T cells are crucial for anti-tumor immunity. We previously demonstrated that Treg cells from CRC patients inhibit transendothelial migration of conventional T cells. However, it remains unclear if local Treg cells affect lymphocyte migration into colonic tumors. By breeding APC(Min/+) mice with depletion of regulatory T cells mice, expressing the diphtheria toxin receptor under the control of the FoxP3 promoter, we were able to selectively deplete Treg cells in tumor-bearing mice, and investigate the impact of these cells on the infiltration of conventional T cells into intestinal tumors. Short-term Treg-cell depletion led to a substantial increase in the frequencies of T cells in the tumors, attributed by both increased infiltration and proliferation of T cells in the Treg-cell-depleted tumors. We also demonstrate a selective increase of the chemokines CXCL9 and CXCL10 in Treg-cell-depleted tumors, which were accompanied by accumulation of CXCR3(+) T cells, and increased IFN-γ mRNA expression. In conclusion, Treg-cell depletion increases the accumulation of conventional T cells in intestinal tumors, and targeting Treg cells could be a possible anti-tumor immunotherapy, which not only affects T-cell effector functions, but also their recruitment to tumors.
Collapse
Affiliation(s)
- Paulina Akeus
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Veronica Langenes
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Jonas Kristensen
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Astrid von Mentzer
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Tim Sparwasser
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Sukanya Raghavan
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Marianne Quiding-Järbrink
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
41
|
Hamilton TD, Leugner D, Kopciuk K, Dixon E, Sutherland FR, Bathe OF. Identification of prognostic inflammatory factors in colorectal liver metastases. BMC Cancer 2014; 14:542. [PMID: 25069793 PMCID: PMC4125702 DOI: 10.1186/1471-2407-14-542] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 07/10/2014] [Indexed: 01/15/2023] Open
Abstract
Background The modified Glasgow Prognostic Score (mGPS) has been reported to be an important prognostic indicator in a number of tumor types, including colorectal cancer (CRC). The features of the inflammatory state thought to accompany elevated C-reactive protein (CRP), a key feature of mGPS, were characterized in patients with colorectal liver metastases. Additional inflammatory mediators that contribute to prognosis were explored. Methods In sera from 69 patients with colorectal liver metastases, a panel of 42 inflammatory mediators were quantified as a function of CRP levels, and as a function of disease-free survival. Multivariate statistical methods were used to determine association of each mediator with elevated CRP and truncated disease-free survival. Results Elevated CRP was confirmed to be a strong predictor of survival (HR 4.00, p = 0.001) and recurrence (HR 3.30, p = 0.002). The inflammatory state associated with elevated CRP was comprised of raised IL-1β, IL-6, IL-12 and IL-15. In addition, elevated IL-8 and PDGF-AB/BB and decreased eotaxin and IP-10 were associated with worse disease-free and overall survival. Conclusions Elevated CRP is associated with a proinflammatory state. The inflammatory state is an important prognostic indicator in CRC liver metastases. The individual contributions of tumor biology and the host to this inflammatory response will require further investigation. Electronic supplementary material The online version of this article (doi:10.1186/1471-2407-14-542) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | - Oliver F Bathe
- Department of Surgery, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
42
|
CXCL10 mRNA expression predicts response to neoadjuvant chemoradiotherapy in rectal cancer patients. Tumour Biol 2014; 35:9683-91. [PMID: 24969558 DOI: 10.1007/s13277-014-2234-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 06/12/2014] [Indexed: 12/24/2022] Open
Abstract
Chemoradiotherapy has been commonly used as neoadjuvant therapy for rectal cancer to allow for less aggressive surgical approaches and to improve quality of life. In cancer, it has been reported that CXCL10 has an anti-tumor function. However, the association between CXCL10 and chemoradiosensitivity has not been fully investigated. We performed this study to investigate the relationship between CXCL10 expression and chemoradiosensitivity in rectal cancer patients. Ninety-five patients with rectal cancer who received neoadjuvant chemoradiotherapy (NCRT) were included. Clinical parameters were compared with the outcome of NCRT and CXCL10 messenger RNA (mRNA) expression between the pathological complete response (pCR) group and non-pathological complete response (npCR) group. CXCL10 mRNA and protein expressions between groups were analyzed using the Student's t test and chi-square test. The mean mRNA level of CXCL10 in the pCR group was significantly higher than that in the npCR group (p = 0.010). In the pCR group, 73.7 % of the patients had high CXCL10 mRNA expression, and 61.4 % of the patients in the npCR group had low CXCL10 mRNA expression. Subjects with high CXCL10 mRNA expression demonstrated a higher sensitivity to NCRT (p = 0.011). The receiver operating characteristic curve showed that the diagnostic performance of CXCL10 mRNA expression had an area under the curve of 0.720 (95 % confidence interval, 0.573-0.867). There were no differences between the pCR and npCR groups in CXCL10 protein expression (p > 0.05). High CXCL10 mRNA expression is associated with a better tumor response to NCRT in rectal cancer patients and may predict the outcome of NCRT in this malignancy.
Collapse
|
43
|
Boissière-Michot F, Lazennec G, Frugier H, Jarlier M, Roca L, Duffour J, Du Paty E, Laune D, Blanchard F, Le Pessot F, Sabourin JC, Bibeau F. Characterization of an adaptive immune response in microsatellite-instable colorectal cancer. Oncoimmunology 2014; 3:e29256. [PMID: 25101223 PMCID: PMC4121339 DOI: 10.4161/onci.29256] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 04/25/2014] [Accepted: 05/16/2014] [Indexed: 12/12/2022] Open
Abstract
Sporadic or hereditary colorectal cancer (CRC) with microsatellite instability (MSI) is frequently characterized by inflammatory lymphocytic infiltration and tends to be associated with a better outcome than microsatellite stable (MSS) CRC, probably reflecting a more effective immune response. We investigated inflammatory mechanisms in 48 MSI CRCs and 62 MSS CRCs by analyzing: (1) the expression of 48 cytokines using Bio-Plex multiplex cytokine assays, and (2) the in situ immune response by immunohistochemical analysis with antibodies against CD3 (T lymphocytes), CD8 (cytotoxic T lymphocytes), CD45RO (memory T lymphocytes), T-bet (Th1 CD4 cells), and FoxP3 (regulatory T cells). MSI CRC exhibited significantly higher expression of CCL5 (RANTES), CXCL8 (IL-8), CXCL9 (MIG), IL-1β, CXCL10 (IP-10), IL-16, CXCL1 (GROα), and IL-1ra, and lower expression of MIF, compared with MSS CRC. Immunohistochemistry combined with image analysis indicated that the density of CD3+, CD8+, CD45RO+, and T-bet+ T lymphocytes was higher in MSI CRC than in MSS CRC, whereas the number of regulatory T cells (FoxP3+) was not statistically different between the groups. These results indicate that MSI CRC is associated with a specific cytokine expression profile that includes CCL5, CXCL10, and CXCL9, which are involved in the T helper type 1 (Th1) response and in the recruitment of memory CD45RO+ T cells. Our findings highlight the major role of adaptive immunity in MSI CRC and provide a possible explanation for the more favorable prognosis of this CRC subtype.
Collapse
Affiliation(s)
- Florence Boissière-Michot
- Pathology Department, Institut régional du Cancer de Montpellier (ICM)-Val d'Aurelle; Montpellier, France
| | | | - Hélène Frugier
- Pathology Department, Institut régional du Cancer de Montpellier (ICM)-Val d'Aurelle; Montpellier, France
| | - Marta Jarlier
- Biostatistics Department, Institut régional du Cancer de Montpellier (ICM)-Val d'Aurelle; Montpellier, France
| | - Lise Roca
- Biostatistics Department, Institut régional du Cancer de Montpellier (ICM)-Val d'Aurelle; Montpellier, France
| | - Jacqueline Duffour
- Oncology Department, Institut régional du Cancer de Montpellier (ICM)-Val d'Aurelle; Montpellier, France
| | | | | | | | | | | | - Frédéric Bibeau
- Pathology Department, Institut régional du Cancer de Montpellier (ICM)-Val d'Aurelle; Montpellier, France
| |
Collapse
|
44
|
Expression of AKR1C3 and CNN3 as markers for detection of lymph node metastases in colorectal cancer. Clin Exp Med 2014; 15:333-41. [PMID: 24934327 PMCID: PMC4522272 DOI: 10.1007/s10238-014-0298-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 05/10/2014] [Indexed: 12/21/2022]
Abstract
The aim of the study was to identify a set of discriminating genes that could be used for the prediction of Lymph node (LN) metastasis in human colorectal cancer (CRC), and for this, we compared the whole genome profiles of two CRC cell lines (the primary cell line SW480 and its LN metastatic variant, SW620) and identified eight genes [S100 calcium-binding protein P; aldo–keto reductase family 1(AKR1), member B1 (aldose reductase; AKR1B1); AKR1, member C3 (AKR1C3); calponin 3, acidic; metastasis associated in colon cancer 1; hemoglobin, epsilon 1; trefoil factor 3; and FGGY carbohydrate kinase domain containing]. These genes were examined by quantitative RT-PCR in tissues and LNs in 14 CRC patients and 11 control patients. The level of AKR1C3 mRNA expression was significantly different between the Dukes’ stage A, B, and C groups and the control group (p < 0.05, p < 0.001, and p < 0.001) and was also significantly different between Dukes’ stage C and A or B groups (p < 0.05 and p < 0.001, respectively). The expression of CNN3 was significantly different between the Dukes’ stage C and B or control groups (p < 0.001 and p < 0.01, respectively). There were significant correlations between the expression levels of AKR1C3 and CNN3. AKR1C3 and CNN3 expressions are more accurate and suitable markers for the diagnosis of LN metastasis than the other six genes examined in this study.
Collapse
|
45
|
Dimberg J, Skarstedt M, Löfgren S, Zar N, Matussek A. Protein expression and gene polymorphism of CXCL10 in patients with colorectal cancer. Biomed Rep 2014; 2:340-343. [PMID: 24748971 PMCID: PMC3990219 DOI: 10.3892/br.2014.255] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 03/04/2014] [Indexed: 12/30/2022] Open
Abstract
Chemokines (chemotactic cytokines) promote leukocyte attraction to sites of inflammation and cancer. Certain chemokines promote and regulate neoplastic progression, including metastasis and angiogenesis. One such chemokine, CXCL10, was found to be expressed in colorectal cancer (CRC) tissue. To gain insight into the prognostic significance of CXCL10, we investigated whether the levels of this chemokine were altered in the colorectal tissue or plasma of CRC patients. Using Luminex technology for protein analyses, we observed a significantly higher CXCL10 protein level in cancer tissue compared to that in paired normal tissue. Moreover, significantly higher plasma levels of CXCL10 were detected in patients compared to those in control subjects and the plasma levels of CXCL10 in disseminated disease were found to be significantly higher compared to those in localized disease. The single-nucleotide polymorphism rs8878, which has been described in exon 4 in the 3′-untranslated region of the CXCL10 gene, was investigated using a TaqMan system. There were significant differences in genotype distribution and allelic frequencies between CRC patients and control subjects. In conclusion, altered CXCL10 protein concentrations in CRC tissues or plasma and the rs8878 genotype variant of CXCL10 may contribute to the prediction of clinical outcome.
Collapse
Affiliation(s)
- Jan Dimberg
- Department of Natural Science and Biomedicine, University College of Health Sciences, Jönköping, Småland SE-55111, Ryhov County Hospital, Jönköping, Småland SE-55185, Sweden
| | - Marita Skarstedt
- Department of Clinical Microbiology, Ryhov County Hospital, Jönköping, Småland SE-55185, Sweden
| | - Sture Löfgren
- Department of Clinical Microbiology, Ryhov County Hospital, Jönköping, Småland SE-55185, Sweden
| | - Niklas Zar
- Department of Surgery, Ryhov County Hospital, Jönköping, Småland SE-55185, Sweden
| | - Andreas Matussek
- Department of Laboratory Services, Ryhov County Hospital, Jönköping, Småland SE-55185, Sweden
| |
Collapse
|
46
|
Comstock SS, Hortos K, Kovan B, McCaskey S, Pathak DR, Fenton JI. Adipokines and obesity are associated with colorectal polyps in adult males: a cross-sectional study. PLoS One 2014; 9:e85939. [PMID: 24465801 PMCID: PMC3895019 DOI: 10.1371/journal.pone.0085939] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 12/03/2013] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Obesity increases the risk of colon cancer. It is also known that most colorectal cancers develop from adenomatous polyps. However, the effects of obesity and adipokines on colonic polyp formation are unknown. METHODS To determine if BMI, waist circumference or adipokines are associated with colon polyps in males, 126 asymptomatic men (48-65 yr) were recruited at time of colonoscopy, and anthropometric measures as well as blood were collected. Odds ratios were determined using polytomous logistic regression for polyp number (0 or ≥3) and polyp type (no polyp, hyperplastic polyp, tubular adenoma). RESULTS 41% of the men in our study were obese (BMI ≥30). The odds of an obese individual having ≥3 polyps was 6.5 (CI: 1.3-33.0) times greater than those of a lean (BMI<25) individual. Additionally, relative to lean individuals, obese individuals were 7.8 (CI: 2.0-30.8) times more likely to have a tubular adenoma than no polyp. As BMI category increased, participants were 2.9 (CI: 1.5-5.4) times more likely to have a tubular adenoma than no polyps. Serum leptin, IP-10 and TNF-α were significantly associated with tubular adenoma presence. Serum leptin and IP-10 were significantly associated with increased likelihood of ≥3 polyps, and TNF-α showed a trend (p = 0.09). CONCLUSIONS Obese men are more likely to have at least three polyps and adenomas. This cross-sectional study provides evidence that colonoscopy should be recommended for obese, white males.
Collapse
Affiliation(s)
- Sarah S. Comstock
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, Michigan, United States of America
| | - Kari Hortos
- College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan, United States of America
| | - Bruce Kovan
- College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan, United States of America
- Tri-County Gastroenterology, Professional Corporation, Clinton Township, Michigan, United States of America
| | - Sarah McCaskey
- College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan, United States of America
| | - Dorothy R. Pathak
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, United States of America
| | - Jenifer I. Fenton
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, Michigan, United States of America
- College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan, United States of America
| |
Collapse
|
47
|
The Continuum of Cancer Immunosurveillance: Prognostic, Predictive, and Mechanistic Signatures. Immunity 2013; 39:11-26. [DOI: 10.1016/j.immuni.2013.07.008] [Citation(s) in RCA: 600] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 07/09/2013] [Indexed: 11/21/2022]
|
48
|
Choi JW, Liu H, Shin DH, Yu GI, Hwang JS, Kim ES, Yun JW. Proteomic and cytokine plasma biomarkers for predicting progression from colorectal adenoma to carcinoma in human patients. Proteomics 2013; 13:2361-74. [PMID: 23606366 DOI: 10.1002/pmic.201200550] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 03/28/2013] [Accepted: 03/30/2013] [Indexed: 12/12/2022]
Abstract
In the present study, we screened proteomic and cytokine biomarkers between patients with adenomatous polyps and colorectal cancer (CRC) in order to improve our understanding of the molecular mechanisms behind turmorigenesis and tumor progression in CRC. To this end, we performed comparative proteomic analysis of plasma proteins using a combination of 2DE and MS as well as profiled differentially regulated cytokines and chemokines by multiplex bead analysis. Proteomic analysis identified 11 upregulated and 13 downregulated plasma proteins showing significantly different regulation patterns with diagnostic potential for predicting progression from adenoma to carcinoma. Some of these proteins have not previously been implicated in CRC, including upregulated leucine-rich α-2-glycoprotein, hemoglobin subunit β, Ig α-2 chain C region, and complement factor B as well as downregulated afamin, zinc-α-2-glycoprotein, vitronectin, and α-1-antichymotrypsin. In addition, plasma levels of three cytokines/chemokines, including interleukin-8, interferon gamma-induced protein 10, and tumor necrosis factor α, were remarkably elevated in patients with CRC compared to those with adenomatous polyps. Although further clinical validation is required, these proteins and cytokines can be established as novel biomarkers for CRC and/or its progression from colon adenoma.
Collapse
Affiliation(s)
- Jung-Won Choi
- Department of Biotechnology, Daegu University, Kyungsan, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
49
|
Murakami T, Kawada K, Iwamoto M, Akagami M, Hida K, Nakanishi Y, Kanda K, Kawada M, Seno H, Taketo MM, Sakai Y. The role of CXCR3 and CXCR4 in colorectal cancer metastasis. Int J Cancer 2013; 132:276-87. [PMID: 22689289 DOI: 10.1002/ijc.27670] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 05/31/2012] [Indexed: 12/12/2022]
Abstract
Chemokines and their receptors play key roles in leukocyte trafficking and are also implicated in cancer metastasis. We previously demonstrated that forced expression of CXCR3 promotes colon cancer metastasis preferentially to the draining lymph nodes (LNs), with poor prognosis. Using clinical colorectal cancer (CRC) samples, here, we show that expressions of CXCR3 and CXCR4 are significantly higher in metastatic foci within LNs and liver compared to primary tumors, whereas ligands for CXCR3 and CXCR4 are not. We also have demonstrated that some human CRC cell lines constitutively express both CXCR3 and CXCR4, and that activation of CXCR3 strengthens the CXCR4-mediated cell migration in vitro in a synergistic manner. By constructing SW620 cell lines with reduced expression of CXCR3 and/or CXCR4 using microRNA, we investigated in vivo metastatic activities in a mouse rectal transplantation model. Six weeks after inoculation, CXCR3-, CXCR4-, and CXCR3/CXCR4 double-knockdowns significantly reduced metastasis to LNs, liver and lungs, compared to the control (p < 0.05). Importantly, its suppressive effect on LN metastasis was significantly stronger in CXCR3- and CXCR3/CXCR4 double-knockdowns. In addition, CXCR3- and CXCR3/CXCR4 double-knockdowns significantly decreased the dissemination of cancer cells to liver and lungs, even after 2 weeks. These results indicate that targeting CXCR3 and CXCR4 can be a promising therapy against CRC metastasis.
Collapse
Affiliation(s)
- Teppei Murakami
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Chaturvedi P, Gilkes DM, Wong CCL, Luo W, Zhang H, Wei H, Takano N, Schito L, Levchenko A, Semenza GL. Hypoxia-inducible factor-dependent breast cancer-mesenchymal stem cell bidirectional signaling promotes metastasis. J Clin Invest 2012; 123:189-205. [PMID: 23318994 DOI: 10.1172/jci64993] [Citation(s) in RCA: 139] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 10/23/2012] [Indexed: 12/17/2022] Open
Abstract
Metastasis involves critical interactions between cancer and stromal cells. Intratumoral hypoxia promotes metastasis through activation of hypoxia-inducible factors (HIFs). We demonstrate that HIFs mediate paracrine signaling between breast cancer cells (BCCs) and mesenchymal stem cells (MSCs) to promote metastasis. In a mouse orthotopic implantation model, MSCs were recruited to primary breast tumors and promoted BCC metastasis to LNs and lungs in a HIF-dependent manner. Coculture of MSCs with BCCs augmented HIF activity in BCCs. Additionally, coculture induced expression of the chemokine CXCL10 in MSCs and the cognate receptor CXCR3 in BCCs, which was augmented by hypoxia. CXCR3 expression was blocked in cocultures treated with neutralizing antibody against CXCL10. Conversely, CXCL10 expression was blocked in MSCs cocultured with BCCs that did not express CXCR3 or HIFs. MSC coculture did not enhance the metastasis of HIF-deficient BCCs. BCCs and MSCs expressed placental growth factor (PGF) and its cognate receptor VEGFR1, respectively, in a HIF-dependent manner, and CXCL10 expression by MSCs was dependent on PGF expression by BCCs. PGF promoted metastasis of BCCs and also facilitated homing of MSCs to tumors. Thus, HIFs mediate complex and bidirectional paracrine signaling between BCCs and MSCs that stimulates breast cancer metastasis.
Collapse
Affiliation(s)
- Pallavi Chaturvedi
- Vascular Program, Johns Hopkins Institute for Cell Engineering, 733 North Broadway, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|