1
|
Wang T, Sheng J, Wang X, Zhu M, Li S, Shen Y, Wu B. CXCL5 Promotes the Malignant Phenotype of Pancreatic Cancer and Is Associated With Immune Infiltration. Clin Med Insights Oncol 2024; 18:11795549241271691. [PMID: 39211563 PMCID: PMC11359438 DOI: 10.1177/11795549241271691] [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: 11/18/2023] [Accepted: 06/27/2024] [Indexed: 09/04/2024] Open
Abstract
Background The significance of CXCL5 in pancreatic cancer is unclear, although it has been implicated in the malignant process of many different types of cancer. Research on the impact of CXCL5 on immune cell infiltration and the malignant phenotype of pancreatic cancer is needed. This study aimed to examine the connection between CXCL5 expression and immune cell infiltration and the malignant phenotype of pancreatic cancer. Methods Tissue samples and clinical information were collected from 90 patients with pancreatic cancer. Tumour tissues and adjacent tissues were made into a tissue microarray and stained for immunohistochemistry analysis. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and Western blot analysis were performed to measure the expression level of CXCL5. CXCL5-overexpressing/CXCL5-knockdown cell lines were constructed via transfection for cytological experiments. CCK-8, cell apoptosis, cell cycle, cell invasion, and cell colony formation assays were used to detect the effect of CXCL5 on the malignant phenotype of pancreatic cancer cells. Finally, a mouse model of pancreatic cancer was constructed for in vivo verification. Results Compared with control cells, pancreatic cancer cells overexpressing CXCL5 exhibited increased proliferation, migration, and invasion but decreased apoptosis. Conversely, knockdown of CXCL5 did not enhance the malignant phenotype of pancreatic cancer cells. Spearman correlation analysis indicated that there was a significant negative correlation between CXCL5 levels and the CD8 IRS. However, there was a significant positive correlation between FOXP3 IRS and CXCL5 levels. Conclusions CXCL5 is highly expressed in pancreatic cancer and promotes the malignant phenotype of pancreatic cancer cells. CXCL5 is associated with immunosuppressive FOXP3 + T-cell infiltration, which facilitates the formation of an immunosuppressive microenvironment (with low CD8 + T-cell infiltration).
Collapse
Affiliation(s)
- Tao Wang
- Graduate School, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jian Sheng
- Department of Science and Education, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Xiaoguang Wang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Minyuan Zhu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Shijun Li
- Graduate School, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yiyu Shen
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Bin Wu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
2
|
Tatsuno R, Komohara Y, Pan C, Kawasaki T, Enomoto A, Jubashi T, Kono H, Wako M, Ashizawa T, Haro H, Ichikawa J. Surface Markers and Chemokines/Cytokines of Tumor-Associated Macrophages in Osteosarcoma and Other Carcinoma Microenviornments-Contradictions and Comparisons. Cancers (Basel) 2024; 16:2801. [PMID: 39199574 PMCID: PMC11353089 DOI: 10.3390/cancers16162801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/01/2024] [Accepted: 08/07/2024] [Indexed: 09/01/2024] Open
Abstract
Osteosarcoma (OS) is the most common primary bone tumor in children and adolescents. Prognosis is improving with advances in multidisciplinary treatment strategies, but the development of new anticancer agents has not, and improvement in prognosis for patients with pulmonary metastases has stalled. In recent years, the tumor microenvironment (TME) has gained attention as a therapeutic target for cancer. The immune component of OS TME consists mainly of tumor-associated macrophages (TAMs). They exhibit remarkable plasticity, and their phenotype is influenced by the TME. In general, surface markers such as CD68 and CD80 show anti-tumor effects, while CD163 and CD204 show tumor-promoting effects. Surface markers have potential value as diagnostic and prognostic biomarkers. The cytokines and chemokines produced by TAMs promote tumor growth and metastasis. However, the role of TAMs in OS remains unclear to date. In this review, we describe the role of TAMs in OS by focusing on TAM surface markers and the TAM-produced cytokines and chemokines in the TME, and by comparing their behaviors in other carcinomas. We found contrary results from different studies. These findings highlight the urgency for further research in this field to improve the stalled OS prognosis percentages.
Collapse
Affiliation(s)
- Rikito Tatsuno
- Department of Orthopaedic Surgery, University of Yamanashi, Yamanashi 400-0016, Japan; (R.T.); (T.J.); (H.K.); (M.W.); (T.A.); (H.H.)
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8555, Japan; (Y.K.); (C.P.)
| | - Cheng Pan
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8555, Japan; (Y.K.); (C.P.)
| | - Tomonori Kawasaki
- Department of Pathology, Saitama Medical University International Medical Center, Saitama 350-1298, Japan;
| | - Atsushi Enomoto
- Department of Pathology, Graduate School of Medicine, Nagoya University, Nagoya 464-8601, Japan;
| | - Takahiro Jubashi
- Department of Orthopaedic Surgery, University of Yamanashi, Yamanashi 400-0016, Japan; (R.T.); (T.J.); (H.K.); (M.W.); (T.A.); (H.H.)
| | - Hiroyuki Kono
- Department of Orthopaedic Surgery, University of Yamanashi, Yamanashi 400-0016, Japan; (R.T.); (T.J.); (H.K.); (M.W.); (T.A.); (H.H.)
| | - Masanori Wako
- Department of Orthopaedic Surgery, University of Yamanashi, Yamanashi 400-0016, Japan; (R.T.); (T.J.); (H.K.); (M.W.); (T.A.); (H.H.)
| | - Tomoyuki Ashizawa
- Department of Orthopaedic Surgery, University of Yamanashi, Yamanashi 400-0016, Japan; (R.T.); (T.J.); (H.K.); (M.W.); (T.A.); (H.H.)
| | - Hirotaka Haro
- Department of Orthopaedic Surgery, University of Yamanashi, Yamanashi 400-0016, Japan; (R.T.); (T.J.); (H.K.); (M.W.); (T.A.); (H.H.)
| | - Jiro Ichikawa
- Department of Orthopaedic Surgery, University of Yamanashi, Yamanashi 400-0016, Japan; (R.T.); (T.J.); (H.K.); (M.W.); (T.A.); (H.H.)
| |
Collapse
|
3
|
Park MN. Therapeutic Strategies for Pancreatic-Cancer-Related Type 2 Diabetes Centered around Natural Products. Int J Mol Sci 2023; 24:15906. [PMID: 37958889 PMCID: PMC10648679 DOI: 10.3390/ijms242115906] [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: 08/25/2023] [Revised: 10/23/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), a highly malignant neoplasm, is classified as one of the most severe and devastating types of cancer. PDAC is a notable malignancy that exhibits a discouraging prognosis and a rising occurrence. The interplay between diabetes and pancreatic cancer exhibits a reciprocal causation. The identified metabolic disorder has been observed to possess noteworthy consequences on health outcomes, resulting in elevated rates of morbidity. The principal mechanisms involve the suppression of the immune system, the activation of pancreatic stellate cells (PSCs), and the onset of systemic metabolic disease caused by dysfunction of the islets. From this point forward, it is important to recognize that pancreatic-cancer-related diabetes (PCRD) has the ability to increase the likelihood of developing pancreatic cancer. This highlights the complex relationship that exists between these two physiological states. Therefore, we investigated into the complex domain of PSCs, elucidating their intricate signaling pathways and the profound influence of chemokines on their behavior and final outcome. In order to surmount the obstacle of drug resistance and eliminate PDAC, researchers have undertaken extensive efforts to explore and cultivate novel natural compounds of the next generation. Additional investigation is necessary in order to comprehensively comprehend the effect of PCRD-mediated apoptosis on the progression and onset of PDAC through the utilization of natural compounds. This study aims to examine the potential anticancer properties of natural compounds in individuals with diabetes who are undergoing chemotherapy, targeted therapy, or immunotherapy. It is anticipated that these compounds will exhibit increased potency and possess enhanced pharmacological benefits. According to our research findings, it is indicated that naturally derived chemical compounds hold potential in the development of PDAC therapies that are both safe and efficacious.
Collapse
Affiliation(s)
- Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemungu, Seoul 05253, Republic of Korea
| |
Collapse
|
4
|
Cheng T, Agwu C, Shim K, Wang B, Jain S, Mahjoub MR. Aberrant centrosome biogenesis disrupts nephron progenitor cell renewal and fate resulting in fibrocystic kidney disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.04.535568. [PMID: 37066373 PMCID: PMC10104032 DOI: 10.1101/2023.04.04.535568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Mutations that disrupt centrosome structure or function cause congenital kidney developmental defects and fibrocystic pathologies. Yet, it remains unclear how mutations in proteins essential for centrosome biogenesis impact embryonic kidney development. Here, we examined the consequences of conditional deletion of a ciliopathy gene, Cep120 , in the two nephron progenitor niches of the embryonic kidney. Cep120 loss led to reduced abundance of both metanephric mesenchyme and ureteric bud progenitor populations. This was due to a combination of delayed mitosis, increased apoptosis, and premature differentiation of progenitor cells. These defects resulted in dysplastic kidneys at birth, which rapidly formed cysts, displayed increased interstitial fibrosis, and decline in filtration function. RNA sequencing of embryonic and postnatal kidneys from Cep120-null mice identified changes in pathways essential for branching morphogenesis, cystogenesis and fibrosis. Our study defines the cellular and developmental defects caused by centrosome dysfunction during kidney development, and identifies new therapeutic targets for renal centrosomopathies. Highlights Defective centrosome biogenesis in nephron progenitors causes:Reduced abundance of metanephric mesenchyme and premature differentiation into tubular structuresAbnormal branching morphogenesis leading to reduced nephron endowment and smaller kidneysChanges in cell-autonomous and paracrine signaling that drive cystogenesis and fibrosisUnique cellular and developmental defects when compared to Pkd1 knockout models.
Collapse
|
5
|
Wang ZZ, Li XT, Li QJ, Zhou JX. Targeting CXCL5 in Pancreatic Cancer Cells Inhibits Cancer Xenograft Growth by Reducing Proliferation and Inhibiting EMT Progression. Dig Dis Sci 2023; 68:841-851. [PMID: 35650416 DOI: 10.1007/s10620-022-07529-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 04/18/2022] [Indexed: 12/09/2022]
Abstract
BACKGROUND Pancreatic cancer (PC) is the most lethal malignant tumor, with average survival period of about 10 months. C-X-C ligand 5 (CXCL5), an important chemokine for immune cell accumulation in tumor tissues, has been reported to be involved in a variety of human cancers. However, the exact role of CXCL5 in PC progression has not been well defined. METHODS The expression of CXCL5 in PC was analyzed based on online databases and clinical specimens immunohistochemical staining, and Western blotting of CXCL5 in PC cell lines and patient samples. The correlation between CXCL5 expression and prognosis in PC was explored. The role of CXCL5 in PC was investigated through in vitro and in vivo experiments. RESULTS The expression of CXCL5 was significantly increased in PC tissues compared with that in pancreas tissues, and CXCL5 high expression predicts poor prognosis in PC patients. Further analyses demonstrated that overexpression of CXCL5 in PC cells was positively related to higher proliferation rate, higher migration ability, and higher EMT markers including SNAI2 and TWIST1 of tumor cells in vitro. Consistently, the knockdown of CXCL5 in PC cells harmed the proliferation rate, migration ability, and expression of EMT indexes of tumor cells in vitro. Importantly, knockdown of CXCL5 inhibited the growth of xenograft tumors in vivo. CONCLUSION CXCL5 high expression predicts poor prognosis in PC patients. CXCL5 promotes PC cell growth and EMT process. Inhibition of CXCL5 may be a potential therapeutic approach for PC.
Collapse
Affiliation(s)
- Zheng-Zheng Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Xiao-Ting Li
- Department of Obstetrics and Gynecology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Qing-Jun Li
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Jin-Xue Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008, China.
| |
Collapse
|
6
|
Zeng J, Peng Y, Wang D, Ayesha K, Chen S. The interaction between osteosarcoma and other cells in the bone microenvironment: From mechanism to clinical applications. Front Cell Dev Biol 2023; 11:1123065. [PMID: 37206921 PMCID: PMC10189553 DOI: 10.3389/fcell.2023.1123065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/10/2023] [Indexed: 05/21/2023] Open
Abstract
Osteosarcoma is a primary bone tumor with a high mortality rate. The event-free survival rate has not improved significantly in the past 30 years, which brings a heavy burden to patients and society. The high heterogeneity of osteosarcoma leads to the lack of specific targets and poor therapeutic effect. Tumor microenvironment is the focus of current research, and osteosarcoma is closely related to bone microenvironment. Many soluble factors and extracellular matrix secreted by many cells in the bone microenvironment have been shown to affect the occurrence, proliferation, invasion and metastasis of osteosarcoma through a variety of signaling pathways. Therefore, targeting other cells in the bone microenvironment may improve the prognosis of osteosarcoma. The mechanism by which osteosarcoma interacts with other cells in the bone microenvironment has been extensively investigated, but currently developed drugs targeting the bone microenvironment have poor efficacy. Therefore, we review the regulatory effects of major cells and physical and chemical properties in the bone microenvironment on osteosarcoma, focusing on their complex interactions, potential therapeutic strategies and clinical applications, to deepen our understanding of osteosarcoma and the bone microenvironment and provide reference for future treatment. Targeting other cells in the bone microenvironment may provide potential targets for the development of clinical drugs for osteosarcoma and may improve the prognosis of osteosarcoma.
Collapse
Affiliation(s)
- Jin Zeng
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yi Peng
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Dong Wang
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Khan Ayesha
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Shijie Chen
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
- *Correspondence: Shijie Chen,
| |
Collapse
|
7
|
Deng J, Jiang R, Meng E, Wu H. CXCL5: A coachman to drive cancer progression. Front Oncol 2022; 12:944494. [PMID: 35978824 PMCID: PMC9376318 DOI: 10.3389/fonc.2022.944494] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Chemokines are a class of pro-inflammatory cytokines that can recruit and activate chemotactic cells. C‐X‐C motif chemokine ligand 5 (CXCL5) is a member of the chemokine family binding CXCR2 (C-X-C Motif Chemokine Receptor 2), a G-protein coupled receptor. Accumulated evidence has shown that dysregulated CXCL5 participates in tumor metastasis and angiogenesis in human malignant tumors. In this review, we summarized the advances in research on CXCL5, including its dysregulation in different tumors and the mechanism associated with tumor behavior (formation of the immunosuppressive microenvironment, promotion of tumor angiogenesis, and metastasis). We also summarized and discussed the perspective about the potential application of CXCL5 in tumor therapy targeting the tumor inflammatory microenvironment.
Collapse
|
8
|
Li H, Wu M, Zhao X. Role of chemokine systems in cancer and inflammatory diseases. MedComm (Beijing) 2022; 3:e147. [PMID: 35702353 PMCID: PMC9175564 DOI: 10.1002/mco2.147] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 12/12/2022] Open
Abstract
Chemokines are a large family of small secreted proteins that have fundamental roles in organ development, normal physiology, and immune responses upon binding to their corresponding receptors. The primary functions of chemokines are to coordinate and recruit immune cells to and from tissues and to participate in regulating interactions between immune cells. In addition to the generally recognized antimicrobial immunity, the chemokine/chemokine receptor axis also exerts a tumorigenic function in many different cancer models and is involved in the formation of immunosuppressive and protective tumor microenvironment (TME), making them potential prognostic markers for various hematologic and solid tumors. In fact, apart from its vital role in tumors, almost all inflammatory diseases involve chemokines and their receptors in one way or another. Modulating the expression of chemokines and/or their corresponding receptors on tumor cells or immune cells provides the basis for the exploitation of new drugs for clinical evaluation in the treatment of related diseases. Here, we summarize recent advances of chemokine systems in protumor and antitumor immune responses and discuss the prevailing understanding of how the chemokine system operates in inflammatory diseases. In this review, we also emphatically highlight the complexity of the chemokine system and explore its potential to guide the treatment of cancer and inflammatory diseases.
Collapse
Affiliation(s)
- Hongyi Li
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of EducationWest China Second HospitalSichuan UniversityChengduChina
| | - Min Wu
- Department of Biomedical Sciences, School of Medicine and Health SciencesUniversity of North DakotaGrand ForksNorth DakotaUSA
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of EducationWest China Second HospitalSichuan UniversityChengduChina
| |
Collapse
|
9
|
A highly branched α-D-glucan facilitates antitumor immunity by reducing cancer cell CXCL5 expression. Int J Biol Macromol 2022; 209:166-179. [PMID: 35390399 DOI: 10.1016/j.ijbiomac.2022.03.217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 03/22/2022] [Accepted: 03/31/2022] [Indexed: 11/22/2022]
Abstract
Tumor immunotherapy has emerged as a major pillar of anticancer therapeutic strategies. Natural polysaccharides, known for their strong immunomodulatory activities with relatively low cost and toxicity, are becoming promising prospects for cancer immunotherapy. In this study, we investigated the antitumor mechanism of JNY2PW, a highly branched α-D-glucan previously purified from the traditional marine Chinese medicine Arca inflata. JNY2PW was shown to enhance the sensitivity of tumor cells to co-culture macrophage supernatants by decreasing cancer cell CXCL5 expression. Furthermore, JNY2PW exerted antitumor effects without obvious toxic side effects in tumor-bearing mice by triggering the Akt/mTOR and ERK/GSK3β/β-catenin pathways and attenuating expression of CXCL5 in cancer cells. Remarkably, JNY2PW reduced tumor proliferation and dampened CXCL5 expression in tumor cells overexpressing CXCL5 both in vitro and in vivo. Additionally, JNY2PW blocked epithelial-mesenchymal transition (EMT) in both CXCL5-overexpressing and wild type tumor cells. Our data therefore uncovered a previously unrecognized antitumor mechanism for JNY2PW, suggesting that JNY2PW is a promising adjuvant as an immunomodulator for cancer immunotherapy.
Collapse
|
10
|
Abstract
Osteosarcoma is the most common primary bone malignancy in adolescents. Its high propensity to metastasize is the leading cause for treatment failure and poor prognosis. Although the research of osteosarcoma has greatly expanded in the past decades, the knowledge and new therapy strategies targeting metastatic progression remain sparse. The prognosis of patients with metastasis is still unsatisfactory. There is resonating urgency for a thorough and deeper understanding of molecular mechanisms underlying osteosarcoma to develop innovative therapies targeting metastasis. Toward the goal of elaborating the characteristics and biological behavior of metastatic osteosarcoma, it is essential to combine the diverse investigations that are performed at molecular, cellular, and animal levels from basic research to clinical translation spanning chemical, physical sciences, and biology. This review focuses on the metastatic process, regulatory networks involving key molecules and signaling pathways, the role of microenvironment, osteoclast, angiogenesis, metabolism, immunity, and noncoding RNAs in osteosarcoma metastasis. The aim of this review is to provide an overview of current research advances, with the hope to discovery druggable targets and promising therapy strategies for osteosarcoma metastasis and thus to overcome this clinical impasse.
Collapse
Affiliation(s)
- Gaohong Sheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Gao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Yang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hua Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
11
|
Ucci A, Cappariello A, Ponzetti M, Tennant F, Loftus AEP, Shefferd K, Maurizi A, Delle Monache S, Teti A, Rucci N. Anti-osteoblastogenic, pro-inflammatory and pro-angiogenic effect of extracellular vesicles isolated from the human osteosarcoma cell line MNNG/HOS. Bone 2021; 153:116130. [PMID: 34329816 DOI: 10.1016/j.bone.2021.116130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 07/15/2021] [Accepted: 07/21/2021] [Indexed: 01/17/2023]
Abstract
Extracellular Vesicles (EVs) are becoming increasingly recognized as integral signaling vehicles in several types of cancers, including bone malignancies. However, the specific mechanisms by which EVs influence osteosarcoma progression have not been fully determined. We evaluated the effects of EVs derived from the human osteosarcoma cell line MNNG/HOS (MNNG/HOS-EVs) on bone resident cells. We found that MNNG/HOS-EVs are internalized by osteoblasts and osteoclasts in vitro, with potent inhibitory effects on osteoblast metabolic activity, cell density and alkaline phosphatase activity. Consistently, MNNG/HOS-EVs reduced the expression of cell cycle and pro-osteoblastogenic genes, whilst increasing transcriptional expression and protein release of pro-osteoclastogenic/inflammatory cytokines (RankL, Il1b, Il6 and Lcn2), pro-tumoral cytokines (CCL2,5,6,12 and CXCL1,2,5) and the metalloproteinase MMP3. MNNG/HOS-EVs did not induce osteoclast differentiation, while promoting in vitro and in vivo angiogenesis. Intriguingly, EVs derived from another osteosarcoma cell line (U2OS) reduced ALP activity but had no other effect on osteoblast phenotype. MNNG/HOS-EVs were also found to dramatically increase Serpin b2 expression in osteoblasts. To evaluate the significance of this finding, osteoblasts were forced to overexpress Serpin b2, which however did not affect osteoblast differentiation, while Il6 and Lcn2 mRNAs were up regulated. Overall, we shed light on the interactions of osteosarcoma EVs with the cells of the bone microenvironment, identifying key anti-osteoblastogenic, pro-inflammatory and pro-angiogenic factors that could contribute to osteosarcoma expansion.
Collapse
Affiliation(s)
- Argia Ucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Alfredo Cappariello
- Oncohematology Department, IRCCS Bambino Gesù Children's Hospital Research Laboratories, Rome, Italy
| | - Marco Ponzetti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Fabianna Tennant
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | | | - Kirsty Shefferd
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Antonio Maurizi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Simona Delle Monache
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Anna Teti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Nadia Rucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| |
Collapse
|
12
|
Jing F, Wang J, Zhou L, Ning Y, Xu S, Zhu Y. Bioinformatics analysis of the role of CXC ligands in the microenvironment of head and neck tumor. Aging (Albany NY) 2021; 13:17789-17817. [PMID: 34247149 PMCID: PMC8312447 DOI: 10.18632/aging.203269] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 05/18/2021] [Indexed: 12/30/2022]
Abstract
Chemokines play a significant role in cancer. CXC-motif chemokine ligands (CXCLs) are associated with the tumorigenesis and progression of head and neck squamous cell carcinoma (HNSC); however, their specific functions in the tumor microenvironment remain unclear. Here, we analyzed the molecular networks and transcriptional data of HNSC patients from the Oncomine, GEPIA, String, cBioPortal, Metascape, TISCH, and TIMER databases. To verify immune functions of CXCLs, their expression was analyzed in different immune cell types. To our knowledge, this is the first report on the correlation between CXCL9-12 and 14 expression and advanced tumor stage. CXCL2, 3, 8, 10, 13, and 16 were remarkably related to tumor immunity. Kaplan-Meier and TIMER survival analyses revealed that high expression of CXCL1, 2, 4, and 6-8 is correlated with low survival in HNSC patients, whereas high expression of CXCL9, 10, 13, 14, and 17 predicts high survival. Only CXCL13 and 14 were associated with overall survival in human papilloma virus (HPV)-negative patients. Single-cell datasets confirmed that CXCLs are associated with HNSC-related immune cells. Thus, CXCL1-6, 8-10, 12-14, and 17 could be prognostic targets for HNSC, and CXCL13 and 14 could be novel biomarkers of HPV-negative HNSC.
Collapse
Affiliation(s)
- Fengyang Jing
- Department of Dental Implant Center, Stomatologic Hospital and College, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui Province, Hefei 230032, China
| | - Jianxiong Wang
- Chief Physician, Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Liming Zhou
- Department of Dental Implant Center, Stomatologic Hospital and College, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui Province, Hefei 230032, China
| | - Yujie Ning
- Department of Dental Implant Center, Stomatologic Hospital and College, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui Province, Hefei 230032, China
| | - Shengqian Xu
- Chief Physician, Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Youming Zhu
- Department of Dental Implant Center, Stomatologic Hospital and College, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui Province, Hefei 230032, China
| |
Collapse
|
13
|
Fang ZA, He Y, Sun C, Zhan L, Zhou G, Wei B, Sun S. Expression and potential role of CXCL5 in the pathogenesis of intrauterine adhesions. J Int Med Res 2021; 49:300060521997718. [PMID: 33752504 PMCID: PMC7995464 DOI: 10.1177/0300060521997718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Objective C-X-C motif chemokine ligand 5 (CXCL5), a member of the chemokine family, is associated with remodeling of connective tissues. However, its role in formation of intrauterine adhesions (IUA) remains unclear. We aimed to investigate the expression and mechanism underlying the role of CXCL5 in IUA. Methods Expression of CXCL5 in IUA was detected by immunohistochemistry in a rat model of IUA and by real-time PCR and western blotting in patients with IUA. The protein levels of matrix metalloproteinase 9 (MMP9) and transcription factor p65 in human endometrial cells were assessed by western blotting after CXCL5 overexpression. Results Protein expression of CXCL5 was significantly decreased in the endometria of IUA rats compared with that of control and sham-operated rats. Real-time PCR and western blotting in patients with IUA showed similar results to those from the rat model. After overexpression, CXCL5 significantly upregulated expression of MMP9 and slightly upregulated expression of p65 in human endometrial cells. Conclusions CXCL5 plays an important role in IUA formation after endometrial injury. We propose a molecular mechanism to explain formation of IUA, including downregulation of MMP9 by low CXCL5 expression. These findings provide valuable information for the prevention and targeted therapy of IUA.
Collapse
Affiliation(s)
- Zi-Ang Fang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yu He
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chao Sun
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lei Zhan
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Guiju Zhou
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Bing Wei
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shiying Sun
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
14
|
Zhang W, Wang H, Sun M, Deng X, Wu X, Ma Y, Li M, Shuoa SM, You Q, Miao L. CXCL5/CXCR2 axis in tumor microenvironment as potential diagnostic biomarker and therapeutic target. Cancer Commun (Lond) 2021; 40:69-80. [PMID: 32237072 PMCID: PMC7163794 DOI: 10.1002/cac2.12010] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 01/31/2020] [Indexed: 12/14/2022] Open
Abstract
The components of the tumor microenvironment (TME) in solid tumors, especially chemokines, are currently attracting much attention from scientists. C-X-C motif chemokine ligand 5 (CXCL5) is one of the important chemokines in TME. Overexpression of CXCL5 is closely related to the survival time, recurrence and metastasis of cancer patients. In TME, CXCL5 binds to its receptors, such as C-X-C motif chemokine receptor 2 (CXCR2), to participate in the recruitment of immune cells and promote angiogenesis, tumor growth, and metastasis. The CXCL5/CXCR2 axis can act as a bridge between tumor cells and host cells in TME. Blocking the transmission of CXCL5/CXCR2 signals can increase the sensitivity and effectiveness of immunotherapy and slow down tumor progression. CXCL5 and CXCR2 are also regarded as biomarkers for predicting prognosis and molecular targets for customizing the treatment. In this review, we summarized the current literature regarding the biological functions and clinical significance of CXCL5/CXCR2 axis in TME. The possibility to use CXCL5 and CXCR2 as potential prognostic biomarkers and therapeutic targets in cancer is also discussed.
Collapse
Affiliation(s)
- Wen Zhang
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, P. R. China
| | - Huishan Wang
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, P. R. China
| | - Mingyang Sun
- Department of Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, P. R. China
| | - Xueting Deng
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, P. R. China
| | - Xueru Wu
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, P. R. China
| | - Yilan Ma
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, P. R. China
| | - Mengjing Li
- Department of Biotherapy, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, P. R. China.,Key Laboratory for Aging and Disease, Nanjing Medical University, Nanjing, 210011, Jiangsu, P. R. China
| | - Said Maisam Shuoa
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, P. R. China
| | - Qiang You
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, P. R. China.,Department of Biotherapy, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, P. R. China.,Key Laboratory for Aging and Disease, Nanjing Medical University, Nanjing, 210011, Jiangsu, P. R. China
| | - Lin Miao
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, P. R. China
| |
Collapse
|
15
|
Tian WJ, Liu SS, Li BR. The Combined Detection of Immune Genes for Predicting the Prognosis of Patients With Non-Small Cell Lung Cancer. Technol Cancer Res Treat 2020; 19:1533033820977504. [PMID: 33256552 PMCID: PMC7711225 DOI: 10.1177/1533033820977504] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Lung cancer is one of the leading causes of cancer-related death. In recent years, there has been an increasing interest in the fields of tumor and immunity. This study focused on the possible prognostic value of immune genes in non-small cell lung cancer patients. We used The Cancer Genome Atlas (TCGA) to download gene expression data and clinical information of lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC). The immune gene list was downloaded from the Immport database. We then constructed immune gene prognostic models on the basis of Cox regression analysis. We further evaluated the clinical significance of the models via survival analysis, receiver operating characteristic (ROC) curves, and independent prognostic factor analysis. Moreover, we analyzed the associations of prognostic models with both mutation burdens and neoantigens. Using the Gene Expression Omnibus (GEO) and Kaplan-Meier plotter databases, we evaluated the validity of the prognostic models. The prognostic model of LUAD included 13 immune genes, and the prognostic model of LUSC contained 10 immune genes. High-risk patients based on prognostic models had a lower 5-year survival rate than did low-risk patients. The ROC curve analysis demonstrated the prediction accuracy of the prognostic models, as the area under the curve (AUC) was 0.742, 0.707, and 0.711 for LUAD, and 0.668, 0.703, and 0.668 for LUSC, when the predicted survival times were 1, 3, and 5 years, respectively. The mutation burden analysis showed that mutation level was associated with the risk score in patients with LUAD. The analysis based on GEO and Kaplan-Meier plotter demonstrated the prognostic validity of the models. Therefore, immune gene-related models of LUAD and LUSC can predict prognosis. Further study of these genes may enable us to better distinguish between LUAD and LUSC and lead to improvement in immunotherapy for lung cancer.
Collapse
Affiliation(s)
- Wen-Juan Tian
- Department of Clinical Laboratory, Second Affiliated Hospital, 117799Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China.,School of Medicine, 117799Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Shan-Shan Liu
- Department of Clinical Laboratory, Second Affiliated Hospital, 117799Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China.,School of Medicine, 117799Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Bu-Rong Li
- Department of Clinical Laboratory, Second Affiliated Hospital, 117799Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| |
Collapse
|
16
|
Wu MY, Shen M, Xu MD, Yu ZY, Tao M. FOLFIRINOX regulated tumor immune microenvironment to extend the survival of patients with resectable pancreatic ductal adenocarcinoma. Gland Surg 2020; 9:2125-2135. [PMID: 33447563 DOI: 10.21037/gs-20-828] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is one of the most deadly malignant tumors worldwide due to its ineffective diagnosis and poor prognosis. The longest median overall survival (OS) to PDAC patients has been provided by FOLFIRINOX. It is essential to identify the mechanisms of FOLFIRINOX to gain new insights for the treatment of PDAC. Methods We compared gene expression levels of PDAC patients who received neoadjuvant FOLFIRINOX prior to surgery with those of patients who received no neoadjuvant chemotherapy. Bioinformatics analysis was applied to screen differentially expressed genes (DEGs). Three microarray data sets were downloaded to analyze gene expression data between PDAC and adjacent non-tumor tissues. Overlapping DEGs were subjected to Kaplan-Meier survival analysis. The genes relating to poor outcomes and would be decreased after FOLFIRINOX were input into the Oncomine, University of Alabama Cancer (UALCAN), and LinkedOmics databases to analyze the gene expression and regulation networks. Results A total of 83 differentially expressed genes (DEGs) were screened and subjected to bioinformatics analysis, which indicated FOLFIRINOX influenced the immune microenvironment of PDAC. Seventy-three genes significantly associated with the OS of PDAC patients. A Venn diagram revealed CXCL5 and PLAU were related to poor outcomes and would decrease after FOLFIRINOX chemotherapy of PDAC patients. It turned out that CXCL5 participated in the immune response-regulating signaling pathway in PDAC patients. Conclusions FOLFIRINOX regulated tumor immunity by reducing expression of the immunosuppressive gene CXCL5, laying a foundation for further study of combination therapy of FOLFIRINOX and immunotherapy.
Collapse
Affiliation(s)
- Meng-Yao Wu
- Department of Oncology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Meng Shen
- Department of Oncology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Meng-Dan Xu
- Department of Oncology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zheng-Yuan Yu
- Department of Oncology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Min Tao
- Department of Oncology, First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
17
|
Xiang D, Li Y, Lin Y. Circular RNA circCCDC66 Contributes to Malignant Phenotype of Osteosarcoma by Sponging miR-338-3p to Upregulate the Expression of PTP1B. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4637109. [PMID: 32851074 PMCID: PMC7439191 DOI: 10.1155/2020/4637109] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 07/21/2020] [Indexed: 12/31/2022]
Abstract
In recent years, the mechanism of cancer research has become hotspots of life science and medicine, especially due to the rapid development of molecular medicine and bioinformatics research. Similarly, the molecular mechanism also has received increasing attention in osteosarcoma (OS) research. Also, a considerable amount of research confirmed that circular RNAs (circRNAs) could regulate cancer cell growth and metastasis. This study aimed to explore the effect of a circRNA, circCCDC66, on OS and reveal its potential molecular mechanism. High circCCDC66 expression level was found in OS patient-derived tissue samples and OS cell lines by qRT-PCR. The abilities cell proliferation and metastatic of U2OS and SW1353 cells were then assessed by Cell Counting Kit-8 and transwell assay, respectively. The interaction between circCCDC66 and its target miRNAs were verified by the dual-luciferase reporter assay. Through functional experiments, we found that circCCDC66 knockdown promoted the inhibition of cell proliferation and metastatic of OS cell lines. From mechanistic perspective, circCCDC66 upregulated PTP1B by sponging miR-338-3p. Collectively, our findings demonstrated that circCCDC66 contributed to malignant behaviors of OS cells by miR-338-3p/PTP1B pathway, which suggested circCCDC66/miR-338-3p/PTP1B axis might be a potential therapeutic target.
Collapse
Affiliation(s)
- Deng Xiang
- Department of Orthopaedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, China
| | - Yugang Li
- Department of Orthopaedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, China
| | - Yanshui Lin
- Department of Orthopaedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, China
| |
Collapse
|
18
|
Jiang X, Huang Y. Curcumin Derivative C086 Combined with Cisplatin Inhibits Proliferation of Osteosarcoma Cells. Med Sci Monit 2020; 26:e924507. [PMID: 32734935 PMCID: PMC7414526 DOI: 10.12659/msm.924507] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background Curcumin derivative C086 (cur C086) is a potential chemotherapeutic agent for patients with osteosarcoma. In this study, the effects of cur C086 combined with cisplatin on the biological processes of osteosarcoma cells were investigated. Material/Methods In this study, expression of BMIL1 was detected by real-time quantitative reverse transcription polymerase chain reaction and Western blotting in MG-63 cells treated with cur C086+cisplatin. Functions of cur C086+cisplatin on proliferation ability, apoptosis response, and metastatic potential of MG-63 cells were determined by MTT, flow cytometry, Hoechst 33258 staining and Transwell assays, respectively. In additionally, expression of P16, E-cadherin, epidermal growth factor (EGFR), and Notch1 was measured by Western blotting. Results Expression of BMIL1 decreased significantly in MG-63 cells treated with cur C086 (20 μM)+cisplatin (1.28 nM). Treatment with cur C086+cisplatin considerably inhibited growth, migration, and invasion potential in MG-63 cells, whereas apoptosis was obviously upregulated. Moreover, cur C086+cisplatin suppressed BMIL1 expression or its potential downstream targets, P16, E-cadherin, EGFR, and Notch1. Conclusions The current results demonstrate that combined treatment with cur C086+cisplatin may be an effective form of chemotherapy for patients with osteosarcoma.
Collapse
Affiliation(s)
- Xi Jiang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), Chongqing University Cancer Hospital, Chongqing, China (mainland)
| | - Yulin Huang
- Department of Clinical Laboratory, The Traditional Chinese Medicine Hospital of Wuxi, Chongqing, China (mainland)
| |
Collapse
|
19
|
Dorr MM, Guignard R, Auger FA, Rochette PJ. The use of tissue-engineered skin to demonstrate the negative effect of CXCL5 on epidermal ultraviolet radiation-induced cyclobutane pyrimidine dimer repair efficiency. Br J Dermatol 2020; 184:123-132. [PMID: 32271940 DOI: 10.1111/bjd.19117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Ultraviolet radiation (UVR) is responsible for keratinocyte cancers through the induction of mutagenic cyclobutane pyrimidine dimers (CPDs). Many factors influence CPD repair in epidermal keratinocytes, and a better understanding of those factors might lead to prevention strategies against skin cancer. OBJECTIVES To evaluate the impact of dermal components on epidermal CPD repair efficiency and to investigate potential factors responsible for the dermal-epidermal crosstalk modulating UVR-induced DNA damage repair in keratinocytes. METHODS A model of self-assembled tissue-engineered skin containing human primary keratinocytes and fibroblasts was used in this study. RESULTS We showed that CPD repair in keratinocytes is positively influenced by the presence of a dermis. We investigated the secretome and found that the cytokine CXCL5 is virtually absent from the culture medium of reconstructed skin, compared with media from fibroblasts and keratinocytes alone. By modulating CXCL5 levels in culture media of keratinocytes, we have shown that CXCL5 is an inhibitor of CPD repair. CONCLUSIONS This work outlines the impact of the secreted dermal components on epidermal UVR-induced DNA damage repair and sheds light on a novel role of CXCL5 in CPD repair.
Collapse
Affiliation(s)
- M M Dorr
- Centre de Recherche du CHU de Québec - Université Laval, Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Québec, QC, Canada.,Centre de Recherche en Organogénèse Expérimentale de l'Université Laval - LOEX, Québec, QC, Canada
| | - R Guignard
- Centre de Recherche du CHU de Québec - Université Laval, Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Québec, QC, Canada.,Centre de Recherche en Organogénèse Expérimentale de l'Université Laval - LOEX, Québec, QC, Canada
| | - F A Auger
- Centre de Recherche du CHU de Québec - Université Laval, Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Québec, QC, Canada.,Centre de Recherche en Organogénèse Expérimentale de l'Université Laval - LOEX, Québec, QC, Canada
| | - P J Rochette
- Centre de Recherche du CHU de Québec - Université Laval, Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Québec, QC, Canada.,Centre de Recherche en Organogénèse Expérimentale de l'Université Laval - LOEX, Québec, QC, Canada.,Université Laval, Faculté de Médecine, Département d'Ophtalmologie, Université Laval, Québec, QC, Canada
| |
Collapse
|
20
|
Zhang R, Liu Q, Peng J, Wang M, Li T, Liu J, Cui M, Zhang X, Gao X, Liao Q, Zhao Y. CXCL5 overexpression predicts a poor prognosis in pancreatic ductal adenocarcinoma and is correlated with immune cell infiltration. J Cancer 2020; 11:2371-2381. [PMID: 32201508 PMCID: PMC7065995 DOI: 10.7150/jca.40517] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 01/20/2020] [Indexed: 02/07/2023] Open
Abstract
Background: C-X-C motif chemokine 5 (CXCL5) is an important attractant for immune cell accumulation in tumor tissues. Recent evidence has shown that CXCL5 could promote carcinogenesis and cancer progression in a variety of cancer types. However, the relationships between CXCL5, immune cell infiltration and pancreatic ductal adenocarcinoma (PDAC) remain largely unknown. This study aimed to explore the role and regulative mechanism of CXCL5 in PDAC carcinogenesis. Materials and Methods: The expression of CXCL5 in PDAC was analyzed based on online databases and tissue microarray staining, and Western blotting of CXCL5 in PDAC cell lines and patient samples. The correlation between CXCL5 expression and clinicopathological features, prognosis and immune cell infiltration in tumor tissues was analyzed. Results: High expression of CXCL5 was observed both in PDAC tumor tissue and PDAC cell lines, compared to normal pancreas tissues and normal ductal epithelium cells. High CXCL5 expression in tumor tissues was positively correlated with an advanced T stage (p=0.036), a positive tumor lymph node metastasis (p=0.014), a poor differentiation status (p=0.003) and a poor prognosis (p=0.001). Combination of CA242 and CXCL5 expression (p<0.0001) served as a better prognostic factor than CA242 alone (p=0.006). In addition, PDAC patients with high CXCL5 expression had more intratumoral M2 polarized macrophages (p=0.0248), neutrophils (p=0.0068) and IgG+ plasma cells (p=0.0133) than patients with low CXCL5 expression. Conclusions: The expression of CXCL5 is elevated in pancreatic cancer cells. High CXCL5 expression is positively correlated with poor survival and the increased infiltration of several types of immune suppressive cells. Thus, CXCL5 could be a promising therapeutic target for PDAC immunotherapy.
Collapse
Affiliation(s)
- Ronghua Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Qiaofei Liu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Junya Peng
- Department of Medical Research Center, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Mengyi Wang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Tong Li
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Jingkai Liu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Ming Cui
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Xiang Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Xiang Gao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Quan Liao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China
| |
Collapse
|
21
|
Pratt HG, Justin EM, Lindsey BA. Applying Osteosarcoma Immunology to Understand Disease Progression and Assess Immunotherapeutic Response. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1258:91-109. [PMID: 32767236 DOI: 10.1007/978-3-030-43085-6_6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Osteosarcoma, the most common malignant bone tumor in children and adolescents, remains a complicated disease to treat; no new treatments have been developed in more than three decades. Due to the importance of the immune system in osteosarcoma disease progression, immunotherapeutic strategies have been explored to potentially improve long-term survival. However, most immunotherapeutics have not reached the level of success hoped would occur in this disease. Understanding the immune system in osteosarcoma will be key to optimizing treatments and improving patient outcomes. Therefore, immunophenotyping can be used as a very powerful tool to help better understand the complexity of the immune response seen in osteosarcoma and in the use of immunotherapy in this malignancy. This book chapter will provide an overview of the known immune responses seen in this disease and potential developments for the future of immunophenotyping. Indeed, it appears that being able to track the immune system throughout the disease and treatment of patients with osteosarcoma could allow for a personalized approach to immunotherapy.
Collapse
Affiliation(s)
- Hillary G Pratt
- West Virginia University School of Medicine, Morgantown, WV, USA
| | - E Markel Justin
- West Virginia University School of Medicine, Morgantown, WV, USA
| | - Brock A Lindsey
- West Virginia University School of Medicine, Morgantown, WV, USA.
| |
Collapse
|
22
|
Abstract
Chemokines are a family of small proteins, subdivided by their conserved cysteine residues and common structural features. Chemokines interact with their cognate G-protein-coupled receptors to elicit downstream signals that result in cell migration, proliferation, and survival. This review presents evidence for how the various CXC and CC subfamily chemokines influence bone hemostasis by acting on osteoclasts, osteoblasts, and progenitor cells. Also discussed are the ways in which chemokines contribute to bone loss as a result of inflammatory diseases such as rheumatoid arthritis, HIV infection, and periodontal infection. Both positive and negative effects of chemokines on bone formation and bone loss are presented. In addition, the role of chemokines in altering the bone microenvironment through effects on angiogenesis and tumor invasion is discussed. Very few therapeutic agents that influence bone formation by targeting chemokines or chemokine receptors are available, although a few are currently being evaluated.
Collapse
Affiliation(s)
- Annette Gilchrist
- Department of Pharmaceutical Sciences, Midwestern University, Downers Grove, IL, USA.
| |
Collapse
|
23
|
Gu Z, Hou Z, Zheng L, Wang X, Wu L, Zhang C. Long noncoding RNA LINC00858 promotes osteosarcoma through regulating miR-139-CDK14 axis. Biochem Biophys Res Commun 2018; 503:1134-1140. [PMID: 29944887 DOI: 10.1016/j.bbrc.2018.06.131] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 06/23/2018] [Indexed: 12/09/2022]
Abstract
Long noncoding RNAs (lncRNAs) have been identified to modulate the tumorigenesis of human cancers. The in-depth of lncRNAs on human osteosarcoma oncogenesis is still ambiguous. In present study, functional and mechanism experiments were conducted to investigate the role of long intergenic non-protein coding RNA 00858 (LINC00858) on human osteosarcoma tumorigenesis. Results demonstrated that LINC00858 expression was significantly upregulated in both osteosarcoma tissues and cell lines. Mechanism assays presented that LINC00858 silencing significantly repressed osteosarcoma cells' proliferation and invasion in vitro, and inhibited the tumor growth in vivo. In further experiments, LINC00858 was identified to sponge miR-139 to form RNA-induced silencing complex (RISC) using luciferase reporter assay and RNA immunoprecipitation (RIP). Besides, CDK14 was validated to be the target protein the miR-139. Rescue experiments confirmed the role of LINC00858/miR-139/CDK14 pathway on osteosarcoma cells' phenotype. In summary, these data prove that LINC00858/miR-139/CDK14 axis promotes the tumorigenesis of osteosarcoma, providing a new mechanism or target for osteosarcoma.
Collapse
Affiliation(s)
- Zenghui Gu
- Department of Orthopedic, The 117 Hospital of The PLA, Hangzhou, Zhejiang, 310012, China.
| | - Zhenhai Hou
- Department of Orthopedic, The 117 Hospital of The PLA, Hangzhou, Zhejiang, 310012, China
| | - Longbao Zheng
- Department of Orthopedic, The 117 Hospital of The PLA, Hangzhou, Zhejiang, 310012, China
| | - Xinqiang Wang
- Department of Orthopedic, The 117 Hospital of The PLA, Hangzhou, Zhejiang, 310012, China
| | - Liangbang Wu
- Department of Orthopedic, The 117 Hospital of The PLA, Hangzhou, Zhejiang, 310012, China
| | - Cheng Zhang
- Department of Orthopedic, The 117 Hospital of The PLA, Hangzhou, Zhejiang, 310012, China
| |
Collapse
|
24
|
Chen G, Wang Q, Yang Q, Li Z, Du Z, Ren M, Zhao H, Song Y, Zhang G. Circular RNAs hsa_circ_0032462, hsa_circ_0028173, hsa_circ_0005909 are predicted to promote CADM1 expression by functioning as miRNAs sponge in human osteosarcoma. PLoS One 2018; 13:e0202896. [PMID: 30153287 PMCID: PMC6112665 DOI: 10.1371/journal.pone.0202896] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 08/07/2018] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Osteosarcoma (OS) is a primary malignant bone tumor with a high fatality rate. Many circRNAs have been proved to play important roles in the pathogenesis of some diseases. However, the occurrence of circRNAs in OS remains little known. METHODS The circular RNA (circRNA) expression file GSE96964 dataset, which included seven osteosarcoma cell lines and one control sample (osteoblast cell line), was downloaded from the Gene Expression Omnibus (GEO) database to explore the potential function of circRNAs in osteosarcoma by competing endogenous RNA (ceRNA) analysis. Three gene expression profiles of OS were downloaded from GEO database and then used for the pathway enrichment analysis, Venn analysis and protein-protein interaction (PPI) network analysis. Real-time qPCR validation and RNA interference were conducted to verify our prediction. RESULTS Differentially expressed circRNAs between OS and control, including 8 up-regulated and 102 down-regulated circRNAs, were generated and ceRNA analysis for 5 most up-regulated or 5 most down-regulated circRNAs in OS were then performed. The pathway enrichment analysis of gene expression profiles indicated differentially expressed genes (DEGs) of three gene profiles significantly enriched in cell cycle pathway, cell adhesion molecules (CAMs) pathway, oxidative phosphorylation pathway, cytokine-cytokine receptor interaction pathway, p53 signaling pathway and proteoglycans in cancer pathway, which were critical important pathways in the pathogenesis of OS. The Venn analysis showed that 2 (one is a pseudogene) up-regulated and 39 down-regulated DEGs were co-expressed in all three gene profiles. Then PPI networks of 41 co-expressed DEGs (up- and down-regulated DEGs) were constructed to predict their functions using the GeneMANIA. The expression levels of these related RNAs also matched our predictions really well. CONCLUSION Ultimately, we found cell adhesion molecule 1 (CADM1) gene was not only a co-expression mRNA of the three mRNA expression profiles of OS, but also are predicted to be regulated by hsa_circ_0032462, hsa_circ_0028173, hsa_circ_0005909 by functioning as miRNAs 'Sponge' in human osteosarcoma. These over-expressed circRNAs may result in the over expression of CADM1 which promote the development of OS. We envision this discovery of these important moleculars, incuding hsa_circ_0032462, hsa_circ_0028173, hsa_circ_0005909 and CADM1 may lead to further development of new concepts, thus allowing for more opportunities in diagnosis and therapy of OS.
Collapse
Affiliation(s)
- Gaoyang Chen
- Department of Orthopedics of the Second Hospital of Jilin University, Changchun, Jilin, China
- Research Centre of the Second Hospital of Jilin University, Changchun, Jilin, China
- The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Changchun, Jilin, China
| | - Qingyu Wang
- Department of Orthopedics of the Second Hospital of Jilin University, Changchun, Jilin, China
- Research Centre of the Second Hospital of Jilin University, Changchun, Jilin, China
- The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Changchun, Jilin, China
| | - Qiwei Yang
- Research Centre of the Second Hospital of Jilin University, Changchun, Jilin, China
- The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Changchun, Jilin, China
| | - Zhaoyan Li
- Department of Orthopedics of the Second Hospital of Jilin University, Changchun, Jilin, China
- The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Changchun, Jilin, China
| | - Zhenwu Du
- Department of Orthopedics of the Second Hospital of Jilin University, Changchun, Jilin, China
- Research Centre of the Second Hospital of Jilin University, Changchun, Jilin, China
- The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Changchun, Jilin, China
| | - Ming Ren
- Department of Orthopedics of the Second Hospital of Jilin University, Changchun, Jilin, China
- The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Changchun, Jilin, China
| | - Haiyue Zhao
- Research Centre of the Second Hospital of Jilin University, Changchun, Jilin, China
- The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Changchun, Jilin, China
| | - Yang Song
- Department of Orthopedics of the Second Hospital of Jilin University, Changchun, Jilin, China
- The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Changchun, Jilin, China
- * E-mail: (GZ); (YS)
| | - Guizhen Zhang
- Department of Orthopedics of the Second Hospital of Jilin University, Changchun, Jilin, China
- Research Centre of the Second Hospital of Jilin University, Changchun, Jilin, China
- The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Changchun, Jilin, China
- * E-mail: (GZ); (YS)
| |
Collapse
|
25
|
Ren Y, Zhu F, Liu Z. Inhibitory effect of icariin on osteosarcoma cell proliferation via the Wnt/β-catenin signaling pathway. Oncol Lett 2018; 16:1405-1410. [PMID: 30008817 PMCID: PMC6036372 DOI: 10.3892/ol.2018.8821] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 11/17/2017] [Indexed: 12/25/2022] Open
Abstract
Osteosarcoma, is a kind of malignant tumor derived from malignant interstitial cells. The pathogenesis of osteosarcoma remains unclear and despite use of chemotherapy drugs, resistance to these drugs affects the success of treatment. The present study was conducted to investigate the effects of icariin (ICA) on osteosarcoma cell proliferation and to investigate the role of the Wnt/β-catenin signaling pathway in the inhibition process of ICA on osteosarcoma cell proliferation. Different concentrations of ICA were selected to treat the osteosarcoma cell line 143B for 24 h, and then the onset concentration of ICA when it inhibited the growth of osteosarcoma cancer cell line 143B was detected via an MTT assay. The effect of ICA on the apoptosis of colon cancer cell line 143B under this concentration was detected using a flow cytometer. RNA in osteosarcoma cell line 143B was extracted, followed by reverse transcription. The expression levels of related and apoptotic proteins in the Wnt/β-catenin signaling pathway using ICA were detected by semi-quantitative PCR and western blot analysis, respectively. The expression quantities of vascular endothelial growth factor (VEGF) and MMP-9 were detected by ELISA. MTT assay showed that ICA inhibited the growth of 143B when its concentration was 5 µM (p<0.01). Flow cytometry showed that the number of apoptotic cells after ICA treatment was significantly higher than that in control group (p<0.01). RNA in osteosarcoma cell line 143B was extracted, followed by reverse transcription. Semi-quantitative PCR and western blot analysis revealed that the expression levels of p-GSK3β, β-catenin, c-Myc and cyclin D1 in cells after ICA treatment were significantly downregulated (p<0.01), while the expression level of caspase-3 was significantly increased (p<0.01). ELISA showed that the expression quantities of VEGF and MMP-9 were significantly decreased (p<0.01). Thus, ICA can significantly inhibit osteosarcoma cell proliferation and promote osteosarcoma cell apoptosis, which may be realized by affecting the expression of the Wnt/β-catenin signaling pathway and blocking the expression of related proteins.
Collapse
Affiliation(s)
- Yuxin Ren
- Department of Spinal Surgery, Dezhou People's Hospital, Dezhou, Shandong 253014, P.R. China
| | - Fuqiang Zhu
- Department of Spinal Surgery, Dezhou People's Hospital, Dezhou, Shandong 253014, P.R. China
| | - Zhendong Liu
- Department of Orthopedics, Dezhou People's Hospital, Dezhou, Shandong 253014, P.R. China
| |
Collapse
|
26
|
Gu W, Zhang E, Song L, Tu L, Wang Z, Tian F, Aikenmu K, Chu G, Zhao J. Long noncoding RNA HOXD-AS1 aggravates osteosarcoma carcinogenesis through epigenetically inhibiting p57 via EZH2. Biomed Pharmacother 2018; 106:890-895. [PMID: 30119259 DOI: 10.1016/j.biopha.2018.06.173] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 06/28/2018] [Accepted: 06/29/2018] [Indexed: 01/26/2023] Open
Abstract
Osteosarcoma is the most common primary malignant bone tumor and long non-coding RNAs (lncRNAs) have been proved to epigenetically regulate the oncogenesis of osteosarcoma. In this research, we investigate the role of lncRNA HOXD-AS1 on the osteosarcoma oncogenesis. Results revealed that HOXD-AS1 expression level was significantly up-regulated in osteosarcoma tissue and cells, moreover, the aberrant overexpression predicted the poor prognosis of osteosarcoma patients. Loss-of-functional experiments indicated that HOXD-AS1 silencing inhibited the osteosarcoma cells proliferation and induced G1/G0 phase arrest in vitro, and repressed tumor cell growth in vivo. Mechanistic investigations showed that HOXD-AS1 epigenetically repressed p57 through recruiting enhancer of zeste homolog 2 (EZH2) to the promoter of p57. Rescue experiments revealed that p57 could recover the oncogenic role of HOXD-AS1 on osteosarcoma. In conclusion, our study confirmed that HOXD-AS1 could interact with EZH2, and then repress p57 expression, to aggravate osteosarcoma oncogenesis. which provide new idea for the osteosarcoma tumorigenesis.
Collapse
Affiliation(s)
- Wenfei Gu
- Department of Spine Surgery of Traditonal Chinese Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830000, China
| | - Enfeng Zhang
- Department of Spine Surgery of Traditonal Chinese Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830000, China
| | - Lijun Song
- Department of Spine Surgery of Traditonal Chinese Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830000, China
| | - Laiyong Tu
- Department of Spine Surgery of Traditonal Chinese Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830000, China
| | - Zhenbin Wang
- Department of Spine Surgery of Traditonal Chinese Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830000, China
| | - Feng Tian
- Department of Spine Surgery of Traditonal Chinese Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830000, China
| | - Kahaer Aikenmu
- Department of Spine Surgery of Traditonal Chinese Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830000, China
| | - Ge Chu
- Department of Spine Surgery of Traditonal Chinese Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830000, China
| | - Jiang Zhao
- Department of Spine Surgery of Traditonal Chinese Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, 830000, China.
| |
Collapse
|
27
|
Qiu WZ, Zhang HB, Xia WX, Ke LR, Yang J, Yu YH, Liang H, Huang XJ, Liu GY, Li WZ, Xiang YQ, Kang TB, Guo X, Lv X. The CXCL5/CXCR2 axis contributes to the epithelial-mesenchymal transition of nasopharyngeal carcinoma cells by activating ERK/GSK-3β/snail signalling. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:85. [PMID: 29665837 PMCID: PMC5905166 DOI: 10.1186/s13046-018-0722-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 02/26/2018] [Indexed: 12/22/2022]
Abstract
Background Distant metastasis is the major cause of treatment failure in patients with nasopharyngeal carcinoma (NPC). Although several biomarkers correlate with metastasis and prognosis, the molecular mechanisms of NPC development and progression remain unclear. Methods Quantitative RT-PCR (qRT-PCR), western blotting, cell growth, foci formation, migration and invasion assays, and xenograft mouse models were utilized to examine the expression levels and functions of the CXCL5/CXCR2 axis in NPC. A luciferase reporter assay, western blotting, immunofluorescence, and migration and invasion assays were used to identify and verify the ERK/GSK-3β/Snail signalling pathway. Results CXCL5 was significantly increased in the sera of NPC patients, and high expression levels of CXCL5/CXCR2 in NPC primary tissues indicated poor survival. CXCL5 and CXCR2 were upregulated in NPC cell lines. Ectopic expression of the CXCL5/CXCR2 axis promoted NPC cell migration and invasion in vitro and the formation of lung metastases in vivo. Mechanistically, the dual overexpression of CXCL5 and CXCR2 promoted cell spreading by inducing the epithelial-mesenchymal transition (EMT) through the activation of the ERK/GSK-3β/Snail signalling pathway. Conclusion The CXCL5/CXCR2 axis contributes to the EMT of NPC cells by activating ERK/GSK-3β/Snail signalling, and this axis may be a potential diagnostic marker and therapeutic target for patients with NPC. Electronic supplementary material The online version of this article (10.1186/s13046-018-0722-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wen-Ze Qiu
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China.,Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China
| | - Hai-Bo Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Wei-Xiong Xia
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China.,Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China
| | - Liang-Ru Ke
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China.,Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China
| | - Jing Yang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, 4365 Kangxin Road, Shanghai, 201321, People's Republic of China
| | - Ya-Hui Yu
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China.,Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China
| | - Hu Liang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China.,Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China
| | - Xin-Jun Huang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China.,Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China
| | - Guo-Ying Liu
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China.,Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China
| | - Wang-Zhong Li
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China.,Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China
| | - Yan-Qun Xiang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China.,Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China
| | - Tie-Bang Kang
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China.
| | - Xiang Guo
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China. .,Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China.
| | - Xing Lv
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China. .,Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, People's Republic of China.
| |
Collapse
|
28
|
Pang Y, Zhao J, Fowdur M, Liu Y, Wu H, He M. To Explore the Mechanism of the GRM4 Gene in Osteosarcoma by RNA Sequencing and Bioinformatics Approach. Med Sci Monit Basic Res 2018; 24:16-25. [PMID: 29339716 PMCID: PMC5782838 DOI: 10.12659/msmbr.908107] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background Glutamate metabotropic receptor 4 (GRM4) has been correlated with the pathogenesis of osteosarcoma. The objective of this study was to explore the underlying molecular mechanism of GRM4 in osteosarcoma. Material/Methods The expression levels of GRM4 in four human osteosarcoma cell lines and hFOB1.19 cells were examined by real-time quantitative PCR (RT-qPCR). The U2OS cells of the highest GRM4 expression were transfected with lentivirus-mediated small interfering RNA (siRNA). The differentially expressed genes (DEGs) after GRM4 gene silencing were screened through RNA sequencing, and analyzed by bioinformatics. Additionally, the transcription factors (TFs) targeting GRM4 were predicted and the downstream protein-protein interaction (PPI) network was constructed using the bioinformatics approach. Results A total of 51 significant DEGs were obtained, including 14 upregulated and 37 downregulated DEGs. The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of the DEGs indicated that four significant enrichment pathways were obtained. A total of six TFs that could be involved in the transcriptional regulation of GRM4 were detected. The results showed that 182 genes in the PPI network were significantly enriched in 14 pathways. The chemokines and chemokine receptors were found to be significantly enriched in three pathways. Conclusions The DEGs in the four significant enrichment pathways might participate in the development and progression of osteosarcoma through GRM4. The results revealed that EGR1 and CTCF are probably involved in the transcriptional regulation of GRM4, which participates in the progress of osteosarcoma by interacting with chemokines and their receptors.
Collapse
Affiliation(s)
- Yunguo Pang
- Division of Spinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland).,Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Jinmin Zhao
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China (mainland).,Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Mitra Fowdur
- Division of Spinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland).,Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Yun Liu
- Division of Spinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Hao Wu
- Division of Spinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Maolin He
- Division of Spinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland).,Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| |
Collapse
|
29
|
He D, Zhang S. UNBS5162 inhibits the proliferation of esophageal cancer squamous cells via the PI3K/AKT signaling pathway. Mol Med Rep 2017; 17:549-555. [PMID: 29115622 DOI: 10.3892/mmr.2017.7893] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/14/2017] [Indexed: 11/06/2022] Open
Abstract
C‑X‑C motif chemokine ligand (CXCL) signaling has been demonstrated to be involved in cancer invasion and migration; therefore, CXCL antagonists may serve as anticancer drugs by preventing tumor proliferation. The present study aimed to investigate whether a pan antagonist of CXCLs, UNBS5162, may inhibit esophageal cancer proliferation and to identify the underlying mechanisms. Cell proliferation and cell colony formation results, which were determined by a Cell Counting Kit‑8 assay and crystal violet staining, respectively, demonstrated that UNBS5162 inhibited esophageal cancer cell proliferation. Following treatment with UNBS5162, Transwell migration and Matrigel invasion assays, and flow cytometry with Annexin V‑fluorescein isothiocyanate and propidium iodide staining, were performed to investigate cell migration, invasion and apoptosis in human esophageal cancer cells. The results indicated that invasion and migration was reduced in UNBS5162‑treated cells, while apoptosis was increased. Western blotting experiments confirmed that UNBS5162 downregulated the protein expression of proteins associated with the phosphatidylinositol 3‑kinase (PI3K)/AKT signaling pathway, including the levels of phosphorylated (p)‑AKT, p‑mechanistic target of rapamycin kinase, ribosomal protein S6 kinase β1 and cyclin D1. In addition, upregulated expression of programed cell death 4 was observed following UNBS5162 treatment. The present study demonstrated that UNBS5162 is a novel naphthalimide that may have potential therapeutic use for the prevention of esophageal cancer proliferation and metastasis via the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Dan He
- Department of Thoracic Surgery, Affiliated Tumor Hospital, Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Suolin Zhang
- Department of Thoracic Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| |
Collapse
|