1
|
Ryan AT, Kim M, Lim K. Immune Cell Migration to Cancer. Cells 2024; 13:844. [PMID: 38786066 PMCID: PMC11120175 DOI: 10.3390/cells13100844] [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: 03/23/2024] [Revised: 04/27/2024] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
Immune cell migration is required for the development of an effective and robust immune response. This elegant process is regulated by both cellular and environmental factors, with variables such as immune cell state, anatomical location, and disease state that govern differences in migration patterns. In all cases, a major factor is the expression of cell surface receptors and their cognate ligands. Rapid adaptation to environmental conditions partly depends on intrinsic cellular immune factors that affect a cell's ability to adjust to new environment. In this review, we discuss both myeloid and lymphoid cells and outline key determinants that govern immune cell migration, including molecules required for immune cell adhesion, modes of migration, chemotaxis, and specific chemokine signaling. Furthermore, we summarize tumor-specific elements that contribute to immune cell trafficking to cancer, while also exploring microenvironment factors that can alter these cellular dynamics within the tumor in both a pro and antitumor fashion. Specifically, we highlight the importance of the secretome in these later aspects. This review considers a myriad of factors that impact immune cell trajectory in cancer. We aim to highlight the immunotherapeutic targets that can be harnessed to achieve controlled immune trafficking to and within tumors.
Collapse
Affiliation(s)
- Allison T. Ryan
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA; (A.T.R.); (M.K.)
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642, USA
| | - Minsoo Kim
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA; (A.T.R.); (M.K.)
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642, USA
| | - Kihong Lim
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA; (A.T.R.); (M.K.)
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642, USA
| |
Collapse
|
2
|
Yang SH, Tey ML, Zhou S, Nitar P, Mariyah H, Sim Y, Kusumawidjaja G, Chay WY, Yong WF, Wong RX. Correlation of Neutrophil-Lymphocyte and Albumin-Globulin Ratios With Outcomes in Patients With Breast Cancer Undergoing Neoadjuvant Chemotherapy or Upfront Surgery. J Breast Cancer 2024; 27:105-120. [PMID: 38529588 PMCID: PMC11065497 DOI: 10.4048/jbc.2023.0242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/27/2023] [Accepted: 01/27/2024] [Indexed: 03/27/2024] Open
Abstract
PURPOSE Higher neutrophil-lymphocyte ratio (NLRs) indicate a pro-inflammatory state and are associated with poor survival. Conversely, higher albumin-globulin ratio (AGRs) may be associated with improved prognosis. We aimed to investigate the association between NLR and AGR and prognosis and survival in patients with breast cancer. METHODS This retrospective study included all patients with stage I-III breast cancer between 2011 and 2017 in Singapore General Hospital and National Cancer Center Singapore. Multivariate logistic regression analysis of NLR, AGR, age, stage, grade, and subtype was performed. Survival data between groups were compared using Cox regression analysis and log-rank tests. RESULTS A total of 1,188 patients were included, of whom 323 received neoadjuvant chemotherapy (NACT) and 865 underwent upfront surgery. In patients who underwent NACT, a higher AGR was significantly associated with a higher pCR rate (cut-off > 1.28; odds ratio [OR], 2.03; 95% confidence interval [CI], 1.13-3.74; p = 0.020), better DFS (cut off > 1.55; hazard ratio [HR], 0.37; 95% CI, 0.16-0.85; p = 0.019), and better CSS (cut off > 1.46; HR, 0.39; 95% CI, 0.17-0.92; p = 0.031). Higher NLR was significantly associated with worse DFS (cut off > 4.09; HR, 1.77; 95% CI, 1.07-2.91; p = 0.026) and worse CSS (cut off > 4.09; HR, 1.98; 95% CI, 1.11-3.53; p = 0.021). In patients who underwent upfront surgery, higher AGR correlated with significantly better OS (cut off > 1.17; HR, 0.54; 95% CI, 0.36-0.82; p = 0.004) and higher NLR correlated with worse OS (cut off > 2.38; HR, 1.63; 95% CI, 1.09-2.44; p = 0.018). CONCLUSION NLR and AGR are useful in predicting the response to NACT as well as prognosis of patients with breast cancer. Further studies are needed to explore their value in clinical decision making.
Collapse
Affiliation(s)
- Shi Hui Yang
- Department of Breast Surgery, Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, Singapore, Singapore.
| | - Min Li Tey
- Department of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Siqin Zhou
- Clinical Trial officer, National Cancer Centre Singapore, Singapore, Singapore
| | - Phyu Nitar
- Cancer Informatics, National Cancer Centre Singapore, Singapore, Singapore
| | - Hanis Mariyah
- Cancer Informatics, National Cancer Centre Singapore, Singapore, Singapore
| | - Yirong Sim
- Department of Breast Surgery, Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Grace Kusumawidjaja
- Department of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Wen Yee Chay
- Department of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Wong Fuh Yong
- Department of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Ru Xin Wong
- Department of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore
| |
Collapse
|
3
|
Huang Z, Xiao Z, Yu L, Liu J, Yang Y, Ouyang W. Tumor-associated macrophages in non-small-cell lung cancer: From treatment resistance mechanisms to therapeutic targets. Crit Rev Oncol Hematol 2024; 196:104284. [PMID: 38311012 DOI: 10.1016/j.critrevonc.2024.104284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/20/2024] [Accepted: 01/31/2024] [Indexed: 02/06/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) remains one of the leading causes of cancer-related deaths worldwide. Different treatment approaches are typically employed based on the stage of NSCLC. Common clinical treatment methods include surgical resection, drug therapy, and radiation therapy. However, with the introduction and utilization of immune checkpoint inhibitors, cancer treatment has entered a new era, completely revolutionizing the treatment landscape for various cancers and significantly improving overall patient survival. Concurrently, treatment resistance often poses a critical challenge, with many patients experiencing disease progression following an initial response due to treatment resistance. Increasing evidence suggests that the tumor microenvironment (TME) plays a pivotal role in treatment resistance. Tumor-associated macrophages (TAMs) within the TME can promote treatment resistance in NSCLC by secreting various cytokines activating signaling pathways, and interacting with other immune cells. Therefore, this article will focus on elucidating the key mechanisms of TAMs in treatment resistance and analyze how targeting TAMs can reduce the levels of treatment resistance in NSCLC, providing a comprehensive understanding of the principles and approaches to overcome treatment resistance in NSCLC.
Collapse
Affiliation(s)
- Zhenjun Huang
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Ziqi Xiao
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Liqing Yu
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Jiayu Liu
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Yihan Yang
- Jiangxi Institute of Respiratory Disease, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang 330006, Jiangxi Province, China.
| | - Wenhao Ouyang
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| |
Collapse
|
4
|
Mirzakhani H, Handy DE, Lu Z, Oppenheimer B, Litonjua AA, Loscalzo J, Weiss ST. Integration of circulating microRNAs and transcriptome signatures identifies early-pregnancy biomarkers of preeclampsia. Clin Transl Med 2023; 13:e1446. [PMID: 37905457 PMCID: PMC10616748 DOI: 10.1002/ctm2.1446] [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: 07/24/2023] [Revised: 09/21/2023] [Accepted: 10/01/2023] [Indexed: 11/02/2023] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) have been implicated in the pathobiology of preeclampsia, a common hypertensive disorder of pregnancy. In a nested matched case-control cohort within the Vitamin D Antenatal Asthma Reduction Trial (VDAART), we previously identified peripheral blood mRNA signatures related to preeclampsia and vitamin D status (≤30 ng/mL) during gestation from 10 to 18 weeks, using differential expression analysis. METHODS Using quantitative PCR arrays, we conducted profiling of circulating miRNAs at 10-18 weeks of gestation in the same VDAART cohort to identify differentially expressed (DE) miRNAs associated with preeclampsia and vitamin D status. For the validation of the expression of circulating miRNA signatures in the placenta, the HTR-8/SVneo trophoblast cell line was used. Targets of circulating miRNA signatures in the preeclampsia mRNA signatures were identified by consensus ranking of miRNA-target prediction scores from four sources. The connected component of target signatures was identified by mapping to the protein-protein interaction (PPI) network and hub targets were determined. As experimental validation, we examined the gene and protein expression of IGF1R, one of the key hub genes, as a target of the DE miRNA, miR-182-5p, in response to a miR-182-5p mimic in HTR-8/SVneo cells. RESULTS Pregnant women with preeclampsia had 16 circulating DE miRNAs relative to normal pregnancy controls that were also DE under vitamin D insufficiency (9/16 = 56% upregulated, FDR < .05). Thirteen miRNAs (13/16 = 81.3%) were detected in HTR-8/SVneo cells. Overall, 16 DE miRNAs had 122 targets, of which 87 were unique. Network analysis demonstrated that the 32 targets of DE miRNA signatures created a connected subnetwork in the preeclampsia module with CXCL8, CXCL10, CD274, MMP9 and IGF1R having the highest connectivity and centrality degree. In an in vitro validation experiment, the introduction of an hsa-miR-182-5p mimic resulted in significant reduction of its target IGF1R gene and protein expression within HTR-8/SVneo cells. CONCLUSIONS The integration of the circulating DE miRNA and mRNA signatures associated preeclampsia added additional insights into the subclinical molecular signature of preeclampsia. Our systems and network biology approach revealed several biological pathways, including IGF-1, that may play a role in the early pathophysiology of preeclampsia. These pathways and signatures also denote potential biomarkers for the early stages of preeclampsia and suggest possible preventive measures.
Collapse
Affiliation(s)
- Hooman Mirzakhani
- Channing Division of Network MedicineDepartment of MedicineHarvard Medical SchoolBrigham and Women's HospitalBostonMassachusettsUSA
| | - Diane E. Handy
- Division of Cardiovascular MedicineDepartment of MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Zheng Lu
- Channing Division of Network MedicineDepartment of MedicineHarvard Medical SchoolBrigham and Women's HospitalBostonMassachusettsUSA
| | - Ben Oppenheimer
- Channing Division of Network MedicineDepartment of MedicineHarvard Medical SchoolBrigham and Women's HospitalBostonMassachusettsUSA
| | - Augusto A. Litonjua
- Division of Pediatric Pulmonary MedicineDepartment of PediatricsGolisano Children's Hospital at StrongUniversity of Rochester Medical CenterRochesterNew YorkUSA
| | - Joseph Loscalzo
- Division of Cardiovascular MedicineDepartment of MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Scott T. Weiss
- Channing Division of Network MedicineDepartment of MedicineHarvard Medical SchoolBrigham and Women's HospitalBostonMassachusettsUSA
| |
Collapse
|
5
|
Qiu J, Li M, Su C, Liang Y, Ou R, Chen X, Huang C, Zhang Y, Ye Y, Liao W, Zhang C. FOXS1 Promotes Tumor Progression by Upregulating CXCL8 in Colorectal Cancer. Front Oncol 2022; 12:894043. [PMID: 35898871 PMCID: PMC9309265 DOI: 10.3389/fonc.2022.894043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 06/03/2022] [Indexed: 11/26/2022] Open
Abstract
Background Forkhead box S1 (FOXS1) is a member of the forkhead box (FOX) transcriptional factor superfamily. The biological roles and underlying regulatory mechanism of FOXS1 in CRC remain unclear. Methods Bioinformatics analysis, Western blotting, real-time PCR, and immunohistochemistry (IHC) were used to detect the expression FOXS1 in CRC. MTT assay, transwell assay, human umbilical vein endothelial cell tube formation assay, and chicken chorioallantoic membrane assay were performed to investigate the effects of FOXS1 on proliferation, invasion, and angiogenesis. Additionally, tumor formation assay and orthotopic implantation assay were used to investigate the effects of FOXS1 on tumor growth and metastasis in vivo. Furthermore, gene set enrichment analysis (GSEA) was used to analyze the correlation between FOXS1 and EMT or angiogenesis. The correlation between FOXS1 and CXCL8 expression was analyzed in clinical CRC samples using IHC. Results The results showed that FOXS1 expression was upregulated in CRC tissues compared with adjacent normal intestine tissues. A high FOXS1 expression is positively correlated with poor survival. FOXS1 promoted the malignant behavior of CRC cancer cells in vitro, including proliferation, invasion, and angiogenesis. In addition, FOXS1 promoted tumor growth and metastasis in nude mice. Mechanistically, FOXS1 upregulated the expression of C–X–C motif chemokine ligand 8 (CXCL8) at the transcriptional level. Knockdown of CXCL8 blocked FOXS1 induced the enhancement of the EMT and angiogenesis. GSEAs in public CRC datasets revealed strong correlations between FOXS1 expression and EMT marker and angiogenesis markers. IHC showed that FOXS1 expression was positively correlated with CXCL8 expression and CD31 expression in clinical CRC samples. Conclusion The results suggest that FOXS1 promotes angiogenesis and metastasis by upregulating CXCL8 in CRC. Interference with the FOXS1/CXCL8 axis may serve as a potential therapeutic target for the treatment of metastatic CRC.
Collapse
Affiliation(s)
- Junfeng Qiu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Department of Pathology, Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Mingzhou Li
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Cailin Su
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Department of Pathology, Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Yihao Liang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ruizhang Ou
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Department of Pathology, Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Xiaoning Chen
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Department of Pathology, Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Chengmei Huang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Department of Pathology, Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Yaxin Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yaping Ye
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Department of Pathology, Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
- *Correspondence: Yaping Ye, ; Wenting Liao, ; Chao Zhang,
| | - Wenting Liao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Department of Pathology, Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- *Correspondence: Yaping Ye, ; Wenting Liao, ; Chao Zhang,
| | - Chao Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- *Correspondence: Yaping Ye, ; Wenting Liao, ; Chao Zhang,
| |
Collapse
|
6
|
Chemokines and NSCLC: Emerging role in prognosis, heterogeneity, and therapeutics. Semin Cancer Biol 2022; 86:233-246. [PMID: 35787939 DOI: 10.1016/j.semcancer.2022.06.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 06/15/2022] [Accepted: 06/24/2022] [Indexed: 12/11/2022]
Abstract
Lung cancer persists to contribute to one-quarter of cancer-associated deaths. Among the different histologies, non-small cell lung cancer (NSCLC) alone accounts for 85% of the cases. The development of therapies involving immune checkpoint inhibitors and angiogenesis inhibitors has increased patients' survival probability and reduced mortality rates. Developing targeted therapies against essential genetic alterations also translates to better treatment strategies. But the benefits still seem farfetched due to the development of drug resistance and refractory tumors. In this review, we have highlighted the interplay of different tumor microenvironment components, essentially discussing the chemokine families (CC, CXC, C, and CX3C) that regulate the tumor biology in NSCLC and promote tumor growth, metastasis, and associated heterogeneity. The development of therapeutics and prognostic markers is a complex and multipronged approach. However, some essential chemokines can act as critical players for being considered potential prognostic markers and therapeutic targets.
Collapse
|
7
|
Bridoux A, Mousa SA. Screening of a Library for Factor VIIa Inhibitors. LETT DRUG DES DISCOV 2022. [DOI: 10.2174/1570180818666211207125903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
As an alternative to the anticoagulant’s strategy using direct or indirect anti-Xa
drugs, considering other targets upstream in the coagulation cascade such as anti-Factor VIIa could represent
an effective and safer strategy in coagulation and pathological angiogenesis.
Objective:
The objective of the study was to assess a high technology methodology composed of virtual
screening, anticoagulant, and anti-angiogenesis assays to identify potent small-molecule FVIIa inhibitors.
Methods:
Chemical databanks were screened to select molecules bearing functional groups that could fit
into the active site of FVIIa, which were then tested. Ligands assigned with the lowest scores were retained
and then biologically assessed.
Results:
From the 500 molecules considered, 8 chemical structures revealed to be effective compounds in
vitro and to inhibit angiogenesis in the chick chorioallantoic membrane (CAM) model.
Conclusion:
New potent small-molecule FVIIa inhibitors have been identified; further biochemical and
chemical developments would be investigated directly from the selected scaffolds.
Collapse
Affiliation(s)
- Alexandre Bridoux
- Vascular Vision Pharmaceuticals, 5 University Place, Rensselaer, NY 12144, USA
- Pharmaceutical Research Institute,
One Discovery Drive, Rensselaer, NY 12144, USA
| | - Shaker A. Mousa
- Vascular Vision Pharmaceuticals, 5 University Place, Rensselaer, NY 12144, USA
- Pharmaceutical Research Institute,
One Discovery Drive, Rensselaer, NY 12144, USA
| |
Collapse
|
8
|
Murashka DI, Tahanovich AD, Kauhanka MM, Gotko OV, Prokhorova VI. On the issue of diagnostic value of determining the level of receptors and their ligands in blood in non-small cell lung cancer. Klin Lab Diagn 2022; 67:277-285. [PMID: 35613346 DOI: 10.51620/0869-2084-2022-67-5-277-285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Non-small cell lung cancer (NSCLC) occupies the first place in the structure of mortality due to oncological diseases. Late diagnosis worsens the effectiveness of its treatment. There are no informative biomarkers that allow us to judge the prevalence of the tumor process, especially in the early stages of NSCLC. To determine the level of CXCL5, CXCL8, CXCR1 and CXCR2 in the peripheral blood of patients with NSCLC to assess the possibility of their use in the diagnosis of the disease. The material was the blood of 218 patients with NSCLC, 19 patients with lung hamartoma and 42 healthy people. The concentration of CXCL5, CXCL8, and SCC in blood serum was determined by enzyme immunoassay, the CYFRA 21-1 level was determined by immunochemiluminescence analysis. The proportion of leukocytes equipped with CXCR1 and CXCR2 receptors and the fluorescence intensity of receptor complexes with antibodies (MFI) in them were measured by flow cytometry. MFI CXCR1 in granulocytes and the proportion of lymphocytes supplied CXCR2, increased in the blood already at stage I of NSCLC and showed an even more significant increase in subsequent stages. The level of these indicators was correlatively related to the stages and characteristics of NSCLC. Measuring the level of MFI CXCR1 in the blood serum makes it possible to diagnose the early stages of NSCLC with a sensitivity of 87.4% (specificity - 73.8%). Determination of the proportion of lymphocytes equipped with CXCR2 demonstrates comparable diagnostic sensitivity (87.2%) and specificity of 66.7% in the detection of stages I-II of NSCLC. MFI CXCR1 in granulocytes can also be used to differentiate stages I and II of NSCLC (diagnostic sensitivity - 75,3%, specificity - 69,6%). The sensitivity of determining for this purpose the proportion of lymphocytes equipped with CXCR2 is 75.0% with a specificity of 71.7%. In 89.7% of patients with stages III-IV NSCLC, the MFI CXCR1 in granulocytes exceeds the threshold value of 47.8 (specificity - 74.8%). Diagnostic sensitivity of determining the proportion of lymphocytes for this purpose was 90.7%.
Collapse
Affiliation(s)
- D I Murashka
- Belarusian stage medical university, Belarusian Ministry of Health
| | - A D Tahanovich
- Belarusian stage medical university, Belarusian Ministry of Health
| | - M M Kauhanka
- Belarusian stage medical university, Belarusian Ministry of Health
| | - O V Gotko
- N.N. Alexandrov Republican Scientific-Practical Centre of Oncology and Medical Radiology, Belarusian Ministry of Health
| | - V I Prokhorova
- N.N. Alexandrov Republican Scientific-Practical Centre of Oncology and Medical Radiology, Belarusian Ministry of Health
| |
Collapse
|
9
|
Taucher E, Mykoliuk I, Lindenmann J, Smolle-Juettner FM. Implications of the Immune Landscape in COPD and Lung Cancer: Smoking Versus Other Causes. Front Immunol 2022; 13:846605. [PMID: 35386685 PMCID: PMC8978964 DOI: 10.3389/fimmu.2022.846605] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/28/2022] [Indexed: 12/30/2022] Open
Abstract
Cigarette smoking is reported in about one third of adults worldwide. A strong relationship between cigarette smoke exposure and chronic obstructive pulmonary disease (COPD) as well as lung cancer has been proven. However, about 15% of lung cancer cases, and between one fourth and one third of COPD cases, occur in never-smokers. The effects of cigarette smoke on the innate as well as the adaptive immune system have been widely investigated. It is assumed that certain immunologic features contribute to lung cancer and COPD development in the absence of smoking as the major risk factor. In this article, we review different immunological aspects of lung cancer and COPD with a special focus on non-smoking related risk factors.
Collapse
Affiliation(s)
- Elisabeth Taucher
- Division of Pulmonology, Department of Internal Medicine, Medical University Graz, Graz, Austria
| | - Iurii Mykoliuk
- Division of Thoracic Surgery, Department of Surgery, Medical University Graz, Graz, Austria
| | - Joerg Lindenmann
- Division of Thoracic Surgery, Department of Surgery, Medical University Graz, Graz, Austria
| | | |
Collapse
|
10
|
Wu Y, Gan D, Leng X, He W, Zhang X, Li C, Gu X, Hu Y, Du S, Han Y. Anti-ageing and Anti-lung Carcinoma Effects of Vulpinic Acid and Usnic Acid Compounds and Biological Investigations with Molecular Modeling Study. J Oleo Sci 2022; 71:247-255. [PMID: 35110467 DOI: 10.5650/jos.ess21276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Disorganization and breakdown of extracellular matrix proteins like fibronectin, collagen, and elastin are key characteristics of skin aging due to the increased activation of important proteolytic enzymes like elastases and collagenase enzymes. Also, inhibition of their enzymatic activities by natural molecules might be a promising factor to prevent extrinsic skin aging. All chemicals were obtained from Sigma-Aldrich unless otherwise stated. The assay employed was based on spectrophotometric methods reported in the literature. The collagenase and elastase inhibition assays of some phenolic compounds were performed according to the previous studies. These compounds showed excellent to good inhibitory activities of vulpinic acid against studied these enzymes with IC50 values of 195.36 µM for collagenase and 25.24 µM for elastase. The molecular docking calculations were conducted to investigate the chemical and biological activity of vulpinic acid and usnic acid against collagenase and elastase. The results indicated that these two compounds can interact with the essential residues of the enzymes and affect their activities. The calculations of binding free energies were also performed to obtain more details about the characteristics and free energies of the ligand-enzyme complexes. Additionally, both compounds exhibited the most potent inhibition in the three lung cancer cells, with an IC50 value of 21-68 µM, indicating that vulpinic acid is more potent than Doxorubicin, which exhibited an IC50 value of 21-29 µM.
Collapse
Affiliation(s)
- Yunhua Wu
- Southwest Medical University.,Thoracic surgery, Chendu Wenjiang District People's Hospital
| | - Dongmei Gan
- Thoracic surgery, Chendu Wenjiang District People's Hospital
| | | | - Wenwu He
- Thoracic surgery, Sichuan Cancer Hospital
| | - Xiuqiong Zhang
- Thoracic surgery, Chendu Wenjiang District People's Hospital
| | - Chong Li
- Thoracic surgery, Chendu Wenjiang District People's Hospital
| | - Xiaobo Gu
- Thoracic surgery, Chendu Wenjiang District People's Hospital
| | - Ying Hu
- Thoracic surgery, Chendu Wenjiang District People's Hospital
| | - Shijian Du
- Thoracic surgery, Chendu Wenjiang District People's Hospital
| | - Yongtao Han
- Southwest Medical University.,Thoracic surgery, Sichuan Cancer Hospital
| |
Collapse
|
11
|
Koltai T, Fliegel L. Role of Silymarin in Cancer Treatment: Facts, Hypotheses, and Questions. J Evid Based Integr Med 2022; 27:2515690X211068826. [PMID: 35018864 PMCID: PMC8814827 DOI: 10.1177/2515690x211068826] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/20/2021] [Accepted: 12/06/2021] [Indexed: 12/14/2022] Open
Abstract
The flavonoid silymarin extracted from the seeds of Sylibum marianum is a mixture of 6 flavolignan isomers. The 3 more important isomers are silybin (or silibinin), silydianin, and silychristin. Silybin is functionally the most active of these compounds. This group of flavonoids has been extensively studied and they have been used as hepato-protective substances for the mushroom Amanita phalloides intoxication and mainly chronic liver diseases such as alcoholic cirrhosis and nonalcoholic fatty liver. Hepatitis C progression is not, or slightly, modified by silymarin. Recently, it has also been proposed for SARS COVID-19 infection therapy. The biochemical and molecular mechanisms of action of these substances in cancer are subjects of ongoing research. Paradoxically, many of its identified actions such as antioxidant, promoter of ribosomal synthesis, and mitochondrial membrane stabilization, may seem protumoral at first sight, however, silymarin compounds have clear anticancer effects. Some of them are: decreasing migration through multiple targeting, decreasing hypoxia inducible factor-1α expression, inducing apoptosis in some malignant cells, and inhibiting promitotic signaling among others. Interestingly, the antitumoral activity of silymarin compounds is limited to malignant cells while the nonmalignant cells seem not to be affected. Furthermore, there is a long history of silymarin use in human diseases without toxicity after prolonged administration. The ample distribution and easy accessibility to milk thistle-the source of silymarin compounds, its over the counter availability, the fact that it is a weed, some controversial issues regarding bioavailability, and being a nutraceutical rather than a drug, has somehow led medical professionals to view its anticancer effects with skepticism. This is a fundamental reason why it never achieved bedside status in cancer treatment. However, in spite of all the antitumoral effects, silymarin actually has dual effects and in some cases such as pancreatic cancer it can promote stemness. This review deals with recent investigations to elucidate the molecular actions of this flavonoid in cancer, and to consider the possibility of repurposing it. Particular attention is dedicated to silymarin's dual role in cancer and to some controversies of its real effectiveness.
Collapse
Affiliation(s)
- Tomas Koltai
- Hospital del Centro Gallego de Buenos Aires, Buenos Aires, Argentina
| | | |
Collapse
|
12
|
Qi D, Liu Y, Li J, Huang JH, Hu X, Wu E. Salinomycin as a potent anticancer stem cell agent: State of the art and future directions. Med Res Rev 2021; 42:1037-1063. [PMID: 34786735 PMCID: PMC9298915 DOI: 10.1002/med.21870] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 10/22/2021] [Accepted: 10/26/2021] [Indexed: 12/11/2022]
Abstract
Cancer stem cells (CSCs) are a small subpopulation of cells within a tumor that can both self‐renew and differentiate into other cell types forming the heterogeneous tumor bulk. Since CSCs are involved in all aspects of cancer development, including tumor initiation, cell proliferation, metastatic dissemination, therapy resistance, and recurrence, they have emerged as attractive targets for cancer treatment and management. Salinomycin, a widely used antibiotic in poultry farming, was identified by the Weinberg group as a potent anti‐CSC agent in 2009. As a polyether ionophore, salinomycin exerts broad‐spectrum activities, including the important anti‐CSC function. Studies on the mechanism of action of salinomycin against cancer have been continuously and rapidly published since then. Thus, it is imperative for us to update its literature of recent research findings in this area. We here summarize the notable work reported on salinomycin's anticancer activities, intracellular binding target(s), effects on tumor microenvironment, safety, derivatives, and tumor‐specific drug delivery; after that we also discuss the translational potential of salinomycin toward clinical application based on current multifaceted understandings.
Collapse
Affiliation(s)
- Dan Qi
- Department of Neurosurgery, Baylor Scott & White Health, Temple, Texas, USA.,Neuroscience Institute, Baylor Scott & White Health, Temple, Texas, USA
| | - Yunyi Liu
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Molecular Engineering for Theranostics, Hunan University, Changsha, China
| | - Juan Li
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Molecular Engineering for Theranostics, Hunan University, Changsha, China
| | - Jason H Huang
- Department of Neurosurgery, Baylor Scott & White Health, Temple, Texas, USA.,Neuroscience Institute, Baylor Scott & White Health, Temple, Texas, USA.,Department of Surgery, Texas A&M University College of Medicine, Temple, Texas, USA
| | - Xiaoxiao Hu
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Molecular Engineering for Theranostics, Hunan University, Changsha, China.,Shenzhen Research Institute, Hunan University, Shenzhen, Guangdong, China
| | - Erxi Wu
- Department of Neurosurgery, Baylor Scott & White Health, Temple, Texas, USA.,Neuroscience Institute, Baylor Scott & White Health, Temple, Texas, USA.,Department of Surgery, Texas A&M University College of Medicine, Temple, Texas, USA.,LIVESTRONG Cancer Institutes and Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA.,Department of Pharmaceutical Sciences, Texas A&M University College of Pharmacy, College Station, Texas, USA
| |
Collapse
|
13
|
Zhang N, Tan Q, Tao D, Song Y, Song W, Wang J, Ma L, Wu D, Feng Y, Yao J, Han X, Shi Y. Cytokines screening identifies MIG (CXCL9) for postoperative recurrence prediction in operated non-small cell lung cancer patients. Cytokine 2021; 149:155759. [PMID: 34775109 DOI: 10.1016/j.cyto.2021.155759] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 09/30/2021] [Accepted: 11/01/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Exploration of reliable biomarkers most likely to identify non-small cell lung cancer (NSCLC) patients at high risk for recurrence after surgery is needed. METHODS Quantibody® Human Cytokine Antibody 6000 was used as screening tool to measure serum levels of 280 cytokines in ten healthy individuals and nine samples from three NSCLC patients before operation, after operation and postoperative recurrence. Selected cytokines were validated in two independent sets (89 patients before surgery, 69 patients after surgery and 40 patients with postoperative recurrence for each set) using ELISA method. The association of the selected cytokine with clinicopathologic features was also evaluated. RESULTS Thirty-six cytokines were declined after surgery and again elevated when recurrence. We selected MIG to be further assessed in 2 validation sets, the mean value of serum MIG levels in 396 NSCLC patients was 253.42 ± 274.48 pg/mL and was significantly higher than the level in 60 healthy controls (47.65 ± 33.23 pg/mL, P < 0.0001). The serum MIG levels were 366.36 ± 324.04 pg/mL pre-operation, 134.04 ± 127.52 pg/mL post-operation and 208.05 ± 239.39 pg/mL in recurrence in NSCLC patients. The serum MIG levels were significant differences among NSCLC patients of pre-operation, post-operation and recurrence and controls (P < 0.0001). Moreover, Serum MIG levels were decreased markedly after operation and notably increased when disease relapsed (P < 0.0005). Serum MIG levels trend to be higher in patients with male gender, older age, smoking habit, poor tumor differentiation, and non-adenocarcinoma histology. CONCLUSIONS These data indicated that MIG might be an indicator of postoperative recurrence and help to identify NSCLC patient who was easy to relapse after surgery.
Collapse
Affiliation(s)
- Ningning Zhang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Qiaoyun Tan
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Dan Tao
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Yuanyuan Song
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Wenya Song
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Jianfei Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Li Ma
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Di Wu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Yun Feng
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Jiarui Yao
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Xiaohong Han
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China; Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 41 Damucang Hutong, Xicheng District, Beijing 100032, China.
| | - Yuankai Shi
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China.
| |
Collapse
|
14
|
Li X, Shi H, Zhang W, Bai C, He M, Ta N, Huang H, Ning Y, Fang C, Qin H, Dong Y. Immunotherapy and Targeting the Tumor Microenvironment: Current Place and New Insights in Primary Pulmonary NUT Carcinoma. Front Oncol 2021; 11:690115. [PMID: 34660264 PMCID: PMC8515126 DOI: 10.3389/fonc.2021.690115] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 09/13/2021] [Indexed: 12/12/2022] Open
Abstract
Primary pulmonary nuclear protein of testis carcinoma is a rare and highly aggressive malignant tumor. It accounts for approximately 0.22% of primary thoracic tumors and is little known, so it is often misdiagnosed as pulmonary squamous cell carcinoma. No effective treatment has been formed yet, and the prognosis is extremely poor. This review aims to summarize the etiology, pathogenesis, diagnosis, treatment, and prognosis of primary pulmonary nuclear protein of testis carcinoma in order to better recognize it and discuss the current and innovative strategies to overcome it. With the increasing importance of cancer immunotherapy and tumor microenvironment, the review also discusses whether immunotherapy and targeting the tumor microenvironment can improve the prognosis of primary pulmonary nuclear protein of testis carcinoma and possible treatment strategies. We reviewed and summarized the clinicopathological features of all patients with primary pulmonary nuclear protein of testis carcinoma who received immunotherapy, including initial misdiagnosis, disease stage, immunohistochemical markers related to tumor neovascularization, and biomarkers related to immunotherapy, such as PD-L1 (programmed death-ligand 1) and TMB (tumor mutational burden). In the meanwhile, we summarized and analyzed the progression-free survival (PFS) and the overall survival (OS) of patients with primary pulmonary nuclear protein of testis carcinoma treated with PD-1 (programmed cell death protein 1)/PD-L1 inhibitors and explored potential population that may benefit from immunotherapy. To the best of our knowledge, this is the first review on the exploration of the tumor microenvironment and immunotherapy effectiveness in primary pulmonary nuclear protein of testis carcinoma.
Collapse
Affiliation(s)
- Xiang Li
- Department of Respiratory and Critical Care Medicine, Changhai Hospital (The First Affiliated Hospital of Naval Medical University), Naval Medical University (Second Military Medical University), Shanghai, China
| | - Hui Shi
- Department of Respiratory and Critical Care Medicine, Changhai Hospital (The First Affiliated Hospital of Naval Medical University), Naval Medical University (Second Military Medical University), Shanghai, China
| | - Wei Zhang
- Department of Respiratory and Critical Care Medicine, Changhai Hospital (The First Affiliated Hospital of Naval Medical University), Naval Medical University (Second Military Medical University), Shanghai, China
| | - Chong Bai
- Department of Respiratory and Critical Care Medicine, Changhai Hospital (The First Affiliated Hospital of Naval Medical University), Naval Medical University (Second Military Medical University), Shanghai, China
| | - Miaoxia He
- Department of Pathology, Changhai Hospital (The First Affiliated Hospital of Naval Medical University), Naval Medical University (Second Military Medical University), Shanghai, China
| | - Na Ta
- Department of Pathology, Changhai Hospital (The First Affiliated Hospital of Naval Medical University), Naval Medical University (Second Military Medical University), Shanghai, China
| | - Haidong Huang
- Department of Respiratory and Critical Care Medicine, Changhai Hospital (The First Affiliated Hospital of Naval Medical University), Naval Medical University (Second Military Medical University), Shanghai, China
| | - Yunye Ning
- Department of Respiratory and Critical Care Medicine, Changhai Hospital (The First Affiliated Hospital of Naval Medical University), Naval Medical University (Second Military Medical University), Shanghai, China
| | - Chen Fang
- Department of Respiratory and Critical Care Medicine, Changhai Hospital (The First Affiliated Hospital of Naval Medical University), Naval Medical University (Second Military Medical University), Shanghai, China
| | - Hao Qin
- Department of Respiratory and Critical Care Medicine, Changhai Hospital (The First Affiliated Hospital of Naval Medical University), Naval Medical University (Second Military Medical University), Shanghai, China
| | - Yuchao Dong
- Department of Respiratory and Critical Care Medicine, Changhai Hospital (The First Affiliated Hospital of Naval Medical University), Naval Medical University (Second Military Medical University), Shanghai, China
| |
Collapse
|
15
|
Saxena S, Singh RK. Chemokines orchestrate tumor cells and the microenvironment to achieve metastatic heterogeneity. Cancer Metastasis Rev 2021; 40:447-476. [PMID: 33959849 PMCID: PMC9863248 DOI: 10.1007/s10555-021-09970-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/22/2021] [Indexed: 01/26/2023]
Abstract
Chemokines, a subfamily of the cell cytokines, are low molecular weight proteins known to induce chemotaxis in leukocytes in response to inflammatory and pathogenic signals. A plethora of literature demonstrates that chemokines and their receptors regulate tumor progression and metastasis. With these diverse functionalities, chemokines act as a fundamental link between the tumor cells and their microenvironment. Recent studies demonstrate that the biology of chemokines and their receptor in metastasis is complex as numerous chemokines are involved in regulating site-specific tumor growth and metastasis. Successful treatment of disseminated cancer is a significant challenge. The most crucial problem for treating metastatic cancer is developing therapy regimes capable of overcoming heterogeneity problems within primary tumors and among metastases and within metastases (intralesional). This heterogeneity of malignant tumor cells can be related to metastatic potential, response to chemotherapy or specific immunotherapy, and many other factors. In this review, we have emphasized the role of chemokines in the process of metastasis and metastatic heterogeneity. Individual chemokines may not express the full potential to address metastatic heterogeneity, but chemokine networks need exploration. Understanding the interplay between chemokine-chemokine receptor networks between the tumor cells and their microenvironment is a novel approach to overcome the problem of metastatic heterogeneity. Recent advances in the understanding of chemokine networks pave the way for developing a potential targeted therapeutic strategy to treat metastatic cancer.
Collapse
Affiliation(s)
- Sugandha Saxena
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198-5900, USA
| | - Rakesh K Singh
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198-5900, USA.
| |
Collapse
|
16
|
Chantharakhit C, Sujaritvanichpong N. Prognostic Impact of the Advanced Lung Cancer Inflammation Index (ALI) in Metastatic Non-Small Cell Lung Cancer Treated with First Line Chemotherapy. Asian Pac J Cancer Prev 2021; 22:1149-1156. [PMID: 33906307 PMCID: PMC8325112 DOI: 10.31557/apjcp.2021.22.4.1149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Indexed: 12/14/2022] Open
Abstract
Background: The advanced lung cancer inflammation index (ALI) has been reported to predict the overall survival in patients with advanced non-small cell lung cancer (NSCLC). However, no previous studies have examined the prognostic significance of ALI in metastatic NSCLC treated with first line chemotherapy. The objective of this study was to explore the relationship between ALI and the prognosis of metastatic NSCLC treated with first line chemotherapy. Materials and Methods: Data of 109 metastatic NSCLC patients who had completed first line treatment with chemotherapy was collected. A multivariate flexible parametric proportional-hazards model with restricted cubic splines (RCS) was used to explore and identify the independent prognostic factors, including clinical potential factors and ALI for the overall survival. Multivariate regression analysis was used to evaluate the potential prognostic factors associated with short survival less than 6 months. The analysis of the restricted mean survival time (RMST) method was used to estimate the event-free time from zero to 18 months. Results: The median OS was 10.9 months (95%CI 9.57-13.18) and median PFS was 7.5 months (95%CI 6.85-8.00).The multivariate survival analyses revealed two prognostic factors for worse survival: Poor ECOG PS (HR46.90; 95%CI 2.90-758.73; p=0.007) and progressive disease after completing the first line chemotherapy treatment (HR 2.85; 95%CI1.18-6.88; p=0.02),whereas a low ALI <11 referred to a non-significant prognostic factor (HR 1.42; 95%CI 0.67-3.01; p=0.364).The results of the multivariate regression analysis revealed that the low ALI and progressive disease status were significantly associated with the short survival outcome (OR 5.12; 95%CI 1.11-23.65; p=0.037; OR 12.57; 95%CI 3.00-52.73; p=0.001). Conclusions: A low ALI was associated with the short survival in metastatic NSCLC treated with chemotherapy. However, using ALI as a prognostic factor only was still too limited. Other considerable clinical prognostic factors should also be used simultaneously, which would have strong significant prognostic impacts.
Collapse
Affiliation(s)
- Chaichana Chantharakhit
- Division of Medical Oncology, Department of Internal Medicine, Buddhasothorn Hospital, Chachoengsao, Thailand
| | - Nantapa Sujaritvanichpong
- Division of Medical Oncology, Department of Internal Medicine, Buddhasothorn Hospital, Chachoengsao, Thailand
| |
Collapse
|
17
|
Gaissmaier L, Christopoulos P. Immune Modulation in Lung Cancer: Current Concepts and Future Strategies. Respiration 2020; 99:1-27. [PMID: 33291116 DOI: 10.1159/000510385] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 07/10/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer immunotherapy represents the most dynamic field of biomedical research currently, with thoracic immuno-oncology as a forerunner. PD-(L)1 inhibitors are already part of standard first-line treatment for both non-small-cell and small-cell lung cancer, while unprecedented 5-year survival rates of 15-25% have been achieved in pretreated patients with metastatic disease. Evolving strategies are mainly aiming for improvement of T-cell function, increase of immune activation in the tumor microenvironment (TME), and supply of tumor-reactive lymphocytes. Several novel therapeutics have demonstrated preclinical efficacy and are increasingly used in rational combinations within clinical trials. Two overarching trends dominate: extension of immunotherapy to earlier disease stages, mainly as neoadjuvant treatment, and a shift of focus towards multivalent, individualized, mutatome-based antigen-specific modalities, mainly adoptive cell therapies and cancer vaccines. The former ensures ample availability of treated and untreated patient samples, the latter facilitates deeper mechanistic insights, and both in combination build an overwhelming force that is accelerating progress and driving the greatest revolution cancer medicine has seen so far. Today, immune modulation represents the most potent therapeutic modality in oncology, the most important topic in clinical and translational cancer research, and arguably our greatest, meanwhile justified hope for achieving cure of pulmonary neoplasms and other malignancies in the next future.
Collapse
Affiliation(s)
- Lena Gaissmaier
- Department of Thoracic Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC-H), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Petros Christopoulos
- Department of Thoracic Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany,
- Translational Lung Research Center Heidelberg (TLRC-H), German Center for Lung Research (DZL), Heidelberg, Germany,
| |
Collapse
|
18
|
Jiang R, Wang T, Zhou F, Yao Y, He J, Xu D. Bioinformatics-based identification of miRNA-, lncRNA-, and mRNA-associated ceRNA networks and potential biomarkers for preeclampsia. Medicine (Baltimore) 2020; 99:e22985. [PMID: 33157942 PMCID: PMC7647555 DOI: 10.1097/md.0000000000022985] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/18/2020] [Accepted: 10/01/2020] [Indexed: 11/29/2022] Open
Abstract
This study aimed to identify significantly altered long non-coding RNAs (lncRNAs), microRNAs (miRNAs), mRNAs, pathways in preeclampsia (PE), and to investigate their targeted relationships and biological functions.GSE96985 from Gene Expression Omnibus database was extracted, involving 3 PE and 4 normal tissues. After the differential expression analysis of miRNAs, lncRNAs, and mRNAs using the limma package, protein-protein interaction (PPI) network and module analyses were performed for differentially expressed mRNAs (dif-mRNAs). Combined with the miRanda and miRWalk tools, a regulatory relationship between dif-miRNAs and dif-mRNAs/lncRNAs (dif-mRNAs/dif-lncRNAs) was predicted. Finally, mRNA-miRNA-lncRNA regulatory network construction was performed using Cytoscape software.A total of 511 dif-mRNAs were screened in PE. The top 5 nodes in the PPI networks included up-regulated complement component 3 (C3), C-X-C motif chemokine ligand 8 (CXCL8), and fibronectin 1 (FN1). Three significant network modules were identified for dif-mRNAs. C3 and CXCL8 were identified in module A, and FN1 was identified in module C. A disintegrin and metalloproteinase with thrombospondin motifs 6 (ADAMTS6) was down-regulated by the miR-210-3p. Therefore, lnc-CTD-2383M3.1 functions as a competing endogenous RNA in ADAMTS6 expression regulation by competitively binding to miR-210-3p during the regulation process of PE.C3, CXCL8, FN1, and ADAMTS6 might be involved in the development of PE. The lnc-CTD-2383M3.1-miR-210-3p-ADAMTS6 axis might be a potential regulatory mechanism in PE.
Collapse
|
19
|
Kikuchi H, Maishi N, Annan DA, Alam MT, Dawood RIH, Sato M, Morimoto M, Takeda R, Ishizuka K, Matsumoto R, Akino T, Tsuchiya K, Abe T, Osawa T, Miyajima N, Maruyama S, Harabayashi T, Azuma M, Yamashiro K, Ameda K, Kashiwagi A, Matsuno Y, Hida Y, Shinohara N, Hida K. Chemotherapy-Induced IL8 Upregulates MDR1/ABCB1 in Tumor Blood Vessels and Results in Unfavorable Outcome. Cancer Res 2020; 80:2996-3008. [PMID: 32536602 DOI: 10.1158/0008-5472.can-19-3791] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 04/10/2020] [Accepted: 05/29/2020] [Indexed: 11/16/2022]
Abstract
Tumor endothelial cells (TEC) lining tumor blood vessels actively contribute to tumor progression and metastasis. In addition to tumor cells, TEC may develop drug resistance during cancer treatment, allowing the tumor cells to survive chemotherapy and metastasize. We previously reported that TECs resist paclitaxel treatment via upregulation of ABCB1. However, whether TEC phenotypes are altered by anticancer drugs remains to be clarified. Here, we show that ABCB1 expression increases after chemotherapy in urothelial carcinoma cases. The ratio of ABCB1-positive TEC before and after first-line chemotherapy in urothelial carcinoma tissues (n = 66) was analyzed by ABCB1 and CD31 immunostaining. In 42 cases (64%), this ratio increased after first-line chemotherapy. Chemotherapy elevated ABCB1 expression in endothelial cells by increasing tumor IL8 secretion. In clinical cases, ABCB1 expression in TEC correlated with IL8 expression in tumor cells after first-line chemotherapy, leading to poor prognosis. In vivo, the ABCB1 inhibitor combined with paclitaxel reduced tumor growth and metastasis compared with paclitaxel alone. Chemotherapy is suggested to cause inflammatory changes in tumors, inducing ABCB1 expression in TEC and conferring drug resistance. Overall, these findings indicate that TEC can survive during chemotherapy and provide a gateway for cancer metastasis. Targeting ABCB1 in TEC represents a novel strategy to overcome cancer drug resistance. SIGNIFICANCE: These findings show that inhibition of ABCB1 in tumor endothelial cells may improve clinical outcome, where ABCB1 expression contributes to drug resistance and metastasis following first-line chemotherapy.
Collapse
Affiliation(s)
- Hiroshi Kikuchi
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido, Japan.,Department of Renal and Genitourinary Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Nako Maishi
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido, Japan.,Department of Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Hokkaido, Japan
| | - Dorcas A Annan
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido, Japan.,Department of Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Hokkaido, Japan
| | - Mohammad Towfik Alam
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Hokkaido, Japan.,Department of Dental Radiology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Hokkaido, Japan
| | - Randa Ibrahim Hassan Dawood
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Masumi Sato
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Masahiro Morimoto
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido, Japan.,Department of Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Hokkaido, Japan
| | - Ryo Takeda
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Keita Ishizuka
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Ryuji Matsumoto
- Department of Renal and Genitourinary Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan.,Department of Urology, Teine Keijinkai Hospital, Sapporo, Hokkaido, Japan
| | - Tomoshige Akino
- Department of Urology, Sapporo City General Hospital, Sapporo, Hokkaido, Japan.,Department of Urology, Tonan Hospital, Sapporo, Hokkaido, Japan
| | - Kunihiko Tsuchiya
- Department of Renal and Genitourinary Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan.,Department of Urology, KKR Sapporo Medical Center, Sapporo, Hokkaido, Japan
| | - Takashige Abe
- Department of Renal and Genitourinary Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Takahiro Osawa
- Department of Renal and Genitourinary Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Naoto Miyajima
- Department of Renal and Genitourinary Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan.,Department of Urology, Teine Keijinkai Hospital, Sapporo, Hokkaido, Japan
| | - Satoru Maruyama
- Department of Renal and Genitourinary Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan.,Department of Urology, Hokkaido Cancer Center, Sapporo, Hokkaido, Japan
| | - Toru Harabayashi
- Department of Urology, Hokkaido Cancer Center, Sapporo, Hokkaido, Japan
| | - Manabu Azuma
- Department of Pathology, Hokkaido Cancer Center, Sapporo, Hokkaido, Japan
| | | | - Kaname Ameda
- Hokkaido Hinyokika Kinen Hospital, Sapporo, Hokkaido, Japan
| | - Akira Kashiwagi
- Department of Urology, Teine Keijinkai Hospital, Sapporo, Hokkaido, Japan
| | - Yoshihiro Matsuno
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Yasuhiro Hida
- Department of Cardiovascular and Thoracic Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Nobuo Shinohara
- Department of Renal and Genitourinary Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Kyoko Hida
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido, Japan. .,Department of Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Hokkaido, Japan
| |
Collapse
|
20
|
Palmer LD, Jordan AT, Maloney KN, Farrow MA, Gutierrez DB, Gant-Branum R, Burns WJ, Romer CE, Tsui T, Allen JL, Beavers WN, Nei YW, Sherrod SD, Lacy DB, Norris JL, McLean JA, Caprioli RM, Skaar EP. Zinc intoxication induces ferroptosis in A549 human lung cells. Metallomics 2020; 11:982-993. [PMID: 30968088 DOI: 10.1039/c8mt00360b] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Zinc (Zn) is an essential trace metal required for all forms of life, but is toxic at high concentrations. While the toxic effects of high levels of Zn are well documented, the mechanism of cell death appears to vary based on the study and concentration of Zn. Zn has been proposed as an anti-cancer treatment against non-small cell lung cancer (NSCLC). The goal of this analysis was to determine the effects of Zn on metabolism and cell death in A549 cells. Here, high throughput multi-omics analysis identified the molecular effects of Zn intoxication on the proteome, metabolome, and transcriptome of A549 human NSCLC cells after 5 min to 24 h of Zn exposure. Multi-omics analysis combined with additional experimental evidence suggests Zn intoxication induces ferroptosis, an iron and lipid peroxidation-dependent programmed cell death, demonstrating the utility of multi-omics analysis to identify cellular response to intoxicants.
Collapse
Affiliation(s)
- Lauren D Palmer
- Vanderbilt Institute for Infection, Immunology and Inflammation and Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Kohsari M, Khadem-Ansari MH, Rasmi Y, Sayyadi H. Serum Levels of Interleukin-8 and Soluble Interleukin-6 Receptor in Patients with Stage-I Multiple Myeloma: A Case-Control Study. Asian Pac J Cancer Prev 2020; 21:127-132. [PMID: 31983174 PMCID: PMC7294003 DOI: 10.31557/apjcp.2020.21.1.127] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Indexed: 11/25/2022] Open
Abstract
Objective: Multiple myeloma (MM) remains an incurable disease that needs better recognition and further research. Previous studies elucidated the interaction between myeloma cells and showed the necessity of bone marrow stromal cells for the initiation and progression of MM. Many chemokines and their receptors including interleukin-8 (IL-8) and soluble interleukin-6 receptor (sIL-6R) play important roles in this interaction. The main purpose of this study is evaluating the serum level of IL-8 and sIL-6R on stage-I of MM patients and healthy controls. Methods: Serum samples from 30 stage-I MM patients (13 males and 17 females) and 30 healthy subjects as controls (13 males and 17 females) were examined in this study. The protein concentrations of serum IL-8 and sIL-6R were assessed by enzyme-linked immunosorbent assay (ELISA). Results: The mean level of IL-8 and sIL-6R were significantly elevated in stage-I MM. The mean levels of IL-8 were 1246.57±279.22 ng/ml in stage-I MM and 902.53± 294.61 ng/ml in controls (P<0.001). The mean levels of sIL-6R were 5.39±1.38 ng/ml and 4.1±1.14 ng/ml in stage-I MM and controls, respectively (P<0.001). The mean levels of IL-8 were 1342.18±193.4 ng/ml in patient females and 859± 278.2ng/ml in control females (P <0.001). The mean levels of sIL-6R were 5.21±1.55 ng/ml and 3.91±1.22 ng/ml in patient females and control females, respectively (P=0.01). The mean level of sIL-6R in patient males and control males were 5.63±1.43 ng/ml and 4.34±1.04 ng/ml, respectively (P=0.01). A significant correlation (Pearson’s correlation = 0.45, P=0.008) was observed in the population of females (patients and controls). Conclusion: The results of study suggest the possible involvement of IL-8 and the sIL-6R at stage-I MM and can better characterize the role of chemokines and their receptors in the disease process, especially in the early stages.
Collapse
Affiliation(s)
- Maryam Kohsari
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | | | - Yousef Rasmi
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Hojjat Sayyadi
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
22
|
Saad MI, Rose-John S, Jenkins BJ. ADAM17: An Emerging Therapeutic Target for Lung Cancer. Cancers (Basel) 2019; 11:E1218. [PMID: 31438559 PMCID: PMC6769596 DOI: 10.3390/cancers11091218] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/07/2019] [Accepted: 08/17/2019] [Indexed: 12/23/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related mortality, which histologically is classified into small-cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC). NSCLC accounts for approximately 85% of all lung cancer diagnoses, with the majority of patients presenting with lung adenocarcinoma (LAC). KRAS mutations are a major driver of LAC, and are closely related to cigarette smoking, unlike mutations in the epidermal growth factor receptor (EGFR) which arise in never-smokers. Although the past two decades have seen fundamental progress in the treatment and diagnosis of NSCLC, NSCLC still is predominantly diagnosed at an advanced stage when therapeutic interventions are mostly palliative. A disintegrin and metalloproteinase 17 (ADAM17), also known as tumour necrosis factor-α (TNFα)-converting enzyme (TACE), is responsible for the protease-driven shedding of more than 70 membrane-tethered cytokines, growth factors and cell surface receptors. Among these, the soluble interleukin-6 receptor (sIL-6R), which drives pro-inflammatory and pro-tumourigenic IL-6 trans-signaling, along with several EGFR family ligands, are the best characterised. This large repertoire of substrates processed by ADAM17 places it as a pivotal orchestrator of a myriad of physiological and pathological processes associated with the initiation and/or progression of cancer, such as cell proliferation, survival, regeneration, differentiation and inflammation. In this review, we discuss recent research implicating ADAM17 as a key player in the development of LAC, and highlight the potential of ADAM17 inhibition as a promising therapeutic strategy to tackle this deadly malignancy.
Collapse
Affiliation(s)
- Mohamed I Saad
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
- Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria 3168, Australia
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-University, D-24098 Kiel, Germany
| | - Brendan J Jenkins
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.
- Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria 3168, Australia.
| |
Collapse
|
23
|
Perumal E, So Youn K, Sun S, Seung-Hyun J, Suji M, Jieying L, Yeun-Jun C. PTEN inactivation induces epithelial-mesenchymal transition and metastasis by intranuclear translocation of β-catenin and snail/slug in non-small cell lung carcinoma cells. Lung Cancer 2019; 130:25-34. [PMID: 30885348 DOI: 10.1016/j.lungcan.2019.01.013] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 12/26/2018] [Accepted: 01/27/2019] [Indexed: 12/11/2022]
|
24
|
Xu M, Jiang H, Wang H, Liu J, Liu B, Guo Z. SB225002 inhibits prostate cancer invasion and attenuates the expression of BSP, OPN and MMP‑2. Oncol Rep 2018; 40:726-736. [PMID: 29917166 PMCID: PMC6072299 DOI: 10.3892/or.2018.6504] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 05/31/2018] [Indexed: 11/14/2022] Open
Abstract
The mechanisms of malignant cell metastasis to secondary sites are complex and multifactorial. Studies have demonstrated that small integrin-binding ligand N-linked glycoproteins (SIBLINGs), particularly bone sialoprotein (BSP) and osteopontin (OPN), are involved in neoplastic growth and metastasis. SIBLINGs promote malignant cell invasion and metastasis by enhancing matrix metalloproteinase 2 (MMP-2) and MMP-9 expression. Moreover, BSP and OPN can combine with integrin, which is located on the tumor cell surface, to further promote the malignant behavior of tumor cells. In the present study, we investigated whether SB225002, a specific CXCR2 receptor antagonist, can inhibit prostate cancer cell expression of BSP and OPN and reduce cancer cell invasion ability. A series of experiments showed that after SB225002 treatment, the proliferation, invasion and migration of two androgen-independent prostate cancer cell lines were inhibited, but this inhibitory effect was not observed on androgen-dependent prostate cancer cells. Western blotting showed that the PI3K signaling pathway could regulate the expression of SIBLING and MMP family proteins, and SB22055 could reduce the expression of BSP, OPN and MMP-2 in prostate cancer cells by inhibiting AKT/mTOR phosphorylation. Finally, in vivo experiments confirmed that SB225002 inhibited the proliferation of prostate cancer cells in vivo, and the expression levels of BSP, OPN and MMP-2 were also inhibited.
Collapse
Affiliation(s)
- Meng Xu
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Huamao Jiang
- Graduate School of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Haiguang Wang
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Jiajie Liu
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Baohao Liu
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Zhongqiang Guo
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| |
Collapse
|
25
|
Liu Q, Li A, Yu S, Qin S, Han N, Pestell RG, Han X, Wu K. DACH1 antagonizes CXCL8 to repress tumorigenesis of lung adenocarcinoma and improve prognosis. J Hematol Oncol 2018; 11:53. [PMID: 29636079 PMCID: PMC5894143 DOI: 10.1186/s13045-018-0597-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 03/26/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND C-X-C motif ligand 8 (CXCL8), known as a proinflammatory chemokine, exerts multiple effects on the proliferation, invasion, and migration of tumor cells via the autocrine or paracrine manner. Conversely, the human Dachshund homologue 1 (DACH1) is recognized as a tumor suppressor which retards the progression of various cancers. In prostate cancer, it has been demonstrated that DACH1 was negatively correlated with the expression of CXCL8 and able to antagonize the effects of CXCL8 on cellular migration. Herein, we explored the mechanisms by which DACH1 regulated the CXCL8 in non-small cell lung cancer (NSCLC). METHODS Public microarray and Kaplan-Meier plotter datasets were analyzed. Blood serum samples from lung adenocarcinoma (ADC) patients were collected for enzyme-linked immunosorbent assay (ELISA) analysis. Immunohistochemical staining was conducted on tissue microarray. Cell lines with stable expression of DACH1 were established, and relative gene expression was measured by Western blot, ELISA, real-time PCR, and human cytokine array. Correspondingly, cell lines transfected with shDACH1 were established, and relative gene expression was measured by real-time PCR and immunofluorescence array. Functional studies were performed by transwell and xenograft mice models. Luciferase reporter gene assay was applied to measure the regulation of DACH1 on CXCL8. RESULTS Our study indicated that CXCL8 both at the mRNA and protein level was associated with the high tumor burden of ADC. Correlational analyses in ADC cell lines and ADC tissues showed that DACH1 was inversely correlated with CXCL8. Meanwhile, patients with high DACH1 expression and low CXCL8 expression had prolonged time to death and recurrence. Moreover, we verified the inhibitory effects of DACH1 on CXCL8 both in vitro and in vivo. Mechanism studies proved that DACH1 transcriptionally repressed CXCL8 promoter activity through activator protein-1 (AP-1) and nuclear transcription factor-kappa B (NF-κB) sites. CONCLUSIONS Our study proved that CXCL8 acted as an unfavorable factor promoting to tumor progression and poor prognosis of ADC, while DACH1 antagonized CXCL8 to provide a favorable survival of ADC patients. Double detection of DACH1 and CXCL8 may provide a precise information for further evaluating the prognosis of ADC patients.
Collapse
Affiliation(s)
- Qian Liu
- 0000 0004 1799 5032grid.412793.aDepartment of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 People’s Republic of China
| | - Anping Li
- grid.412633.1Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 People’s Republic of China
| | - Shengnan Yu
- 0000 0004 1799 5032grid.412793.aDepartment of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 People’s Republic of China
| | - Shuang Qin
- 0000 0004 1799 5032grid.412793.aDepartment of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 People’s Republic of China
| | - Na Han
- 0000 0004 1799 5032grid.412793.aDepartment of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 People’s Republic of China
| | - Richard G. Pestell
- Pennsylvania Cancer and Regenerative Medicine Research Center, Wynnewood, PA 19096 USA
| | - Xinwei Han
- grid.412633.1Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 People’s Republic of China
| | - Kongming Wu
- 0000 0004 1799 5032grid.412793.aDepartment of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 People’s Republic of China
| |
Collapse
|
26
|
Rubina KA, Semina EV, Tkachuk VA. Guidance molecules and chemokines in angiogenesis and vascular remodeling. J EVOL BIOCHEM PHYS+ 2017. [DOI: 10.1134/s0022093017050015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
27
|
Cancer-promoting mechanisms of tumor-associated neutrophils. Am J Surg 2017; 214:938-944. [DOI: 10.1016/j.amjsurg.2017.08.003] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 08/03/2017] [Accepted: 08/07/2017] [Indexed: 11/20/2022]
|
28
|
Dai X, Kaluz S, Jiang Y, Shi L, Mckinley D, Wang Y, Wang B, Van Meir EG, Tan C. A novel small-molecule arylsulfonamide causes energetic stress and suppresses breast and lung tumor growth and metastasis. Oncotarget 2017; 8:99245-99260. [PMID: 29245898 PMCID: PMC5725089 DOI: 10.18632/oncotarget.22104] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 10/10/2017] [Indexed: 12/19/2022] Open
Abstract
Neoplastic cells display reprogrammed metabolism due to the heightened energetic demands and the need for biomass synthesis of a growing tumor. Targeting metabolic vulnerabilities is thus an important goal for cancer therapy. Here, we describe a novel small-molecule arylsulfonamide (N-cyclobutyl-N-((2,2-dimethyl-2H-pyrano[3,2-b]pyridin-6-yl)methyl)-3,4-dimethoxybenzenesulfonamide) that exerts potent cytotoxicity and energetic stress on tumor cells while largely sparing non-cancerous human cells. In tumor cells, it stimulates glycolysis and accelerates glucose consumption. Consequently, intracellular ATP levels plummet, triggering activation of AMP-activated protein kinase (AMPK), and diminishing the mammalian target of rapamycin complex 1 (mTORC1) and hypoxia-inducible factor 1 (HIF-1) signaling. In orthotopic triple-negative breast cancer and subcutaneous lung cancer mouse models, this arylsulfonamide robustly suppresses primary tumor growth, inhibits the formation of distant metastases to the lung, and extends mouse survival while being very well tolerated. These therapeutic effects are further potentiated by co-administration of 2-deoxy-D-glucose (2-DG), a glucose analog and glycolysis inhibitor. Collectively, our findings provide preclinical proof of concept for the further development of this arylsulfonamide in combination with 2-DG towards cancer treatment.
Collapse
Affiliation(s)
- Xin Dai
- Department of Pharmaceutics and Drug Delivery, University of Mississippi, Oxford, MS, USA
| | - Stefan Kaluz
- Department of Neurosurgery, Emory University, Atlanta, GA, USA.,Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Ying Jiang
- Department of Pharmaceutical Sciences, Mercer University, Atlanta, GA, USA
| | - Lei Shi
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | - DeAngelo Mckinley
- Department of Pharmaceutical Sciences, Mercer University, Atlanta, GA, USA
| | - Yingzhe Wang
- Department of Pharmaceutical Sciences, Mercer University, Atlanta, GA, USA
| | - Binghe Wang
- Department of Chemistry, Georgia State University, Atlanta, GA, USA
| | - Erwin G Van Meir
- Department of Neurosurgery, Emory University, Atlanta, GA, USA.,Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA.,Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Chalet Tan
- Department of Pharmaceutics and Drug Delivery, University of Mississippi, Oxford, MS, USA
| |
Collapse
|
29
|
Richter HE, Moalli P, Amundsen CL, Malykhina AP, Wallace D, Rogers R, Myers D, Paraiso M, Albo M, Shi H, Nolen T, Meikle S, Word RA. Urinary Biomarkers in Women with Refractory Urgency Urinary Incontinence Randomized to Sacral Neuromodulation versus OnabotulinumtoxinA Compared to Controls. J Urol 2017; 197:1487-1495. [PMID: 28089729 PMCID: PMC5433900 DOI: 10.1016/j.juro.2017.01.037] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/31/2016] [Indexed: 11/21/2022]
Abstract
PURPOSE We measured urinary biomarker levels in women with refractory urgency urinary incontinence and controls at baseline and 6 months after treatment with sacral neuromodulation or intradetrusor injection of onabotulinumtoxinA. We also assessed the association of baseline biomarkers with posttreatment urgency urinary incontinence episodes and overactive bladder symptom bother outcomes. MATERIALS AND METHODS First morning urine samples were collected from consented trial participants and age matched women without urgency urinary incontinence. Biomarkers reflecting general inflammation, neuroinflammation, afferent neurotransmitters and tissue remodeling were measured using standardized enzyme-linked immunosorbent assay and activity assays as appropriate. Symptom bother was assessed by the overactive bladder questionnaire and urgency urinary incontinence episodes were determined by bladder diary. Linear models were used to examine differences in mean biomarker levels and the change in urgency urinary incontinence episodes and symptom bother between baseline and 6 months. Modest evidence of a potential association was represented by p ≤0.01 and p ≤0.004 represented moderate evidence of an association with outcomes. RESULTS Baseline biomarker levels differed little between cases and controls except tropoelastin (p = 0.001) and N-terminal telopeptide collagen type 1 (p <0.001). Changes in biomarker levels 6 months after intervention included decreases in collagenase (p <0.001) in both treatment groups and increases in interleukin-8 (p = 0.002) and matrix metalloprotease-9 (p <0.001) in the onabotulinumtoxinA group. Higher baseline calcitonin gene-related peptide across both treatments (p = 0.007) and nerve growth factor in the onabotulinumtoxinA arm (p = 0.007) were associated with less reduction in overactive bladder symptom bother. CONCLUSIONS Refractory urgency urinary incontinence is a complex condition. These data suggest that matrix remodeling and neuropeptide mediation may be involved in its pathophysiological mechanisms and response to treatment.
Collapse
Affiliation(s)
| | | | | | | | - Dennis Wallace
- RTI International, Research Triangle Park, North Carolina
| | | | | | | | - Michael Albo
- University of California-San Diego, San Diego, California
| | - Haolin Shi
- University of Texas Southwestern, Dallas, Texas
| | - Tracy Nolen
- RTI International, Research Triangle Park, North Carolina
| | - Susie Meikle
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Baltimore, Maryland
| | - R Ann Word
- University of Texas Southwestern, Dallas, Texas
| |
Collapse
|
30
|
Cao Y, Huang H, Wang Z, Zhang G. The Inflammatory CXC Chemokines, GROα high, IP-10 low, and MIG low, in Tumor Microenvironment Can Be Used as New Indicators for Non-small Cell Lung Cancer Progression. Immunol Invest 2017; 46:361-374. [PMID: 28375674 DOI: 10.1080/08820139.2017.1280052] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
AIMS To investigate the clinical significance of tumor tissue-infiltrating chemokines expression in non-small cell lung cancer (NSCLC) microenvironment. MATERIALS AND METHODS Fresh tissue samples were acquired from 50 patients with NSCLC after operation. Then, we quantified the total protein with the BCA Protein Assay Kit and tested 13 chemotactic factors in paired samples including tumor tissues, tumor adjacent tissues, and normal tissues with the CBA Kit. RESULTS We found that the chemokine CC subfamily of MCP-1, MIP-1α, MIP-1β, and MIP-3α and the chemokine CXC subfamily of IL-8, GROα, IP-10, and MIG expressions in tumor tissues were significantly higher than those in tumor-adjacent tissues and normal tissues. However, regulated upon activation normal T cell expressed and secreted (RANTES), human thymus activation regulated chemokine (TARC), chemokine (C-C motif) ligand 11 (CCL11), interferon-inducible T cell alpha chemoattractant (I-TAC), and ENA-78 expressions did not show significant difference. Analyzing the influence of chemokine expression level in tumor tissues on disease progression, we found the median progression-free survival (mPFS) of patients with GROαhigh was significantly lower than those with GROαlow; mPFS of patients with IP-10low was significantly lower than those with IP-10high; and mPFS of patients with MIGlow was significantly lower than those with MIGhigh. However, MCP-1, MIP-1α, MIP-1β, MIP-3α, and IL-8 had no significant value to elevate the mPFS of patients with NSCLC. CONCLUSION In summary, tumor tissue-infiltrating CXC chemokines, GROαhigh, IP-10low, and MIGlow in the tumor microenvironment can be used as potential indicators for the progression of NSCLC.
Collapse
Affiliation(s)
- Ya Cao
- a Jiangsu Key Laboratory of Clinical Immunology, The First Affiliated Hospital of Soochow University , Suzhou , China.,b Department of Medical Oncology , The First Affiliated Hospital of Soochow University , Suzhou , China
| | - Haitao Huang
- b Department of Medical Oncology , The First Affiliated Hospital of Soochow University , Suzhou , China.,c Department of Thoracic Surgery , The First Affiliated Hospital of Soochow University , Suzhou , China
| | - Zhenxin Wang
- a Jiangsu Key Laboratory of Clinical Immunology, The First Affiliated Hospital of Soochow University , Suzhou , China.,b Department of Medical Oncology , The First Affiliated Hospital of Soochow University , Suzhou , China
| | - Guangbo Zhang
- a Jiangsu Key Laboratory of Clinical Immunology, The First Affiliated Hospital of Soochow University , Suzhou , China
| |
Collapse
|
31
|
Hattar K, Reinert CP, Sibelius U, Gökyildirim MY, Subtil FSB, Wilhelm J, Eul B, Dahlem G, Grimminger F, Seeger W, Grandel U. Lipoteichoic acids from Staphylococcus aureus stimulate proliferation of human non-small-cell lung cancer cells in vitro. Cancer Immunol Immunother 2017; 66:799-809. [PMID: 28314957 PMCID: PMC5445152 DOI: 10.1007/s00262-017-1980-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 02/24/2017] [Indexed: 12/22/2022]
Abstract
Pulmonary infections are frequent complications in lung cancer and may worsen its outcome and survival. Inflammatory mediators are suspected to promote tumor growth in non-small-cell lung cancer (NSCLC). Hence, bacterial pathogens may affect lung cancer growth by activation of inflammatory signalling. Against this background, we investigated the effect of purified lipoteichoic acids (LTA) of Staphylococcus aureus (S. aureus) on cellular proliferation and liberation of interleukin (IL)-8 in the NSCLC cell lines A549 and H226. A549 as well as H226 cells constitutively expressed TLR-2 mRNA. Even in low concentrations, LTA induced a prominent increase in cellular proliferation of A549 cells as quantified by automatic cell counting. In parallel, metabolic activity of A549 cells was enhanced. The increase in proliferation was accompanied by an increase in IL-8 mRNA expression and a dose- and time-dependent release of IL-8. Cellular proliferation as well as the release of IL-8 was dependent on specific ligation of TLR-2. Interestingly, targeting IL-8 by neutralizing antibodies completely abolished the LTA-induced proliferation of A549 cells. The pro-proliferative effect of LTA could also be reproduced in the squamous NSCLC cell line H226. In summary, LTA of S. aureus induced proliferation of NSCLC cell lines of adeno- and squamous cell carcinoma origin. Ligation of TLR-2 followed by auto- or paracrine signalling by endogenously synthesized IL-8 is centrally involved in LTA-induced tumor cell proliferation. Therefore, pulmonary infections may exert a direct pro-proliferative effect on lung cancer growth.
Collapse
Affiliation(s)
- Katja Hattar
- Department of Internal Medicine IV/V, University of Giessen and Marburg Lung Center (UGMLC), Klinikstrasse 33, Giessen, Germany
| | - Christian P Reinert
- Department of Internal Medicine IV/V, University of Giessen and Marburg Lung Center (UGMLC), Klinikstrasse 33, Giessen, Germany
| | - Ulf Sibelius
- Department of Internal Medicine IV/V, University of Giessen and Marburg Lung Center (UGMLC), Klinikstrasse 33, Giessen, Germany
| | - Mira Y Gökyildirim
- Department of Internal Medicine IV/V, University of Giessen and Marburg Lung Center (UGMLC), Klinikstrasse 33, Giessen, Germany
| | | | - Jochen Wilhelm
- Department of Internal Medicine II, University of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
| | - Bastian Eul
- Department of Internal Medicine IV/V, University of Giessen and Marburg Lung Center (UGMLC), Klinikstrasse 33, Giessen, Germany
| | - Gabriele Dahlem
- Department of Internal Medicine IV/V, University of Giessen and Marburg Lung Center (UGMLC), Klinikstrasse 33, Giessen, Germany
| | - Friedrich Grimminger
- Department of Internal Medicine IV/V, University of Giessen and Marburg Lung Center (UGMLC), Klinikstrasse 33, Giessen, Germany
| | - Werner Seeger
- Department of Internal Medicine II, University of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany.,Max-Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Ulrich Grandel
- Department of Internal Medicine IV/V, University of Giessen and Marburg Lung Center (UGMLC), Klinikstrasse 33, Giessen, Germany. .,Asklepios Klinik Lich, Lich, Germany.
| |
Collapse
|
32
|
Lesage J, Suarez‐Carmona M, Neyrinck‐Leglantier D, Grelet S, Blacher S, Hunziker W, Birembaut P, Noël A, Nawrocki‐Raby B, Gilles C, Polette M. Zonula occludens‐1/NF‐κB/CXCL8: a new regulatory axis for tumor angiogenesis. FASEB J 2017; 31:1678-1688. [DOI: 10.1096/fj.201600890r] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 01/03/2017] [Indexed: 01/18/2023]
Affiliation(s)
- Julien Lesage
- INSERM, Unité Mixte de Recherche–S 903, Structure Fédérative de Recherche Champagne‐Ardennes Picardie Santé (SFR CAP)University of Reims Champagne‐Ardenne Reims France
| | - Meggy Suarez‐Carmona
- Laboratory of Tumor and Development Biology, Grappe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)‐CancerUniversity of Liège Liège Belgium
| | - Deborah Neyrinck‐Leglantier
- INSERM, Unité Mixte de Recherche–S 903, Structure Fédérative de Recherche Champagne‐Ardennes Picardie Santé (SFR CAP)University of Reims Champagne‐Ardenne Reims France
| | - Simon Grelet
- Department of Biochemistry and Molecular BiologyMedical University of South Carolina Charleston South Carolina USA
| | - Silvia Blacher
- Laboratory of Tumor and Development Biology, Grappe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)‐CancerUniversity of Liège Liège Belgium
| | - Walter Hunziker
- Epithelial Cell Biology LaboratoryInstitute of Molecular and Cell Biology Singapore Singapore
| | - Philippe Birembaut
- INSERM, Unité Mixte de Recherche–S 903, Structure Fédérative de Recherche Champagne‐Ardennes Picardie Santé (SFR CAP)University of Reims Champagne‐Ardenne Reims France
- Laboratory of BiopathologyCentres Hospitaliers Universitaires Reims France
| | - Agnes Noël
- Laboratory of Tumor and Development Biology, Grappe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)‐CancerUniversity of Liège Liège Belgium
| | - Béatrice Nawrocki‐Raby
- INSERM, Unité Mixte de Recherche–S 903, Structure Fédérative de Recherche Champagne‐Ardennes Picardie Santé (SFR CAP)University of Reims Champagne‐Ardenne Reims France
| | - Christine Gilles
- Laboratory of Tumor and Development Biology, Grappe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)‐CancerUniversity of Liège Liège Belgium
| | - Myriam Polette
- INSERM, Unité Mixte de Recherche–S 903, Structure Fédérative de Recherche Champagne‐Ardennes Picardie Santé (SFR CAP)University of Reims Champagne‐Ardenne Reims France
- Laboratory of BiopathologyCentres Hospitaliers Universitaires Reims France
| |
Collapse
|
33
|
Liu Q, Li A, Tian Y, Wu JD, Liu Y, Li T, Chen Y, Han X, Wu K. The CXCL8-CXCR1/2 pathways in cancer. Cytokine Growth Factor Rev 2016; 31:61-71. [PMID: 27578214 PMCID: PMC6142815 DOI: 10.1016/j.cytogfr.2016.08.002] [Citation(s) in RCA: 443] [Impact Index Per Article: 49.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 08/13/2016] [Accepted: 08/23/2016] [Indexed: 02/07/2023]
Abstract
Persistent infection or chronic inflammation contributes significantly to tumourigenesis and tumour progression. C-X-C motif ligand 8 (CXCL8) is a chemokine that acts as an important multifunctional cytokine to modulate tumour proliferation, invasion and migration in an autocrine or paracrine manner. Studies have suggested that CXCL8 and its cognate receptors, C-X-C chemokine receptor 1 (CXCR1) and CX-C chemokine receptor 2 (CXCR2), mediate the initiation and development of various cancers including breast cancer, prostate cancer, lung cancer, colorectal carcinoma and melanoma. CXCL8 also integrates with multiple intracellular signalling pathways to produce coordinated effects. Neovascularisation, which provides a basis for fostering tumour growth and metastasis, is now recognised as a critical function of CXCL8 in the tumour microenvironment. In this review, we summarize the biological functions and ficlinical significance of the CXCL8 signalling axis in cancer. We also propose that CXCL8 may be a potential therapeutic target for cancer treatment
Collapse
Affiliation(s)
- Qian Liu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Anping Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yijun Tian
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jennifer D Wu
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Yu Liu
- Department of Geriatric, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Tengfei Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yuan Chen
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
34
|
Abstract
The human body combats infection and promotes wound healing through the remarkable process of inflammation. Inflammation is characterized by the recruitment of stromal cell activity including recruitment of immune cells and induction of angiogenesis. These cellular processes are regulated by a class of soluble molecules called cytokines. Based on function, cell target, and structure, cytokines are subdivided into several classes including: interleukins, chemokines, and lymphokines. While cytokines regulate normal physiological processes, chronic deregulation of cytokine expression and activity contributes to cancer in many ways. Gene polymorphisms of all types of cytokines are associated with risk of disease development. Deregulation RNA and protein expression of interleukins, chemokines, and lymphokines have been detected in many solid tumors and hematopoetic malignancies, correlating with poor patient prognosis. The current body of literature suggests that in some tumor types, interleukins and chemokines work against the human body by signaling to cancer cells and remodeling the local microenvironment to support the growth, survival, and invasion of primary tumors and enhance metastatic colonization. Some lymphokines are downregulated to suppress tumor progression by enhancing cytotoxic T cell activity and inhibiting tumor cell survival. In this review, we will describe the structure/function of several cytokine families and review our current understanding on the roles and mechanisms of cytokines in tumor progression. In addition, we will also discuss strategies for exploiting the expression and activity of cytokines in therapeutic intervention.
Collapse
Affiliation(s)
- M Yao
- University of Kansas Medical Center, Kansas City, KS, United States
| | - G Brummer
- University of Kansas Medical Center, Kansas City, KS, United States
| | - D Acevedo
- University of Kansas Medical Center, Kansas City, KS, United States
| | - N Cheng
- University of Kansas Medical Center, Kansas City, KS, United States.
| |
Collapse
|
35
|
Whiteford JR, De Rossi G, Woodfin A. Mutually Supportive Mechanisms of Inflammation and Vascular Remodeling. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 326:201-78. [PMID: 27572130 DOI: 10.1016/bs.ircmb.2016.05.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chronic inflammation is often accompanied by angiogenesis, the development of new blood vessels from existing ones. This vascular response is a response to chronic hypoxia and/or ischemia, but is also contributory to the progression of disorders including atherosclerosis, arthritis, and tumor growth. Proinflammatory and proangiogenic mediators and signaling pathways form a complex and interrelated network in these conditions, and many factors exert multiple effects. Inflammation drives angiogenesis by direct and indirect mechanisms, promoting endothelial proliferation, migration, and vessel sprouting, but also by mediating extracellular matrix remodeling and release of sequestered growth factors, and recruitment of proangiogenic leukocyte subsets. The role of inflammation in promoting angiogenesis is well documented, but by facilitating greater infiltration of leukocytes and plasma proteins into inflamed tissues, angiogenesis can also propagate chronic inflammation. This review examines the mutually supportive relationship between angiogenesis and inflammation, and considers how these interactions might be exploited to promote resolution of chronic inflammatory or angiogenic disorders.
Collapse
Affiliation(s)
- J R Whiteford
- William Harvey Research Institute, Barts and London School of Medicine and Dentistry, Queen Mary College, University of London, London, United Kingdom
| | - G De Rossi
- William Harvey Research Institute, Barts and London School of Medicine and Dentistry, Queen Mary College, University of London, London, United Kingdom
| | - A Woodfin
- Cardiovascular Division, King's College, University of London, London, United Kingdom.
| |
Collapse
|
36
|
Khan MN, Wang B, Wei J, Zhang Y, Li Q, Luan X, Cheng JW, Gordon JR, Li F, Liu H. CXCR1/2 antagonism with CXCL8/Interleukin-8 analogue CXCL8(3-72)K11R/G31P restricts lung cancer growth by inhibiting tumor cell proliferation and suppressing angiogenesis. Oncotarget 2016; 6:21315-27. [PMID: 26087179 PMCID: PMC4673267 DOI: 10.18632/oncotarget.4066] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 05/25/2015] [Indexed: 12/27/2022] Open
Abstract
CXCR1 and CXCR2 together with cognate chemokines are significantly upregulated in a number of cancers, where they act as key regulators of tumor cell proliferation, metastasis, and angiogenesis. We have previously reported a mutant protein of CXCL8/Interleukin-8, CXCL8(3–72)K11R/G31P (G31P), which can act as a selective antagonist towards CXCR1/2 with therapeutic efficacy in both inflammatory diseases and malignancies. In this study, we investigated the effect of this ELR-CXC chemokine antagonist G31P on human non-small cell lung cancer cells and lung tumor progression in an orthotopic xenograft model. We report increased mRNA levels of CXCR1 and CXCR2 in human lung cancer tissues compared to normal counterparts. Expression levels of CXCR1/2 cognate ligands was determined by ELISA. CXCR1/2 receptor antagonism via G31P leads to decreased H460 and A549 cell proliferation and migration in a dose-dependent manner. G31P also enhanced apoptosis in lung cancer cells as determined by elevated levels of cleaved PARP, Caspase-8, and Bax, together with a reduced expression of the anti-apoptotic protein Bcl-2. In an in vivo orthotopic xenograft mouse model of human lung cancer, G31P treatment suppressed tumor growth, metastasis, and angiogenesis. At the molecular level, G31P treatment was correlated with decreased expression of VEGF and NFкB-p65, in addition to reduced phosphorylation of ERK1/2 and AKT. Our results suggest that G31P blockage of CXCR1 and CXCR2 can inhibit human lung cancer cell growth and metastasis, which offers potential therapeutic opportunities.
Collapse
Affiliation(s)
- Muhammad Noman Khan
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.,Department of Immunology, Dalian Medical University, Dalian, China
| | - Bing Wang
- Department of Immunology, Dalian Medical University, Dalian, China
| | - Jing Wei
- Department of Immunology, Dalian Medical University, Dalian, China
| | - Yingqiu Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Qiang Li
- Jilin Medical College, Jilin, China
| | - Xuelin Luan
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Jya-Wei Cheng
- Institute of Biotechnology, Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | - John R Gordon
- The Division of Respirology, Critical Care and Sleep Medicine, Royal University Hospital, University of Saskatchewan, Saskatoon, Canada
| | - Fang Li
- Department of Immunology, Dalian Medical University, Dalian, China
| | - Han Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| |
Collapse
|
37
|
Liu TT, Mu LQ, Dai W, Wang CB, Liu XY, Xiang DX. Preparation, characterization, and evaluation of antitumor effect of Brucea javanica oil cationic nanoemulsions. Int J Nanomedicine 2016; 11:2515-29. [PMID: 27330293 PMCID: PMC4898033 DOI: 10.2147/ijn.s101918] [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] [Indexed: 01/09/2023] Open
Abstract
The purpose of this study was to prepare Brucea javanica oil cationic nanoemulsions (BJO-CN) with BJO as drug as well as oil phase and chitosan as cationic inducer, to explore the practical suitability of using cationic nanoemulsions for oral delivery of mixed oil, and to test its bioavailability and antitumor effect. BJO-CN was prepared by chitosan solution stirring method and then characterized physicochemically. The obtained BJO-CN had a spherical morphology with a positive zeta potential of 18.9 mV and an average particle size of 42.36 nm, showing high colloidal stability. The drug loading of BJO-CN was 91.83 mg·mL−1, determined by high-performance liquid chromatography with precolumn derivatization. Pharmacokinetic studies revealed that, compared with BJO emulsion (BJO-E) (the dosage of BJO-CN and BJO-E was equal to 505 mg·kg−1, calculated by oleic acid), BJO-CN exhibited a significant increase in the area under the plasma drug concentration–time curve over the period of 24 hours and relative bioavailability was 1.6-fold. Furthermore, the antitumor effect of BJO-CN in the orthotopic mouse model of lung cancer was evaluated by recording the median survival time and the weight of lung tissue with tumor, hematoxylin and eosin staining, and immunohistochemical technique. Results of anticancer experiments illustrated that, even though the administrated dosage in the BJO-CN group was half of that in the BJO-E group, BJO-CN exhibited similar antitumor effect to BJO-E. Moreover, BJO-CN had good synergistic effect in combination therapy with vinorelbine. These results suggested that cationic nanoemulsions are an effective and promising delivery system to enhance the oral bioavailability and anticancer effect of BJO.
Collapse
Affiliation(s)
- Ting-Ting Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China; Institute of Clinical Pharmacy, Central South University, Changsha, People's Republic of China; Key Laboratory for New Technology of Chinese Medicine Preparations of Hunan Province, Changsha, People's Republic of China
| | - Li-Qiu Mu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China; Institute of Clinical Pharmacy, Central South University, Changsha, People's Republic of China; Key Laboratory for New Technology of Chinese Medicine Preparations of Hunan Province, Changsha, People's Republic of China
| | - Wei Dai
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China; Institute of Clinical Pharmacy, Central South University, Changsha, People's Republic of China; Key Laboratory for New Technology of Chinese Medicine Preparations of Hunan Province, Changsha, People's Republic of China
| | - Chuan-Bang Wang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China; Institute of Clinical Pharmacy, Central South University, Changsha, People's Republic of China; Key Laboratory for New Technology of Chinese Medicine Preparations of Hunan Province, Changsha, People's Republic of China
| | - Xin-Yi Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China; Institute of Clinical Pharmacy, Central South University, Changsha, People's Republic of China; Key Laboratory for New Technology of Chinese Medicine Preparations of Hunan Province, Changsha, People's Republic of China
| | - Da-Xiong Xiang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China; Institute of Clinical Pharmacy, Central South University, Changsha, People's Republic of China; Key Laboratory for New Technology of Chinese Medicine Preparations of Hunan Province, Changsha, People's Republic of China
| |
Collapse
|
38
|
Jiang YN, Yan HQ, Huang XB, Wang YN, Li Q, Gao FG. Interleukin 6 trigged ataxia-telangiectasia mutated activation facilitates lung cancer metastasis via MMP-3/MMP-13 up-regulation. Oncotarget 2015; 6:40719-33. [PMID: 26528698 PMCID: PMC4747364 DOI: 10.18632/oncotarget.5825] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 09/23/2015] [Indexed: 12/24/2022] Open
Abstract
Our previous studies show that the phosphorylation of ataxia-telangiectasia mutated (ATM) induced by interleukin 6 (IL-6) treatment contributes to multidrug resistance formation in lung cancer cells, but the exact role of ATM activation in IL-6 increased metastasis is still elusive. In the present study, matrix metalloproteinase-3 (MMP-3) and MMP-13 were firstly demonstrated to be involved in IL-6 correlated cell migration. Secondly, IL-6 treatment not only increased MMP-3/MMP-13 expression but also augmented its activities. Thirdly, the inhibition of ATM phosphorylation efficiently abolished IL-6 up-regulating MMP-3/MMP-13 expression and increasing abilities of cell migration. Most importantly, the in vivo test showed that the inhibition of ATM abrogate the effect of IL-6 on lung cancer metastasis via MMP-3/MMP-13 down-regulation. Taken together, these findings demonstrate that IL-6 inducing ATM phosphorylation increases the expression of MMP-3/MMP-13, augments the abilities of cell migration, and promotes lung cancer metastasis, indicating that ATM is a potential target molecule to overcome IL-6 correlated lung cancer metastasis.
Collapse
Affiliation(s)
- Yi Na Jiang
- Department of Immunology, Basic Medicine Science, Medical College, Xiamen University, Xiamen 361005, People's Republic of China
| | - Hong Qiong Yan
- Department of Immunology, Basic Medicine Science, Medical College, Xiamen University, Xiamen 361005, People's Republic of China
| | - Xiao Bo Huang
- Department of Immunology, Basic Medicine Science, Medical College, Xiamen University, Xiamen 361005, People's Republic of China
| | - Yi Nan Wang
- Department of Immunology, Basic Medicine Science, Medical College, Xiamen University, Xiamen 361005, People's Republic of China
| | - Qing Li
- Department of Immunology, Basic Medicine Science, Medical College, Xiamen University, Xiamen 361005, People's Republic of China
| | - Feng Guang Gao
- Department of Immunology, Basic Medicine Science, Medical College, Xiamen University, Xiamen 361005, People's Republic of China
- State Key Laboratory of Oncogenes and Related Genes, Shang Hai Jiao Tong University, Shanghai 200032, People's Republic of China
| |
Collapse
|
39
|
Chemopreventive effect of leflunomide against Ehrlich's solid tumor grown in mice: Effect on EGF and EGFR expression and tumor proliferation. Life Sci 2015; 141:193-201. [DOI: 10.1016/j.lfs.2015.10.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 08/07/2015] [Accepted: 10/02/2015] [Indexed: 02/07/2023]
|
40
|
|
41
|
Choi S, Coonrod S, Estroff L, Fischbach C. Chemical and physical properties of carbonated hydroxyapatite affect breast cancer cell behavior. Acta Biomater 2015; 24:333-42. [PMID: 26072364 DOI: 10.1016/j.actbio.2015.06.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 05/06/2015] [Accepted: 06/01/2015] [Indexed: 01/04/2023]
Abstract
Breast microcalcifications are routinely explored for mammographic detection of breast cancer and are primarily composed of non-stoichiometric hydroxyapatite (Ca10-x(PO4)6-x(CO3)x(OH)2-x) (HA). Interestingly, HA morphology and carbonate substitution vary in malignant vs. benign lesions. However, whether or not these changes (i) are functionally linked and (ii) impact malignancy remains unclear due in part to lack of model systems that permit evaluating these possibilities. Here, we have adapted a 96 well-based mineralized culture platform to investigate breast cancer cell behavior in response to systematic changes in the chemical and physical properties of HA. By adjusting the carbonate content of the simulated body fluid (SBF) solutions used during growth, we can control the morphology and carbonate substitution of the deposited HA. Our results suggest that both the combined and individual effects of these differences alter breast cancer cell growth and secretion of tumorigenic interleukin-8 (IL-8). Consequently, changes in both HA carbonate incorporation and morphology impact the behavior of breast cancer cells. Collectively, our data underline the importance of biomineralized culture platforms to evaluate the functional contribution of HA material properties to the pathogenesis of breast cancer. STATEMENT OF SIGNIFICANCE Breast microcalcifications are small mineral deposits primarily composed of hydroxyapatite (HA). HA physicochemical properties have been of considerable interest, as these are often altered during breast cancer progression and linked to malignancy. However, the functional relationship between these changes and malignancy remains unclear due in part to lack of model systems. Here, we have adapted a previously developed a 96 well-based culture platform to evaluate breast cancer cell behavior in response to systematic changes in HA properties. Our results demonstrate that changes in HA morphology and carbonate content influence breast cancer cell growth and interleukin-8 secretion, and suggest that characterizing the effect of HA properties on breast cancer cells may improve our understanding of breast cancer development and progression.
Collapse
|
42
|
Rivas-Fuentes S, Salgado-Aguayo A, Pertuz Belloso S, Gorocica Rosete P, Alvarado-Vásquez N, Aquino-Jarquin G. Role of Chemokines in Non-Small Cell Lung Cancer: Angiogenesis and Inflammation. J Cancer 2015; 6:938-52. [PMID: 26316890 PMCID: PMC4543754 DOI: 10.7150/jca.12286] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 06/23/2015] [Indexed: 12/12/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is one of the most common types of aggressive cancer. The tumor tissue, which shows an active angiogenesis, is composed of neoplastic and stromal cells, and an abundant inflammatory infiltrate. Angiogenesis is important to support tumor growth, while infiltrating cells contribute to the tumor microenvironment through the secretion of growth factors, cytokines and chemokines, important molecules in the progression of the disease. Chemokines are important in development, activation of the immune response, and physiological angiogenesis. Chemokines have emerged as important regulators in the pathophysiology of cancer. These molecules are involved in the angiogenesis/angiostasis balance and in the recruitment of tumor infiltrating hematopoietic cells. In addition, chemokines promote tumor cell survival, as well as the directing and establishment of tumor cells to metastasis sites. The findings summarized here emphasize the central role of chemokines as modulators of tumor angiogenesis and their potential role as therapeutic targets in the inflammatory process of NSCLC angiogenesis.
Collapse
Affiliation(s)
- Selma Rivas-Fuentes
- 1. Department of Biochemistry Research, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Alfonso Salgado-Aguayo
- 2. Laboratory of Research on Rheumatic Diseases, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Silvana Pertuz Belloso
- 3. Department of Comparative Biology, Faculty of Sciences, National Autonomous University of Mexico, Mexico City, Mexico
| | - Patricia Gorocica Rosete
- 1. Department of Biochemistry Research, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Noé Alvarado-Vásquez
- 1. Department of Biochemistry Research, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Guillermo Aquino-Jarquin
- 4. Laboratory of Research on Genomics, Genetics and Bioinformatics. Tower of Haemato-oncology, Children´s Hospital of Mexico “Federico Gomez”, Mexico City, Mexico
| |
Collapse
|
43
|
DelNero P, Lane M, Verbridge SS, Kwee B, Kermani P, Hempstead B, Stroock A, Fischbach C. 3D culture broadly regulates tumor cell hypoxia response and angiogenesis via pro-inflammatory pathways. Biomaterials 2015; 55:110-8. [PMID: 25934456 DOI: 10.1016/j.biomaterials.2015.03.035] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 03/16/2015] [Accepted: 03/20/2015] [Indexed: 01/08/2023]
Abstract
Oxygen status and tissue dimensionality are critical determinants of tumor angiogenesis, a hallmark of cancer and an enduring target for therapeutic intervention. However, it is unclear how these microenvironmental conditions interact to promote neovascularization, due in part to a lack of comprehensive, unbiased data sets describing tumor cell gene expression as a function of oxygen levels within three-dimensional (3D) culture. Here, we utilized alginate-based, oxygen-controlled 3D tumor models to study the interdependence of culture context and the hypoxia response. Microarray gene expression analysis of tumor cells cultured in 2D versus 3D under ambient or hypoxic conditions revealed striking interdependence between culture dimensionality and hypoxia response, which was mediated in part by pro-inflammatory signaling pathways. In particular, interleukin-8 (IL-8) emerged as a major player in the microenvironmental regulation of the hypoxia program. Notably, this interaction between dimensionality and oxygen status via IL-8 increased angiogenic sprouting in a 3D endothelial invasion assay. Taken together, our data suggest that pro-inflammatory pathways are critical regulators of tumor hypoxia response within 3D environments that ultimately impact tumor angiogenesis, potentially providing important therapeutic targets. Furthermore, these results highlight the importance of pathologically relevant tissue culture models to study the complex physical and chemical processes by which the cancer microenvironment mediates new vessel formation.
Collapse
Affiliation(s)
- Peter DelNero
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Maureen Lane
- Department of Medicine, Weill Cornell Medical School, New York, NY 10065, USA
| | - Scott S Verbridge
- School of Biomedical Engineering and Sciences, Virginia Tech-Wake Forest University, Blacksburg, VA 24061, USA
| | - Brian Kwee
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Pouneh Kermani
- Department of Medicine, Weill Cornell Medical School, New York, NY 10065, USA
| | - Barbara Hempstead
- Department of Medicine, Weill Cornell Medical School, New York, NY 10065, USA
| | - Abraham Stroock
- School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA; Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY 14853, USA
| | - Claudia Fischbach
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA; Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
44
|
Liu X, Dai LI, Zhou R. Association between preeclampsia and the CXC chemokine family (Review). Exp Ther Med 2015; 9:1572-1576. [PMID: 26136860 DOI: 10.3892/etm.2015.2337] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 02/18/2015] [Indexed: 12/30/2022] Open
Abstract
Preeclampsia is a major cause of maternal and perinatal mortality and morbidity, characterized by gestational hypertension, proteinuria, systemic endothelial cell activation and an exaggerated inflammatory response. The precise cause of preeclampsia is not currently known; however, it is widely accepted that the pathogenesis of preeclampsia involves inadequate trophoblast invasion, leading to generalized endothelial dysfunction and an exaggerated inflammatory response. Chemokines are a superfamily of structurally similar proteins that mediate cell recruitment, angiogenesis, immunity and stem cell trafficking. CXC chemokines are a family of cytokines, unique in their ability to behave in a disparate manner in the regulation of angiogenesis. The CXC chemokine family further divides into two subfamilies; CXC ELR+, which promotes angiogenesis, and CXC ELR-, which inhibits angiogenesis. Furthermore, CXC chemokines are involved in the pathogenesis of various conditions, including malignant tumors, wound repair, chronic inflammation, atherosclerosis and potentially preeclampsia.
Collapse
Affiliation(s)
- Xijing Liu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - L I Dai
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Rong Zhou
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
45
|
Song Y, Baba T, Li YY, Furukawa K, Tanabe Y, Matsugo S, Sasaki S, Mukaida N. Gemcitabine-induced CXCL8 expression counteracts its actions by inducing tumor neovascularization. Biochem Biophys Res Commun 2015; 458:341-6. [PMID: 25646691 DOI: 10.1016/j.bbrc.2015.01.112] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 01/23/2015] [Indexed: 11/16/2022]
|
46
|
CXCL8, overexpressed in colorectal cancer, enhances the resistance of colorectal cancer cells to anoikis. Cancer Lett 2015; 361:22-32. [PMID: 25687885 DOI: 10.1016/j.canlet.2015.02.021] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Revised: 02/10/2015] [Accepted: 02/10/2015] [Indexed: 12/25/2022]
Abstract
Anoikis is a form of apoptosis which occurs when anchorage-dependent cells either show loss of adhesion or inappropriate adhesion. Only a few cancer cells that detach from the primary site of the tumor acquire the ability to resist anoikis and form metastasis. The mechanism underlying the resistance of colorectal cancer (CRC) cells to anoikis remains unclear. Interleukin-8 (alternatively known as CXCL8) is associated with CRC angiogenesis and progression. Here, we found that a high abundance of CXCL8 or TOPK strongly correlated with poor overall and disease-free survival of 186 patients with CRC. A combination of high CXCL8 and high TOPK expressions had the worst prognosis. We showed that CXCL8 expression was negatively correlated with anoikis in CRC cells. CXCL8 treatment enhanced the resistance of CRC cells to apoptosis, which was accompanied by the increase of TOPK, and the activation of AKT and ERK. Moreover, we demonstrated that the inhibition of either ERK or AKT by specific chemical inhibitors attenuated the CXCL8-mediated resistance to anoikis. Treatment with AKT inhibitor abolished the effects of CXCL8 on TOPK expression, suggesting that TOPK was downstream of AKT in the process of anoikis. Taken together, we demonstrated that CXCL8 is strongly implicated in the resistance of CRC cells to anoikis, and that the AKT, TOPK and ERK pathway may be a potential therapeutic target for CRC.
Collapse
|
47
|
MyD88 expression is associated with paclitaxel resistance in lung cancer A549 cells. Oncol Rep 2014; 32:1837-44. [PMID: 25175786 DOI: 10.3892/or.2014.3433] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 06/03/2014] [Indexed: 11/05/2022] Open
Abstract
The purpose of the present study was to investigate the relationship between myeloid differentiation primary response gene 88 (MyD88) expression and the resistance to paclitaxel of A549 lung cancer cells. In order to achieve MyD88 gene overexpression or knockdown in the A549 cell line, the cells were infected with lentivirus carrying MyD88 gene or shRNA to MyD88. MyD88 mRNA and protein expression was measured by RT-qPCR, immunohistochemistry and western blotting after infection for confirmation. Cell prolife-ration was detected by the WST-1 assay. Flow cytometry was used to measure the cell cycle and apoptosis. The transwell migration assay was used to observe the change of migration of transfected cells. The results showed that the overexpression of MyD88 increased the resistance of lung cancer A549 cells to paclitaxel, while the suppression of MyD88 increased the sensitivity of A549 lung cancer cells to paclitaxel. Following the paclitaxel treatment, a decreased apoptosis and G2 phase ratio, an increased cell migration ratio, and an increased production of IL-8 were found in MyD88‑overexpressed A549 cells. The western blot analysis revealed that Myd88 overexpression resulted in an increased level of Bcl-2 but a decreased level of Bax in A549 cells. These findings suggested that the expression level of MyD88 is closely associated with paclitaxel resistance in A549 lung cancer cells. Thus, the downregulation of MyD88 in A549 cells increased its sensitivity to paclitaxel treatment, whereas the upregulation of MyD88 substantiates its paclitaxel resistance.
Collapse
|
48
|
Bauerle KT, Schweppe RE, Lund G, Kotnis G, Deep G, Agarwal R, Pozdeyev N, Wood WM, Haugen BR. Nuclear factor κB-dependent regulation of angiogenesis, and metastasis in an in vivo model of thyroid cancer is associated with secreted interleukin-8. J Clin Endocrinol Metab 2014; 99:E1436-44. [PMID: 24758177 PMCID: PMC4121024 DOI: 10.1210/jc.2013-3636] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
CONTEXT Development of novel strategies in the treatment of advanced thyroid cancer are needed. Our laboratory has previously identified a role for nuclear factor κB (NF-κB) signaling in human thyroid cancer cell growth, survival, and invasion. OBJECTIVE Our goal was to establish the role of NF-κB signaling on thyroid cancer growth and metastases in vivo and to begin to dissect mechanisms regulating this effect. SETTING AND DESIGN We examined tumor formation of five thyroid cancer cell lines in an in vivo model of thyroid cancer and observed tumor establishment in two of the cell lines (8505C and BCPAP). RESULTS Inhibition of NF-κB signaling by overexpression of a dominant-negative IκBα (mIκBα) significantly inhibited thyroid tumor growth in tumors derived from both cell lines. Further studies in an experimental metastasis model demonstrated that NF-κB inhibition impaired growth of tumor metastasis and prolonged mouse survival. Proliferation (mitotic index) was decreased in 8505C tumors, but not in BCPAP tumors, while in vitro angiogenesis and in vivo tumor vascularity were significantly inhibited by mIkBα only in the BCPAP cells. Cytokine antibody array analysis demonstrated that IL-8 secretion was blocked by mIκBα expression. Interestingly, basal NF-κB activity and IL-8 levels were significantly higher in the two tumorigenic cell lines compared with the nontumorigenic lines. Furthermore, IL-8 transcript levels were elevated in high-risk human tumors, suggesting that NF-κB and IL-8 are associated with more aggressive tumor behavior. CONCLUSIONS These studies suggest that NF-κB signaling is a key regulator of angiogenesis and growth of primary and metastatic thyroid cancer, and that IL-8 may be an important downstream mediator of NF-κB signaling in advanced thyroid cancer growth and progression.
Collapse
Affiliation(s)
- Kevin T Bauerle
- Departments of Medicine and Pathology, Division of Endocrinology, Metabolism, and Diabetes (K.T.B., R.E.S., G.L., N.P., W.M.W., B.R.H.), University of Colorado School of Medicine, Aurora, Colorado 80045; University of Colorado Cancer Center (R.E.S., G.D., R.A., W.M.W., B.R.H.), University of Colorado School of Medicine, Aurora, Colorado 80045; CCF Pathology and Laboratory Medicine Institute (G.K.), Cleveland, Ohio 44120; and Skaggs School of Pharmacy and Pharmaceutical Sciences (G.K., R.A.), University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado 80045
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Vyas D, Laput G, Vyas AK. Chemotherapy-enhanced inflammation may lead to the failure of therapy and metastasis. Onco Targets Ther 2014; 7:1015-23. [PMID: 24959088 PMCID: PMC4061164 DOI: 10.2147/ott.s60114] [Citation(s) in RCA: 221] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The lack of therapy and the failure of existing therapy has been a challenge for clinicians in treating various cancers. Doxorubicin, 5-fluorouracil, cisplatin, and paclitaxel are the first-line therapy in various cancers; however, toxicity, resistance, and treatment failure limit their clinical use. Their status leads us to discover and investigate more targeted therapy with more efficacy. In this article, we dissect literature from the patient perspective, the tumor biology perspective, therapy-induced metastasis, and cell data generated in the laboratory.
Collapse
Affiliation(s)
- Dinesh Vyas
- College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - Gieric Laput
- College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - Arpitak K Vyas
- College of Human Medicine, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
50
|
CXC and CC chemokines as angiogenic modulators in nonhaematological tumors. BIOMED RESEARCH INTERNATIONAL 2014; 2014:768758. [PMID: 24971349 PMCID: PMC4058128 DOI: 10.1155/2014/768758] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 04/08/2014] [Indexed: 12/26/2022]
Abstract
Chemokines are a superfamily of structurally homologous heparin-binding proteins that includes potent inducers and inhibitors of angiogenesis. The imbalance between angiogenic and angiostatic chemokine activities can lead to abnormalities, such as chronic inflammation, dysplastic transformation, and even tumor development and spreading. In this review, we summarize the current literature regarding the role of chemokines as modulators of tumor angiogenesis and their potential role as therapeutic targets in patients with nonhaematological tumors.
Collapse
|