1
|
Zhou X, Liang W, Hong L, Gong S, Liu Z, Li W, Cao N, Tian Y, Xu D, Li B. Transcriptome analysis reveals the alleviating effect of Polysaccharide of Atractylodes macrocephala Koidz on thymic involution in Magang geese. Poult Sci 2025; 104:105155. [PMID: 40245540 DOI: 10.1016/j.psj.2025.105155] [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: 02/05/2025] [Revised: 04/03/2025] [Accepted: 04/11/2025] [Indexed: 04/19/2025] Open
Abstract
Thymic involution is one of the important causes of decreased immunity in the body. Noncoding RNAs (miRNAs and lncRNAs) play crucial roles in regulating organ growth and development. Polysaccharide of Atractylodes macrocephala Koidz (PAMK) is widely acknowledged for its anti-oxidant, anti-aging, and immune-enhancing effects. However, its potential application in preventing the age-related thymic involution of Magang geese has not been previously reported. In this study, 54 4-month-old Magang geese were randomly divided into 3 groups, the thymus and serum of 18 geese were collected aseptically after 3 days of prefeeding period, and the remaining geese were randomly divided into control and PAMK groups (3 replicates per group and 6 Magang geese per replicate). Geese in the control group were fed a basal diet, and geese in the PAMK group were fed a basal diet supplemented with 400 mg/kg PAMK. The thymus and serum were collected 1 month later. The results of thymus index measurement showed that PAMK could alleviate thymus index. Furthermore, compared with the M5-Control group, HE staining showed that PAMK made the proportion of thymus medulla increased, and the boundary between cortex and medulla was clearer. Antioxidant function and cytokine content detection showed that, compared with the M5-Control group, PAMK increased T-AOC and GSH-Px levels in thymus, increased T-AOC level and SOD activity in serum, decreased MDA content in thymus and serum, and decreased IL-1β, IL-6 and TNF-α levels. To further explore the mechanism, 3 samples from the control and PAMK groups were selected for RNA-Seq. Through transcriptome analysis and prediction, a triple regulatory ceRNA network of 9 mRNAs, 11 miRNAs and 32 lncRNAs associated with alleviating thymic involution was constructed. Moreover, these genes were respectively enriched in the PPAR, Cytokine-cytokine receptor interaction, WNT, Apelin and MAPK signaling pathways. In summary, PAMK may alleviate age-related thymic involution in Magang geese by alleviating the thymus index, increasing the antioxidant level and regulating the cytokine content, potentially via the PPAR, Cytokine-cytokine receptor interaction, WNT, Apelin, and MAPK signaling pathways.
Collapse
Affiliation(s)
- Xiang Zhou
- Science & Technology Innovation Platform of Guangdong Waterfowl, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Weijun Liang
- Science & Technology Innovation Platform of Guangdong Waterfowl, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Longsheng Hong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Shuying Gong
- Science & Technology Innovation Platform of Guangdong Waterfowl, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Zhuokun Liu
- Science & Technology Innovation Platform of Guangdong Waterfowl, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Wanyan Li
- Science & Technology Innovation Platform of Guangdong Waterfowl, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Nan Cao
- Science & Technology Innovation Platform of Guangdong Waterfowl, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Yunbo Tian
- Science & Technology Innovation Platform of Guangdong Waterfowl, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Danning Xu
- Science & Technology Innovation Platform of Guangdong Waterfowl, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Bingxin Li
- Science & Technology Innovation Platform of Guangdong Waterfowl, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China.
| |
Collapse
|
2
|
Jiang J, Cheng R, Song A, Lou Y, Fan G. Multi-omics analysis reveals mechanism of Schisandra chinensis lignans and acteoside on EMT in hepatoma cells via ERK1/2 pathway. Funct Integr Genomics 2024; 24:112. [PMID: 38849609 DOI: 10.1007/s10142-024-01351-w] [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: 12/31/2023] [Revised: 03/08/2024] [Accepted: 03/28/2024] [Indexed: 06/09/2024]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC), a globally common cancer, often presents late and shows high resistance to chemotherapy, resulting in suboptimal treatment efficacy. Components from traditional Chinese medicines have been recognized for their anti-cancer properties. OBJECTIVE Exploring the mechanism of Schisandra chinensis lignans and acteoside in suppressing Epithelial-Mesenchymal Transition (EMT) in hepatoma cells through the Extracellular signal-Regulated Kinases (ERK)1/2 pathway and identifying biomarkers, molecular subtypes, and targets via multi-omics for precision oncology. METHODS Proliferation was assessed using cell counting kit-8 (CCK-8) assays, with scratch and transwell assays for evaluating invasion and migration. Flow cytometry quantified apoptosis rates. Expression levels of CCL20, p-ERK1/2, c-Myc, Vimentin, and E-cadherin/N-cadherin were analyzed by real-time PCR and Western blot. Tumor volume was calculated with a specific formula, and growth. RESULTS The Schisandra chinensis lignans and acteoside combination decreased CCL20 expression, inhibited hepatoma proliferation and migration, and enhanced apoptosis in a dose- and time-dependent manner. Molecular analysis revealed increased E-cadherin and decreased N-cadherin, p-ERK1/2, c-Myc, and Vimentin expression, indicating ERK1/2 pathway modulation. In vivo, treated nude mice showed significantly reduced tumor growth and volume. CONCLUSION Schisandra chinensis lignans and acteoside potentially counteract CCL20-induced EMT, invasion, and migration in hepatocellular carcinoma cells via the ERK1/2 pathway, enhancing apoptosis. Multi-omics analysis further aids in pinpointing novel biomarkers for precision cancer therapy.
Collapse
Affiliation(s)
- Jingjing Jiang
- Department of Pharmacy, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
| | - Ru Cheng
- Department of Pharmacy, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
| | - Aoqi Song
- Department of Pharmacy, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
| | - Yuefen Lou
- Department of Pharmacy, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China.
| | - Guorong Fan
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
3
|
Wu Z, Sun L, Xu Y, Huang H, Wu Z, Qiu B, Yan J, Yin X. The Value of Chemokine and Chemokine Receptors in Diagnosis, Prognosis, and Immunotherapy of Hepatocellular Carcinoma. Cancer Manag Res 2024; 16:403-420. [PMID: 38736589 PMCID: PMC11086648 DOI: 10.2147/cmar.s450959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/20/2024] [Indexed: 05/14/2024] Open
Abstract
Background Chemokines and chemokine receptors (CCRs) are involved in a variety of anti-tumour and pro-tumour immune processes in vivo, such as angiogenesis, metastasis, proliferation and invasiveness, and influence patient prognosis and response to therapy. Methods CCRs differentially expressed in HCC and associated with prognosis were extracted from TCGA and GEO databases, and the obtained CCRs were then used to construct signature genes, and the signature gene were selected for expression validation as well as functional experiments to explore the role of CCRs in the treatment and prognosis of HCC. Results We constructed a prognostic model including five CCRs (CCL20, CCL23, CCR3, CCR10, and CXCR3) and validated the expression of signature genes. The model's risk score is an independent prognostic factor for HCC. We have also developed prognostic model nomograms for clinical use. In addition, we validated that CCR3 expression is associated with poor prognosis in HCC, and the proliferation and migration ability of HCC cells was significantly inhibited after interfering with the expression of CCR3 in MHCC-LM3. We also looked at differences in pathway enrichment, immune infiltration and immune checkpoints. Finally, we found that risk scores were also correlated with drug sensitivity, the high-risk group had a better sensitivity to sorafenib. Conclusion The CCRs-related gene signature may better assess HCC prognosis and response to immunotherapy and tyrosine kinase inhibitors such as sorafenib in HCC, providing prospective solutions for diagnosis and treatment.
Collapse
Affiliation(s)
- Zhengyi Wu
- Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People’s Republic of China
| | - Liang Sun
- Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People’s Republic of China
| | - Yongkang Xu
- Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People’s Republic of China
| | - He Huang
- Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People’s Republic of China
| | - Zhipeng Wu
- Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People’s Republic of China
| | - Bingbing Qiu
- Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People’s Republic of China
| | - Jinlong Yan
- Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People’s Republic of China
| | - Xiangbao Yin
- Hepatobiliary Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People’s Republic of China
| |
Collapse
|
4
|
Zhai H, Zhang N, Mo D, Qin T. CCL20 is a potential therapeutic target associated with immune infiltration in breast cancer. J Int Med Res 2023; 51:3000605231171762. [PMID: 37571985 PMCID: PMC10423453 DOI: 10.1177/03000605231171762] [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: 08/10/2022] [Accepted: 04/07/2023] [Indexed: 08/14/2023] Open
Abstract
OBJECTIVES CCL20 is a chemotactic factor that is involved in immune cell recruitment and cancer progression. However, the role of CCL20 in the prognosis of breast cancer remains unclear. This study analyzed correlations between CCL20 expression and immune infiltration, clinicopathological parameters, and prognosis in breast cancer patients. METHODS Correlations between CCL20 expression and clinicopathological parameters, prognosis, and immune infiltration in breast cancer were determined using the TIMER, UALCAN, and PrognoScan databases. Furthermore, gene-gene and protein-protein interactions were determined using GeneMANIA and STING network construction, respectively. RESULTS CCL20 expression was significantly upregulated in breast cancer and had significant associations with clinicopathological features, including race, sex, age, menopause status, cancer stage, cancer subclass, and nodal metastasis; moreover, patients with higher CCL20 expression exhibited poor prognosis. Meanwhile, CCL20 expression was significantly correlated with the infiltration of immune cells in breast cancer, including monocytes, neutrophils, tumor-associated macrophages, Th1 cells, regulatory T cells, and exhausted T cells. Moreover, the network of CCL20 expression showed the majority genes and proteins were associated with immune reactions. CONCLUSIONS CCL20 is a prognosis-related biomarker in breast cancer on the basis of its correlation with immune infiltration levels and has potential to also be a therapeutic target.
Collapse
Affiliation(s)
- Hongyan Zhai
- Department of Oncology, Linfen People's Hospital, Linfen, China
| | - Na Zhang
- Department of Oncology, Linfen People's Hospital, Linfen, China
| | - Dan Mo
- Department of Galactophore, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Tingting Qin
- Department of Integrated Traditional Chinese and Western Medicine, Wuhan Third Hospital, Wuhan, China
| |
Collapse
|
5
|
Zhang Y, Liu D, Vithran DTA, Kwabena BR, Xiao W, Li Y. CC chemokines and receptors in osteoarthritis: new insights and potential targets. Arthritis Res Ther 2023; 25:113. [PMID: 37400871 PMCID: PMC10316577 DOI: 10.1186/s13075-023-03096-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/23/2023] [Indexed: 07/05/2023] Open
Abstract
Osteoarthritis (OA) is a prevalent degenerative disease accompanied by the activation of innate and adaptive immune systems-associated inflammatory responses. Due to the local inflammation, the expression of various cytokines was altered in affected joints, including CC motif chemokine ligands (CCLs) and their receptors (CCRs). As essential members of chemokines, CCLs and CCRs played an important role in the pathogenesis and treatment of OA. The bindings between CCLs and CCRs on the chondrocyte membrane promoted chondrocyte apoptosis and the release of multiple matrix-degrading enzymes, which resulted in cartilage degradation. In addition, CCLs and CCRs had chemoattractant functions to attract various immune cells to osteoarthritic joints, further leading to the aggravation of local inflammation. Furthermore, in the nerve endings of joints, CCLs and CCRs, along with several cellular factors, contributed to pain hypersensitivity by releasing neurotransmitters in the spinal cord. Given this family's diverse and complex functions, targeting the functional network of CCLs and CCRs is a promising strategy for the prognosis and treatment of OA in the future.
Collapse
Affiliation(s)
- Yuchen Zhang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Di Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | | | - Bosomtwe Richmond Kwabena
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Wenfeng Xiao
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
6
|
Gao FY, Li XT, Xu K, Wang RT, Guan XX. c-MYC mediates the crosstalk between breast cancer cells and tumor microenvironment. Cell Commun Signal 2023; 21:28. [PMID: 36721232 PMCID: PMC9887805 DOI: 10.1186/s12964-023-01043-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 01/08/2023] [Indexed: 02/01/2023] Open
Abstract
The MYC oncogenic family is dysregulated in diverse tumors which is generally linked to the poor prognosis of tumors. The members in MYC family are transcription factors which are responsible for the regulation of various genes expression. Among them, c-MYC is closely related to the progression of tumors. Furthermore, c-MYC aberrations is tightly associated with the prevalence of breast cancer. Tumor microenvironment (TME) is composed of many different types of cellular and non-cellular factors, mainly including cancer-associated fibroblasts, tumor-associated macrophages, vascular endothelial cells, myeloid-derived suppressor cells and immune cells, all of which can affect the diagnosis, prognosis, and therapeutic efficacy of breast cancer. Importantly, the biological processes occurred in TME, such as angiogenesis, immune evasion, invasion, migration, and the recruition of stromal and tumor-infiltrating cells are under the modulation of c-MYC. These findings indicated that c-MYC serves as a critical regulator of TME. Here, we aimed to summarize and review the relevant research, thus to clarify c-MYC is a key mediator between breast cancer cells and TME. Video Abstract.
Collapse
Affiliation(s)
- Fang-yan Gao
- grid.412676.00000 0004 1799 0784Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Xin-tong Li
- grid.412676.00000 0004 1799 0784Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Kun Xu
- grid.412676.00000 0004 1799 0784Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Run-tian Wang
- grid.412676.00000 0004 1799 0784Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Xiao-xiang Guan
- grid.412676.00000 0004 1799 0784Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| |
Collapse
|
7
|
Xiong J, Bao J, Hu W, Shang M, Zhang L. Whole-genome resequencing reveals genetic diversity and selection characteristics of dairy goat. Front Genet 2023; 13:1044017. [PMID: 36685859 PMCID: PMC9852865 DOI: 10.3389/fgene.2022.1044017] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 12/13/2022] [Indexed: 01/09/2023] Open
Abstract
The dairy goat is one of the earliest dairy livestock species, which plays an important role in the economic development, especially for developing countries. With the development of agricultural civilization, dairy goats have been widely distributed across the world. However, few studies have been conducted on the specific characteristics of dairy goat. In this study, we collected the whole-genome data of 89 goat individuals by sequencing 48 goats and employing 41 publicly available goats, including five dairy goat breeds (Saanen, Nubian, Alpine, Toggenburg, and Guanzhong dairy goat; n = 24, 15, 11, 6, 6), and three goat breeds (Guishan goat, Longlin goat, Yunshang Black goat; n = 6, 15, 6). Through compared the genomes of dairy goat and non-dairy goat to analyze genetic diversity and selection characteristics of dairy goat. The results show that the eight goats could be divided into three subgroups of European, African, and Chinese indigenous goat populations, and we also found that Australian Nubian, Toggenburg, and Australian Alpine had the highest linkage disequilibrium, the lowest level of nucleotide diversity, and a higher inbreeding coefficient, indicating that they were strongly artificially selected. In addition, we identified several candidate genes related to the specificity of dairy goat, particularly genes associated with milk production traits (GHR, DGAT2, ELF5, GLYCAM1, ACSBG2, ACSS2), reproduction traits (TSHR, TSHB, PTGS2, ESR2), immunity traits (JAK1, POU2F2, LRRC66). Our results provide not only insights into the evolutionary history and breed characteristics of dairy goat, but also valuable information for the implementation and improvement of dairy goat cross breeding program.
Collapse
|
8
|
Ma Y, Zhou Y, Zou SS, Sun Y, Chen XF. OUP accepted manuscript. Mol Hum Reprod 2022; 28:6516534. [PMID: 35088858 DOI: 10.1093/molehr/gaac002] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/17/2022] [Indexed: 11/14/2022] Open
Affiliation(s)
- Yi Ma
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yan Zhou
- Department of Central Lab, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | | | - Yun Sun
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Xiang-Feng Chen
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
- Shanghai Human Sperm Bank, Shanghai, China
| |
Collapse
|
9
|
CCL20 induces colorectal cancer neoplastic epithelial cell proliferation, migration, and further CCL20 production through autocrine HGF-c-Met and MSP-MSPR signaling pathways. Oncotarget 2021; 12:2323-2337. [PMID: 34853656 PMCID: PMC8629403 DOI: 10.18632/oncotarget.28131] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 09/28/2021] [Indexed: 12/16/2022] Open
Abstract
CCL20-CCR6 interactions promote colorectal cancer through direct effects on neoplastic epithelial cells and through modulating the tumor microenvironment. The mechanism of these effects on neoplastic epithelial cells is poorly understood. This study demonstrates that CCL20 induces secretion of hepatocyte growth factor (HGF) and phosphorylation of HGF’s cognate receptor c-Met in HT29 and HCT116 colorectal cancer cell lines both in concentration- and time-dependent manners. Similar to CCL20, HGF induces migration, autofeedback CCL20 secretion, and ERK1/2 phosphorylation in the colon cancer cells. CCL20-dependent ERK1/2 phosphorylation is blocked by HGF inhibition, and CCL20-dependent migration and CCL20 secretion are blocked by inhibition of HGF or ERK. Interestingly, unlike CCL20, HGF does not induce proliferation of colon cancer cells, and CCL20-dependent cell proliferation is not blocked by direct HGF inhibition. CCL20-dependent proliferation, however, is blocked by the multi-tyrosine kinase inhibitor crizotinib. Exploring this effect, it was found that CCL20 also induces production of MSP and phosphorylation of MSP’s receptor MSPR by the colorectal cancer cells. CCL20-dependent cell proliferation is inhibited by directly blocking MSP-MSPR interactions. Thus, CCL20-mediated migration and CCL20 secretion are regulated through a pathway involving HGF, c-Met, and ERK, while CCL20-mediated proliferation is instead regulated through MSP and its receptor MSPR.
Collapse
|
10
|
Recruitment and Expansion of Tregs Cells in the Tumor Environment-How to Target Them? Cancers (Basel) 2021; 13:cancers13081850. [PMID: 33924428 PMCID: PMC8069615 DOI: 10.3390/cancers13081850] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/04/2021] [Accepted: 04/08/2021] [Indexed: 12/22/2022] Open
Abstract
Simple Summary The immune response against cancer is generated by effector T cells, among them cytotoxic CD8+ T cells that destroy cancer cells and helper CD4+ T cells that mediate and support the immune response. This antitumor function of T cells is tightly regulated by a particular subset of CD4+ T cells, named regulatory T cells (Tregs), through different mechanisms. Even if the complete inhibition of Tregs would be extremely harmful due to their tolerogenic role in impeding autoimmune diseases in the periphery, the targeted blockade of their accumulation at tumor sites or their targeted depletion represent a major therapeutic challenge. This review focuses on the mechanisms favoring Treg recruitment, expansion and stabilization in the tumor microenvironment and the therapeutic strategies developed to block these mechanisms. Abstract Regulatory T cells (Tregs) are present in a large majority of solid tumors and are mainly associated with a poor prognosis, as their major function is to inhibit the antitumor immune response contributing to immunosuppression. In this review, we will investigate the mechanisms involved in the recruitment, amplification and stability of Tregs in the tumor microenvironment (TME). We will also review the strategies currently developed to inhibit Tregs’ deleterious impact in the TME by either inhibiting their recruitment, blocking their expansion, favoring their plastic transformation into other CD4+ T-cell subsets, blocking their suppressive function or depleting them specifically in the TME to avoid severe deleterious effects associated with Treg neutralization/depletion in the periphery and normal tissues.
Collapse
|
11
|
Shen D, Tian L, Yang F, Li J, Li X, Yao Y, Lam EWF, Gao P, Jin B, Wang R. ADO/hypotaurine: a novel metabolic pathway contributing to glioblastoma development. Cell Death Discov 2021; 7:21. [PMID: 33483477 PMCID: PMC7822925 DOI: 10.1038/s41420-020-00398-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/09/2020] [Accepted: 12/15/2020] [Indexed: 01/07/2023] Open
Abstract
Significant advance has been made towards understanding glioblastoma metabolism through global metabolomic profiling. However, hitherto little is known about the role by which altered metabolism plays in driving the aggressive glioma phenotype. We have previously identified hypotaurine as one of the top-ranked metabolites for differentiating low- and high-grade tumors, and that there is also a strong association between the levels of intratumoral hypotaurine and expression of its biosynthetic enzyme, cysteamine (2-aminoethanethiol) dioxygenase (ADO). Using transcription profiling, we further uncovered that the ADO/hypotaurine axis targets CCL20 secretion through activating the NF-κB pathway to drive the self-renewal and maintenance of glioma 'cancer stem cells' or glioma cancer stem-like cells. Conversely, abrogating the ADO/hypotaurine axis using CRISPR/Cas9-mediated gene editing limited glioblastoma cell proliferation and self-renewal in vitro and tumor growth in vivo in an orthotopical mouse model, indicating that this metabolic pathway is a potential key therapeutic target. Collectively, our results unveil a targetable metabolic pathway, which contributes to the growth and progression of aggressive high-grade gliomas, as well as a novel predictive marker for glioblastoma diagnosis and therapy.
Collapse
Affiliation(s)
- Dachuan Shen
- Department of Oncology, Affiliated Zhongshan Hospital of Dalian University, 116001, Dalian, Liaoning, P.R. China
| | - Lili Tian
- Department of Oncology, First Affiliated Hospital of Dalian Medical University, 116011, Dalian, Liaoning, P.R. China
| | - Fangyu Yang
- Department of Neurosurgery, General Hospital of Northern Theater Command, 110015, Shenyang, Liaoning, P.R. China
| | - Jun Li
- Department of Neurosurgery, First Affiliated Hospital of Dalian Medical University, 116011, Dalian, Liaoning, P.R. China
| | - Xiaodong Li
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, 116044, Dalian, Liaoning, P.R. China
| | - Yiqun Yao
- Department of Thyroid and Breast Surgery, Affiliated Zhongshan Hospital of Dalian University, 116001, Dalian, Liaoning, P.R. China
| | - Eric W-F Lam
- Department of Surgery and Cancer, Imperial College London, London, W12 0NN, UK
| | - Peng Gao
- Clinical Laboratory, Dalian Sixth People's Hospital, 116031, Dalian, Liaoning, P.R. China.
| | - Bilian Jin
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, 116044, Dalian, Liaoning, P.R. China.
| | - Ruoyu Wang
- Department of Oncology, Affiliated Zhongshan Hospital of Dalian University, 116001, Dalian, Liaoning, P.R. China.
| |
Collapse
|
12
|
Wang N, Wang S, Wang X, Zheng Y, Yang B, Zhang J, Pan B, Gao J, Wang Z. Research trends in pharmacological modulation of tumor-associated macrophages. Clin Transl Med 2021; 11:e288. [PMID: 33463063 PMCID: PMC7805405 DOI: 10.1002/ctm2.288] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/27/2020] [Accepted: 12/29/2020] [Indexed: 02/06/2023] Open
Abstract
As one of the most abundant immune cell populations in the tumor microenvironment (TME), tumor-associated macrophages (TAMs) play important roles in multiple solid malignancies, including breast cancer, prostate cancer, liver cancer, lung cancer, ovarian cancer, gastric cancer, pancreatic cancer, and colorectal cancer. TAMs could contribute to carcinogenesis, neoangiogenesis, immune-suppressive TME remodeling, cancer chemoresistance, recurrence, and metastasis. Therefore, reprogramming of the immune-suppressive TAMs by pharmacological approaches has attracted considerable research attention in recent years. In this review, the promising pharmaceutical targets, as well as the existing modulatory strategies of TAMs were summarized. The chemokine-chemokine receptor signaling, tyrosine kinase receptor signaling, metabolic signaling, and exosomal signaling have been highlighted in determining the biological functions of TAMs. Besides, both preclinical research and clinical trials have suggested the chemokine-chemokine receptor blockers, tyrosine kinase inhibitors, bisphosphonates, as well as the exosomal or nanoparticle-based targeting delivery systems as the promising pharmacological approaches for TAMs deletion or reprogramming. Lastly, the combined therapies of TAMs-targeting strategies with traditional treatments or immunotherapies as well as the exosome-like nanovesicles for cancer therapy are prospected.
Collapse
Affiliation(s)
- Neng Wang
- The Research Center for Integrative MedicineSchool of Basic Medical SciencesGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Shengqi Wang
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Xuan Wang
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Yifeng Zheng
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Bowen Yang
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Juping Zhang
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Bo Pan
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Jianli Gao
- Academy of Traditional Chinese MedicineZhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Zhiyu Wang
- The Research Center for Integrative MedicineSchool of Basic Medical SciencesGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| |
Collapse
|
13
|
Wang Y, Ren S, Wang Z, Wang Z, Zhu N, Cai D, Ye Z, Ruan J. Chemokines in bone-metastatic breast cancer: Therapeutic opportunities. Int Immunopharmacol 2020; 87:106815. [PMID: 32711376 DOI: 10.1016/j.intimp.2020.106815] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022]
Abstract
Due to non-response to chemotherapy, incomplete surgical resection, and resistance to checkpoint inhibitors, breast cancer with bone metastasis is notoriously difficult to cure. Therefore, the development of novel, efficient strategies to tackle bone metastasis of breast cancer is urgently needed. Chemokines, which induce directed migration of immune cells and act as guide molecules between diverse cells and tissues, are small proteins indispensable in immunity. These complex chemokine networks play pro-tumor roles or anti-tumor roles when produced by breast cancer cells in the tumor microenvironment. Additionally, chemokines have diverse roles when secreted by various immune cells in the tumor microenvironment of breast cancer, which can be roughly divided into immunosuppressive effects and immunostimulatory effects. Recently, targeting chemokine networks has been shown to have potential for use in treatment of metastatic malignancies, including bone-metastatic breast cancer. In this review, we focus on the role of chemokines networks in the biology of breast cancer and metastasis to the bone. We also discuss the therapeutic opportunities and future prospects of targeting chemokine networks, in combination with other current standard therapies, for the treatment of bone-metastatic breast cancer.
Collapse
Affiliation(s)
| | - Shihong Ren
- First People's Hospital of Wenling, Wenling, China
| | - Zhan Wang
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zenan Wang
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ning Zhu
- Hebei North University, Zhangjiakou, China
| | | | - Zhaoming Ye
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | | |
Collapse
|
14
|
MEGF11 is related to tumour recurrence in triple negative breast cancer via chemokine upregulation. Sci Rep 2020; 10:8060. [PMID: 32415115 PMCID: PMC7229019 DOI: 10.1038/s41598-020-64950-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 09/23/2019] [Indexed: 01/10/2023] Open
Abstract
Our previous study demonstrated that upregulation of multiple epidermal growth factor-like domains 11 (MEGF11) gene expression is involved in the mechanism by which recurrence of Triple Negative Breast Cancer (TNBC) occurs. Our aim was to elucidate the role of MEGF11 expression in TNBC cells, both in vitro and in vivo, and in human tissue. Following MEGF11 gene knockdown (∆MEGF11) or over-expression in MDA-MB-231 and MB-468 cells, cell growth and chemokine gene expression were evaluated. In vivo, tumour growth of implanted human TNBC cells and the number of circulating 4T1 mouse tumour cells were measured. There was a significant decrease in cell growth via inhibition of AKT, NF-kB, CREB and AP-1 activation in ∆MEGF11 MDA-MB-231 and 468 cells. This also resulted, in vivo, in a suppression of tumour growth and a decrease in the number of mouse circulating 4T1 breast cancer cells. Surprisingly, overexpression of MEGF11 upregulated the expression of various chemokines and proinflammatory cytokines via AKT activation, but there was no increase in cell proliferation. MEGF11 was found to cross-talk positively with IL-17A signalling. Patients with tumours that over-expressed MEGF11 had a poorer prognosis. We conclude that MEGF11 plays an important role in tumour survival and that overexpression of MEGF11 induces both a cytokine and a chemokine cascade, which will favour the tumour microenvironment in terms of distant metastasis. MEGF11 might be a potential therapeutic target for preventing TNBC recurrence.
Collapse
|
15
|
Huang C, Foster SR, Shah AD, Kleifeld O, Canals M, Schittenhelm RB, Stone MJ. Phosphoproteomic characterization of the signaling network resulting from activation of the chemokine receptor CCR2. J Biol Chem 2020; 295:6518-6531. [PMID: 32241914 DOI: 10.1074/jbc.ra119.012026] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 03/19/2020] [Indexed: 12/13/2022] Open
Abstract
Leukocyte recruitment is a universal feature of tissue inflammation and regulated by the interactions of chemokines with their G protein-coupled receptors. Activation of CC chemokine receptor 2 (CCR2) by its cognate chemokine ligands, including CC chemokine ligand 2 (CCL2), plays a central role in recruitment of monocytes in several inflammatory diseases. In this study, we used phosphoproteomics to conduct an unbiased characterization of the signaling network resulting from CCL2 activation of CCR2. Using data-independent acquisition MS analysis, we quantified both the proteome and phosphoproteome in FlpIn-HEK293T cells stably expressing CCR2 at six time points after activation with CCL2. Differential expression analysis identified 699 significantly regulated phosphorylation sites on 441 proteins. As expected, many of these proteins are known to participate in canonical signal transduction pathways and in the regulation of actin cytoskeleton dynamics, including numerous guanine nucleotide exchange factors and GTPase-activating proteins. Moreover, we identified regulated phosphorylation sites in numerous proteins that function in the nucleus, including several constituents of the nuclear pore complex. The results of this study provide an unprecedented level of detail of CCR2 signaling and identify potential targets for regulation of CCR2 function.
Collapse
Affiliation(s)
- Cheng Huang
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia.,Monash Proteomics and Metabolomics Facility, Monash Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
| | - Simon R Foster
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
| | - Anup D Shah
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia.,Monash Proteomics and Metabolomics Facility, Monash Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia.,Monash Bioinformatics Platform, Monash Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
| | - Oded Kleifeld
- Faculty of Biology, Technion-Israel Institute of Technology, Technion City, Haifa 3200003, Israel
| | - Meritxell Canals
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, United Kingdom.,Centre of Membrane Protein and Receptors, Universities of Birmingham and Nottingham, The Midlands NG7 2UH, United Kingdom
| | - Ralf B Schittenhelm
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia .,Monash Proteomics and Metabolomics Facility, Monash Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
| | - Martin J Stone
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
| |
Collapse
|
16
|
Pawar AS, Eirin A, Tang H, Zhu XY, Lerman A, Lerman LO. Upregulated tumor necrosis factor-α transcriptome and proteome in adipose tissue-derived mesenchymal stem cells from pigs with metabolic syndrome. Cytokine 2020; 130:155080. [PMID: 32240922 PMCID: PMC7529712 DOI: 10.1016/j.cyto.2020.155080] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 03/04/2020] [Accepted: 03/21/2020] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Mesenchymal stem cells (MSCs) have endogenous reparative properties, and may constitute an exogenous therapeutic intervention in patients with chronic kidney disease. The microenvironment of metabolic syndrome (MetS) induces fat inflammation, with abundant expression of tumor necrosis factor (TNF)-α. MetS may also alter the content of adipose tissue-derived MSCs, and we hypothesized that the inflammatory profile of MetS manifests via upregulating MSC mRNAs and proteins of the TNF-α pathway. METHODS Domestic pigs were fed a 16-week Lean or MetS diet (n = 4 each). MSCs were harvested from abdominal subcutaneous fat, and their extracellular vesicles (EVs) isolated. Expression profiles of mRNAs and proteins in MSCs and EVs were obtained by high-throughput sequencing and proteomics. Nuclear translocation of the pro-inflammatory transcription factor (NF)-kB was evaluated in MSC and in pig renal tubular cells (TEC) co-incubated with EVs. RESULTS We found 13 mRNAs and 4 proteins in the TNF-α pathway upregulated in MetS- vs. Lean-MSCs (fold-change > 1.4, p < 0.05), mostly via TNF-α receptor-1 (TNF-R1) signaling. Three mRNAs were upregulated in MetS-EVs. MetS-MSCs, as well as TECs co-incubated with MetS-EVs, showed increased nuclear translocation of NF-kB. Using qPCR, JUNB, MAP2K7 and TRAF2 genes followed the same direction of RNA-sequencing findings. CONCLUSIONS MetS upregulates the TNF-α transcriptome and proteome in swine adipose tissue-derived MSCs, which are partly transmitted to their EV progeny, and are associated with activation of NF-kB in target cells. Hence, the MetS milieu may affect the profile of endogenous MSCs and their paracrine vectors and limit their use as an exogenous regenerative therapy. Anti-inflammatory strategies targeting the TNF-α pathway might be a novel strategy to restore MSC phenotype, and in turn function.
Collapse
Affiliation(s)
- Aditya S Pawar
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Alfonso Eirin
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Hui Tang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Xiang-Yang Zhu
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Amir Lerman
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, United States
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States; Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, United States.
| |
Collapse
|
17
|
Kudryavtseva AV, Lukyanova EN, Kharitonov SL, Nyushko KM, Krasheninnikov AA, Pudova EA, Guvatova ZG, Alekseev BY, Kiseleva MV, Kaprin AD, Dmitriev AA, Snezhkina AV, Krasnov GS. Bioinformatic identification of differentially expressed genes associated with prognosis of locally advanced lymph node-positive prostate cancer. J Bioinform Comput Biol 2020; 17:1950003. [PMID: 30866732 DOI: 10.1142/s0219720019500033] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Prostate cancer (PCa) is one of the primary causes of cancer-related mortality in men worldwide. Patients with locally advanced PCa with metastases in regional lymph nodes are usually marked as a high-risk group. One of the chief concerns for this group is to make an informed decision about the necessity of conducting adjuvant androgen deprivation therapy after radical surgical treatment. During the oncogenic transformation and progression of the disease, the expression of many genes is altered. Some of these genes can serve as markers for diagnosis, predicting the prognosis or effectiveness of drug therapy, as well as possible therapeutic targets. We undertook bioinformatic analysis of the RNA-seq data deposited in The Cancer Genome Atlas consortium database to identify possible prognostic markers. We compared the groups with favorable and unfavorable prognosis for the cohort of patients with PCa showing lymph node metastasis (pT2N1M0, pT3N1M0, and pT4N1M0) and for the most common molecular type carrying the fusion transcript TMPRSS2-ERG. For the entire cohort, we revealed at least six potential markers (IDO1, UGT2B15, IFNG, MUC6, CXCL11, and GBP1). Most of these genes are involved in the positive regulation of immune response. For the TMPRSS2-ERG subtype, we also identified six genes, the expression of which may be associated with prognosis: TOB1, GALNT7, INAFM1, APELA, RAC3, and NNMT. The identified genes, after additional studies and validation in the extended cohort, could serve as a prognostic marker of locally advanced lymph node-positive PCa.
Collapse
Affiliation(s)
- Anna V Kudryavtseva
- * Laboratory of Postgenomic Research, Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Vavilova 32, Moscow 119991, Russian Federation
| | - Elena N Lukyanova
- * Laboratory of Postgenomic Research, Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Vavilova 32, Moscow 119991, Russian Federation
| | - Sergey L Kharitonov
- * Laboratory of Postgenomic Research, Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Vavilova 32, Moscow 119991, Russian Federation
| | - Kirill M Nyushko
- † Federal State Budgetary Institution, National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 4 Korolev Str., Obninsk 249036, Russian Federation
| | - Alexey A Krasheninnikov
- † Federal State Budgetary Institution, National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 4 Korolev Str., Obninsk 249036, Russian Federation
| | - Elena A Pudova
- * Laboratory of Postgenomic Research, Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Vavilova 32, Moscow 119991, Russian Federation
| | - Zulfiya G Guvatova
- * Laboratory of Postgenomic Research, Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Vavilova 32, Moscow 119991, Russian Federation
| | - Boris Y Alekseev
- † Federal State Budgetary Institution, National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 4 Korolev Str., Obninsk 249036, Russian Federation
| | - Marina V Kiseleva
- † Federal State Budgetary Institution, National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 4 Korolev Str., Obninsk 249036, Russian Federation
| | - Andrey D Kaprin
- † Federal State Budgetary Institution, National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, 4 Korolev Str., Obninsk 249036, Russian Federation
| | - Alexey A Dmitriev
- * Laboratory of Postgenomic Research, Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Vavilova 32, Moscow 119991, Russian Federation
| | - Anastasiya V Snezhkina
- * Laboratory of Postgenomic Research, Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Vavilova 32, Moscow 119991, Russian Federation
| | - George S Krasnov
- * Laboratory of Postgenomic Research, Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Vavilova 32, Moscow 119991, Russian Federation
| |
Collapse
|
18
|
Chen W, Qin Y, Liu S. CCL20 Signaling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1231:53-65. [PMID: 32060846 DOI: 10.1007/978-3-030-36667-4_6] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
CCL20, as a chemokine, plays an important role in rheumatoid arthritis, psoriasis, and other diseases by binding to its receptor CCR6. Recent 10 years' research has demonstrated that CCL20 also contributes to the progression of many cancers, such as liver cancer, colon cancer, breast cancer, pancreatic cancer, and gastric cancer. This article reviews and discusses the previous studies on CCL20 roles in cancers from the aspects of its specific effects on various cancers, its remodeling on tumor microenvironment (TME), its synergistic effects with other cytokines in tumor microenvironment, and the specific mechanisms of CCL20 signal activation, illustrating CCL20 signaling in TME from multiple directions.
Collapse
Affiliation(s)
- Weilong Chen
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, Shanghai Medical College, Shanghai, China.,Key Laboratory of Breast Cancer in Shanghai, Innovation Center for Cell Signaling Network, Cancer Institute, Shanghai, China.,Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yuanyuan Qin
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, Shanghai Medical College, Shanghai, China.,Key Laboratory of Breast Cancer in Shanghai, Innovation Center for Cell Signaling Network, Cancer Institute, Shanghai, China.,Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Suling Liu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, Shanghai Medical College, Shanghai, China. .,Key Laboratory of Breast Cancer in Shanghai, Innovation Center for Cell Signaling Network, Cancer Institute, Shanghai, China. .,Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
| |
Collapse
|
19
|
Ge X, Zhao Y, Chen C, Wang J, Sun L. Cancer Immunotherapies Targeting Tumor-Associated Regulatory T Cells. Onco Targets Ther 2019; 12:11033-11044. [PMID: 31997881 PMCID: PMC6917600 DOI: 10.2147/ott.s231052] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 12/02/2019] [Indexed: 12/18/2022] Open
Abstract
Tumor-associated regulatory T cells (Tregs) are important effectors in the tumor microenvironment (TME), acting as accomplices in the promotion of tumor progression. Currently, the importance of removing the immunosuppressive activity in the TME has received its due attention, and Tregs have been focused on. The cytokine-receptor axes are among the essential signaling pathways in immunocytes, and tumor-associated Tregs are no exception. Therefore, manipulating cytokine-receptor pathways may be a promising effective strategy for treating various malignancies. Here, we summarize the classification, immunosuppressive mechanisms, existing immunotherapies, and potential biomarkers related to tumor-infiltrating Tregs to guide the development of effective cancer immunotherapies.
Collapse
Affiliation(s)
- Xiaoxu Ge
- Department of Colorectal Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Hangzhou, People's Republic of China.,Key Laboratory of Molecular Biology in Medical Sciences, Hangzhou, People's Republic of China.,The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Yamei Zhao
- Department of Colorectal Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Hangzhou, People's Republic of China.,Key Laboratory of Molecular Biology in Medical Sciences, Hangzhou, People's Republic of China.,The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Chao Chen
- Department of Colorectal Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Hangzhou, People's Republic of China.,Key Laboratory of Molecular Biology in Medical Sciences, Hangzhou, People's Republic of China.,The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Jian Wang
- Department of Colorectal Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Hangzhou, People's Republic of China.,Key Laboratory of Molecular Biology in Medical Sciences, Hangzhou, People's Republic of China.,The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Lifeng Sun
- Department of Colorectal Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Hangzhou, People's Republic of China.,Key Laboratory of Molecular Biology in Medical Sciences, Hangzhou, People's Republic of China.,The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| |
Collapse
|
20
|
Guo W, Li H, Liu H, Ma X, Yang S, Wang Z. DEPDC1 drives hepatocellular carcinoma cell proliferation, invasion and angiogenesis by regulating the CCL20/CCR6 signaling pathway. Oncol Rep 2019; 42:1075-1089. [PMID: 31322256 PMCID: PMC6667871 DOI: 10.3892/or.2019.7221] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 07/03/2019] [Indexed: 12/16/2022] Open
Abstract
DEP domain containing 1 (DEPDC1) functions as an oncogene in hepatocellular carcinoma (HCC). However, the underlying mechanism of DEPDC1 remains largely unknown. The present study revealed that DEPDC1 knockdown inhibited HCC cell proliferation, colony formation and invasion in vitro and suppressed the growth of HCC xenografts in vivo. Furthermore, DEPDC1 overexpression promoted HCC cell proliferation, colony formation and invasion. DNA microarray, reverse transcription-quantitative-PCR and western blotting results demonstrated that DEPDC1 knockdown in Huh-7 significantly inhibited the expression of chemokine (C-C motif) ligand 20 (CCL20) and chemokine (C-C motif) receptor 6 (CCR6). In addition, the expression of CCL20 and CCR6 were upregulated in HCC tissues and cell lines, and were positively correlated with DEPDC1 expression. CCL20 or CCR6 knockdown via small interfering RNA reversed the effects of DEPDC1 overexpression in HCC cells. Furthermore, it was revealed that conditioned medium from DEPDC1 upregulated Li-7 and Hep3B cells led to angiogenesis in vitro, whereas CCL20 knockdown in Li-7 and Hep3B cells or CCR6 knockdown in human umbilical vein endothelial cells reversed the angiogenic effect of DEPDC1 overexpression. In conclusion, DEPDC1 facilitated cell proliferation, invasion and angiogenesis via the CCL20/CCR6 pathway in HCC.
Collapse
Affiliation(s)
- Wubin Guo
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Hui Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Huan Liu
- Research Laboratory of Biomedical Engineering, The TCM Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Xin Ma
- Department of General Surgery, The TCM Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Sijin Yang
- Department of Cardiovascular Medicine, The TCM Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Ziwei Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
21
|
Yang DW, Qian GB, Jiang MJ, Wang P, Wang KZ. Inhibition of microRNA-495 suppresses chondrocyte apoptosis through activation of the NF-κB signaling pathway by regulating CCL4 in osteoarthritis. Gene Ther 2019; 26:217-229. [PMID: 30940879 DOI: 10.1038/s41434-019-0068-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 03/07/2019] [Accepted: 03/08/2019] [Indexed: 12/13/2022]
Abstract
As a common form of arthritis, osteoarthritis (OA) represents a degenerative disease, characterized by articular cartilage damage and synovium inflammation. Recently, the role of various microRNAs (miRs) and their specific expression in OA has been highlighted. Therefore, the aim of the current study was to elucidate the role by which miR-495 and chemokine ligand 4 (CCL4) influence the development and progression of OA. OA mice models were established, after which the CCL4 and collagen levels as well as cell apoptosis were determined in cartilage tissue of OA mice. The chondrocytes of the OA mice models were subsequently treated with a series of miR-495 mimic, inhibitor, and siRNA against CCL4. Afterwards, miR-495 expressions as well as the levels of CCL4, p50, p65, and IkBa and the extent of IkBa phosphorylation in addition to the luciferase activity of NF-kB were measured accordingly. Finally, cell apoptosis and cell cycle distribution were detected. miR-495 was highly expressed while NF-κB, CCL4, and collagen II were poorly expressed. Cell apoptosis was elevated in the cartilage tissue of the OA mice. CCL4 was a potential target gene of miR-495. Downregulation of miR-495 led to accelerated chondrocyte proliferation accompanied by diminished cell apoptosis among the OA mice. Taken together, the results of the current study demonstrated that inhibition of miR-495 suppressed chondrocyte apoptosis and promoted its proliferation through activation of the NF-κB signaling pathway by up-regulation of CCL4 in OA.
Collapse
Affiliation(s)
- Da-Wei Yang
- Department of Orthopaedics, The Fourth Affiliated Hospital of Harbin Medical University, 150001, Harbin, China
| | - Gui-Bin Qian
- Department of Orthopaedics, The Fourth Affiliated Hospital of Harbin Medical University, 150001, Harbin, China
| | - Ming-Jiu Jiang
- Department of Orthopaedics, The Fourth Affiliated Hospital of Harbin Medical University, 150001, Harbin, China
| | - Peng Wang
- Department of Orthopaedics, The Fourth Affiliated Hospital of Harbin Medical University, 150001, Harbin, China
| | - Kun-Zheng Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Xi'an Jiaotong University, 710000, Xi'an, China.
| |
Collapse
|
22
|
CC chemokines are differentially expressed in Breast Cancer and are associated with disparity in overall survival. Sci Rep 2019; 9:4014. [PMID: 30850664 PMCID: PMC6408438 DOI: 10.1038/s41598-019-40514-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 02/18/2019] [Indexed: 12/21/2022] Open
Abstract
Despite recent advances, breast cancer (BrCa) still affects many women and the impact is disproportional in African Americans (AA) compared to European Americans (EA). Addressing socioeconomic and behavioral status has not been enough to reduce disparity, suggesting contribution of biological differences in BrCa disparity. Our laboratory was first to show involvement of CC chemokines in BrCa. In this study, using ONCOMINE, TCGA, bc-GenExMiner and KMplotter, we examined the association of CC chemokines in BrCa outcomes and disparity. We show over-expression of CCL5, -7, -11, -17, -20, -22 and -25 in BrCa tissues. High mRNA levels of CCL7, -8, -17, -20 and -25 predicted a decrease in overall survival (OS). CCL7 and CCL8 were associated with decreased relapse-free survival. Expression of CCL17 and CCL25 was associated with decreased OS in AA. In EA, CCL8 was associated with decreased OS. Expression of CCL5, -7, -8, -17, -20 and -25 was highest in TNBC. Expression of CCL11 and CCL22 was associated with HER2. CCL7, -8, -17, -20 and -25 were elevated in AAs. In conclusion, our analysis suggests significant association of CC-chemokines in BrCa progression, OS and disparate disease outcome in AA compared to EA patients.
Collapse
|
23
|
Antonaci G, Cossa LG, Muscella A, Vetrugno C, De Pascali SA, Fanizzi FP, Marsigliante S. [Pt( O,O'-acac)(γ-acac)(DMS)] Induces Autophagy in Caki-1 Renal Cancer Cells. Biomolecules 2019; 9:biom9030092. [PMID: 30845773 PMCID: PMC6468382 DOI: 10.3390/biom9030092] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 02/23/2019] [Accepted: 02/26/2019] [Indexed: 01/12/2023] Open
Abstract
We have demonstrated the cytotoxic effects of [Pt(O,O′-acac)(γ-acac)(dimethyl sulfide (DMS))] on various immortalized cell lines, in primary cultures, and in murine xenograft models in vivo. Recently, we also showed that [Pt(O,O′-acac)(γ-acac)(DMS)] is able to kill Caki-1 renal cells both in vivo and in vitro. In the present paper, apoptotic and autophagic effects of [Pt(O,O′-acac)(γ-acac)(DMS)] and cisplatin were studied and compared using Caki-1 cancerous renal cells. The effects of cisplatin include activation of caspases, proteolysis of enzyme poly ADP ribose polymerase (PARP), control of apoptosis modulators B-cell lymphoma 2 (Bcl-2), Bcl-2-associated X protein (Bax), and BH3-interacting domain death agonist (Bid), and cell cycle arrest in G2/M phase. Conversely, [Pt(O,O′-acac)(γ-acac)(DMS)] did not induce caspase activation, nor chromatin condensation or DNA fragmentation. The effects of [Pt(O,O′-acac)(γ-acac)(DMS)] include microtubule-associated proteins 1A/1B light chain 3B (LC3)-I to LC3-II conversion, Beclin-1 and Atg-3, -4, and -5 increase, Bcl-2 decrease, and monodansylcadaverine accumulation in autophagic vacuoles. [Pt(O,O′-acac)(γ-acac)(DMS)] also modulated various kinases involved in intracellular transduction regulating cell fate. [Pt(O,O′-acac)(γ-acac)(DMS)] inhibited the phosphorylation of mammalian target of rapmycin (mTOR), p70S6K, and AKT, and increased the phosphorylation of c-Jun N-terminal kinase (JNK1/2), a kinase activity pattern consistent with autophagy induction. In conclusion, while in past reports the high cytotoxicity of [Pt(O,O′-acac)(γ-acac)(DMS)] was always attributed to its ability to trigger an apoptotic process, in this paper we show that Caki-1 cells die as a result of the induction of a strong autophagic process.
Collapse
Affiliation(s)
- Giovanna Antonaci
- Laboratory of Cell Physiology, Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, 73100 Lecce, Italy.
| | - Luca Giulio Cossa
- Laboratory of Cell Physiology, Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, 73100 Lecce, Italy.
| | - Antonella Muscella
- Laboratory of Cell Pathology, Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, 73100 Lecce, Italy.
| | - Carla Vetrugno
- Laboratory of Cell Pathology, Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, 73100 Lecce, Italy.
| | - Sandra Angelica De Pascali
- Laboratory of General Inorganic Chemistry, Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, 73100 Lecce, Italy.
| | - Francesco Paolo Fanizzi
- Laboratory of General Inorganic Chemistry, Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, 73100 Lecce, Italy.
| | - Santo Marsigliante
- Laboratory of Cell Physiology, Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, 73100 Lecce, Italy.
| |
Collapse
|
24
|
Kriseman M, Monsivais D, Agno J, Masand RP, Creighton CJ, Matzuk MM. Uterine double-conditional inactivation of Smad2 and Smad3 in mice causes endometrial dysregulation, infertility, and uterine cancer. Proc Natl Acad Sci U S A 2019; 116:3873-3882. [PMID: 30651315 PMCID: PMC6397514 DOI: 10.1073/pnas.1806862116] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
SMAD2 and SMAD3 are downstream proteins in the transforming growth factor-β (TGF β) signaling pathway that translocate signals from the cell membrane to the nucleus, bind DNA, and control the expression of target genes. While SMAD2/3 have important roles in the ovary, we do not fully understand the roles of SMAD2/3 in the uterus and their implications in the reproductive system. To avoid deleterious effects of global deletion, and given previous data showing redundant function of Smad2 and Smad3, a double-conditional knockout was generated using progesterone receptor-cre (Smad2/3 cKO) mice. Smad2/3 cKO mice were infertile due to endometrial hyperproliferation observed as early as 6 weeks of postnatal life. Endometrial hyperplasia worsened with age, and all Smad2/3 cKO mice ultimately developed bulky endometrioid-type uterine cancers with 100% mortality by 8 months of age. The phenotype was hormone-dependent and could be prevented with removal of the ovaries at 6 weeks of age but not at 12 weeks. Uterine tumor epithelium was associated with decreased expression of steroid biosynthesis genes, increased expression of inflammatory response genes, and abnormal expression of cell cycle checkpoint genes. Our results indicate the crucial role of SMAD2/3 in maintaining normal endometrial function and confirm the hormone-dependent nature of SMAD2/3 in the uterus. The hyperproliferation of the endometrium affected both implantation and maintenance of pregnancy. Our findings generate a mouse model to study the roles of SMAD2/3 in the uterus and serve to provide insight into the mechanism by which the endometrium can escape the plethora of growth regulatory proteins.
Collapse
Affiliation(s)
- Maya Kriseman
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030
- Reproductive Endocrinology and Infertility, Baylor College of Medicine/Texas Children's Hospital Women's Pavilion, Houston, TX 77030
| | - Diana Monsivais
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030
| | - Julio Agno
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030
| | - Ramya P Masand
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030
| | - Chad J Creighton
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030
| | - Martin M Matzuk
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030;
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
25
|
Küçükköse C, Yalçin Özuysal Ö. Effects of Notch signalling on the expression of SEMA3C, HMGA2, CXCL14, CXCR7, and CCL20 in breast cancer. ACTA ACUST UNITED AC 2019; 43:70-76. [PMID: 30930637 PMCID: PMC6426645 DOI: 10.3906/biy-1808-58] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Metastasis is the main reason for death in breast cancer. Understanding the molecular players in metastasis is crucial for diagnostic and therapeutic purposes. Notch signalling plays an oncogenic role in breast tumorigenesis and is involved in metastasis. Downstream mediators of Notch signalling in prometastatic processes are not yet fully discovered. Here we aimed to investigate whether Notch signalling regulates the expression of SEMA3C, HMGA2, CXCL14, CXCR7, and CCL20, which are involved in prometastatic processes, in breast cell lines. To this end, expression of the selected genes was analysed following Notch activation by overexpression of the Notch1 intracellular domain in the normal breast epithelial cell line MCF10A, and inhibition by silencing of the Notch transcriptional mediator RBPjκ in the breast cancer cell line MDA MB 231. SEMA3C and HMGA2 mRNA were decreased, while CXCL14 and CXCR7 mRNA were increased significantly in response to Notch activation in MCF10A cells. Notch inhibition in MDA MB 231 cells significantly decreased HMGA2 and CCL20 mRNA. Protein levels were not significantly altered by Notch modulation. In conclusion, we showed that Notch signalling regulates expression of SEMA3C, CXCL14, CCL20, CXCR7, and HMGA2, which are prominent candidate genes that might function downstream of Notch to induce prometastatic processes.
Collapse
Affiliation(s)
- Cansu Küçükköse
- Department of Molecular Biology and Genetics, Faculty of Science, İzmir Institute of Technology , İzmir , Turkey
| | - Özden Yalçin Özuysal
- Department of Molecular Biology and Genetics, Faculty of Science, İzmir Institute of Technology , İzmir , Turkey
| |
Collapse
|
26
|
Ranasinghe R, Eri R. Modulation of the CCR6-CCL20 Axis: A Potential Therapeutic Target in Inflammation and Cancer. ACTA ACUST UNITED AC 2018; 54:medicina54050088. [PMID: 30453514 PMCID: PMC6262638 DOI: 10.3390/medicina54050088] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 11/01/2018] [Accepted: 11/13/2018] [Indexed: 12/23/2022]
Abstract
Prototypical functions of the chemokine receptor CCR6 include immune regulation by maneuvering cell chemotaxis and selective delimiting of the pro-inflammatory TH17 and regulatory Treg subsets during chronic or acute systemic inflammation. Inhibition of CCR6 is proposed to attenuate disease symptoms and promote recuperation of multiple inflammatory and autoimmune disorders. Prescription medicines with pharmacodynamics involving the inhibition of the chemokine axis CCR6–CCL20 are very limited. The development of such therapeutics is still at an early experimental stage and has mostly involved the utilization of pre-clinical models and neutralizing mono or polyclonal antibodies against either partner (CCR6 or CCL20). Other methods include the constitutive use of small molecules as peptide inhibitors or small interfering ribonucleic acid (siRNA) to interfere with transcription at the nuclear level. In our review, we aim to introduce the wide array of potential CCR6–CCL20 inhibitors with an emphasis on attendant immune-modulator capacity that have been tested in the research field to date and are immensely promising compounds as forerunners of future curatives. Sixteen different tractable inhibitors of the CCR6–CCL20 duo have been identified as possessing high medicinal potential by drug developers worldwide to treat autoimmune and inflammatory diseases as shown in Figure 1. A multitude of antibody preparations are already available in the current pharmaceutical market as patented treatments for diseases in which the CCR6–CCL20 axis is operative, yet they must be used only as supplements with existing routinely prescribed medication as they collectively produce adverse side effects. Novel inhibitors are needed to evaluate this invaluable therapeutic target which holds much promise in the research and development of complaisant remedies for inflammatory diseases.
Collapse
Affiliation(s)
- Ranmali Ranasinghe
- School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Tasmania 7248, Australia.
| | - Rajaraman Eri
- School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Tasmania 7248, Australia.
| |
Collapse
|
27
|
Huang Y, Peng Q, Li HY, Jia ZD, Li Y, Gao Y. Novel sericin-based hepatocyte serum-free medium and sericin’s effect on hepatocyte transcriptome. World J Gastroenterol 2018; 24:3398-3413. [PMID: 30122879 PMCID: PMC6092578 DOI: 10.3748/wjg.v24.i30.3398] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 06/17/2018] [Accepted: 06/28/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To develop a novel hepatocyte serum-free medium based on sericin, and to explore the effect of sericin on the hepatocyte transcriptome.
METHODS A controlled trial comparing novel serum-free medium and other media: C3A cells were cultured in our novel serum-free medium, HepatoZYME, complete medium (DMEM/F12 with 100 mL/L FBS), and DMEM/F12, and then cell attachment, proliferation, and function as well as the biocompatibility of the media were assessed. A comparative study of serum-free media with or without 2 mg/mL sericin: the effect of sericin on C3A growth was assessed by cell viability and proliferation, the effect of sericin on C3A cell cycle distribution was determined by flow cytometry, and the effect of sericin on the C3A transcriptome was assessed by gene-chip array and RT-qPCR.
RESULTS More C3A cells attached to the plate containing our serum-free medium than to those containing HepatoZYME and DMEM/F12 at 24 h post-seeding. Both the viability and proliferation rate of C3A cells in sericin-based serum-free medium were superior to those of cells in HepatoZYME and DMEM/F12 (P < 0.001). The content of albumin and urea in our serum-free medium was significantly higher than that in HepatoZYME and DMEM/F12 throughout the whole culture period (P < 0.001) and was similar to that in complete medium at day 3, 4, and 5. In part 2, cell viability and proliferation were greater in the presence of 2 mg/mL sericin (P < 0.001), as was the proportion of cells in S phase (16.21% ± 0.98% vs 12.61% ± 0.90%, P < 0.01). Gene-chip array analysis indicated that the expression of CCR6, EGFR, and FOS were up-regulated by 2 mg/mL sericin, and RT-qPCR revealed that the expression of CCR6, EGFR, FOS, AKT1, JNK1, NFkB1, MMP-9, MEK2, ERK1/2 and MYC was up-regulated by 2 mg/mL sericin (P < 0.05).
CONCLUSION We developed a novel hepatocyte serum-free medium. Sericin probably enhances cell attachment through the CCR6-Akt-JNK-NF-κB pathway and promotes cell proliferation through CCR6-mediated activation of the ERK1/2-MAPK pathway.
Collapse
Affiliation(s)
- Yun Huang
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, Guangdong Province, China
| | - Qing Peng
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, Guangdong Province, China
| | - Hai-Yan Li
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, Guangdong Province, China
| | - Zhi-Dong Jia
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, Guangdong Province, China
| | - Yang Li
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, Guangdong Province, China
| | - Yi Gao
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, Guangdong Province, China
- State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| |
Collapse
|
28
|
CCL20 triggered by chemotherapy hinders the therapeutic efficacy of breast cancer. PLoS Biol 2018; 16:e2005869. [PMID: 30052635 PMCID: PMC6082578 DOI: 10.1371/journal.pbio.2005869] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 08/08/2018] [Accepted: 07/12/2018] [Indexed: 12/22/2022] Open
Abstract
Chemotherapeutic resistance in triple-negative breast cancer (TNBC) has brought great challenges to the improvement of patient survival. The mechanisms of taxane chemoresistance in TNBC have not been well investigated. Our results illustrated C-C motif chemokine ligand 20 (CCL20) was significantly elevated during taxane-containing chemotherapy in breast cancer patients with nonpathologic complete response. Furthermore, CCL20 promoted the self-renewal and maintenance of breast cancer stem cells (BCSCs) or breast cancer stem-like cells through protein kinase Cζ (PKCζ) or p38 mitogen-activated protein kinase (MAPK)-mediated activation of p65 nuclear factor kappa B (NF-κB) pathway, significantly increasing the frequency and taxane resistance of BCSCs. Moreover, CCL20-promoted NF-κB activation increased ATP-binding cassette subfamily B member 1 (ABCB1)/multidrug resistance 1 (MDR1) expression, leading to the extracellular efflux of taxane. These results suggested that chemotherapy-induced CCL20 mediated chemoresistance via up-regulating ABCB1. In addition, NF-κB activation increased CCL20 expression, forming a positive feedback loop between NF-κB and CCL20 pathways, which provides sustained impetus for chemoresistance in breast cancer cells. Our results suggest that CCL20 can be a novel predictive marker for taxane response, and the blockade of CCL20 or its downstream pathway might reverse the taxane resistance in breast cancer patients.
Collapse
|
29
|
Muscella A, Cossa LG, Vetrugno C, Antonaci G, Marsigliante S. Adenosine diphosphate regulates MMP2 and MMP9 activity in malignant mesothelioma cells. Ann N Y Acad Sci 2018; 1431:72-84. [PMID: 29984433 DOI: 10.1111/nyas.13922] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 05/22/2018] [Accepted: 06/13/2018] [Indexed: 12/19/2022]
Abstract
Although an association between cancer progression and matrix metalloproteinase (MMP) 2 and MPP9 expression has been known, the expression, nuclear localization, and physiologically controlled activation of these two MMPs have not been investigated in malignant mesothelioma cells. We examined the expression and intracellular localization of MMP2/9 in ZL55 malignant mesothelioma cells, as well as their regulation by ADP. Using real-time PCR, we showed that activation of the P2Y1 receptor by ADP increased the expression of MMP2/9 mRNAs; MMP2/9 collected from conditioned media also showed an increase in activity; and ADP induced the nuclear localization of MMP2/9. The effects of ADP on transcription of the MMPs were due to activation of c-Src, Akt, and NF-κB, while ERK1/2 phosphorylation was needed for the increase in enzymatic activity and the regulation of nuclear import. We also showed that the nuclear localization of MMP2/9 induced by ADP causes the cleavage and inactivation of poly-ADP-ribose polymerase-1. These findings may help to elucidate the mechanisms regulating MMP2/9 activation in ZL55 human epithelioid mesothelioma cells, and perhaps other cells. Therapeutic approaches that promote ADP accumulation in a tumor environment may constitute an effective means to induce anticancer activity.
Collapse
Affiliation(s)
- Antonella Muscella
- Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, Lecce, Italy
| | - Luca Giulio Cossa
- Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, Lecce, Italy
| | - Carla Vetrugno
- Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, Lecce, Italy
| | - Giovanna Antonaci
- Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, Lecce, Italy
| | - Santo Marsigliante
- Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, Lecce, Italy
| |
Collapse
|
30
|
Jin P, Shin SH, Chun YS, Shin HW, Shin YJ, Lee Y, Kim D, Nam DH, Park JW. Astrocyte-derived CCL20 reinforces HIF-1-mediated hypoxic responses in glioblastoma by stimulating the CCR6-NF-κB signaling pathway. Oncogene 2018. [PMID: 29535421 DOI: 10.1038/s41388-018-0182-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
During tumor development, stromal cells are co-opted to the tumor milieu and provide favorable conditions for the tumor. Hypoxia stimulates cancer cells to acquire a more malignant phenotype via activation of hypoxia-inducible factor 1 (HIF-1). Given that cancer cells and astrocytes in glioblastomas coexist in a hypoxic microenvironment, we examined whether astrocytes affect the adaptation of glioblastoma cells to hypoxia. Immunoblotting, reporter assays, quantitative RT-PCR, and chromatin immunoprecipitation were performed to evaluate HIF-1 signaling in glioblastoma cells. Astrocyte-derived chemokine C-C motif ligand 20 (CCL20) was identified using cytokine arrays, and its role in glioblastoma development was evaluated in orthotopic xenografts. Astrocytes augmented HIF-1α expression in glioblastoma cells under hypoxia. The expression of HIF-1 downstream genes, cancer colony formation, and Matrigel invasion of glioblastoma cells were stimulated by conditioned medium from astrocytes pre-exposed to hypoxia. CCL20 was secreted in a hypoxia-dependent manner from astrocytes and busted the hypoxic induction of HIF-1α in glioblastoma cells. Mechanistically, the CCL20/CCR6 signaling pathway upregulates HIF-1α by stimulating nuclear factor kappa B-driven transactivation of the HIF1A gene. Compared with the control tumors, CCR6-deficient glioblastoma xenografts grew more slowly, with poor vascularization, and expressed lower levels of HIF-1α and its downstream proteins. Furthermore, CCR6 expression was correlated with HIF-1α expression in GEO and TCGA datasets from human glioblastoma tissues. These results suggest that glioblastoma cells adapt well to hypoxic stress by virtue of CCL20 derived from neighboring astrocytes.
Collapse
Affiliation(s)
- Peng Jin
- Department of Biomedical Sciences, BK21-plus education program, Seoul National University College of Medicine, Seoul, Korea.,Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea.,Ischemic/Hypoxic Disease Institute and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Seung-Hyun Shin
- Department of Biomedical Sciences, BK21-plus education program, Seoul National University College of Medicine, Seoul, Korea.,Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea.,Ischemic/Hypoxic Disease Institute and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Yang-Sook Chun
- Department of Biomedical Sciences, BK21-plus education program, Seoul National University College of Medicine, Seoul, Korea.,Ischemic/Hypoxic Disease Institute and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hyun-Woo Shin
- Department of Biomedical Sciences, BK21-plus education program, Seoul National University College of Medicine, Seoul, Korea.,Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea.,Ischemic/Hypoxic Disease Institute and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Yong Jae Shin
- Institute for Refractory Cancer Research, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea.,Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yeri Lee
- Institute for Refractory Cancer Research, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Donggeon Kim
- Institute for Refractory Cancer Research, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Do-Hyun Nam
- Institute for Refractory Cancer Research, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea.,Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jong-Wan Park
- Department of Biomedical Sciences, BK21-plus education program, Seoul National University College of Medicine, Seoul, Korea. .,Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea. .,Ischemic/Hypoxic Disease Institute and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
31
|
Liu Y, Wang J, Ni T, Wang L, Wang Y, Sun X. CCL20 mediates RANK/RANKL-induced epithelial-mesenchymal transition in endometrial cancer cells. Oncotarget 2018; 7:25328-39. [PMID: 27015366 PMCID: PMC5041907 DOI: 10.18632/oncotarget.8291] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 03/10/2016] [Indexed: 12/16/2022] Open
Abstract
RANK/RANKL facilitates migration/invasion via epithelial-mesenchymal transition (EMT) in certain malignant tumors. The relationship and mechanism between RANK/RANKL and EMT in endometrial cancer (EC) cells, however, remain unclear. In this study, we firstly showed that RANK/RANKL activation was correlated with EC staging and EMT markers in human EC tissue specimen. RANK/RANKL promoted migration/invasion and initiated EMT of EC cell lines. Then, protein chip analysis and enzyme-linked immunosorbent assay (ELISA) revealed that the expression and secretion of chemokine ligand 20 (CCL20) was dramatically enhanced in RANKL-treated RANK over-expressed EC cells. Moreover, the higher level of CCL20 in both serum and tumor tissue was detected in orthotopic transplantation mouse models. Finally, we confirmed that CCL20 contributed to invasion and EMT of RANK over-expressed EC cells. In summary, all data supported the hypothesis that RANK/RANKL elevated the expression and secretion of CCL20 in EC cells, which promoted cancer progression through EMT.
Collapse
Affiliation(s)
- Yao Liu
- Department of Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Wang
- Department of Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ting Ni
- Department of Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lihua Wang
- Department of Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yudong Wang
- Department of Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao Sun
- Laboratory for Gynecologic Oncology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
32
|
Chen SC, Chen FW, Hsu YL, Kuo PL. Systematic Analysis of Transcriptomic Profile of Renal Cell Carcinoma under Long-Term Hypoxia Using Next-Generation Sequencing and Bioinformatics. Int J Mol Sci 2017; 18:ijms18122657. [PMID: 29215599 PMCID: PMC5751259 DOI: 10.3390/ijms18122657] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 11/25/2017] [Accepted: 12/04/2017] [Indexed: 01/28/2023] Open
Abstract
Patients with clear cell renal cell carcinoma (ccRCC) are often diagnosed with both von Hippel-Lindau (VHL) mutations and the constitutive activation of hypoxia-inducible factor-dependent signaling. In this study, we investigated the effects of long-term hypoxia in 786-O, a VHL-defective renal cell carcinoma cell line, to identify potential genes and microRNAs associated with tumor malignancy. The transcriptomic profiles of 786-O under normoxia, short-term hypoxia and long-term hypoxia were analyzed using next-generation sequencing. The results showed that long-term hypoxia promoted the ability of colony formation and transwell migration compared to normoxia. In addition, the differentially expressed genes induced by long-term hypoxia were involved in various biological processes including cell proliferation, the tumor necrosis factor signaling pathway, basal cell carcinoma and cancer pathways. The upregulated (L1CAM and FBN1) and downregulated (AUTS2, MAPT, AGT and USH1C) genes in 786-O under long-term hypoxia were also observed in clinical ccRCC samples along with malignant grade. The expressions of these genes were significantly correlated with survival outcomes in patients with renal cancer. We also found that long-term hypoxia in 786-O resulted in decreased expressions of hsa-mir-100 and hsa-mir-378 and this effect was also observed in samples of metastatic ccRCC compared to samples of non-metastatic ccRCC. These findings may provide a new direction for the study of potential molecular mechanisms associated with the progression of ccRCC.
Collapse
Affiliation(s)
- Szu-Chia Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Internal Medicine, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung 812, Taiwan.
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Feng-Wei Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Ya-Ling Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Po-Lin Kuo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Institute of Medical Science and Technology, National Sun Yat-Sen University, Kaohsiung 804, Taiwan.
| |
Collapse
|
33
|
Th9 cells promote antitumor immunity via IL-9 and IL-21 and demonstrate atypical cytokine expression in breast cancer. Int Immunopharmacol 2017; 52:163-167. [DOI: 10.1016/j.intimp.2017.08.031] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 08/29/2017] [Accepted: 08/31/2017] [Indexed: 11/19/2022]
|
34
|
Xia X, Chiu PWY, Lam PK, Chin WC, Ng EKW, Lau JYW. Secretome from hypoxia-conditioned adipose-derived mesenchymal stem cells promotes the healing of gastric mucosal injury in a rodent model. Biochim Biophys Acta Mol Basis Dis 2017; 1864:178-188. [PMID: 28993190 DOI: 10.1016/j.bbadis.2017.10.009] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 08/08/2017] [Accepted: 10/05/2017] [Indexed: 12/17/2022]
Abstract
Studies have indicated that the definitive engraftment and transdifferentiation potential of stem cells do not seem crucial for its property of tissue repair. Our previous study showed that transplantation of adipose-derived mesenchymal stem cells (ADMSCs) enhanced the healing of sutured gastric perforation. This study aimed to investigate the paracrine role of ADMSCs in the experimental gastric mucosal injury. Normoxia-conditioned medium (Nor CM) and hypoxia (HPO) CM were obtained after culturing ADMSCs in 20% O2 and 5% O2 for 48h. Cell migration, proliferation, viability, and angiogenesis in vitro were significantly enhanced upon incubation with CM, especially the HPO CM. Experiments in vivo using a rodent model of gastric ulcer demonstrated that HPO CM treatment significantly accelerated wound healing by suppressing inflammation and promoting neovascularization and re-epithelization. Meanwhile, the infusion of HPO CM activated the COX2-PGE2 axis both in vitro and in vivo. And the upregulation of COX2 was further dependent on the activation of ErK1/2-MAPK pathway. In addition, vascular endothelial growth factor, tissue inhibitors of metalloproteinases-1, and chemokine (C-C motif) ligand 20 (CCL-20) were analyzed as being highly abundant factors secreted by ADMSCs under hypoxic condition. Notably, the blockade of CCL-20 abrogated the HPO CM-induced COX2 signaling in the primary gastric mucosal epithelial cells, while incubation with recombinant CCL-20 increased the expression of COX2. In conclusion, the secretome from hypoxia-conditioned ADMSCs facilitates the repair of gastric mucosal injury through the enhancement of angiogenesis and re-epithelization, as well as the activation of COX2-PGE2 axis with a paracrine activity involving CCL-20 factor.
Collapse
Affiliation(s)
- Xianfeng Xia
- Department of Surgery, Institute of Digestive Disease, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong; Chow Yuk Ho Technology Center for Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Philip Wai Yan Chiu
- Department of Surgery, Institute of Digestive Disease, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong; Chow Yuk Ho Technology Center for Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong.
| | - Ping Kuen Lam
- Chow Tai Fook-Cheng Yu Tung Surgical Stem Cell Research Centre, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Wai Ching Chin
- Chow Tai Fook-Cheng Yu Tung Surgical Stem Cell Research Centre, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Enders Kwok Wai Ng
- Department of Surgery, Institute of Digestive Disease, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - James Yun Wong Lau
- Department of Surgery, Institute of Digestive Disease, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong; Chow Yuk Ho Technology Center for Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| |
Collapse
|
35
|
Muscella A, Cossa LG, Vetrugno C, Antonaci G, Marsigliante S. Inhibition of ZL55 cell proliferation by ADP via PKC-dependent signalling pathway. J Cell Physiol 2017; 233:2526-2536. [PMID: 28777435 DOI: 10.1002/jcp.26128] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 08/01/2017] [Indexed: 11/09/2022]
Abstract
Extracellular nucleotides can regulate cell proliferation in both normal and tumorigenic tissues. Here, we studied how extracellular nucleotides regulate the proliferation of ZL55 cells, a mesothelioma-derived cell line obtained from bioptic samples of asbestos-exposed patients. ADP and 2-MeS-ADP inhibited ZL55 cell proliferation, whereas ATP, UTP, and UDP were inactive. The nucleotide potency profile and the blockade of the ADP-mediated inhibitory effect by the phospholipase C inhibitor U-73122 suggest that P2Y1 receptor controls ZL55 cell proliferation. The activation of P2Y1 receptor by ADP leads to activation of intracellular transduction pathways involving [Ca2+ ]i , PKC-δ/PKC-α, and MAPKs, ERK1/2 and JNK1/2. Cell treatment with ADP or 2-MeS-ADP also provokes the activation of p53, causing an accumulation of the G1 cyclin-dependent kinase inhibitors p21WAF1 and p27Kip . Inhibition of ZL55 cell proliferation by ADP was completely reversed by inhibiting MEK1/2, or JNK1/2, or PKC-δ, and PKC-α. Through the inhibition of ADP-activated transductional kinases it was found that PKC-δ was responsible for JNK1/2 activation. JNK1/2 has a role in transcriptional up-regulation of p53, p21WAF1/CIP1 , and p27kip1 . Conversely, the ADP-activated PKC-α provoked ERK1/2 phosphorylation. ERK1/2 increased p53 stabilization, required to G1 arrest of ZL55 cells. Concluding, the importance of the study is twofold: first, results shed light on the mechanism of cell cycle inhibition by ADP; second, results suggest that extracellular ADP may inhibit mesothelioma progression.
Collapse
Affiliation(s)
- Antonella Muscella
- Dipartimento di Scienze e Tecnologie Biologiche e Ambientali (Di.S.Te.B.A.), Universita' del Salento, Lecce, Italy
| | - Luca G Cossa
- Dipartimento di Scienze e Tecnologie Biologiche e Ambientali (Di.S.Te.B.A.), Universita' del Salento, Lecce, Italy
| | - Carla Vetrugno
- Dipartimento di Scienze e Tecnologie Biologiche e Ambientali (Di.S.Te.B.A.), Universita' del Salento, Lecce, Italy
| | - Giovanna Antonaci
- Dipartimento di Scienze e Tecnologie Biologiche e Ambientali (Di.S.Te.B.A.), Universita' del Salento, Lecce, Italy
| | - Santo Marsigliante
- Dipartimento di Scienze e Tecnologie Biologiche e Ambientali (Di.S.Te.B.A.), Universita' del Salento, Lecce, Italy
| |
Collapse
|
36
|
Apoptosis by [Pt(O,O'-acac)(γ-acac)(DMS)] requires PKC-δ mediated p53 activation in malignant pleural mesothelioma. PLoS One 2017; 12:e0181114. [PMID: 28704484 PMCID: PMC5507537 DOI: 10.1371/journal.pone.0181114] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 06/25/2017] [Indexed: 12/29/2022] Open
Abstract
Mesothelioma cancer cells have epithelioid or sarcomatoid morphology. The worst prognosis is associated with sarcomatoid phenotype and resistance to therapy is affected by cells heterogeneity. We recently showed that in ZL55 mesothelioma cell line of epithelioid origin [Pt(O,O'-acac)(γ-acac)(DMS)] (Ptac2S) has an antiproliferative effect in vitro and in vivo. Aim of this work was to extend the study on the effects of Ptac2S on ZL34 cell line, representative of sarcomatoid mesothelioma. ZL34 cells were used to assay the antitumor activity of Ptac2S in a mouse xenograft model in vivo. Then, both ZL34 and ZL55 cells were used in order to assess the involvement of p53 protein in (a) the processes underlying the sensitivity to chemotherapy and (b) the activation of various transduction proteins involved in apoptosis/survival processes. Ptac2S increases ZL34 cell death in vivo compared with cisplatin and, in vitro, Ptac2S was more efficacious than cisplatin in inducing apoptosis. In Ptac2S-treated ZL34 and ZL55 cells, p53 regulated gene products of apoptotic BAX and anti-apoptotic Bcl-2 proteins via transcriptional activation. Ptac2S activated PKC-δ and PKC-ε; their inhibition by PKC-siRNA decreased the apoptotic death of cells. PKC-δ was responsible for JNK1/2 activation that has a role in p53 activation. In addition, PKC-ε activation provoked phosphorylation of p38MAPK, concurring to apoptosis. In ZL34 cells, Ptac2S also activated PKC-α thus provoking ERK1/2 activation; inhibition of PKC-α, or ERK1/2, increased Ptac2S cytotoxicity. Results confirm that Ptac2S is a promising therapeutic agent for malignant mesothelioma, giving a substantial starting point for its further validation.
Collapse
|
37
|
Muscella A, Vetrugno C, Marsigliante S. CCL20 promotes migration and invasiveness of human cancerous breast epithelial cells in primary culture. Mol Carcinog 2017; 56:2461-2473. [PMID: 28618084 DOI: 10.1002/mc.22693] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 05/25/2017] [Accepted: 06/13/2017] [Indexed: 11/06/2022]
Abstract
The relation between the tumor and its microenvironment is one of the most interesting and less understood issues. Recently, we showed a role of CCL20 chemokine in proning the healthy tissue neighboring the tumor to carcinogenesis. Besides, tumor-secreted CCL20 induced proliferation, migration, and EMT of healthy cells. In this context, we have studied here if CCL20 had effects on the migration of cancer cells and the intracellular pathways used in breast epithelial cells in primary culture. Using molecular (siRNA) and pharmacological (inhibitors) techniques, we found multiple signaling kinases to be activated and involved in CCL20-induced tumor breast cell migration. CCL20 provoked a 2.5-fold increase of cell migration and invasion; CCL20 also enhanced MMP- 2 and MMP-9 mRNAs/protein expression and activities. Cell migration and invasiveness due to CCL20 significantly decreased when MMP-2 and MMP-9 were inhibited in CCL20-stimulated cells. CCL20 controlled MMP-2 expression through the JAK2/STAT3 pathway, while the expression of MMP-9 occurred by PKC-α that activated, consequently, c-Src, Akt, and finally NF-kB. These results reveal a role for CCL20 also in tumor breast cell and point to CCL20 as a novel therapeutic target in cancer.
Collapse
Affiliation(s)
- Antonella Muscella
- Dipartimento di Scienze e Tecnologie Biologiche e Ambientali (Di.S.Te.B.A.), Università del Salento, Lecce, Italy
| | - Carla Vetrugno
- Dipartimento di Scienze e Tecnologie Biologiche e Ambientali (Di.S.Te.B.A.), Università del Salento, Lecce, Italy
| | - Santo Marsigliante
- Dipartimento di Scienze e Tecnologie Biologiche e Ambientali (Di.S.Te.B.A.), Università del Salento, Lecce, Italy
| |
Collapse
|
38
|
Lin J, Kato M, Nagata K, Okuwaki M. Efficient DNA binding of NF-κB requires the chaperone-like function of NPM1. Nucleic Acids Res 2017; 45:3707-3723. [PMID: 28003476 PMCID: PMC5397172 DOI: 10.1093/nar/gkw1285] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 11/21/2016] [Accepted: 12/13/2016] [Indexed: 12/11/2022] Open
Abstract
NPM1/nucleophosmin is frequently overexpressed in various tumors, although the oncogenic role of NPM1 remains unclear. Here we revealed the link between NPM1 and nuclear factor-κB (NF-κB), a master regulator of inflammation. We found that NPM1 knockdown decreased NF-κB-mediated transcription of selected target genes by decreasing the recruitment of NF-κB p65 to the gene promoters. NPM1 is directly associated with the DNA binding domain of p65 to enhance its DNA binding activity without being a part of the DNA-NF-κB complex. This result suggests that NF-κB requires the chaperone-like function of NPM1 for DNA binding. Furthermore, we demonstrated that NPM1 was required for efficient inflammatory gene expression induced by tumor necrosis factor alpha (TNF-α) and lipopolysaccharide in fibroblasts and macrophages. The NF-κB-mediated invasion of breast cancer cells was significantly decreased by NPM1 knockdown. Our study suggests a novel mechanistic insight into the NF-κB-mediated transcription and an oncogenic role of NPM1 in both tumor cells and the tumor micro-environment through the regulation of NF-κB.
Collapse
Affiliation(s)
- Jianhuang Lin
- PhD Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, 1-1-1, Tennodai, Tsukuba 305-8575 Japan
- Department of Infection Biology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba 305-8575 Japan
| | - Mitsuyasu Kato
- PhD Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, 1-1-1, Tennodai, Tsukuba 305-8575 Japan
- Department of Experimental Pathology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba 305-8575 Japan
| | - Kyosuke Nagata
- Department of Infection Biology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba 305-8575 Japan
| | - Mitsuru Okuwaki
- PhD Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, 1-1-1, Tennodai, Tsukuba 305-8575 Japan
- Department of Infection Biology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba 305-8575 Japan
| |
Collapse
|
39
|
Lin B, Zhang L, Li D, Sun H. MED23 in endocrinotherapy for breast cancer. Oncol Lett 2017; 13:4679-4684. [PMID: 28588722 PMCID: PMC5452902 DOI: 10.3892/ol.2017.6036] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 02/21/2017] [Indexed: 12/29/2022] Open
Abstract
We investigated the role of the transcriptional mediator subunit 23 (MED23) in everolimus drug resistance, invasion and metastasis during breast cancer treatment and its molecular mechanism. We also evaluated the endocrinotherapy and prevention method for breast cancer. Breast cancer cell strains were established that can continuously express MED23, as well as inducible MED23-shRNA expression plasmids. The inductive agent, doxycycline (Dox), was added to the water for long-term silencing of MED23 in intratumoral cells. We conducted experiments on the role of MED23 in the regulation of invasion and metastasis of breast cancer using cell culture, western blotting, MTT proliferation experiment, fluorescent quantitative PCR and chromatin immunoprecipitation (ChIP). The silencing of MED23 significantly inhibited cellular growth and proliferation as well as soft agar cloning. Silencing of MED23 strengthened the sensitivity of the everolimus-resistant breast cancer cell strains BT474 and MCF-7/ADM cells to everolimus medication. The silencing of MED23, in combination with everolimus, inhibits the cell cycle progress of breast cancer cells. ChIP indicated that the mutual regulation of HER2 and MED23 also participates in the formation of the everolimus drug resistance mechanism. Therefore, MED23 plays an important role in everolimus drug resistance, invasion, and metastasis of breast cancer. As a potential molecular therapeutic target of breast cancer, MED23 overcomes drug resistance in clinical endocrinotherapy and controls the distal relapse and metastasis in breast cancer by the targeted silencing of MED23.
Collapse
Affiliation(s)
- Benrui Lin
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Lan Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Dinuo Li
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Hongzhi Sun
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| |
Collapse
|
40
|
Phospholipase Cε deficiency delays the early stage of cutaneous wound healing and attenuates scar formation in mice. Biochem Biophys Res Commun 2017; 484:144-151. [PMID: 28093232 DOI: 10.1016/j.bbrc.2017.01.054] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/09/2017] [Accepted: 01/11/2017] [Indexed: 12/31/2022]
Abstract
This study aimed to investigate the role of phospholipase Cε (PLCε) in the skin wound healing process. PLCε, an effect factor of Ras/Rap small G protein, plays a crucial role in skin inflammation by regulating inflammatory cytokines. Inflammatory responses are closely associated with wound healing. Full-thickness skin wounds were made in the PLCε knockout (KO) and wild-type (WT) mice, and the healing process was analyzed. The macroscopic wound closure rate declined in the PLCε KO mice on days 3, 4, and 5 after wounding, following the decreased expression of interleukin (IL)-6, chemokine (C-X-C motif) ligand (Cxcl)-1, Cxcl-2, and chemokine (C-C motif) ligand (Ccl) 20. The proliferation rate of epidermal keratinocytes was not affected by PLCε, but silencing of PLCε resulted in the delayed migration of keratinocytes. Moreover, the scars were found to be much smaller in the PLCε KO mice than in the WT mice. The mRNA expression of Ccl20, collagen (Col) 6a1, and Col17a1 decreased in the PLCε KO mice. These results were in agreement with a previous hypothesis that PLCε might delay the early stage of cutaneous wound healing by inhibiting the migration of keratinocytes, and decrease the expression of Col6a1, Col17a1, and Ccl20 by inhibiting the inflammatory response to reduce scar formation. This study shed light on a novel role of PLCε in wound healing and provided new therapeutic approaches to target PLCε for diminishing scar formation after injury.
Collapse
|
41
|
King J, Mir H, Singh S. Association of Cytokines and Chemokines in Pathogenesis of Breast Cancer. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 151:113-136. [DOI: 10.1016/bs.pmbts.2017.07.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
42
|
Luo J, Xiang G, Pan C. Discovery of microRNAs and Transcription Factors Co-Regulatory Modules by Integrating Multiple Types of Genomic Data. IEEE Trans Nanobioscience 2017; 16:51-59. [DOI: 10.1109/tnb.2017.2649560] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
43
|
Lacalle RA, Blanco R, Carmona-Rodríguez L, Martín-Leal A, Mira E, Mañes S. Chemokine Receptor Signaling and the Hallmarks of Cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 331:181-244. [PMID: 28325212 DOI: 10.1016/bs.ircmb.2016.09.011] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The chemokines are a family of chemotactic cytokines that mediate their activity by acting on seven-transmembrane-spanning G protein-coupled receptors. Both the ability of the chemokines and their receptors to form homo- and heterodimers and the promiscuity of the chemokine-chemokine receptor interaction endow this protein family with enormous signaling plasticity and complexity that are not fully understood at present. Chemokines were initially identified as essential regulators of homeostatic and inflammatory trafficking of innate and adaptive leucocytes from lymphoid organs to tissues. Chemokines also mediate the host response to cancer. Nevertheless, chemokine function in this response is not limited to regulating leucocyte infiltration into the tumor microenvironment. It is now known that chemokines and their receptors influence most-if not all-hallmark processes of cancer; they act on both neoplastic and untransformed cells in the tumor microenvironment, including fibroblasts, endothelial cells (blood and lymphatic), bone marrow-derived stem cells, and, obviously, infiltrating leucocytes. This review begins with an overview of chemokine and chemokine receptor structure, to better define how chemokines affect the proliferation, survival, stemness, and metastatic potential of neoplastic cells. We also examine the main mechanisms by which chemokines regulate tumor angiogenesis and immune cell infiltration, emphasizing the pro- and antitumorigenic activity of this protein superfamily in these interrelated processes.
Collapse
Affiliation(s)
- R A Lacalle
- Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - R Blanco
- Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | | | - A Martín-Leal
- Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - E Mira
- Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - S Mañes
- Centro Nacional de Biotecnología/CSIC, Madrid, Spain.
| |
Collapse
|
44
|
Girard BJ, Knutson TP, Kuker B, McDowell L, Schwertfeger KL, Ostrander JH. Cytoplasmic Localization of Proline, Glutamic Acid, Leucine-rich Protein 1 (PELP1) Induces Breast Epithelial Cell Migration through Up-regulation of Inhibitor of κB Kinase ϵ and Inflammatory Cross-talk with Macrophages. J Biol Chem 2016; 292:339-350. [PMID: 27881676 DOI: 10.1074/jbc.m116.739847] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 11/22/2016] [Indexed: 01/06/2023] Open
Abstract
Cytoplasmic localization of proline, glutamic acid, leucine-rich protein 1 (PELP1) is observed in ∼40% of women with invasive breast cancer. In mouse models, PELP1 overexpression in the mammary gland leads to premalignant lesions and eventually mammary tumors. In preliminary clinical studies, cytoplasmic localization of PELP1 was seen in 36% of women at high risk of developing breast cancer. Here, we investigated whether cytoplasmic PELP1 signaling promotes breast cancer initiation in models of immortalized human mammary epithelial cells (HMECs). Global gene expression analysis was performed on HMEC lines expressing vector control, PELP1-wt, or mutant PELP1 in which the nuclear localization sequence was altered, resulting in cytoplasmic localization of PELP1 (PELP1-cyto). Global gene expression analysis identified that PELP1-cyto expression in HMECs induced NF-κB signaling pathways. Western blotting analysis of PELP1-cyto HMECs showed up-regulation of inhibitor of κB kinase ϵ (IKKϵ) and increased phosphorylation of the NF-κB subunit RelB. To determine whether secreted factors produced by PELP1-cyto HMECs promote macrophage activation, THP-1 macrophages were treated with HMEC-conditioned medium (CM). PELP1-cyto CM induced changes in THP-1 gene expression as compared with control cell CM. Double conditioned medium (DCM) from the activated THP-1 cells was then applied to HMECs to determine whether paracrine signaling from PELP1-cyto-activated macrophages could in turn promote migration of HMECs. PELP1-cyto DCM induced robust HMEC migration, which was reduced in DCM from PELP1-cyto HMECs expressing IKKϵ shRNA. Our findings suggest that cytoplasmic localization of PELP1 up-regulates pro-tumorigenic IKKϵ and secreted inflammatory signals, which through paracrine macrophage activation regulates the migratory phenotype associated with breast cancer initiation.
Collapse
Affiliation(s)
| | | | | | | | - Kathryn L Schwertfeger
- From the Masonic Cancer Center and.,Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota 55455
| | | |
Collapse
|
45
|
Mejía-Rangel J, Córdova E, Orozco L, Ventura-Gallegos JL, Mitre-Aguilar I, Escalona-Guzmán A, Vadillo F, Vázquez-Prado J, Gariglio P, Zentella-Dehesa A. Pro-adhesive phenotype of normal endothelial cells responding to metastatic breast cancer cell conditioned medium is linked to NFκB-mediated transcriptomic regulation. Int J Oncol 2016; 49:2173-2185. [DOI: 10.3892/ijo.2016.3705] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 07/26/2016] [Indexed: 11/06/2022] Open
|
46
|
Doucet M, Jayaraman S, Swenson E, Tusing B, Weber KL, Kominsky SL. CCL20/CCR6 Signaling Regulates Bone Mass Accrual in Mice. J Bone Miner Res 2016; 31:1381-90. [PMID: 26890063 DOI: 10.1002/jbmr.2813] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 02/03/2016] [Accepted: 02/13/2016] [Indexed: 11/10/2022]
Abstract
CCL20 is a member of the macrophage inflammatory protein family and is reported to signal monogamously through the receptor CCR6. Although studies have identified the genomic locations of both Ccl20 and Ccr6 as regions important for bone quality, the role of CCL20/CCR6 signaling in regulating bone mass is unknown. By micro-computed tomography (μCT) and histomorphometric analysis, we show that global loss of Ccr6 in mice significantly decreases trabecular bone mass coincident with reduced osteoblast numbers. Notably, CCL20 and CCR6 were co-expressed in osteoblast progenitors and levels increased during osteoblast differentiation, indicating the potential of CCL20/CCR6 signaling to influence osteoblasts through both autocrine and paracrine actions. With respect to autocrine effects, CCR6 was found to act as a functional G protein-coupled receptor in osteoblasts and although its loss did not appear to affect the number or proliferation rate of osteoblast progenitors, differentiation was significantly inhibited as evidenced by delays in osteoblast marker gene expression, alkaline phosphatase activity, and mineralization. In addition, CCL20 promoted osteoblast survival concordant with activation of the PI3K-AKT pathway. Beyond these potential autocrine effects, osteoblast-derived CCL20 stimulated the recruitment of macrophages and T cells, known facilitators of osteoblast differentiation and survival. Finally, we generated mice harboring a global deletion of Ccl20 and found that Ccl20(-/-) mice exhibit a reduction in bone mass similar to that observed in Ccr6(-/-) mice, confirming that this phenomenon is regulated by CCL20 rather than alternate CCR6 ligands. Collectively, these data indicate that CCL20/CCR6 signaling may play an important role in regulating bone mass accrual, potentially by modulating osteoblast maturation, survival, and the recruitment of osteoblast-supporting cells. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Michele Doucet
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Swaathi Jayaraman
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Emily Swenson
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Brittany Tusing
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kristy L Weber
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Scott L Kominsky
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
47
|
Li Q, Ma Z, Liu Y, Kan X, Wang C, Su B, Li Y, Zhang Y, Wang P, Luo Y, Na D, Wang L, Zhang G, Zhu X, Wang L. Low doses of paclitaxel enhance liver metastasis of breast cancer cells in the mouse model. FEBS J 2016; 283:2836-52. [DOI: 10.1111/febs.13767] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 05/13/2016] [Accepted: 05/30/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Qi Li
- Institute of Chinese Materia Medica; China Academy of Chinese Medical Sciences; Beijing China
| | - Zhuang Ma
- Department of Immunology; Center for Human Disease Genomics; School of Basic Medical Science; Peking University Health Science Centre; Beijing China
| | - Yinhua Liu
- Surgery Department; Peking University First Hospital; Beijing China
| | - Xiaoxi Kan
- Institute of Chinese Materia Medica; China Academy of Chinese Medical Sciences; Beijing China
| | - Changjun Wang
- Department of Immunology; Center for Human Disease Genomics; School of Basic Medical Science; Peking University Health Science Centre; Beijing China
| | - Bingnan Su
- Department of Immunology; Center for Human Disease Genomics; School of Basic Medical Science; Peking University Health Science Centre; Beijing China
| | - Yuchen Li
- Department of Immunology; Center for Human Disease Genomics; School of Basic Medical Science; Peking University Health Science Centre; Beijing China
| | - Yingmei Zhang
- Department of Immunology; Center for Human Disease Genomics; School of Basic Medical Science; Peking University Health Science Centre; Beijing China
| | - Pingzhang Wang
- Department of Immunology; Center for Human Disease Genomics; School of Basic Medical Science; Peking University Health Science Centre; Beijing China
| | - Yang Luo
- Department of Immunology; Center for Human Disease Genomics; School of Basic Medical Science; Peking University Health Science Centre; Beijing China
| | - Daxiang Na
- Department of Immunology; Center for Human Disease Genomics; School of Basic Medical Science; Peking University Health Science Centre; Beijing China
| | - Lanlan Wang
- Department of Immunology; Center for Human Disease Genomics; School of Basic Medical Science; Peking University Health Science Centre; Beijing China
| | - Guoying Zhang
- Department of Immunology; Center for Human Disease Genomics; School of Basic Medical Science; Peking University Health Science Centre; Beijing China
| | - Xiaoxin Zhu
- Institute of Chinese Materia Medica; China Academy of Chinese Medical Sciences; Beijing China
| | - Lu Wang
- Department of Immunology; Center for Human Disease Genomics; School of Basic Medical Science; Peking University Health Science Centre; Beijing China
| |
Collapse
|
48
|
Fluctuations in Blood Marginal Zone B-Cell Frequencies May Reflect Migratory Patterns Associated with HIV-1 Disease Progression Status. PLoS One 2016; 11:e0155868. [PMID: 27203285 PMCID: PMC4874627 DOI: 10.1371/journal.pone.0155868] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 05/05/2016] [Indexed: 12/29/2022] Open
Abstract
We have previously shown that overexpression of BLyS/BAFF was associated with increased relative frequencies of innate “precursor” marginal zone (MZ)-like B-cells in the blood of HIV-1-infected rapid and classic progressors. However, along with relatively normal BLyS/BAFF expression levels, these cells remain unaltered in elite-controllers (EC), rather, percentages of more mature MZ-like B-cells are decreased in the blood of these individuals. Fluctuations in frequencies of blood MZ-like B-cell populations may reflect migratory patterns associated with disease progression status, suggesting an important role for these cells in HIV-1 pathogenesis. We have therefore longitudinally measured plasma levels of B-tropic chemokines by ELISA-based technology as well as their ligands by flow-cytometry on blood B-cell populations of HIV-1-infected individuals with different rates of disease progression and uninfected controls. Migration potential of B-cell populations from these individuals were determined by chemotaxis assays. We found important modulations of CXCL13-CXCR5, CXCL12-CXCR4/CXCR7, CCL20-CCR6 and CCL25-CCR9 chemokine-axes and increased cell migration patterns in HIV progressors. Interestingly, frequencies of CCR6 expressing cells were significantly elevated within the precursor MZ-like population, consistent with increased migration in response to CCL20. Although we found little modulation of chemokine-axes in EC, cell migration was greater than that observed for uninfected controls, especially for MZ-like B-cells. Overall the immune response against HIV-1 may involve recruitment of MZ-like B-cells to peripheral sites. Moreover, our findings suggest that “regulated” attraction of these cells in a preserved BLyS/BAFF non-inflammatory environment, such as encountered in EC could be beneficial to the battle and even control of HIV.
Collapse
|
49
|
Wang B, Shi L, Sun X, Wang L, Wang X, Chen C. Production of CCL20 from lung cancer cells induces the cell migration and proliferation through PI3K pathway. J Cell Mol Med 2016; 20:920-9. [PMID: 26968871 PMCID: PMC4831357 DOI: 10.1111/jcmm.12781] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 12/07/2015] [Indexed: 11/28/2022] Open
Abstract
Tumour inflammatory microenvironment is considered to play a role in the sensitivity of tumour cells to therapies and prognosis of patients with lung cancer. The expression of CCL20, one of the critical chemoattractants responsible for inflammation cells recruitment, has been shown overexpressed in variety of tumours. This study aimed at investigating potential mechanisms of CCL20 function and production in human non-small cell lung cancer (NSCLC). Expression of CCL20 gene and protein in lung tissues of patients with NSCLC and NSCLC cells (A549) were determined. The interleukin (IL)-1β-induced signal pathways in A549 and the effect of CCL20-induced A549 cell migration and proliferation were determined using migration assays and cell-alive monitoring system. Mechanisms of signal pathways involved in the migration of CCL20 were also studied. We initially found that NSCLC tumour tissues markedly overexpressed CCL20 in comparison with normal lung samples. In addition, IL-1β could directly promote CCL20 production in lung cancer cells, which was inhibited by extracellular signal-regulated kinase (ERK)1/2 inhibitor, p38 mitogen-activated protein kinase (p38 MARP) inhibitor or PI3K inhibitors. CCL20 promoted lung cancer cells migration and proliferation in an autocrine manner via activation of ERK1/2-MAPK and PI3K pathways. Our data indicated that IL-1β could stimulate CCL20 production from lung cancer cells through the activation of MAPKs and PI3K signal pathways, and the auto-secretion of CCL20 could promote lung cancer cell migration and proliferation through the activation of ERK and PI3K signal pathways. Our results may provide a novel evidence that CCL20 could be a new therapeutic target for lung cancer.
Collapse
Affiliation(s)
- Beibei Wang
- Department of Lung Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Lin Shi
- Zhongshan Hospital Biomedical Research Center, Shanghai Institute of Clinical Bioinformatics, Fudan University Center for Clinical Bioinformatics, Fudan University Shanghai Medical College, Shanghai, China
| | - Xiaoru Sun
- Department of Lung Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Lingyan Wang
- Zhongshan Hospital Biomedical Research Center, Shanghai Institute of Clinical Bioinformatics, Fudan University Center for Clinical Bioinformatics, Fudan University Shanghai Medical College, Shanghai, China
| | - Xiangdong Wang
- Department of Lung Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
- Zhongshan Hospital Biomedical Research Center, Shanghai Institute of Clinical Bioinformatics, Fudan University Center for Clinical Bioinformatics, Fudan University Shanghai Medical College, Shanghai, China
| | - Chengshui Chen
- Department of Lung Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
50
|
Khalil BD, Hsueh C, Cao Y, Abi Saab WF, Wang Y, Condeelis JS, Bresnick AR, Backer JM. GPCR Signaling Mediates Tumor Metastasis via PI3Kβ. Cancer Res 2016; 76:2944-53. [PMID: 27013201 DOI: 10.1158/0008-5472.can-15-1675] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 02/23/2016] [Indexed: 12/16/2022]
Abstract
Inappropriate activation of PI3K signaling has been implicated strongly in human cancer. Although studies on the role of PI3K signaling in breast tumorigenesis and progression have focused most intensively on PI3Kα, a role for PI3Kβ has begun to emerge. The PI3Kβ isoform is unique among class IA PI3K enzymes in that it is activated by both receptor tyrosine kinases and G-protein-coupled receptors (GPCR). In previous work, we identified a mutation that specifically abolishes PI3Kβ binding to Gβγ (p110(526KK-DD)). Expression of this mutant in p110β-silenced breast cancer cells inhibits multiple steps of the metastatic cascade in vitro and in vivo and causes a cell autonomous defect in invadopodial matrix degradation. Our results identify a novel link between GPCRs and PI3Kβ in mediating metastasis, suggesting that disruption of this link might offer a novel therapeutic target to prevent the development of metastatic disease. Cancer Res; 76(10); 2944-53. ©2016 AACR.
Collapse
Affiliation(s)
- Bassem D Khalil
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York
| | - Christine Hsueh
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York
| | - Yanyan Cao
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York
| | - Widian F Abi Saab
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York
| | - Yarong Wang
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York
| | - John S Condeelis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Anne R Bresnick
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York.
| | - Jonathan M Backer
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York. Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York.
| |
Collapse
|