1
|
Khalighfard S, Khori V, Esmati E, Ahmadi F, Amiriani T, Poorkhani A, Sadani S, Khodayari S, Khodayari H, Kalhori MR, Keshavarz P, Alizadeh AM. Breast tumor metastasis following filgrastim administration due to the SDF-1/CXCR4 pathway. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2023; 40:74. [PMID: 36609711 DOI: 10.1007/s12032-022-01935-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/07/2022] [Indexed: 01/07/2023]
Abstract
Filgrastim, a recombinant type of granulocyte-colony stimulating factor (G-CSF), has a high potential to manage chemotherapy-induced leukopenia. It can increase stromal cell-derived factor 1 (SDF-1) which may stimulate C-X-C chemokine receptor type 4 (CXCR4) to migrate bone marrow-derived stem/progenitor cells to the bloodstream. Here, we aimed to investigate in vitro and in vivo effects of filgrastim on cell migration, invasion, and metastasis. A lentivirus vector of the anti-CXCR4 receptor was first used for the CXCR4 knockout. Effects of filgrastim on cell proliferation and migration were then investigated on 4T1 cells by Transwell migration and wound healing assay. At last, the effects of filgrastim on cell metastasis and the possible involved mechanisms have been investigated in a metastatic murine breast tumor. The knockout of the CXCR4 receptor could lead to a decrease in cell proliferation, migration, and invasion of the 4T1 cells. Filgrastim could directly target SDF-1 and upregulate the expression of the CXCR4 receptor. The knockout of the CXCR4 receptor reduced cell metastasis in an animal model of breast cancer. CXCR4 receptor stimulation by the filgrastim-affected pathways is a conserved evolutionary response that could increase cancer cell proliferation and consequent cell metastasis. Our results suggest that the activation of the CXCR4 receptor is a conserved evolutionary response that can increase cell proliferation, migration, and consequent metastasis. It seems that filgrastim may increase the chance of cancer cell metastasis in people continuously receiving it to increase the number of neutrophils. Filgrastim induces the SDF-1/CXCR4 axis on tumor cell growth. SDF-1 and its receptor CXCR4 are vital targets for filgrastim. The CXCR4 can stimulate the PI3K/AKT, NF-κB, and JAK/STAT signaling pathways. The SDF-1/CXCR4 pathway promotes cell chemotaxis and proliferation via MAPKs signaling. It also enhances cell survival, proliferation, and angiogenesis, increasing tumor cell metastasis. The STAT3-mediated inflammation is essential for tumorigenesis processes, and Akt, Wnt, STAT3, and CXCR4 signaling pathways are all correlated. CXCR4 = C-X-C chemokine receptor type 4, SDF-1 = stromal-derived-factor-1, MAPK = mitogen activated protein kinase; NF-κB = nuclear factor-κB, PI3K = phosphoinositide 3-kinase, JAK = Janus kinase, STAT = signal transducer and activator of transcription, PLC = phospholipase C, PKC = Protein kinase C, GRK = G protein-coupled receptor kinase.
Collapse
Affiliation(s)
- Solmaz Khalighfard
- Radiation Oncology Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran.,Research Center on Developing Advanced Technologies, Tehran, Iran
| | - Vahid Khori
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ebrahim Esmati
- Radiation Oncology Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Farahnazsadat Ahmadi
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Taghi Amiriani
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Amirhoushang Poorkhani
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Somayeh Sadani
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Saeed Khodayari
- International Center for Personalized Medicine (ICPM), Düsseldorf, Germany
| | - Hamid Khodayari
- International Center for Personalized Medicine (ICPM), Düsseldorf, Germany
| | - Mohammad Reza Kalhori
- Regenerative Medicine Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Pedram Keshavarz
- Department of Radiology, Tbilisi State Medical University (TSMU), Tbilisi, Georgia
| | - Ali Mohammad Alizadeh
- Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Marques CS, Santos AR, Gameiro A, Correia J, Ferreira F. CXCR4 and its ligand CXCL12 display opposite expression profiles in feline mammary metastatic disease, with the exception of HER2-overexpressing tumors. BMC Cancer 2018; 18:741. [PMID: 30012106 PMCID: PMC6048851 DOI: 10.1186/s12885-018-4650-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 06/29/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The receptor CXCR4 and its ligand CXCL12 play crucial roles in breast cancer. Despite the fact that the spontaneous feline mammary carcinoma (FMC) is considered a suitable model for breast cancer studies, the importance of the CXCR4/CXCL12 axis in FMC is completely unknown. Therefore, this work aims to elucidate the role of CXCR4 and its ligand in the progression of FMC and metastatic disease. METHODS CXCR4 and CXCL12 expression was analyzed by immunohistochemistry and immunofluorescence on primary tumors (PT), regional and distant metastases of female cats with mammary carcinoma and correlated with serum CXCL12 levels, tumor molecular subtypes and clinicopathological features. RESULTS CXCR4 was more expressed in PT than in metastases (p = 0.0067), whereas CXCL12 was highly expressed in metastatic lesions located in liver and lung (p < 0.0001), as reported for human breast cancer. Moreover, cats with CXCR4 positive PT exhibited significantly lower serum CXCL12 levels than cats with CXCR4 negative mammary carcinomas (p = 0.0324). At metastatic lesions, HER2-overexpressing tumors presented higher CXCR4 expression than the other molecular tumor subtypes (p = 0.012) and significant differences in overall (p = 0.0147) and disease-free survival (p = 0.0279) curves between the cats with CXCL12 positive and CXCL12 negative tumors were found. Indeed, CXCL12 negative PT were associated with unfavorable prognosis in cats with HER2-overexpressing tumors. CONCLUSIONS This work exposes part of the complex interaction between CXCR4 and CXCL12 in PT, but also in metastases of a breast cancer model. These findings could uncover novel therapeutic tools to be used in cats and humans.
Collapse
Affiliation(s)
- Cláudia S. Marques
- Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, 1300-477 Lisbon, Portugal
| | - Ana Rita Santos
- Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, 1300-477 Lisbon, Portugal
| | - Andreia Gameiro
- Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, 1300-477 Lisbon, Portugal
| | - Jorge Correia
- Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, 1300-477 Lisbon, Portugal
| | - Fernando Ferreira
- Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, 1300-477 Lisbon, Portugal
| |
Collapse
|
3
|
Wang J, Huang Y, Zhang J, Xing B, Xuan W, Wang H, Huang H, Yang J, Tang J. High co-expression of the SDF1/CXCR4 axis in hepatocarcinoma cells is regulated by AnnexinA7 in vitro and in vivo. Cell Commun Signal 2018; 16:22. [PMID: 29783989 PMCID: PMC5963093 DOI: 10.1186/s12964-018-0234-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 05/15/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND SDF1/CXCR4 and AnnexinA7 play important roles in many physiological and pathological conditions, but the molecular association between them in cancer cells has not been studied thus far. METHODS The expression changes of SDF1/CXCR4 were detected by gene transcriptome sequencing, qRT-PCR, Western blotting, cytoimmunofluorescence and immunohistochemistry in mouse hepatocarcinoma F/P cells, AnnexinA7 downregulated expression F (FA7DOWN) cells, AnnexinA7 overexpression P (PA7UP) cells, AnnexinA7 unrelated sequence F (FSHUS) cells, empty vector P (PNCEV) cells and normal liver cells in vitro and in vivo. RESULTS SDF1 and CXCR4 were co-expressed in hepatocarcinoma cells. SDF1 was localized mainly in the cytoplasm of cells, while CXCR4 was mainly localized in the cell membrane. Both in vitro and in vivo, expression levels of SDF1/CXCR4 in F and P cells were higher than in normal liver cells, and expression levels of SDF1/CXCR4 in F cells with high lymphatic metastatic potential were higher than those in P cells with low lymphatic metastatic potential. Expression of SDF1 was higher than that of CXCR4 in P cells and normal liver cells, while expression of CXCR4 was higher than that of SDF1 in F cells. Expression levels of SDF1/CXCR4 were completely consistent with AnnexinA7 regulation. After the AnnexinA7 gene was downregulated or upregulated, expression levels of SDF1/CXCR4 in FA7DOWN/PA7UP cells were lower or higher than those in FSHUS/PNCEV cells. Furthermore, CXCR4 was more sensitively modulated by AnnexinA7 regulation than SDF1. CONCLUSIONS High co-expression of SDF1/CXCR4 is a molecular characteristic of hepatocarcinoma cells, especially those with high lymphatic metastatic potential. AnnexinA7 positively regulates expression levels of SDF1/CXCR4, in particular CXCR4, and AnnexinA7 is a functional regulator of SDF1/CXCR4.
Collapse
Affiliation(s)
- Jingwen Wang
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis and Intervention of Liaoning Province, 9 West, Lvshun Southern Road, Dalian, 116044, Liaoning, China
| | - Yuhong Huang
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis and Intervention of Liaoning Province, 9 West, Lvshun Southern Road, Dalian, 116044, Liaoning, China
| | - Jun Zhang
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis and Intervention of Liaoning Province, 9 West, Lvshun Southern Road, Dalian, 116044, Liaoning, China
| | - Boyi Xing
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis and Intervention of Liaoning Province, 9 West, Lvshun Southern Road, Dalian, 116044, Liaoning, China
| | - Wei Xuan
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis and Intervention of Liaoning Province, 9 West, Lvshun Southern Road, Dalian, 116044, Liaoning, China
| | - Honghai Wang
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis and Intervention of Liaoning Province, 9 West, Lvshun Southern Road, Dalian, 116044, Liaoning, China
| | - He Huang
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis and Intervention of Liaoning Province, 9 West, Lvshun Southern Road, Dalian, 116044, Liaoning, China
| | | | - Jianwu Tang
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis and Intervention of Liaoning Province, 9 West, Lvshun Southern Road, Dalian, 116044, Liaoning, China.
| |
Collapse
|
4
|
Xu L, Liang Z, Li S, Ma J. Signaling via the CXCR5/ERK pathway is mediated by CXCL13 in mice with breast cancer. Oncol Lett 2018; 15:9293-9298. [PMID: 29844827 DOI: 10.3892/ol.2018.8510] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 01/10/2018] [Indexed: 01/01/2023] Open
Abstract
Breast cancer is the most common cause of cancer-associated mortality and the most frequently diagnosed type of cancer in women worldwide. It has been revealed that the chemokine C-X-C motif chemokine ligand 13 (CXCL13) serves a pivotal role in breast cancer growth and is associated with lymph node metastasis. However, to the best of our knowledge, the mechanism by which CXCL13 mediates breast cancer growth remains uncharacterized. Female BALB/c mice were used in this study. Tumor volume was calculated and changes of gross tumor morphology were observed by hematoxylin and eosin staining. The expression of CXCL13, C-X-C motif chemokine receptor 5 (CXCR5) and extracellular signaling-related kinase (ERK) mRNA and protein expression were detected by reverse transcriptase quantitative-polymerase chain reaction and western blot analysis. Simultaneously, the production of cytokines [interleukin-1β (IL-1β), tumor necrosis factor (TNF) and tumor growth factor β1 (TGF-β1)] was detected by an ELISA. The CXCL13 inhibitor reduced tumor volume and growth, and reduced the mRNA and protein expression levels of key members of the CXCR5/ERK signaling pathway: CXCL13, CXCR5 and ERK. Furthermore, the detectable concentration of the cytokines IL-1β and TNF decreased following CXCL13 inhibition, whereas the concentration of TGF-β1 was increased. The attenuation of tumor growth resulting from CXCL13 inhibition may be associated with the CXCR5/ERK signaling pathway. This study provides a theoretical basis for treating breast cancer through CXCL13 inhibition in clinical trials.
Collapse
Affiliation(s)
- Licheng Xu
- Department of Breast Surgery, Yantaishan Hospital, Yantai, Shandong 264002, P.R. China
| | - Zhi Liang
- Department of Breast Surgery, Yantaishan Hospital, Yantai, Shandong 264002, P.R. China
| | - Shuyan Li
- Department of Breast Surgery, Yantaishan Hospital, Yantai, Shandong 264002, P.R. China
| | - Jianjun Ma
- Department of Medical Oncology, The People's Liberation Army 107th Hospital, Yantai, Shandong 264002, P.R. China
| |
Collapse
|
5
|
Reker D, Schneider P, Schneider G. Multi-objective active machine learning rapidly improves structure-activity models and reveals new protein-protein interaction inhibitors. Chem Sci 2016; 7:3919-3927. [PMID: 30155037 PMCID: PMC6013791 DOI: 10.1039/c5sc04272k] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 02/27/2016] [Indexed: 11/21/2022] Open
Abstract
Active machine learning puts artificial intelligence in charge of a sequential, feedback-driven discovery process. We present the application of a multi-objective active learning scheme for identifying small molecules that inhibit the protein-protein interaction between the anti-cancer target CXC chemokine receptor 4 (CXCR4) and its endogenous ligand CXCL-12 (SDF-1). Experimental design by active learning was used to retrieve informative active compounds that continuously improved the adaptive structure-activity model. The balanced character of the compound selection function rapidly delivered new molecular structures with the desired inhibitory activity and at the same time allowed us to focus on informative compounds for model adjustment. The results of our study validate active learning for prospective ligand finding by adaptive, focused screening of large compound repositories and virtual compound libraries.
Collapse
Affiliation(s)
- D Reker
- Department of Chemistry and Applied Biosciences , ETH Zürich , Vladimir-Prelog Weg 4 , 8093 Zürich , Switzerland .
| | - P Schneider
- Department of Chemistry and Applied Biosciences , ETH Zürich , Vladimir-Prelog Weg 4 , 8093 Zürich , Switzerland .
| | - G Schneider
- Department of Chemistry and Applied Biosciences , ETH Zürich , Vladimir-Prelog Weg 4 , 8093 Zürich , Switzerland .
| |
Collapse
|
6
|
Immunohistochemical expression of CXCR4 on breast cancer and its clinical significance. Anal Cell Pathol (Amst) 2015; 2015:891020. [PMID: 26161302 PMCID: PMC4486754 DOI: 10.1155/2015/891020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 06/03/2015] [Accepted: 06/08/2015] [Indexed: 12/26/2022] Open
Abstract
Many tumor cells express chemokines and chemokine receptors, and, for this reason, these molecules can affect the tumor progression. It is known that breast cancer is a complex and heterogeneous neoplasia comprising distinct diseases, histological characteristics, and clinical outcomes. The most studied role for CXCL12 chemokine and its receptor CXCR4 in breast cancer pathogenesis is the metastasis event, although several reports have demonstrated its involvement in other processes, such as angiogenesis and tumor growth. It has been found that CXCR4 is required for breast cancer cell migration to other sites such as lung, bone, and lymph nodes, which express high levels of CXCL12 chemokine. Therefore, CXCR4 is being considered a prognostic marker in breast cancer. Within this context, this review summarizes established studies involving expression of CXCR4 on breast cancer, focusing on its clinical significance.
Collapse
|