1
|
Yang L, Ren S, Lou L, He J, Huang Q, Wu X, Zhao R. A bioinformatics analysis and experimental validation of PDGFD as a promising diagnostic biomarker for acute myeloid leukemia. Sci Rep 2025; 15:14862. [PMID: 40295666 PMCID: PMC12037738 DOI: 10.1038/s41598-025-99038-0] [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: 10/04/2024] [Accepted: 04/16/2025] [Indexed: 04/30/2025] Open
Abstract
Acute myeloid leukemia (AML) is a malignant blood cancer resulting from leukemia stem cells (LSCs) supplanting normal stem cells. Platelet-derived growth factors (PDGFs) are important for LSCs but have not been studied in the development of AML. In this study, transcriptome data of PDGFs were sourced from The Cancer Genome Atlas (TCGA) and GTEx databases, and relevant differential expression and prognosis analysis were performed using R software packages and online tools (UCSC-Xena Shiny tools, GEPIA2, Kaplan-Meier Plotter databases, etc.). Then, we focused on PDGFD expression in AML, along with its clinical and diagnostic importance, drug resistance studies, and association with immunotherapy. The real-time quantitative polymerase chain reaction (RT-qPCR) was performed to verify the expression and clinical characteristics of PDGFD. Analyses of public data and clinical samples revealed that PDGFD expression was upregulated compared with other PDGF genes, and only this upregulation was associated with poor prognosis in AML. High expression of PDGFD showed a significant positive correlation with intermediate-high cytogenetic risk, NPM1 mutation, FLT3-ITD mutation, and unfavorable prognosis. ROC curve analysis indicated that PDGFD holds substantial diagnostic potential for AML patients. Functional enrichment analysis revealed the role of PDGFD in calcium and Rap1 signaling pathways. Additionally, PDGFD expression exhibited a significant positive correlation with natural killer cells and dendritic cells. Furthermore, we propose that MiR-203-3p targeting PDGFD has potential anti-leukemic effects in AML. In conclusion, PDGFD serves as a possible diagnostic and prognostic biomarker, as well as a target for cellular immunotherapy in AML.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/mortality
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Computational Biology/methods
- Nucleophosmin
- Prognosis
- Platelet-Derived Growth Factor/genetics
- Platelet-Derived Growth Factor/metabolism
- Lymphokines/genetics
- Lymphokines/metabolism
- Male
- Female
- Gene Expression Regulation, Leukemic
- Middle Aged
- Gene Expression Profiling
- Mutation
Collapse
Affiliation(s)
- Lulu Yang
- Department of Hematology, Ninghai First Hospital, Ningbo, 315600, Zhejiang, China
| | - Shuangshuang Ren
- Department of Ultrasound, Dongyang People's Hospital, Jinhua, 322100, Zhejiang, China
| | - Lijiang Lou
- Department of Hematology, Ninghai First Hospital, Ningbo, 315600, Zhejiang, China
| | - Jiasu He
- Department of Anesthesiology, Ninghai First Hospital, Ningbo, 315600, Zhejiang, China
| | - Qianlei Huang
- Department of Hematology, The First Affiliated Hospital of Hainan Medical College, Haikou, 570102, Hainan, China
| | - Xiaojin Wu
- Department of Hematology, National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Ranran Zhao
- Department of Hematology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China.
| |
Collapse
|
2
|
Ballato M, Germanà E, Ricciardi G, Giordano WG, Tralongo P, Buccarelli M, Castellani G, Ricci-Vitiani L, D’Alessandris QG, Giuffrè G, Pizzimenti C, Fiorentino V, Zuccalà V, Ieni A, Caffo M, Fadda G, Martini M. Understanding Neovascularization in Glioblastoma: Insights from the Current Literature. Int J Mol Sci 2025; 26:2763. [PMID: 40141406 PMCID: PMC11943220 DOI: 10.3390/ijms26062763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/15/2025] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
Glioblastomas (GBMs), among the most aggressive and resilient brain tumors, characteristically exhibit high angiogenic potential, leading to the formation of a dense yet aberrant vasculature, both morphologically and functionally. With these premises, numerous expectations were initially placed on anti-angiogenic therapies, soon dashed by their limited efficacy in concretely improving patient outcomes. Neovascularization in GBM soon emerged as a complex, dynamic, and heterogeneous process, hard to manage with the classical standard of care. Growing evidence has revealed the existence of numerous non-canonical strategies of angiogenesis, variously exploited by GBM to meet its ever-increasing metabolic demand and differently involved in tumor progression, recurrence, and escape from treatments. In this review, we provide an accurate description of each neovascularization mode encountered in GBM tumors to date, highlighting the molecular players and signaling cascades primarily involved. We also detail the key architectural and functional aspects characteristic of the GBM vascular compartment because of an intricate crosstalk between the different angiogenic networks. Additionally, we explore the repertoire of emerging therapies against GBM that are currently under study, concluding with a question: faced with such a challenging scenario, could combined therapies, tailored to the patient's genetic signatures, represent an effective game changer?
Collapse
Affiliation(s)
- Mariagiovanna Ballato
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, 98125 Messina, Italy; (M.B.); (E.G.); (G.R.); (W.G.G.); (P.T.)
| | - Emanuela Germanà
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, 98125 Messina, Italy; (M.B.); (E.G.); (G.R.); (W.G.G.); (P.T.)
| | - Gabriele Ricciardi
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, 98125 Messina, Italy; (M.B.); (E.G.); (G.R.); (W.G.G.); (P.T.)
- Istituto Clinico Polispecialistico C.O.T. Cure Ortopediche Traumatologiche s.pa., 98124 Messina, Italy
| | - Walter Giuseppe Giordano
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, 98125 Messina, Italy; (M.B.); (E.G.); (G.R.); (W.G.G.); (P.T.)
| | - Pietro Tralongo
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, 98125 Messina, Italy; (M.B.); (E.G.); (G.R.); (W.G.G.); (P.T.)
| | - Mariachiara Buccarelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (G.C.); (L.R.-V.)
| | - Giorgia Castellani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (G.C.); (L.R.-V.)
| | - Lucia Ricci-Vitiani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.B.); (G.C.); (L.R.-V.)
| | | | - Giuseppe Giuffrè
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (G.G.); (V.F.); (V.Z.); (A.I.); (G.F.)
| | | | - Vincenzo Fiorentino
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (G.G.); (V.F.); (V.Z.); (A.I.); (G.F.)
| | - Valeria Zuccalà
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (G.G.); (V.F.); (V.Z.); (A.I.); (G.F.)
| | - Antonio Ieni
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (G.G.); (V.F.); (V.Z.); (A.I.); (G.F.)
| | - Maria Caffo
- Biomedical and Dental Sciences and Morphofunctional Imaging, Unit of Neurosurgery, University of Messina, 98122 Messina, Italy;
| | - Guido Fadda
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (G.G.); (V.F.); (V.Z.); (A.I.); (G.F.)
| | - Maurizio Martini
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (G.G.); (V.F.); (V.Z.); (A.I.); (G.F.)
| |
Collapse
|
3
|
Huang XY, Fu FY, Qian K, Feng QL, Cao S, Wu WY, Luo YL, Chen WJ, Zhang Z, Huang SC. CircHAT1 regulates the proliferation and phenotype switch of vascular smooth muscle cells in lower extremity arteriosclerosis obliterans through targeting SFRS1. Mol Cell Biochem 2025; 480:203-215. [PMID: 38409514 DOI: 10.1007/s11010-024-04932-2] [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: 08/02/2023] [Accepted: 01/05/2024] [Indexed: 02/28/2024]
Abstract
This study aimed to decipher the mechanism of circular ribonucleic acids (circRNAs) in lower extremity arteriosclerosis obliterans (LEASO). First, bioinformatics analysis was performed for screening significantly down-regulated cardiac specific circRNA-circHAT1 in LEASO. The expression of circHAT1 in LEASO clinical samples was detected by quantitative real-time polymerase chain reaction (qRT-PCR). The protein expression of splicing factor arginine/serine-rich 1 (SFRS1), α-smooth muscle actin (α-SMA), Calponin (CNN1), cyclin D1 (CNND1) and smooth muscle myosin heavy chain 11 (SMHC) in vascular smooth muscle cells (VSMCs) was detected by Western blotting. Cell Counting Kit-8 (CCK-8), 5-ethynyl-2'-deoxyuridine (EdU) and Transwell assays were used to evaluate cell proliferation and migration, respectively. RNA immunoprecipitation (RNA-IP) and RNA pulldown verified the interaction between SFRS1 and circHAT1. By reanalyzing the dataset GSE77278, circHAT1 related to VSMC phenotype conversion was screened, and circHAT1 was found to be significantly reduced in peripheral blood mononuclear cells (PBMCs) of LEASO patients compared with healthy controls. Knockdown of circHAT1 significantly promoted the proliferation and migration of VSMC cells and decreased the expression levels of contractile markers. However, overexpression of circHAT1 induced the opposite cell phenotype and promoted the transformation of VSMCs from synthetic to contractile. Besides, overexpression of circHAT1 inhibited platelet-derived growth factor-BB (PDGF-BB)-induced phenotype switch of VSMC cells. Mechanistically, SFRS1 is a direct target of circHAT1 to mediate phenotype switch, proliferation and migration of VSMCs. Overall, circHAT1 regulates SFRS1 to inhibit the cell proliferation, migration and phenotype switch of VSMCs, suggesting that it may be a potential therapeutic target for LEASO.
Collapse
Affiliation(s)
- Xian-Ying Huang
- Division of Vascular and Interventional Radiology, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Fang-Yong Fu
- Division of Vascular and Interventional Radiology, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Kai Qian
- Division of Vascular and Interventional Radiology, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Qiao-Li Feng
- Division of Vascular and Interventional Radiology, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Sai Cao
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Wei-Yu Wu
- Division of Vascular and Interventional Radiology, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Yuan-Lin Luo
- Division of Vascular and Interventional Radiology, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Wei-Jie Chen
- Department of Vascular Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong Province, China
| | - Zhi Zhang
- Department of Vascular Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong Province, China.
| | - Shui-Chuan Huang
- Division of Vascular and Interventional Radiology, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China.
| |
Collapse
|
4
|
Skapinker E, Aucoin EB, Kombargi HL, Yaish AM, Li Y, Baghaie L, Szewczuk MR. Contemporaneous Inflammatory, Angiogenic, Fibrogenic, and Angiostatic Cytokine Profiles of the Time-to-Tumor Development by Cancer Cells to Orchestrate Tumor Neovascularization, Progression, and Metastasis. Cells 2024; 13:1739. [PMID: 39451257 PMCID: PMC11506673 DOI: 10.3390/cells13201739] [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: 09/21/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
Cytokines can promote various cancer processes, such as angiogenesis, epithelial to mesenchymal transition (EMT), invasion, and tumor progression, and maintain cancer stem-cell-like (CSCs) cells. The mechanism(s) that continuously promote(s) tumors to progress in the TME still need(s) to be investigated. The data in the present study analyzed the inflammatory, angiogenic, fibrogenic, and angiostatic cytokine profiles in the host serum during tumor development in a mouse model of human pancreatic cancer. Pancreatic MiaPaCa-2-eGFP cancer cells were subcutaneously implanted in RAG2xCγ double mutant mice. Blood samples were collected before cancer cell implantation and every week until the end point of the study. The extracted serum from the blood of each mouse at different time points during tumor development was analyzed using a Bio-Plex microarray analysis and a Bio-Plex 200 system for proinflammatory (IL-1β, IL-10, IFN-γ, and TNF-α) and angiogenic and fibrogenic (IL-15, IL-18, basic FGF, LIF, M-CSF, MIG, MIP-2, PDGF-BB, and VEGF) cytokines. Here, we find that during cancer cell colonization for tumor development, host angiogenic, fibrogenic, and proinflammatory cytokine profiling in the tumor-bearing mice has been shown to significantly reduce host angiostatic and proinflammatory cytokines that restrain tumor development and increase those for tumor growth. The proinflammatory cytokines IL-15, IL-18, and IL-1β profiles reveal a significant host serum increase after day 35 when the tumor began to progress in growth. In contrast, the angiostatic cytokine profiles of TNFα, MIG, M-CSF, IL-10, and IFNγ in the host serum revealed a dramatic and significant decrease after day 5 post-implantation of cancer cells. OP treatment of tumor-bearing mice on day 35 maintained high levels of angiostatic and fibrogenic cytokines. The data suggest an entirely new regulation by cancer cells for tumor development. The findings identify for the first time how pancreatic cancer cells use host cytokine profiling to orchestrate the initiation of tumor development.
Collapse
Affiliation(s)
- Elizabeth Skapinker
- Faculty of Arts and Science, Queen’s University, Kingston, ON K7L 3N9, Canada; (E.S.); (Y.L.)
| | - Emilyn B. Aucoin
- Faculty of Science, Biology (Biomedical Science), York University, Toronto, ON M3J 1P3, Canada;
| | - Haley L. Kombargi
- Faculty of Health Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada; (H.L.K.); (A.M.Y.)
| | - Abdulrahman M. Yaish
- Faculty of Health Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada; (H.L.K.); (A.M.Y.)
| | - Yunfan Li
- Faculty of Arts and Science, Queen’s University, Kingston, ON K7L 3N9, Canada; (E.S.); (Y.L.)
| | - Leili Baghaie
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada;
| | - Myron R. Szewczuk
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada;
| |
Collapse
|
5
|
Wang K, Li C, Chen H, Gu P, Lu J, Zhao H, Zhuo X. Increased Expression of PDGFA Is Associated With Poor Prognosis and Immune Infiltration in Head and Neck Squamous Cell Carcinoma. J Oral Pathol Med 2024; 53:584-594. [PMID: 39295197 DOI: 10.1111/jop.13579] [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: 09/28/2023] [Revised: 06/11/2024] [Accepted: 08/02/2024] [Indexed: 09/21/2024]
Abstract
BACKGROUND Platelet-derived growth factor A (PDGFA) has been shown to be upregulated in several tumors, contributing to their malignant phenotypes. However, its expression and function in head and neck squamous cell carcinoma (HNSC) are not clearly understood. Thus, we aimed to evaluate this issue using bioinformatic analyses and primary experimental validation. METHODS The expression of PDGFA was analyzed using popular bio-databases and further validated by RT-PCR and immunohistochemical staining. Survival analyses were then performed. The association between PDGFA expression levels and immune cell infiltration in the immune microenvironment was assessed. RESULTS PDGFA has been found to be significantly upregulated in a variety of cancers, including HNSC, and increased PDGFA expression may be an independent prognostic factor associated with immune cell infiltration in HNSC. CONCLUSION Overexpression of PDGFA in HNSC is significantly associated with poor prognosis and immune cell infiltration in the tumor microenvironment (TME). PDGFA has potential as a molecular indicator for diagnosis, prognosis, and immune processes in HNSC.
Collapse
Affiliation(s)
- Kaiqin Wang
- Department of Otolaryngology-Head and Neck Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Changya Li
- Department of Otolaryngology-Head and Neck Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Huarong Chen
- Department of Otolaryngology-Head and Neck Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Ping Gu
- Department of Otolaryngology-Head and Neck Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Jiafeng Lu
- Department of Otolaryngology-Head and Neck Surgery, Anshun People's Hospital, Anshun, Guizhou Province, China
| | - Houyu Zhao
- Department of Otolaryngology-Head and Neck Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Xianlu Zhuo
- Department of Otolaryngology-Head and Neck Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
6
|
Ayoub NM, Sardiah S, Al-Share QY, Alkader MS. Exploring angiogenic pathways in breast cancer: Clinicopathologic correlations and prognostic implications based on gene expression profiles from a large-scale genomic dataset. PLoS One 2024; 19:e0310557. [PMID: 39302921 DOI: 10.1371/journal.pone.0310557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 09/03/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Angiogenesis inhibitors targeting VEGF, or its receptors have consistently produced disappointing clinical outcomes in breast cancer. Therefore, there is an urgent need to explore alternative angiogenic pathways in breast cancer. This study aimed to describe the gene expression of pivotal pro-angiogenic genes in breast cancer and to further analyze the associations with the clinicopathologic tumor features, prognostic factors, and overall survival. Such findings would expand the understanding of the role of different angiogenic pathways in breast cancer pathogenesis and identify patients at risk of more aggressive disease who could be eligible for intense treatment regimens. Additionally, exploring angiogenic pathways helps identify new potential drug targets for breast cancer. METHODS The mRNA expression levels for eight pro-angiogenic genes [VEGFA, HGF, FGF1, FGF2, ANGPT1, ANGPT2, PDGFA, and PDGFB] were obtained from the METABRIC (Molecular Taxonomy of Breast Cancer International Consortium) dataset available at cBioPortal public domain. Pertinent demographic and tumor information were retrieved. RESULTS VEGFA and ANGPT2 genes had the highest expression levels with average mRNA log intensities of 7.18±0.7 and 7.11±0.53, respectively. VEGFA expression was not correlated with the expression of other pro-angiogenic genes, the clinicopathologic tumor features, and the overall survival of patients. FGF1, ANGPT1, and PDGFA mRNA levels were negatively correlated with the age of patients at diagnosis. The expression of FGF1 and FGF2 correlated inversely with tumor size and the Nottingham Prognostic Index (p = 0.03 and p = 0.002, respectively). Expression of HGF was significantly associated with advanced tumor stage (p<0.05). Expression of ANGPT1 and ANGPT2 was associated with hormone receptor-negative status and the non-luminal subtypes. PDGFB expression was significantly higher in patients with high-grade disease and HER2-positive status. Patients with high expression status of ANGPT2 and PDGFB had significantly reduced overall survival compared to those with low expression levels of these genes (p = 0.004 and p = 0.0001, respectively). CONCLUSIONS In this dataset of patients with breast cancer, the expression levels of 8 different pro-angiogenic genes revealed remarkable differences in terms of their association with clinicopathologic tumor characteristics and prognosis. The expression of ANGPTs and PDGFs was associated with adverse tumor features, worse prognosis, and reduced survival in patients. Targeting ANGPTs and PDGF pathways could provide new insights for effective anti-angiogenic drugs in breast cancer.
Collapse
Affiliation(s)
- Nehad M Ayoub
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Salam Sardiah
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Qusai Y Al-Share
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Mohammad S Alkader
- Department of Medical Oncology, Military Cancer Center, Jordanian Royal Medical Services, Amman, Jordan
| |
Collapse
|
7
|
Tang M, Zhang Z, Wang P, Zhao F, Miao L, Wang Y, Li Y, Li Y, Gao Z. Advancements in precision nanomedicine design targeting the anoikis-platelet interface of circulating tumor cells. Acta Pharm Sin B 2024; 14:3457-3475. [PMID: 39220884 PMCID: PMC11365446 DOI: 10.1016/j.apsb.2024.04.034] [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] [Received: 12/28/2023] [Revised: 03/10/2024] [Accepted: 03/13/2024] [Indexed: 09/04/2024] Open
Abstract
Tumor metastasis, the apex of cancer progression, poses a formidable challenge in therapeutic endeavors. Circulating tumor cells (CTCs), resilient entities originating from primary tumors or their metastases, significantly contribute to this process by demonstrating remarkable adaptability. They survive shear stress, resist anoikis, evade immune surveillance, and thwart chemotherapy. This comprehensive review aims to elucidate the intricate landscape of CTC formation, metastatic mechanisms, and the myriad factors influencing their behavior. Integral signaling pathways, such as integrin-related signaling, cellular autophagy, epithelial-mesenchymal transition, and interactions with platelets, are examined in detail. Furthermore, we explore the realm of precision nanomedicine design, with a specific emphasis on the anoikis‒platelet interface. This innovative approach strategically targets CTC survival mechanisms, offering promising avenues for combatting metastatic cancer with unprecedented precision and efficacy. The review underscores the indispensable role of the rational design of platelet-based nanomedicine in the pursuit of restraining CTC-driven metastasis.
Collapse
Affiliation(s)
- Manqing Tang
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhijie Zhang
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ping Wang
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Feng Zhao
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Lin Miao
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yuming Wang
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yingpeng Li
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yunfei Li
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhonggao Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
8
|
Vargová D, Dargaj J, Dohál M, Fraňová S, Ľupták J, Škorňová I, Švihra J, Briš L, Slávik P, Šutovská M. Immune analysis of urine and plasma samples from patients with clear cell renal cell carcinoma. Oncol Lett 2024; 27:281. [PMID: 38736737 PMCID: PMC11082642 DOI: 10.3892/ol.2024.14414] [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] [Received: 10/31/2023] [Accepted: 03/25/2024] [Indexed: 05/14/2024] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the third most common type of urological malignancy worldwide, and it is associated with a silent progression and late manifestation. Patients with a metastatic form of ccRCC have a poor prognosis; however, when the disease is diagnosed early, it is largely curable. Currently, there are no biomarkers available in clinical practice for ccRCC. Thus, the aim of the present study was to measure 27 biologically relevant cytokines in preoperative and postoperative urine samples, and in preoperative plasma samples from 34 patients with ccRCC, and to evaluate their diagnostic significance. The concentrations of cytokines were assessed by multiplex immune assay. The results showed significantly higher levels of IL-1 receptor antagonist, IL-6, IL-15, chemokine (C-C motif) ligand (CCL)2, CCL3, CCL4, C-X-C motif ligand (CXCL)10, granulocyte-macrophage colony stimulating factor (GM-CSF) and platelet-derived growth factor-BB (PDGF-BB), and lower levels of granulocyte colony stimulating factor (G-CSF) in urine samples from patients prior to surgery compared with those in the controls. Notably, the urine levels of G-CSF, IL-5 and vascular endothelial growth factor differed following tumor removal compared with the preoperative urine levels. In addition, urinary G-CSF, GM-CSF, IL-6, CXCL10, CCL5 and PDGF-BB appeared to be potential markers of tumor grade. Plasma from patients with ccRCC contained significantly higher levels of IL-6 and lower levels of CCL2 than control plasma. In conclusion, the present findings indicated that urinary and circulating cytokines may represent a promising novel tool for the early diagnosis of ccRCC and/or prediction of tumor grade.
Collapse
Affiliation(s)
- Daniela Vargová
- Department of Pharmacology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Ján Dargaj
- Department of Urology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava and University Hospital Martin, 036 01 Martin, Slovakia
| | - Matúš Dohál
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Soňa Fraňová
- Department of Pharmacology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Ján Ľupták
- Department of Urology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava and University Hospital Martin, 036 01 Martin, Slovakia
| | - Ingrid Škorňová
- Department of Hematology and Transfusiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava and University Hospital Martin, 036 01 Martin, Slovakia
| | - Ján Švihra
- Department of Urology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava and University Hospital Martin, 036 01 Martin, Slovakia
| | - Lukáš Briš
- Department of Urology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava and University Hospital Martin, 036 01 Martin, Slovakia
| | - Pavol Slávik
- Department of Pathological Anatomy, Jessenius Faculty of Medicine in Martin, Comenius University, 036 01 Martin, Slovakia
| | - Martina Šutovská
- Department of Pharmacology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| |
Collapse
|
9
|
Wen Z, Zhang Y, Wang X, Wu Y, Mao J, Li Q, Gong S. THBS1-Mediated Degradation of Collagen via the PI3K/AKT Pathway Facilitates the Metastasis and Poor Prognosis of OSCC. Int J Mol Sci 2023; 24:13312. [PMID: 37686118 PMCID: PMC10488045 DOI: 10.3390/ijms241713312] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/12/2023] [Accepted: 08/17/2023] [Indexed: 09/10/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a prevalent form of malignant tumor, characterized by a persistently high incidence and mortality rate. The extracellular matrix (ECM) plays a crucial role in the initiation, progression, and diverse biological behaviors of OSCC, facilitated by mechanisms such as providing structural support, promoting cell migration and invasion, regulating cell morphology, and modulating signal transduction. This study investigated the involvement of ECM-related genes, particularly THBS1, in the prognosis and cellular behavior of OSCC. The analysis of ECM-related gene data from OSCC samples identified 165 differentially expressed genes forming two clusters with distinct prognostic outcomes. Seventeen ECM-related genes showed a significant correlation with survival. Experimental methods were employed to demonstrate the impact of THBS1 on proliferation, migration, invasion, and ECM degradation in OSCC cells. A risk-prediction model utilizing four differentially prognostic genes demonstrated significant predictive value in overall survival. THBS1 exhibited enrichment of the PI3K/AKT pathway, indicating its potential role in modulating OSCC. In conclusion, this study observed and verified that ECM-related genes, particularly THBS1, have the potential to influence the prognosis, biological behavior, and immunotherapy of OSCC. These findings hold significant implications for enhancing survival outcomes and providing guidance for precise treatment of OSCC.
Collapse
Affiliation(s)
- Zhihao Wen
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.W.); (Y.Z.); (X.W.); (Y.W.); (J.M.)
| | - Yuxiao Zhang
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.W.); (Y.Z.); (X.W.); (Y.W.); (J.M.)
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Xiangyao Wang
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.W.); (Y.Z.); (X.W.); (Y.W.); (J.M.)
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Yaxin Wu
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.W.); (Y.Z.); (X.W.); (Y.W.); (J.M.)
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Jing Mao
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.W.); (Y.Z.); (X.W.); (Y.W.); (J.M.)
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Qilin Li
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.W.); (Y.Z.); (X.W.); (Y.W.); (J.M.)
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Shiqiang Gong
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.W.); (Y.Z.); (X.W.); (Y.W.); (J.M.)
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| |
Collapse
|
10
|
Age-associated microenvironmental changes highlight the role of PDGF-C in ER + breast cancer metastatic relapse. NATURE CANCER 2023; 4:468-484. [PMID: 36914817 PMCID: PMC10132974 DOI: 10.1038/s43018-023-00525-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 02/07/2023] [Indexed: 03/16/2023]
Abstract
Patients with estrogen receptor (ER)-positive breast cancer are at risk of metastatic relapse for decades after primary tumor resection and treatment, a consequence of dormant disseminated tumor cells (DTCs) reawakening at secondary sites. Here we use syngeneic ER+ mouse models in which DTCs display a dormant phenotype in young mice but accelerated metastatic outgrowth in an aged or fibrotic microenvironment. In young mice, low-level Pdgfc expression by ER+ DTCs is required for their maintenance in secondary sites but is insufficient to support development of macrometastases. By contrast, the platelet-derived growth factor (PDGF)-Chi environment of aging or fibrotic lungs promotes DTC proliferation and upregulates tumor cell Pdgfc expression stimulating further stromal activation, events that can be blocked by pharmacological inhibition of PDGFRα or with a PDGF-C-blocking antibody. These results highlight the role of the changing microenvironment in regulating DTC outgrowth and the opportunity to target PDGF-C signaling to limit metastatic relapse in ER+ breast cancer.
Collapse
|
11
|
Mei C, Gong W, Wang X, Lv Y, Zhang Y, Wu S, Zhu C. Anti-angiogenic therapy in ovarian cancer: Current understandings and prospects of precision medicine. Front Pharmacol 2023; 14:1147717. [PMID: 36959862 PMCID: PMC10027942 DOI: 10.3389/fphar.2023.1147717] [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: 01/19/2023] [Accepted: 02/23/2023] [Indexed: 03/09/2023] Open
Abstract
Ovarian cancer (OC) remains the most fatal disease of gynecologic malignant tumors. Angiogenesis refers to the development of new vessels from pre-existing ones, which is responsible for supplying nutrients and removing metabolic waste. Although not yet completely understood, tumor vascularization is orchestrated by multiple secreted factors and signaling pathways. The most central proangiogenic signal, vascular endothelial growth factor (VEGF)/VEGFR signaling, is also the primary target of initial clinical anti-angiogenic effort. However, the efficiency of therapy has so far been modest due to the low response rate and rapidly emerging acquiring resistance. This review focused on the current understanding of the in-depth mechanisms of tumor angiogenesis, together with the newest reports of clinical trial outcomes and resistance mechanism of anti-angiogenic agents in OC. We also emphatically summarized and analyzed previously reported biomarkers and predictive models to describe the prospect of precision therapy of anti-angiogenic drugs in OC.
Collapse
Affiliation(s)
- Chao Mei
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weijing Gong
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, China
| | - Xu Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongning Lv
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sanlan Wu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, China
| | - Chunqi Zhu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
12
|
Lee YE, Go GY, Koh EY, Yoon HN, Seo M, Hong SM, Jeong JH, Kim JC, Cho D, Kim TS, Kim SC, Jun E, Jang M. Synergistic therapeutic combination with a CAF inhibitor enhances CAR-NK-mediated cytotoxicity via reduction of CAF-released IL-6. J Immunother Cancer 2023; 11:e006130. [PMID: 36849201 PMCID: PMC9972461 DOI: 10.1136/jitc-2022-006130] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2023] [Indexed: 03/01/2023] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) in the tumor microenvironment (TME) contribute to an impaired functionality of natural killer (NK) cells that have emerged as a promising therapeutic modality. The interaction between CAFs and NK cells within the TME exerts major inhibitory effects on immune responses, indicating CAF-targeted therapies as potential targets for effective NK-mediated cancer killing. METHODS To overcome CAF-induced NK dysfunction, we selected an antifibrotic drug, nintedanib, for synergistic therapeutic combination. To evaluate synergistic therapeutic efficacy, we established an in vitro 3D Capan2/patient-derived CAF spheroid model or in vivo mixed Capan2/CAF tumor xenograft model. The molecular mechanism of NK-mediated synergistic therapeutic combination with nintedanib was revealed through in vitro experiments. In vivo therapeutic combination efficacy was subsequently evaluated. Additionally, the expression score of target proteins was measured in patient-derived tumor sections by the immunohistochemical method. RESULTS Nintedanib blocked the platelet-derived growth factor receptor β (PDGFRβ) signaling pathway and diminished the activation and growth of CAFs, markedly reducing CAF-secreted IL-6. Moreover, coadministration of nintedanib improved the mesothelin (MSLN) targeting chimeric antigen receptor-NK-mediated tumor killing abilities in CAF/tumor spheroids or a xenograft model. The synergistic combination resulted in intense NK infiltration in vivo. Nintedanib alone exerted no effects, whereas blockade of IL-6 trans-signaling ameliorated the function of NK cells. The combination of the expression of MSLN and the PDGFRβ+-CAF population area, a potential prognostic/therapeutic marker, was associated with inferior clinical outcomes. CONCLUSION Our strategy against PDGFRβ+-CAF-containing pancreatic cancer allows improvements in the therapy of pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Young Eun Lee
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, Korea (the Republic of)
- Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea (the Republic of)
| | - Ga-Yeon Go
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, Korea (the Republic of)
| | - Eun-Young Koh
- Department of Convergence Medicine, Asan Institute for Life Sciences, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea (the Republic of)
| | - Han-Na Yoon
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, Korea (the Republic of)
| | - Minkoo Seo
- Corporate Research & Development Center, UCI therapeutics, Seoul, Korea (the Republic of)
| | - Seung-Mo Hong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea (the Republic of)
| | - Ji Hye Jeong
- Department of Convergence Medicine, Asan Institute for Life Sciences, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea (the Republic of)
| | - Jin-Chul Kim
- Natural Product Research Center, Institute of Natural Products, Korea Institute of Science and Technology, Gangneung, Korea (the Republic of)
| | - Duck Cho
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea (the Republic of)
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University, Seoul, Korea (the Republic of)
| | - Tae Sung Kim
- Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea (the Republic of)
| | - Song Cheol Kim
- Division of Hepato-Biliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea (the Republic of)
| | - Eunsung Jun
- Department of Convergence Medicine, Asan Institute for Life Sciences, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea (the Republic of)
- Division of Hepato-Biliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea (the Republic of)
| | - Mihue Jang
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, Korea (the Republic of)
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, Korea (the Republic of)
| |
Collapse
|
13
|
Sadovskaya AV, Petinati NA, Kapranov NM, Drize NI, Vasil'eva AN, Aleshina OA, Parovichnikova EN. Dynamics of Changes in the Properties of Multipotent Mesenchymal Stromal Cells in Patients with Acute Leukemia. Bull Exp Biol Med 2023; 174:556-563. [PMID: 36894815 DOI: 10.1007/s10517-023-05747-9] [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: 09/12/2022] [Indexed: 03/11/2023]
Abstract
In acute leukemia, the stromal microenvironment of the bone marrow that regulates hematopoiesis is modified under the influence of malignant cells. Chemotherapy also adversely affects stromal cells. Multipotent mesenchymal stromal cells (MSC) are involved in the formation of the stromal microenvironment and in the regulation of normal and tumor hematopoietic cells. The properties of MSC from the bone marrow of patients with acute myeloid and lymphoid leukemia were studied at the onset of the disease and after achieving remission. The immunophenotype and the level of gene expression were analyzed in MSC of 34 patients. In MSC from patients with acute leukemia, the expression of CD105 and CD274 was significantly reduced in comparison with MSC from healthy donors. At the onset of the disease, the expression of IL6, JAG1, PPARG, IGF1, and PDGFRA was enhanced, while the expression of IL1B, IL8, SOX9, ANG1, and TGFB was reduced. All these changes affect the course of the disease in patients and can be the targets of therapeutic intervention.
Collapse
Affiliation(s)
- A V Sadovskaya
- National Medical Research Center of Hematology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - N A Petinati
- National Medical Research Center of Hematology, Ministry of Health of the Russian Federation, Moscow, Russia.
| | - N M Kapranov
- National Medical Research Center of Hematology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - N I Drize
- National Medical Research Center of Hematology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - A N Vasil'eva
- National Medical Research Center of Hematology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - O A Aleshina
- National Medical Research Center of Hematology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - E N Parovichnikova
- National Medical Research Center of Hematology, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
14
|
Hu Z, Wen S, Huo Z, Wang Q, Zhao J, Wang Z, Chen Y, Zhang L, Zhou F, Guo Z, Liu H, Zhou S. Current Status and Prospects of Targeted Therapy for Osteosarcoma. Cells 2022; 11:3507. [PMID: 36359903 PMCID: PMC9653755 DOI: 10.3390/cells11213507] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 09/26/2023] Open
Abstract
Osteosarcoma (OS) is a highly malignant tumor occurring in bone tissue with a high propensity to metastasize, and its underlying mechanisms remain largely elusive. The OS prognosis is poor, and improving the survival of OS patients remains a challenge. Current treatment methods such as surgical approaches, chemotherapeutic drugs, and immunotherapeutic drugs remain ineffective. As research progresses, targeted therapy is gradually becoming irreplaceable. In this review, several treatment modalities for osteosarcoma, such as surgery, chemotherapy, and immunotherapy, are briefly described, followed by a discussion of targeted therapy, the important targets, and new technologies for osteosarcoma treatment.
Collapse
Affiliation(s)
- Zunguo Hu
- Department of Joint Surgery, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang 261061, China
| | - Shuang Wen
- Department of Joint Surgery, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang 261061, China
| | - Zijun Huo
- Department of Histology and Embryology, School of Basic Medical Sciences, Weifang Medical University, Weifang 261053, China
| | - Qing Wang
- Neurologic Disorders and Regenerative Repair Laboratory, Weifang Medical University, Weifang 261053, China
| | - Jiantao Zhao
- Department of Histology and Embryology, School of Basic Medical Sciences, Weifang Medical University, Weifang 261053, China
| | - Zihao Wang
- Department of Joint Surgery, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang 261061, China
| | - Yanchun Chen
- Department of Histology and Embryology, School of Basic Medical Sciences, Weifang Medical University, Weifang 261053, China
| | - Lingyun Zhang
- Neurologic Disorders and Regenerative Repair Laboratory, Weifang Medical University, Weifang 261053, China
| | - Fenghua Zhou
- Neurologic Disorders and Regenerative Repair Laboratory, Weifang Medical University, Weifang 261053, China
| | - Zhangyu Guo
- Neurologic Disorders and Regenerative Repair Laboratory, Weifang Medical University, Weifang 261053, China
| | - Huancai Liu
- Department of Joint Surgery, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang 261061, China
| | - Shuanhu Zhou
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
15
|
Jiang S, Ito-Hirano R, Shen TNY, Fujimura S, Mizuno H, Tanaka R. Effect of MNCQQ Cells on Migration of Human Dermal Fibroblast in Diabetic Condition. Biomedicines 2022; 10:biomedicines10102544. [PMID: 36289806 PMCID: PMC9599466 DOI: 10.3390/biomedicines10102544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/05/2022] [Accepted: 10/09/2022] [Indexed: 11/18/2022] Open
Abstract
A major symptom of diabetes mellitus (DM) is unfit hyperglycemia, which leads to impaired wound healing. It has been reported that the migration of fibroblasts can be suppressed under high glucose (HG) conditions. In our previous study, we introduced a serum-free culture method for mononuclear cells (MNCs) called quantity and quality control culture (QQc), which could improve the vasculogenic and tissue regeneration ability of MNCs. In this study, we described a culture model in which we applied a high glucose condition in human dermal fibroblasts to simulate the hyperglycemia condition in diabetic patients. MNC-QQ cells were cocultured with fibroblasts in this model to evaluate its role in improving fibroblasts dysfunction induced by HG and investigate its molecular mechanism. It was proven in this study that the impaired migration of fibroblasts induced by high glucose could be remarkably enhanced by coculture with MNC-QQ cells. PDGF B is known to play important roles in fibroblasts migration. Quantitative PCR revealed that MNC-QQ cells enhanced the gene expressions of PDGF B in fibroblasts under HG. Taken with these results, our data suggested a possibility that MNC-QQ cells accelerate wound healing via improving the fibroblasts migration and promote the gene expressions of PDGF B under diabetic conditions.
Collapse
Affiliation(s)
- Sen Jiang
- Division of Regenerative Therapy, Graduate School of Medicine, Juntendo University, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Rie Ito-Hirano
- Division of Regenerative Therapy, Graduate School of Medicine, Juntendo University, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Center for Genomic and Regenerative Medicine, Graduate School of Medicine, Juntendo University, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Tsubame Nishikai-Yan Shen
- Division of Regenerative Therapy, Graduate School of Medicine, Juntendo University, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Plastic and Reconstructive Surgery, School of Medicine, Juntendo University, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Satoshi Fujimura
- Division of Regenerative Therapy, Graduate School of Medicine, Juntendo University, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Hiroshi Mizuno
- Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Plastic and Reconstructive Surgery, School of Medicine, Juntendo University, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Rica Tanaka
- Division of Regenerative Therapy, Graduate School of Medicine, Juntendo University, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Plastic and Reconstructive Surgery, School of Medicine, Juntendo University, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Correspondence:
| |
Collapse
|
16
|
Dabravolski SA, Markin AM, Andreeva ER, Eremin II, Orekhov AN, Melnichenko AA. Molecular Mechanisms Underlying Pathological and Therapeutic Roles of Pericytes in Atherosclerosis. Int J Mol Sci 2022; 23:11663. [PMID: 36232962 PMCID: PMC9570222 DOI: 10.3390/ijms231911663] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/17/2022] Open
Abstract
Pericytes are multipotent mesenchymal stromal cells playing an active role in angiogenesis, vessel stabilisation, maturation, remodelling, blood flow regulation and are able to trans-differentiate into other cells of the mesenchymal lineage. In this review, we summarised recent data demonstrating that pericytes play a key role in the pathogenesis and development of atherosclerosis (AS). Pericytes are involved in lipid accumulation, inflammation, growth, and vascularization of the atherosclerotic plaque. Decreased pericyte coverage, endothelial and pericyte dysfunction is associated with intraplaque angiogenesis and haemorrhage, calcification and cholesterol clefts deposition. At the same time, pericytes can be used as a novel therapeutic target to promote vessel maturity and stability, thus reducing plaque vulnerability. Finally, we discuss recent studies exploring effective AS treatments with pericyte-mediated anti-atherosclerotic, anti-inflammatory and anti-apoptotic effects.
Collapse
Affiliation(s)
- Siarhei A. Dabravolski
- Department of Biotechnology Engineering, ORT Braude College, Snunit 51, P.O. Box 78, Karmiel 2161002, Israel
| | - Alexander M. Markin
- Petrovsky National Research Center of Surgery, Abrikosovsky Lane, 2, 119991 Moscow, Russia
| | - Elena R. Andreeva
- Laboratory of Cell Physiology, Institute of Biomedical Problems, Russian Academy of Sciences, Khoroshevskoye Shosse, 76a, 123007 Moscow, Russia
| | - Ilya I. Eremin
- Petrovsky National Research Center of Surgery, Abrikosovsky Lane, 2, 119991 Moscow, Russia
| | - Alexander N. Orekhov
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia
| | | |
Collapse
|
17
|
Wu R, Gandhi S, Tokumaru Y, Asaoka M, Oshi M, Yan L, Ishikawa T, Takabe K. Intratumoral PDGFB gene predominantly expressed in endothelial cells is associated with angiogenesis and lymphangiogenesis, but not with metastasis in breast cancer. Breast Cancer Res Treat 2022; 195:17-31. [PMID: 35793004 PMCID: PMC12118772 DOI: 10.1007/s10549-022-06661-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/15/2022] [Indexed: 11/02/2022]
Abstract
PURPOSE Platelet-derived growth factor B (PDGFB) is known to play essential roles in angiogenesis and lymphangiogenesis during development, and tumor growth and vessel stabilization in experimental models. However, whether these findings could be translated to breast cancer patients remains unclear. We hypothesized that PDGFB gene expression is associated with angiogenesis, cell proliferation, and clinical outcomes in breast cancer patients. METHODS A total of 7635 primary breast cancer patients with full transcriptome and clinical data available from 13 independent cohorts were analyzed using in silico approach. The median value was used to divide each cohort into high and low PDGFB expression groups. RESULTS High PDGFB gene expression was associated with increased expression of angiogenesis-related genes, higher amount of vascular cell infiltrations, and with enrichment of angiogenesis gene set, lymphangiogenesis-related gene expressions, lymphangiogenesis-related cell infiltrations, and enrichmentof lymphangiogenesis gene set in GSE96058 and validated by TCGA cohorts; however, not with lymphatic metastasis. PDGFB expression was neither associated with cell proliferation as assessed by Ki67 expression nor with Nottingham histological grade, or response to neoadjuvant chemotherapy. We found that PDGFB was most extensively expressed by endothelial and perivascular-like cells in the tumor microenvironment, and minimally by cancer cells consistently in two single-cell sequence cohorts. High PDGFB expression enriched TGFβ, epithelial-mesenchymal transition, hypoxia, and cancer stem cell-associated pathways. However, no association with distant metastasis was observed. Disease-specific and disease-free survival were worse in the high PDGFB expression group consistently in TCGA and METABRIC cohorts. CONCLUSION PDGFB is predominantly expressed in endothelial cells and is associated with angiogenesis and lymphangiogenesis, but not with cellular proliferation or metastasis in breast cancer.
Collapse
Affiliation(s)
- Rongrong Wu
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, 14263, USA
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo, 160-8402, Japan
| | - Shipra Gandhi
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Yoshihisa Tokumaru
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, 14263, USA
- Department of Surgical Oncology, Graduate School of Medicine, Gifu University, Gifu, 501-1193, Japan
| | - Mariko Asaoka
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo, 160-8402, Japan
| | - Masanori Oshi
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, 14263, USA
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, 236-0004, Japan
| | - Li Yan
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Takashi Ishikawa
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo, 160-8402, Japan
| | - Kazuaki Takabe
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, 14263, USA.
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo, 160-8402, Japan.
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, 236-0004, Japan.
- Department of Surgery, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY, 14263, USA.
- Department of Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.
- Department of Breast Surgery, Fukushima Medical University, Fukushima, Japan.
| |
Collapse
|
18
|
Guo H, Qian Y, Yu Y, Bi Y, Jiao J, Jiang H, Yu C, Wu H, Shi Y, Kong X. An Immunity-Related Gene Model Predicts Prognosis in Cholangiocarcinoma. Front Oncol 2022; 12:791867. [PMID: 35847907 PMCID: PMC9283581 DOI: 10.3389/fonc.2022.791867] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 05/31/2022] [Indexed: 12/11/2022] Open
Abstract
The prognosis of patients with cholangiocarcinoma (CCA) is closely related to both immune cell infiltration and mRNA expression. Therefore, we aimed at conducting multi-immune-related gene analyses to improve the prediction of CCA recurrence. Immune-related genes were selected from the Gene Expression Omnibus (GEO), The Cancer Genome Atlas (TCGA), and the Immunology Database and Analysis Portal (ImmPort). The least absolute shrinkage and selection operator (LASSO) regression model was used to establish the multi-gene model that was significantly correlated with the recurrence-free survival (RFS) in two test series. Furthermore, compared with single genes, clinical characteristics, tumor immune dysfunction and exclusion (TIDE), and tumor inflammation signature (TIS), the 8-immune-related differentially expressed genes (8-IRDEGs) signature had a better prediction value. Moreover, the high-risk subgroup had a lower density of B-cell, plasma, B-cell naïve, CD8+ T-cell, CD8+ T-cell naïve, and CD8+ T-cell memory infiltration, as well as more severe immunosuppression and higher mutation counts. In conclusion, the 8-IRDEGs signature was a promising biomarker for distinguishing the prognosis and the molecular and immune features of CCA, and could be beneficial to the individualized immunotherapy for CCA patients.
Collapse
Affiliation(s)
- Han Guo
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Clinical Immunology, Department of Liver Diseases, Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Yihan Qian
- Institute of Clinical Immunology, Department of Liver Diseases, Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Yeping Yu
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuting Bi
- Institute of Clinical Immunology, Department of Liver Diseases, Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Junzhe Jiao
- Institute of Clinical Immunology, Department of Liver Diseases, Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Haocheng Jiang
- Institute of Clinical Immunology, Department of Liver Diseases, Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Chang Yu
- Institute of Clinical Immunology, Department of Liver Diseases, Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Hailong Wu
- Shanghai Key Laboratory for Molecular Imaging, Collaborative Research Center, Shanghai University of Medicine and Health Sciences, Shanghai, China
- *Correspondence: Xiaoni Kong, ; Yanjun Shi, ; Hailong Wu,
| | - Yanjun Shi
- Department of Hepatobiliary and Pancreas Surgery , The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Xiaoni Kong, ; Yanjun Shi, ; Hailong Wu,
| | - Xiaoni Kong
- Institute of Clinical Immunology, Department of Liver Diseases, Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
- *Correspondence: Xiaoni Kong, ; Yanjun Shi, ; Hailong Wu,
| |
Collapse
|
19
|
Antiangiogenic Strategies in Epithelial Ovarian Cancer: Mechanism, Resistance, and Combination Therapy. JOURNAL OF ONCOLOGY 2022; 2022:4880355. [PMID: 35466318 PMCID: PMC9019437 DOI: 10.1155/2022/4880355] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/23/2022] [Indexed: 12/24/2022]
Abstract
Angiogenesis is one of the hallmarks of cancer and plays a crucial role in carcinogenesis and progression of epithelial ovarian cancer. Antiangiogenic agent is the first approved targeted agent in ovarian cancer. Anti-angiogenic agents mainly include agents target VEGF/VEGFR pathway, such as bevacizumab and agents target receptor tyrosine kinase, and non-VEGF/VEGFR targets of angiogenesis. Antiangiogenic agents demonstrate certain effects in ovarian cancer treatment either as monotherapy or combined with chemotherapy. Unfortunately, antiangiogenic agents, such as bevacizumab, integrated into the ovarian cancer treatment paradigm do not increase cures. Thus, the benefits of anti-angiogenic agents must be carefully weighed against the cost and associated toxicities. Antiangiogenic agents drug resistance and short of predictive biomarkers are main obstacles in ovarian cancer treatment. A combination of poly (ADP-ribose) polymerase inhibitors or immune checkpoint inhibitors might be great strategies to overcome resistance as well as enhance anti-tumor activity of anti-angiogenic drugs. Predictive biomarkers of antiangiogenic agents are in urgent need.
Collapse
|
20
|
Markwell SM, Ross JL, Olson CL, Brat DJ. Necrotic reshaping of the glioma microenvironment drives disease progression. Acta Neuropathol 2022; 143:291-310. [PMID: 35039931 DOI: 10.1007/s00401-021-02401-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/13/2022]
Abstract
Glioblastoma is the most common primary brain tumor and has a dismal prognosis. The development of central necrosis represents a tipping point in the evolution of these tumors that foreshadows aggressive expansion, swiftly leading to mortality. The onset of necrosis, severe hypoxia and associated radial glioma expansion correlates with dramatic tumor microenvironment (TME) alterations that accelerate tumor growth. In the past, most have concluded that hypoxia and necrosis must arise due to "cancer outgrowing its blood supply" when rapid tumor growth outpaces metabolic supply, leading to diffusion-limited hypoxia. However, growing evidence suggests that microscopic intravascular thrombosis driven by the neoplastic overexpression of pro-coagulants attenuates glioma blood supply (perfusion-limited hypoxia), leading to TME restructuring that includes breakdown of the blood-brain barrier, immunosuppressive immune cell accumulation, microvascular hyperproliferation, glioma stem cell enrichment and tumor cell migration outward. Cumulatively, these adaptations result in rapid tumor expansion, resistance to therapeutic interventions and clinical progression. To inform future translational investigations, the complex interplay among environmental cues and myriad cell types that contribute to this aggressive phenotype requires better understanding. This review focuses on contributions from intratumoral thrombosis, the effects of hypoxia and necrosis, the adaptive and innate immune responses, and the current state of targeted therapeutic interventions.
Collapse
Affiliation(s)
- Steven M Markwell
- Department of Pathology, Northwestern Medicine Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave. Ward 3-140, Chicago, IL, USA
| | - James L Ross
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
| | - Cheryl L Olson
- Department of Pathology, Northwestern Medicine Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave. Ward 3-140, Chicago, IL, USA
| | - Daniel J Brat
- Department of Pathology, Northwestern Medicine Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave. Ward 3-140, Chicago, IL, USA.
| |
Collapse
|
21
|
Ayoub NM, Jaradat SK, Al-Shami KM, Alkhalifa AE. Targeting Angiogenesis in Breast Cancer: Current Evidence and Future Perspectives of Novel Anti-Angiogenic Approaches. Front Pharmacol 2022; 13:838133. [PMID: 35281942 PMCID: PMC8913593 DOI: 10.3389/fphar.2022.838133] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/03/2022] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis is a vital process for the growth and dissemination of solid cancers. Numerous molecular pathways are known to drive angiogenic switch in cancer cells promoting the growth of new blood vessels and increased incidence of distant metastasis. Several angiogenesis inhibitors are clinically available for the treatment of different types of advanced solid cancers. These inhibitors mostly belong to monoclonal antibodies or small-molecule tyrosine kinase inhibitors targeting the classical vascular endothelial growth factor (VEGF) and its receptors. Nevertheless, breast cancer is one example of solid tumors that had constantly failed to respond to angiogenesis inhibitors in terms of improved survival outcomes of patients. Accordingly, it is of paramount importance to assess the molecular mechanisms driving angiogenic signaling in breast cancer to explore suitable drug targets that can be further investigated in preclinical and clinical settings. This review summarizes the current evidence for the effect of clinically available anti-angiogenic drugs in breast cancer treatment. Further, major mechanisms associated with intrinsic or acquired resistance to anti-VEGF therapy are discussed. The review also describes evidence from preclinical and clinical studies on targeting novel non-VEGF angiogenic pathways in breast cancer and several approaches to the normalization of tumor vasculature by targeting pericytes, utilization of microRNAs and extracellular tumor-associate vesicles, using immunotherapeutic drugs, and nanotechnology.
Collapse
Affiliation(s)
- Nehad M. Ayoub
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology (JUST), Irbid, Jordan
- *Correspondence: Nehad M. Ayoub,
| | - Sara K. Jaradat
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology (JUST), Irbid, Jordan
| | - Kamal M. Al-Shami
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
| | - Amer E. Alkhalifa
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology (JUST), Irbid, Jordan
| |
Collapse
|
22
|
Jiang H, Zhao Y, Feng P, Liu Y. Sulfiredoxin-1 Inhibits PDGF-BB-Induced Vascular Smooth Muscle Cell Proliferation and Migration by Enhancing the Activation of Nrf2/ARE Signaling. Int Heart J 2022; 63:113-121. [PMID: 35034915 DOI: 10.1536/ihj.21-213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Sulfiredoxin1 (Srxn1), an endogenous antioxidant protein, is involved in cardiovascular diseases. In this study, we aimed to investigate the role of Srxn1 in VSMCs and its molecular mechanism. The murine vascular smooth muscle cells MOVAS were treated with different doses of platelet-derived growth factor-BB (PDGF-BB); then, Srxn1 expression was detected using reverse transcription-quantitative polymerase chain reaction and western blot analysis. MTT and wound healing assay were used to examine the effect of Srxn1 on MOVAS cell proliferation and migration. Reactive oxygen species (ROS) production, malondialdehyde (MDA) level, and superoxide dismutase (SOD) activity in MOVAS cells were detected using corresponding commercial kits. Moreover, the expression of proliferating cell nuclear antigen (PCNA), matrix metalloproteinase 2 (MMP-2), and nuclear factor erythroid-2-related factor 2 (Nrf2) /antioxidant response element (ARE) signaling-related proteins was detected using western blot analysis. In our study, PDGF-BB dose-dependently increased Srxn1 expression in MOVAS cells, and Srxn1 expression was increased with time dependence in PDGF-BB-treated MOVAS cells. The knockdown of Srxn1 increased PDGF-BB-induced the proliferation, migration, ROS production, MDA level, and the protein expression of PCNA and MMP-2, as well as decreased SOD activity and the expression of Nrf2/ARE signaling-related proteins in PDGF-BB-stimulated MOVAS cells. However, the overexpression of Srxn1 showed the opposite results to those of knockdown of Srxn1. Moreover, the inhibitory effects of Srxn1 overexpression on PDGF-BB induced proliferation, migration, ROS production, and MDA level and the promotion of Srxn1 overexpression on PDGF-BB induced SOD activity were partially reversed by the knockdown of Nrf2. Srxn1 inhibited PDGF-BB-induced proliferation, migration, and oxidative stress through activating Nrf2/ARE signaling.
Collapse
Affiliation(s)
- Haijie Jiang
- Department of Cardiology, The Second Affiliated Hospital of Dalian Medical University
| | - Yueyan Zhao
- Department of Cardiology, The Second Affiliated Hospital of Dalian Medical University
| | - Panyang Feng
- Department of Cardiology, The Second Affiliated Hospital of Dalian Medical University
| | - Yan Liu
- Department of Cardiology, The Second Affiliated Hospital of Dalian Medical University
| |
Collapse
|
23
|
Zhou J, Shao L, Yu J, Huang J, Feng Q. PDGF-BB promotes vascular smooth muscle cell migration by enhancing Pim-1 expression via inhibiting miR-214. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1728. [PMID: 35071422 PMCID: PMC8743727 DOI: 10.21037/atm-21-5638] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/16/2021] [Indexed: 12/13/2022]
Abstract
Background Several studies have indicated that the platelet-derived growth factor/platelet-derived growth factor receptor (PDGF/PDGFR) pathway is involved in the process of atherosclerosis. However, its underlying mechanism remains to be further elucidated. Serine/threonine-protein kinase pim-1 (Pim-1), a member of serine/threonine-specific kinases, is a pro-oncogene published to be related to cell proliferation, apoptosis, and metastasis of cancer cells. Whether Pim-1 is involved in PDGF/PDGFR pathway-mediated coronary atherosclerotic heart disease remains to be elucidated. Methods We established a cell model of PDGF-BB-stimulated smooth muscle cells using A7r5 cells. Transwell assay was used to detect the potential of cell migration and invasion. The targeted regulation of Pim-1 by miR-214 was confirmed by luciferase assay. Rescue experiments were performed to determine the role of the PDGF-BB/miR-214/Pim-1 axis on the cell migration of smooth muscle cells by including PDGF-BB treatment, and the overexpression of miR-214 and Pim-1. Quantitative polymerase chain reaction (qPCR) was used to examine the gene expression and western blot was performed to detect the protein expression. Results Our data indicated that PDGF-BB could effectively enhance smooth muscle cell migration. We also showed Pim-1 was a target of miR-214 in A7r5 cells. The expression of Pim-1 was shown to be upregulated by PDGF-BB via suppression of the expression of miR-214. Moreover, overexpression miR-214 inhibited PDGF-BB-stimulated Pim-1 expression and smooth muscle cell migration via modulating epithelial-mesenchymal transition (EMT), but no change on cell cycle. However, overexpression of Pim-1 reversed miR-214-blocked cell migration by promoting the activation of the STAT3, AKT, and ERK signaling pathways. Conclusions Our data suggested that the PDGF/miR-214/Pim-1 axis could be a potential target for coronary atherosclerotic heart disease.
Collapse
Affiliation(s)
- Jinshan Zhou
- Department of Cardiothoracic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Lifang Shao
- Department of Cardiothoracic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Jianghao Yu
- Department of Cardiothoracic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Junchao Huang
- Department of Cardiothoracic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Qiang Feng
- Department of Cardiothoracic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| |
Collapse
|
24
|
Wang Y, Wu H, Deng R. Angiogenesis as a potential treatment strategy for rheumatoid arthritis. Eur J Pharmacol 2021; 910:174500. [PMID: 34509462 DOI: 10.1016/j.ejphar.2021.174500] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 08/04/2021] [Accepted: 09/06/2021] [Indexed: 12/15/2022]
Abstract
Angiogenesis is an early and key event in the pathogenesis of rheumatoid arthritis (RA) and is crucial for the proliferation of synovial tissue and the formation of pannus. This process is regulated by both angiogenesis-stimulating factors and angiogenesis inhibitors, the basis for the "on-off hypothesis of angiogenesis." In RA, inflammation, immune imbalance, and hypoxia can further turn on the switch for blood vessel formation and induce angiogenesis. The new vasculature can recruit white blood cells, induce immune imbalance, and aggravate inflammation. At the same time, it also can provide oxygen and nutrients for the proliferating synovial tissue, which can accelerate the process of RA. The current therapies for RA mainly target the inflammatory response of autoimmune activation. Although these therapies have been greatly improved, there are still many patients whose RA is difficult to treat or who do not fully respond to treatment. Therefore, new innovative therapies are still urgently needed. This review covers the mechanism of synovial angiogenesis in RA, including the detailed process of angiogenesis and the relationship between inflammation, immune imbalance, hypoxia, and synovial angiogenesis, respectively. At the same time, in the context of the development of angiogenesis inhibition therapy for cancer, we also discuss similar treatment strategies for RA, especially the combination of targeted angiogenesis inhibition therapy and immunotherapy.
Collapse
Affiliation(s)
- Yan Wang
- College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei, 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230012, China
| | - Hong Wu
- College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei, 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230012, China.
| | - Ran Deng
- College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei, 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230012, China
| |
Collapse
|
25
|
Wang J, Fu MS, Zhou MW, Ke BL, Zhang ZH, Xu X. Potential effects of angiogenesis-related factors on the severity of APAC and surgical outcomes of trabeculectomy. BMC Ophthalmol 2021; 21:297. [PMID: 34384366 PMCID: PMC8359530 DOI: 10.1186/s12886-021-02051-w] [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] [Received: 12/09/2020] [Accepted: 07/26/2021] [Indexed: 11/10/2022] Open
Abstract
Background EPO (erythropoietin) and PDGF (platelet derived growth factor) families are thought to be associated with angiogenesis under hypoxic condition. The sharp rise of intraocular pressure in acute primary angle closure (APAC) results in an inefficient supply of oxygen and nutrients. We aimed to measure the expression of EPO and PDGF family members in APAC eyes and demonstrate their associations with APAC’s surgical success rate. Methods Concentrations of EPO, PDGF-AA, -BB, -CC and -DD collected in aqueous humor samples of 55 patients recruited were measured. Before operations, correlations between target proteins and IOP (intraocular pressure) were detected between APAC (acute primary angle closure) and cataract patients. Based on the post-operative follow-up, the effects of EPO and PDGF family members on the successful rate of trabeculectomy were tested. Results The levels of EPO, PDGF-CC and -DD were significantly elevated in the APAC group compared to the cataract group. During the post-operative follow-up, EPO, PDGF-CC and -DD showed significant differences between the success and failure groups. In multivariable linear regression analyses, failed filtration surgery was more likely in APAC eyes with higher EPO level. The Kaplan-Meier survival plot suggested that the success rate in eyes with low EPO level was significantly higher than that in eyes with high EPO level. Conclusion The levels of EPO, PDGF-CC and -DD were significantly elevated in failure group. EPO level correlated with preoperative IOP and numbers of eyedrops, and higher EPO level in aqueous humor is a risk factor for trabeculectomy failure. It can be a biomarker to estimate the severity of APAC and the success rate of surgery. The investigation of mechanism of EPO in APAC a may have potential clinical applications for the surgical treatment of APAC.
Collapse
Affiliation(s)
- Jing Wang
- National Clinical Research Center for Eye Diseases, Shanghai, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.,Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, China.,Eye & ENT Hospital, Fudan University, 83 Fenyang Rd, Shanghai, 20000, China
| | - Ming-Shui Fu
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.,Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, China.,Eye & ENT Hospital, Fudan University, 83 Fenyang Rd, Shanghai, 20000, China.,Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Min-Wen Zhou
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.,Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, China.,Eye & ENT Hospital, Fudan University, 83 Fenyang Rd, Shanghai, 20000, China.,Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Bi-Lian Ke
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.,Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, China.,Eye & ENT Hospital, Fudan University, 83 Fenyang Rd, Shanghai, 20000, China.,Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhi-Hua Zhang
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China. .,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China. .,Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, China. .,Eye & ENT Hospital, Fudan University, 83 Fenyang Rd, Shanghai, 20000, China. .,Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China.
| | - Xun Xu
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.,Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, China.,Eye & ENT Hospital, Fudan University, 83 Fenyang Rd, Shanghai, 20000, China.,Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
26
|
Chen YJ, Chang JT, You GR, Huang CY, Fan KH, Cheng AJ. Panel biomarkers associated with cancer invasion and prognostic prediction for head-neck cancer. Biomark Med 2021; 15:861-877. [PMID: 34032473 DOI: 10.2217/bmm-2021-0213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 04/28/2021] [Indexed: 11/21/2022] Open
Abstract
Aim: Cell invasion leading to metastasis is a major cause of treatment failure in head-neck cancers (HNCs). Identifying prognostic molecules associated with invasiveness is imperative for clinical applications. Materials & methods: A systemic approach was used to globally survey invasion-related genes, including transcriptomic profiling, pathway analysis, data mining and prognostic assessment using TCGA-HNSC dataset. Results: Six functional pathways and six hub molecules (LAMA3, LAMC2, THBS1, IGF1R, PDGFB and TGFβ1) were identified that significantly contributed to cell invasion, leading to poor survival in HNC patients. Combinations of multiple biomarkers substantially increased the probability of accurately predicting prognosis. Conclusion: Our six defined invasion-related molecules may be used as a panel signature in precision medicine for prognostic indicators or molecular therapeutic targets for HNC.
Collapse
Affiliation(s)
- Yin-Ju Chen
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei, 11031, Taiwan
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Joseph T Chang
- Department of Radiation Oncology, Chang Gung Memorial Hospital-Linkou, Taoyuan, 33333, Taiwan
- Department of Medical School, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
| | - Guo-Rung You
- Department of Medical Biotechnology & Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
| | - Chun-Yu Huang
- School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
| | - Kang-Hsing Fan
- Department of Radiation Oncology, Chang Gung Memorial Hospital-Linkou, Taoyuan, 33333, Taiwan
- Department of Radiation Oncology, New Taipei Municipal TuCheng Hospital, New Taipei City, 236017, Taiwan
| | - Ann-Joy Cheng
- Department of Radiation Oncology, Chang Gung Memorial Hospital-Linkou, Taoyuan, 33333, Taiwan
- Department of Medical Biotechnology & Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
| |
Collapse
|
27
|
Lu JF, Hu ZQ, Yang MX, Liu WY, Pan GF, Ding JB, Liu JZ, Tang L, Hu B, Li HC. Downregulation of PDGF-D Inhibits Proliferation and Invasion in Breast Cancer MDA-MB-231 Cells. Clin Breast Cancer 2021; 22:e173-e183. [PMID: 34272173 DOI: 10.1016/j.clbc.2021.06.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 05/06/2021] [Accepted: 06/04/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND The platelet derived growth factor-D (PDGF-D) plays an important role in breast tumor aggressiveness. However, limited study has investigated the effect of silencing PDGF-D on the biological function of breast cancer. The purpose of this study is to clarify the potential value of PDGF-D as a target for breast cancer treatment. METHODS Reverse transcription-polymerase chain reaction and western blot were used to detect PDGF-D expression in 5 different breast cancer cells. The lentiviral vector was usd to silence PDGF-D in MDA-MB-231 cells. Then, Methyl Thiazolyl Tetrazolium was used to detect cell viability, 5-Ethynyl-2'- deoxyuridine and a soft agar assay were used to detect cell proliferation and clonality. Additionally, cell apoptosis after PDGF-D knockdown was measured by Annexin V/ Prodium Iodide staining, and cell migration was detected by trans-well assay. Survival rate and tumor size were measured by nude mice transplantation. RESULTS The MDA-MB-231 and SK-BR-3 cell lines showed higher PDGF-D expression than the MCF7 cell lines (P<.05). After the PDGF-D gene was silenced, the growth and colony forming abilitys ignificantly decreased (P<.05) together with the induction of apoptosis in MDA-MB-231 cells (P<.05). Moreover, MDA-MB-231 cells with PDGF-D silencing showed significantly diminished aggressive migration and invasion potential compared to other cells (P<.05). In vivo experiments also indicated that PDGF-D silencing inhibited tumor growth and improved the survival rate of tumor-bearing mice. CONCLUSION Downregulation of PDGF-D had dramatic effects on breast cancer cell proliferation, apoptosis and migration, which indicates that it plays an important role in breast cancer development and progression.
Collapse
Affiliation(s)
- Jing-Feng Lu
- Department of General Surgery, Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201100, China
| | - Zhi-Qiu Hu
- Department of General Surgery, Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201100, China
| | - Meng-Xuan Yang
- Department of General Surgery, Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201100, China
| | - Wei-Yan Liu
- Department of General Surgery, Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201100, China
| | - Gao-Feng Pan
- Department of General Surgery, Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201100, China
| | - Jun-Bin Ding
- Department of General Surgery, Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201100, China
| | - Jia-Zhe Liu
- Department of General Surgery, Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201100, China
| | - Lang Tang
- Department of Ultrasonography Department, Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201100, China
| | - Bin Hu
- Department of Ultrasonography Department, Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201100, China.
| | - Hong-Chang Li
- Department of General Surgery, Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201100, China.
| |
Collapse
|
28
|
Xelwa N, Candy GP, Devar J, Omoshoro-Jones J, Smith M, Nweke EE. Targeting Growth Factor Signaling Pathways in Pancreatic Cancer: Towards Inhibiting Chemoresistance. Front Oncol 2021; 11:683788. [PMID: 34195085 PMCID: PMC8236623 DOI: 10.3389/fonc.2021.683788] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/24/2021] [Indexed: 12/15/2022] Open
Abstract
Pancreatic cancer is one of the most deadly cancers, ranking amongst the top leading cause of cancer related deaths in developed countries. Features such as dense stroma microenvironment, abnormal signaling pathways, and genetic heterogeneity of the tumors contribute to its chemoresistant characteristics. Amongst these features, growth factors have been observed to play crucial roles in cancer cell survival, progression, and chemoresistance. Here we review the role of the individual growth factors in pancreatic cancer chemoresistance. Importantly, the interplay between the tumor microenvironment and chemoresistance is explored in the context of pivotal role played by growth factors. We further describe current and future potential therapeutic targeting of these factors.
Collapse
|
29
|
Aberrant lncRNA Profiles Are Associated With Chronic Benzene Poisoning and Acute Myelocytic Leukemia. J Occup Environ Med 2021; 62:e308-e317. [PMID: 32730034 DOI: 10.1097/jom.0000000000001875] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
OBJECTIVE This study investigates the mechanisms of benzene hematotoxicity. METHODS We used microarray to detect expression profiles of long non-coding RNAs (lncRNAs) and mRNAs in peripheral lymphocytes from chronic benzene poisoning, acute myelocytic leukemia, and healthy controls. The lncRNAs and mRNAs were validated using real-time quantitative PCR (RT-qPCR). Cytokinesis-block micronucleus assay was used to analyze chromosomal aberration. RESULTS We found 173 upregulated and 258 downregulated lncRNAs, and 695 upregulated and 804 downregulated mRNAs. The lncRNA CUST_40243 and mRNA PDGFC and CDKN1A associated with chronic benzene poisoning. Relevant inflammatory response, hematopoietic cell lineage, and cell cycle may be important pathways for the sifted lncRNAs and mRNAs. Furthermore, micronuclei frequency was significantly higher in off-post chronic benzene poisoning patients. CONCLUSIONS Chromosomal aberration induced by benzene exposure is irreversible. The lncRNA CUST_40243 and mRNA PDGFC and CDKN1A are related to chronic benzene poisoning.
Collapse
|
30
|
Han N, Zhang YY, Zhang ZM, Zhang F, Zeng TY, Zhang YB, Zhao WC. High expression of PDGFA predicts poor prognosis of esophageal squamous cell carcinoma. Medicine (Baltimore) 2021; 100:e25932. [PMID: 34011067 PMCID: PMC8137088 DOI: 10.1097/md.0000000000025932] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 04/17/2021] [Indexed: 01/05/2023] Open
Abstract
Platelet-derived growth factor A (PDGFA), the most known member of PDGF family, plays a crucial role in occurrence and progression of different tumors. However, PDGFA expression and its clinical significance in esophageal squamous cell carcinoma (ESCC) are not clear. The present study aimed to assess the expression and prognostic value of PDGFA in ESCC.The Gene Expression Omnibus databases (GSE53625, GSE23400, and GSE67269) and fresh clinical samples were employed for detecting PDGFA messenger RNA expression in ESCC. The associations of PDGFA expression with clinicopathological characteristics were evaluated by chi-square test. Kaplan-Meier analysis and Cox proportional hazard regression model were performed to determine the prognostic value of PDGFA in ESCC patients. PDGFA-related signaling pathways were defined by gene set enrichment analysis based on Gene Expression Omnibus databases.The PDGFA messenger RNA expression was upregulated in ESCC tissues compared with paired adjacent noncancerous tissues (P < .05) and was positively correlated with T stage (P < .05). Kaplan-Meier survival analysis suggested that ESCC patients with high PDGFA expression were associated with poorer overall survival compared to those with low PDGFA expression (P < .05), especially in advanced T stage (P < .05). Cox analyses showed that high expression of PDGFA was an independent predictor for poor prognosis in ESCC patients. Gene set enrichment analysis identified 3 signaling pathways (extracellular matrix receptor interaction, focal adhesion, and glycosaminoglycan biosynthesis chondroitin sulfate) that were enriched in PDGFA high expression phenotype (all P < .01).PDGFA may serve as an oncogene in ESCC and represent an independent molecular biomarker for prognosis of ESCC patients.
Collapse
Affiliation(s)
- Na Han
- Department of Oncology, The Second Affiliated Hospital of Zhengzhou University
| | - Yan-Yan Zhang
- Department of Oncology, The Second Affiliated Hospital of Zhengzhou University
| | - Zhong-Mian Zhang
- Department of Oncology, The Second Affiliated Hospital of Zhengzhou University
| | - Fang Zhang
- Department of Oncology, The Second Affiliated Hospital of Zhengzhou University
| | | | | | - Wen-Chao Zhao
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, PR China
| |
Collapse
|
31
|
Wang J, You J, Gong D, Xu Y, Yang B, Jiang C. PDGF-BB induces conversion, proliferation, migration, and collagen synthesis of oral mucosal fibroblasts through PDGFR-β/PI3K/ AKT signaling pathway. Cancer Biomark 2021; 30:407-415. [PMID: 33492283 DOI: 10.3233/cbm-201681] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To explore the pathogenesis of oral submucosal fibrosis (OSF) by analyzing the impact of Platelet Derived Growth Factor (PDGF)-BB on oral mucosal fibroblasts (FB) and PDGFR-β/Phosphoinositide 3-kinase (PI3K)/serine/threonine protein kinase (AKT) signaling pathway. METHODS The isolated and purified oral mucosal fibroblasts were divided into four groups: the control group (CON, 10% FBS DMEM), the PDGF-BB group (40 ng/ml PDGF-BB), the PDGF-BB+IMA group (40 ng/ml PDGF-BB and 60 μmol/L IMA), and the PDGF-BB+LY294002 group (40 ng/ml PDGF-BB and 48 μmol/L LY294002). Primary human FB cells were isolated and cultured for detecting the effects of PDGF-BB on α-smooth muscle actin (α-SMA) by indirect immunofluorescence. 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide, Thiazolyl Blue Tetrazolium Bromide (MTT) method and scratch test were used to detect the proliferation and migration of FB. Western blots were used to detect the synthesis of type I collagen (Col I) and the expression of PDGFR-β/PI3K/AKT signaling pathway-related proteins. The effects of PDGFR-β inhibitor and PI3K inhibitor were observed. RESULTS Compared with group CON, group IMA, and group LY294002, α-SMA was upregulated in group PDGF-BB (p< 0.05), with higher OD490 nm value (p< 0.05), narrower average scratch width, and higher relative cell migration rate (p< 0.05). The expression levels of Col I, p-PDGFR-β, p-PI3K, and p-AKT were higher in group PDGF-BB (p< 0.05). CONCLUSIONS PDGF-BB induces FB to transform into myofibroblasts (MFB) through the PDGFR-β/PI3K/AKT signaling pathway, and promotes the proliferation, migration, and collagen synthesis.
Collapse
Affiliation(s)
- Jie Wang
- Department of Oral and Maxillofacial Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Immunology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jialing You
- Department of Immunology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Ding Gong
- Department of Immunology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Ying Xu
- Department of Immunology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Bo Yang
- Department of Immunology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Canhua Jiang
- Department of Oral and Maxillofacial Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
32
|
Neuropilin-2 and Its Transcript Variants Correlate with Clinical Outcome in Bladder Cancer. Genes (Basel) 2021; 12:genes12040550. [PMID: 33918816 PMCID: PMC8070368 DOI: 10.3390/genes12040550] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/26/2021] [Accepted: 04/07/2021] [Indexed: 12/24/2022] Open
Abstract
Urothelial bladder cancer ranks among the 10 most frequently diagnosed cancers worldwide. In our previous study, the transmembrane protein neuropilin-2 (NRP2) emerged as a predictive marker in patients with bladder cancer. NRP2 consists of several splice variants; the most abundant of these, NRP2a and NRP2b, are reported to have different biological functions in lung cancer progression. For other cancer types, there are no published data on the role of these transcript variants in cancer progression and the clinical outcome. Here, we correlate NRP2 and its two most abundant transcript variants, NRP2A and NRP2B, with the clinical outcome using available genomic data with subsequent validation in our own cohort of patients with muscle-invasive bladder cancer. In addition to NRP2, NRP1 and the NRP ligands PDGFC and PDGFD were studied. Only NRP2A emerged as an independent prognostic marker for shorter cancer-specific survival in muscle-invasive bladder cancer in our cohort of 102 patients who underwent radical cystectomy between 2008 and 2014 with a median follow-up time of 82 months. Additionally, we demonstrate that high messenger expression of NRP2, NRP1, PDGFC and PDGFD associates with a more aggressive disease (i.e., a high T stage, positive lymph node status and reduced survival).
Collapse
|
33
|
Li S, Zhao X, Cheng S, Li J, Bai X, Meng X. Downregulating long non-coding RNA PVT1 expression inhibited the viability, migration and phenotypic switch of PDGF-BB-treated human aortic smooth muscle cells via targeting miR-27b-3p. Hum Cell 2020; 34:335-348. [PMID: 33106979 DOI: 10.1007/s13577-020-00452-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 10/14/2020] [Indexed: 11/30/2022]
Abstract
Long non-coding RNA Plasmacytoma Variant Translocation 1 (LncRNA PVT1) was involved in various human diseases, but its role in aortic dissection (AD) remained to be fully examined. In this study, the viability and migration of human aortic smooth muscle cells (HASMCs) were respectively measured by MTT assay and wound-healing assay. Relative phenotypic switch-related protein expressions were measured with quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot as needed. An AD model was established in animals and hematoxylin-eosin (H&E) staining was used for pathological examination. We found that, in HASMCs, microRNA (miR)-27b-3p could competitively bind with PVT1. In AD, PVT1 expression was upregulated, yet that of miR-27b-3p was downregulated. Downregulating PVT1 reversed the effects of growth factor-BB (PDGF-BB) treatment on PVT1, miR-27b-3p and expressions of phenotypic switch-related markers, and cell viability and migration, while downregulating miR-27b-3p reversed the effects of downregulating PVT1. Moreover, downregulating PVT1 suppressed the effects of upregulated PVT1 and downregulated miR-27b-3p induced by AD as well as media degeneration in vivo. In conclusion, downregulating PVT1 expression suppressed the proliferation, migration and phenotypic switch of HASMCs treated by PDGF-BB via targeting miR-27b-3p.
Collapse
Affiliation(s)
- Shouming Li
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, No.107, West Wenhua Road, Jinan, 250012, Shandong, China
| | - Xin Zhao
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, No.107, West Wenhua Road, Jinan, 250012, Shandong, China.
| | - Shaopeng Cheng
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, No.107, West Wenhua Road, Jinan, 250012, Shandong, China
| | - Jialiang Li
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, No.107, West Wenhua Road, Jinan, 250012, Shandong, China
| | - Xiao Bai
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, No.107, West Wenhua Road, Jinan, 250012, Shandong, China
| | - Xiangbin Meng
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, No.107, West Wenhua Road, Jinan, 250012, Shandong, China
| |
Collapse
|
34
|
Iacobas DA. Powerful quantifiers for cancer transcriptomics. World J Clin Oncol 2020; 11:679-704. [PMID: 33033692 PMCID: PMC7522543 DOI: 10.5306/wjco.v11.i9.679] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 06/06/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023] Open
Abstract
Every day, investigators find a new link between a form of cancer and a particular alteration in the sequence or/and expression level of a key gene, awarding this gene the title of “biomarker”. The clinician may choose from numerous available panels to assess the type of cancer based on the mutation or expression regulation (“transcriptomic signature”) of “driver” genes. However, cancer is not a “one-gene show” and, together with the alleged biomarker, hundreds other genes are found as mutated or/and regulated in cancer samples. Regardless of the platform, a well-designed transcriptomic study produces three independent features for each gene: Average expression level, expression variability and coordination with expression of each other gene. While the average expression level is used in all studies to identify what genes were up-/down-regulated or turn on/off, the other two features are unfairly ignored. We use all three features to quantify the transcriptomic change during the progression of the disease and recovery in response to a treatment. Data from our published microarray experiments on cancer nodules and surrounding normal tissue from surgically removed tumors prove that the transcriptomic topologies are not only different in histopathologically distinct regions of a tumor but also dynamic and unique for each human being. We show also that the most influential genes in cancer nodules [the Gene Master Regulators (GMRs)] are significantly less influential in the normal tissue. As such, “smart” manipulation of the cancer GMRs expression may selectively kill cancer cells with little consequences on the normal ones. Therefore, we strongly recommend a really personalized approach of cancer medicine and present the experimental procedure and the mathematical algorithm to identify the most legitimate targets (GMRs) for gene therapy.
Collapse
Affiliation(s)
- Dumitru Andrei Iacobas
- Personalized Genomics Laboratory, CRI Center for Computational Systems Biology, Roy G Perry College of Engineering, Prairie View A&M University, Prairie View, TX 77446, United States
| |
Collapse
|
35
|
Zoledronic Acid Abrogates Restraint Stress-Induced Macrophage Infiltration, PDGF-AA Expression, and Ovarian Cancer Growth. Cancers (Basel) 2020; 12:cancers12092671. [PMID: 32962103 PMCID: PMC7563308 DOI: 10.3390/cancers12092671] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 09/16/2020] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Biobehavioral disorders can negatively impact patients with ovarian cancer. Growing evidence suggests that chronic stress can promote tumor progression, the release of inflammatory mediators, and macrophage infiltration into the tumor. However, the role of stress hormones in regulating cancer cell/macrophage crosstalk remains unclear. This study aimed to assess the role of stress hormone-stimulated macrophages in modulating inflammatory networks and ovarian cancer biology. Our data show that stress hormones induced secretion of inflammatory proteins in ovarian cancer cell/macrophage co-cultures. Furthermore, we show that restraint stress leads to cancer growth, macrophage infiltration, and PDGF-AA protein expression in animal models of ovarian cancer. Conversely, zoledronic acid was able to prevent the effects of restraint stress on ovarian cancer growth. Overall, our data suggest a role for stress hormone-stimulated macrophages in ovarian cancer progression and suggest the involvement of PDGF-AA as a key mediator of this process. Abstract Multiple studies suggest that chronic stress accelerates the growth of existing tumors by activating the sympathetic nervous system. Data suggest that sustained adrenergic signaling can induce tumor growth, secretion of pro-inflammatory cytokines, and macrophage infiltration. Our goal was to study the role of adrenergic-stimulated macrophages in ovarian cancer biology. Cytokine arrays were used to assess the effect of adrenergic stimulation in pro-tumoral cytokine networks. An orthotopic model of ovarian cancer was used to assess the in vivo effect of daily restraint stress on tumor growth and adrenergic-induced macrophages. Cytokine analyses showed that adrenergic stimulation modulated pro-inflammatory cytokine secretion in a SKOV3ip1 ovarian cancer cell/U937 macrophage co-culture system. Among these, platelet-derived growth factor AA (PDGF-AA), epithelial cell-derived neutrophil-activating peptide (ENA-78), Angiogenin, vascular endothelial growth factor (VEGF), granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-5 (IL-5), Lipocalin-2, macrophage migration inhibitory factor (MIF), and transferrin receptor (TfR) were upregulated. Enriched biological processes included cytokine-mediated signaling pathways and positive regulation of cell proliferation. In addition, daily restraint stress increased ovarian cancer growth, infiltration of CD68+ macrophages, and expression of PDGF-AA in orthotopic models of ovarian cancer (SKOV3ip1 and HeyT30), while zoledronic acid, a macrophage-depleting agent, abrogated this effect. Furthermore, in ovarian cancer patients, high PDGFA expression correlated with worse outcomes. Here, it is shown that the adrenergic regulation of macrophages and PDGFA might play a role in ovarian cancer progression.
Collapse
|
36
|
Ming J, Cronin SJF, Penninger JM. Targeting the RANKL/RANK/OPG Axis for Cancer Therapy. Front Oncol 2020; 10:1283. [PMID: 32850393 PMCID: PMC7426519 DOI: 10.3389/fonc.2020.01283] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022] Open
Abstract
RANKL and RANK are expressed in different cell types and tissues throughout the body. They were originally described for their essential roles in bone remodeling and the immune system but have subsequently been shown to provide essential signals from regulating mammary gland homeostasis during pregnancy to modulating tumorigenesis. The success of RANKL/RANK research serves as a paragon for translational research from the laboratory to the bedside. The case in point has been the development of Denosumab, a RANKL-blocking monoclonal antibody which has already helped millions of patients suffering from post-menopausal osteoporosis and skeletal related events in cancer. Here we will provide an overview of the pathway from its origins to its clinical relevance in disease, with a special focus on emerging evidence demonstrating the therapeutic value of targeting the RANKL/RANK/OPG axis not only in breast cancer but also as an addition to the cancer immunotherapy arsenal.
Collapse
Affiliation(s)
- Jie Ming
- Department of Breast and Thyroid Surgery, Wuhan Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Shane J F Cronin
- Institute of Molecular Biotechnology of the Austrian Academy of Science, Vienna Biocenter, Vienna, Austria
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Science, Vienna Biocenter, Vienna, Austria.,Department of Medical Genetics, Life Science Institute, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
37
|
Na L, Bai Y, Sun Y, Wang Z, Wang W, Yuan L, Zhao C. Identification of 9-Core Immune-Related Genes in Bladder Urothelial Carcinoma Prognosis. Front Oncol 2020; 10:1142. [PMID: 32733809 PMCID: PMC7360854 DOI: 10.3389/fonc.2020.01142] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 06/05/2020] [Indexed: 12/15/2022] Open
Abstract
Background: Immune microenvironment within tumors affects initiation, progression and clinical outcome of human cancers. Here we explored an immune-related gene signature associated with prognosis of patients with bladder urothelial carcinoma. Method: The Cancer Genome Atlas (TCGA) database was interrogated for expressions of immune-related genes in bladder urothelial carcinomas. Integrated bioinformatics analyses were performed to identify prognostic factors. Results: Twenty-seven immune-related genes were revealed significantly associated with patient's overall survival (OS) by univariate Cox proportional hazards regression analysis. Nine-core immune-related genes including MMP9, PDGFRA, AHNAK, OLR1, RAC3, IGF1, PGF, OAS1, and SH3BP2 were selected to construct a risk score model by multivariate Cox proportional hazards regression analysis. Bioinformatics analyses further validated that risk score could be used as an important independent factor in evaluating prognosis. Conclusion: We established a prognostic immune signature for patients with bladder urothelial carcinoma, which may provide novel targets for prediction and therapy of these patients.
Collapse
Affiliation(s)
- Lei Na
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China.,Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu Bai
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China.,Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu Sun
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Zhuo Wang
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Wei Wang
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Lin Yuan
- Liaoning Branch of China Telecom, Shenyang, China
| | - Chenghai Zhao
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| |
Collapse
|
38
|
Saini H, Rahmani Eliato K, Veldhuizen J, Zare A, Allam M, Silva C, Kratz A, Truong D, Mouneimne G, LaBaer J, Ros R, Nikkhah M. The role of tumor-stroma interactions on desmoplasia and tumorigenicity within a microengineered 3D platform. Biomaterials 2020; 247:119975. [PMID: 32278213 DOI: 10.1016/j.biomaterials.2020.119975] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 03/11/2020] [Accepted: 03/14/2020] [Indexed: 12/14/2022]
Abstract
The tumor microenvironment has been demonstrated to play a crucial role in modulating cancer progression. Amongst various cell types within the tumor microenvironment, cancer associated fibroblasts (CAFs) are in abundance, serving to modulate the biophysical properties of the stromal matrix, through excessive deposition of extracellular matrix (ECM) proteins that leads to enhanced tumor progression. There is still a critical need to develop a fundamental framework on the role of tumor-stromal cell interactions on desmoplasia and tumorigenicity. Herein, we developed a 3D microengineered organotypic tumor-stroma model incorporated with breast cancer cells surrounded by CAF-embedded collagen matrix. We further integrated our platform with atomic force microscopy (AFM) to study the dynamic changes in stromal stiffness during active tumor invasion. Our findings primarily demonstrated enhanced tumor progression in the presence of CAFs. Furthermore, we highlighted the crucial role of crosstalk between tumor cells and CAFs on stromal desmoplasia, where we identified the role of tumor-secreted PDGF-AA/-BB on elevated matrix stiffness. Inhibition of the activity of PDGFRs in CAFs led to attenuation of stromal stiffness. Overall, our work presents a well-controlled tumor microenvironment model capable of dissecting specific biophysical and biochemical signaling cues which lead to stromal desmoplasia and tumor progression.
Collapse
|
39
|
Gao X, Yang J, Chen Y. Identification of a four immune-related genes signature based on an immunogenomic landscape analysis of clear cell renal cell carcinoma. J Cell Physiol 2020; 235:9834-9850. [PMID: 32452055 DOI: 10.1002/jcp.29796] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 05/06/2020] [Indexed: 12/13/2022]
Abstract
Renal clear cell carcinoma (ccRCC) is the most common type of renal cell carcinoma, which has strong immunogenicity. A comprehensive study of the role of immune-related genes (IRGs) in ccRCC is of great significance in finding ccRCC treatment targets and improving patient prognosis. In this study, we comprehensively analyzed the expression of IRGs in ccRCC based on The Cancer Genome Atlas datasets. The mechanism of differentially expressed IRGs in ccRCC was analyzed by bioinformatics. In addition, Cox regression analysis was used to screen prognostic related IRGs from differentially expressed IRGs. We also identified a four IRGs signature consisting of four IRGs (CXCL2, SEMA3G, PDGFD, and UCN) through lasso regression and multivariate Cox regression analysis. Further analysis results showed that the four IRGs signature could effectively predict the prognosis of patients with ccRCC, and its predictive power is independent of other clinical factors. In addition, the correlation analysis of immune cell infiltration showed that this four IRGs signature could effectively reflect the level of immune cell infiltration of ccRCC. We also found that the expression of immune checkpoint genes CTLA-4, LAG3, and PD-1 in the high-risk group was higher than that in the low-risk group. Our research revealed the role of IRGs in ccRCC, and developed a four IRGs signature that could be used to evaluate the prognosis of patients with ccRCC, which will help to develop personalized treatment strategies for patients with ccRCC and improve their prognosis. In addition, these four IRGs may be effective therapeutic targets for ccRCC.
Collapse
Affiliation(s)
- Xin Gao
- Clinical Laboratory, The First People's Hospital of Huaihua, Huaihua, Hunan, China
| | - Jinlian Yang
- Clinical Laboratory, The First People's Hospital of Huaihua, Huaihua, Hunan, China
| | - Yinyi Chen
- Clinical Laboratory, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| |
Collapse
|
40
|
Dai S, Zhang J, Xu Z. Silencing CCL8 inhibited the proliferation and migration of PDGF-BB-stimulated human aortic smooth muscle cells. Biosci Biotechnol Biochem 2020; 84:1585-1593. [PMID: 32432500 DOI: 10.1080/09168451.2020.1762160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
C-C motif Chemokine ligand 8 (CCL8) has been found in diseases' pathogenesis. But its molecular mechanism in atherosclerosis (AS) remains to be elucidated. Human aortic smooth muscle cells (HASMCs) were stimulated by PDGF-BB to establish cell model. α-SMA in PDGF-BB-stimulated HASMCs was measured by immunofluorescence staining. Relative gene expressions in PDGF-BB-stimulated HASMCs were detected by quantitative real-time polymerase chain reaction and western blot. HASMCs proliferation, migration, and cell cycle were assessed by cell counting kit-8, wound-healing assay, and flow cytometry. HASMCs viability was increased after PDGF-BB stimulation, with α-SMA downregulation yet CCL8 upregulation. Silencing CCL8 inhibited PDGF-BB-stimulated HASMCs proliferation and migration, and increased cells percentage in G1 phases but decreased those in S phase. Also, silencing CCL8 decreased OPN and cyclinD1 expressions and AKT and ERK1/2 phosphorylation while increased those of α-SMA and Sm22α. However, upregulating CCL8 led to opposite effects, suggesting CCL8 could be an atherosclerosis therapeutic target.
Collapse
Affiliation(s)
- Shipeng Dai
- Department of Cardiology II, Cangzhou Teaching Hospital of Tianjin Medical University , Tianjin, China
| | - Jiangang Zhang
- Department of Cardiology II, Cangzhou Teaching Hospital of Tianjin Medical University , Tianjin, China
| | - Zesheng Xu
- Department of Cardiology II, Cangzhou Teaching Hospital of Tianjin Medical University , Tianjin, China
| |
Collapse
|
41
|
Thies KA, Hammer AM, Hildreth BE, Steck SA, Spehar JM, Kladney RD, Geisler JA, Das M, Russell LO, Bey JF, Bolyard CM, Pilarski R, Trimboli AJ, Cuitiño MC, Koivisto CS, Stover DG, Schoenfield L, Otero J, Godbout JP, Chakravarti A, Ringel MD, Ramaswamy B, Li Z, Kaur B, Leone G, Ostrowski MC, Sizemore ST, Sizemore GM. Stromal Platelet-Derived Growth Factor Receptor-β Signaling Promotes Breast Cancer Metastasis in the Brain. Cancer Res 2020; 81:606-618. [PMID: 32327406 DOI: 10.1158/0008-5472.can-19-3731] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/25/2020] [Accepted: 04/20/2020] [Indexed: 11/16/2022]
Abstract
Platelet-derived growth factor receptor-beta (PDGFRβ) is a receptor tyrosine kinase found in cells of mesenchymal origin such as fibroblasts and pericytes. Activation of this receptor is dependent on paracrine ligand induction, and its preferred ligand PDGFB is released by neighboring epithelial and endothelial cells. While expression of both PDGFRβ and PDGFB has been noted in patient breast tumors for decades, how PDGFB-to-PDGFRβ tumor-stroma signaling mediates breast cancer initiation, progression, and metastasis remains unclear. Here we demonstrate this paracrine signaling pathway that mediates both primary tumor growth and metastasis, specifically, metastasis to the brain. Elevated levels of PDGFB accelerated orthotopic tumor growth and intracranial growth of mammary tumor cells, while mesenchymal-specific expression of an activating mutant PDGFRβ (PDGFRβD849V) exerted proproliferative signals on adjacent mammary tumor cells. Stromal expression of PDGFRβD849V also promoted brain metastases of mammary tumor cells expressing high PDGFB when injected intravenously. In the brain, expression of PDGFRβD849V was observed within a subset of astrocytes, and aged mice expressing PDGFRβD849V exhibited reactive gliosis. Importantly, the PDGFR-specific inhibitor crenolanib significantly reduced intracranial growth of mammary tumor cells. In a tissue microarray comprised of 363 primary human breast tumors, high PDGFB protein expression was prognostic for brain metastases, but not metastases to other sites. Our results advocate the use of mice expressing PDGFRβD849V in their stromal cells as a preclinical model of breast cancer-associated brain metastases and support continued investigation into the clinical prognostic and therapeutic use of PDGFB-to-PDGFRβ signaling in women with breast cancer. SIGNIFICANCE: These studies reveal a previously unknown role for PDGFB-to-PDGFRβ paracrine signaling in the promotion of breast cancer brain metastases and support the prognostic and therapeutic clinical utility of this pathway for patients.See related article by Wyss and colleagues, p. 594.
Collapse
Affiliation(s)
- Katie A Thies
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Radiation Oncology, The Ohio State University, Columbus, Ohio
| | - Anisha M Hammer
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Blake E Hildreth
- O'Neal Comprehensive Cancer Center, University of Alabama-Birmingham, Birmingham, Alabama.,Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama-Birmingham, Birmingham, Alabama
| | - Sarah A Steck
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Radiation Oncology, The Ohio State University, Columbus, Ohio
| | - Jonathan M Spehar
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Radiation Oncology, The Ohio State University, Columbus, Ohio
| | - Raleigh D Kladney
- Department of Medicine, Molecular Oncology Division, Washington University School of Medicine, St. Louis, Missouri
| | - Jennifer A Geisler
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Radiation Oncology, The Ohio State University, Columbus, Ohio
| | - Manjusri Das
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Radiation Oncology, The Ohio State University, Columbus, Ohio
| | - Luke O Russell
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Neurological Surgery, The Ohio State University, Columbus, Ohio
| | - Jerome F Bey
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Chelsea M Bolyard
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Neurological Surgery, The Ohio State University, Columbus, Ohio
| | - Robert Pilarski
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Division of Human Genetics, The Ohio State University, Columbus, Ohio
| | - Anthony J Trimboli
- The Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina.,Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Maria C Cuitiño
- The Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina.,Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Christopher S Koivisto
- The Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina.,Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Daniel G Stover
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Lynn Schoenfield
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Pathology, The Ohio State University, Columbus, Ohio
| | - Jose Otero
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Pathology, The Ohio State University, Columbus, Ohio
| | - Jonathan P Godbout
- Department of Neuroscience, The Ohio State University, Columbus, Ohio.,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio
| | - Arnab Chakravarti
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Radiation Oncology, The Ohio State University, Columbus, Ohio
| | - Matthew D Ringel
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Bhuvaneswari Ramaswamy
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Zaibo Li
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Pathology, The Ohio State University, Columbus, Ohio
| | - Balveen Kaur
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Gustavo Leone
- The Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina.,Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Michael C Ostrowski
- The Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina.,Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Steven T Sizemore
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Radiation Oncology, The Ohio State University, Columbus, Ohio
| | - Gina M Sizemore
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio. .,Department of Radiation Oncology, The Ohio State University, Columbus, Ohio
| |
Collapse
|
42
|
Jørgensen CLT, Forsare C, Bendahl PO, Falck AK, Fernö M, Lövgren K, Aaltonen K, Rydén L. Expression of epithelial-mesenchymal transition-related markers and phenotypes during breast cancer progression. Breast Cancer Res Treat 2020; 181:369-381. [PMID: 32300922 PMCID: PMC7188722 DOI: 10.1007/s10549-020-05627-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 04/03/2020] [Indexed: 12/12/2022]
Abstract
Purpose The study aimed to investigate expression of epithelial-to-mesenchymal transition (EMT)-related proteins and phenotypes during breast cancer progression and to relate this to patient outcome. Methods Protein expression patterns of E-cadherin, N-cadherin, twist, and vimentin were examined by immunohistochemistry on formalin-fixed paraffin-embedded samples from primary tumors (PTs) (n = 419), synchronous lymph node metastases (LNMs) (n = 131) and recurrences (n = 34) from patients included in an observational prospective primary breast cancer study. Markers were evaluated individually and combined as defined EMT phenotypes (epithelial, mesenchymal, partial EMT, and negative). EMT profiles were compared between matched tumor progression stages, and related to clinicopathological data and distant recurrence-free interval (DRFi). Results N-cadherin-positivity, vimentin-positivity, mesenchymal and partial EMT phenotypes were associated with more aggressive tumor characteristics such as triple-negative subtype. Single EMT markers and phenotype discordance rates between paired tumor samples were observed in the range of 2–35%. Non-epithelial phenotypes were more frequently identified in recurrences compared to PTs, however, no skewness of expression or phenotype was detected between PTs and matched LNMs or between PTs and matched recurrences (Exact McNemar test). Interestingly, patients with a twist positive PT had shorter DRFi, compared to patients with a twist negative PT (hazard ratio (HR) 2.4, 95% confidence interval (CI) 1.2–5.1, P = 0.02). Essentially, the same effect was seen in multivariable analysis (HR 2.5, 95% CI 0.97–6.6, P = 0.06). Conclusion The epithelial phenotype was indicated to be lost between PTs and recurrences as a reflection of tumor progression. Twist status of the PT was related to long-term prognosis warranting further investigation in larger cohorts. Electronic supplementary material The online version of this article (10.1007/s10549-020-05627-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Charlotte Levin Tykjær Jørgensen
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Medicon Village, Building 404, 22381, Lund, Sweden.
| | - Carina Forsare
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Medicon Village, Building 404, 22381, Lund, Sweden
| | - Pär-Ola Bendahl
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Medicon Village, Building 404, 22381, Lund, Sweden
| | - Anna-Karin Falck
- Department of Surgery, Helsingborg Hospital, Helsingborg, Sweden
| | - Mårten Fernö
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Medicon Village, Building 404, 22381, Lund, Sweden
| | - Kristina Lövgren
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Medicon Village, Building 404, 22381, Lund, Sweden
| | - Kristina Aaltonen
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Lisa Rydén
- Division of Surgery, Department of Clinical Sciences, Lund, Lund University, Lund, Sweden.,Department of Surgery, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
43
|
Li L, Wang XL, Lei Q, Sun CZ, Xi Y, Chen R, He YW. Comprehensive immunogenomic landscape analysis of prognosis-related genes in head and neck cancer. Sci Rep 2020; 10:6395. [PMID: 32286381 PMCID: PMC7156482 DOI: 10.1038/s41598-020-63148-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 03/26/2020] [Indexed: 02/06/2023] Open
Abstract
Head and neck cancer is the sixth most common malignancy around the world, and 90% of cases are squamous cell carcinomas. In this study, we performed a systematic investigation of the immunogenomic landscape to identify prognostic biomarkers for head and neck squamous cell carcinoma (HNSCC). We analyzed the expression profiles of immune-related genes (IRGs) and clinical characteristics by interrogating RNA-seq data from 527 HNSCC patients in the cancer genome atlas (TCGA) dataset, including 41 HPV+ and 486 HPV- samples. We found that differentially expressed immune genes were closely associated with patient prognosis in HNSCC by comparing the differences in gene expression between cancer and normal samples and performing survival analysis. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed to annotate the biological functions of the differentially expressed immunogenomic prognosis-related genes. Two additional cohorts from the Oncomine database were used for validation. 65, 56 differentially expressed IRGs was associated with clinical prognosis in total and HPV- samples, respectively. Furthermore, we extracted 10, 11 prognosis-related IRGs from 65, 56 differentially expressed IRGs, respectively. They were significantly correlated with clinical prognosis and used to construct the prognosis prediction models. The multivariable ROC curves (specifically, the AUC) were used to measure the accuracy of the prognostic models. These genes were mainly enriched in several gene ontology (GO) terms related to immunocyte migration and receptor and ligand activity. KEGG pathway analysis revealed enrichment of pathways related to cytokine-cytokine receptor interactions, which are primarily involved in biological processes. In addition, we identified 63 differentially expressed transcription factors (TFs) from 4784 differentially expressed genes, and 16 edges involving 18 nodes were formed in the regulatory network between differentially expressed TFs and the high-risk survival-associated IRGs. B cell and CD4 T cell infiltration levels were significantly negatively correlated with the expression of prognosis-related immune genes regardless of HPV status. In conclusion, this comprehensive analysis identified the prognostic IRGs as potential biomarkers, and the model generated in this study may enable an accurate prediction of survival.
Collapse
Affiliation(s)
- Lei Li
- Department of Head and Neck Surgery Section II, the Third Affiliated Hospital of Kunming Medical University, 519 Kunzhou Road, Kunming, China
| | - Xiao-Li Wang
- Radiation Therapy Center, the Third Affiliated Hospital of Kunming Medical University, 519 Kunzhou Road, Kunming, China
| | - Qian Lei
- Department of Head and Neck Surgery Section II, the Third Affiliated Hospital of Kunming Medical University, 519 Kunzhou Road, Kunming, China
| | - Chuan-Zheng Sun
- Department of Head and Neck Surgery Section II, the Third Affiliated Hospital of Kunming Medical University, 519 Kunzhou Road, Kunming, China
| | - Yan Xi
- Department of Head and Neck Surgery Section II, the Third Affiliated Hospital of Kunming Medical University, 519 Kunzhou Road, Kunming, China
| | - Ran Chen
- Department of Head and Neck Surgery Section II, the Third Affiliated Hospital of Kunming Medical University, 519 Kunzhou Road, Kunming, China
| | - Yong-Wen He
- Department of Dental Research, The Affiliated Stomatological Hospital of Kunming Medical University, Yunnan, China.
| |
Collapse
|
44
|
Knockdown of LINC00467 contributed to Axitinib sensitivity in hepatocellular carcinoma through miR-509-3p/PDGFRA axis. Gene Ther 2020; 28:634-645. [DOI: 10.1038/s41434-020-0137-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 02/15/2020] [Accepted: 02/20/2020] [Indexed: 12/18/2022]
|
45
|
Jin J, Wang L, Tao Z, Zhang J, Lv F, Cao J, Hu X. PDGFD induces ibrutinib resistance of diffuse large B‑cell lymphoma through activation of EGFR. Mol Med Rep 2020; 21:2209-2219. [PMID: 32186759 PMCID: PMC7115192 DOI: 10.3892/mmr.2020.11022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 01/17/2020] [Indexed: 12/20/2022] Open
Abstract
Ibrutinib, an FDA approved, orally administered BTK inhibitor, has demonstrated high response rates to diffuse large B-cell lymphoma (DLBCL), however, complete responses are infrequent and acquired resistance to BTK inhibition can emerge. The present study investigated the role of the platelet-derived growth factor D (PDGFD) gene and the ibrutinib resistance of DLBCL in relation to epidermal growth factor receptor (EGFR). Bioinformatics was used to screen and analyze differentially expressed genes (DEGs) in complete response (CR), partial response (PR) and stable disease (SD) in DLBCL treatment with ibrutinib, and Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed to analyze enriched the signaling pathways increasing DEGs. The Search Tool for Interactions of Chemicals database was used to analyze the target genes of ibrutinib. An interaction network of DEGs, disease-related genes and ibrutinib was constructed. The expression of PDGFD in tissues that were resistant or susceptible to DLBCL/ibrutinib was detected via immunohistochemistry (IHC), and the expression of PDGFD in DLBCL/ibrutinib-resistant strains and their parental counterparts were examined via reverse transcription-quantitative PCR and western blot analyses. Subsequently, a drug-resistant cell model of DLBCL/ibrutinib in which PDGFD was silenced was constructed. The apoptosis of the DLBCL/ibrutinib-resistant strains was examined using MTT and flow cytometry assays. EGFR gene expression was then assessed. At the same time, a PDGFD-interfering plasmid and an EGFR overexpression plasmid were transfected into the DLBCL drug-resistant cells (TMD8-ibrutinib, HBL1-ibrutinib) separately or together. MTT was used to measure cell proliferation and changes in the IC50 of ibrutinib. A total of 86 DEGs that increased in the CR, PR and SD tissues were screened, and then evaluated with GO and KEGG. The interaction network diagram showed that there was a regulatory relationship between PDGFD and disease-related genes, and that PDGFD could indirectly target the ibrutinib target gene EGFR, indicating that PDGFD could regulate DLBCL via EGFR. IHC results showed high expression of PDGFD in diffuse large B-cell lymphoma tissues with ibrutinib tolerance. PDGFD expression in ibrutinib-resistant DLBCL cells was higher compared with in parental cells. Following interference with PDGFD expression in ibrutinib-resistant DLBCL cells, the IC50 value of ibrutinib decreased, the rate of apoptosis increased and EGFR expression decreased. In brief, EGFR overexpression can reverse the resistance of DLBCL to ibrutinib via PDGFD interference, and PDGFD induces the resistance of DLBCL to ibrutinib via EGFR.
Collapse
Affiliation(s)
- Jia Jin
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - Leiping Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - Zhonghua Tao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - Jian Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - Fangfang Lv
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - Junning Cao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - Xichun Hu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| |
Collapse
|
46
|
Liu H, Chen H, Deng X, Peng Y, Zeng Q, Song Z, He W, Zhang L, Xiao T, Gao G, Li B. Knockdown of TRIM28 inhibits PDGF-BB-induced vascular smooth muscle cell proliferation and migration. Chem Biol Interact 2019; 311:108772. [PMID: 31351049 DOI: 10.1016/j.cbi.2019.108772] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 07/04/2019] [Accepted: 07/24/2019] [Indexed: 11/18/2022]
Abstract
Atherosclerosis is a common type of cardiovascular disease (CVD), remaining one of the leading causes of global death. Tripartite motif-containing 28 (TRIM28) is a member of TRIM family that has been found to be involved in atherosclerosis. However, the role of TRIM28 in atherosclerosis remains unknown. This study aimed to investigate the effects of TRIM28 on the phenotypic switching of human aortic smooth muscle cells (HASMCs), which is considered as a fundamental event during the development of atherosclerosis. The results showed that TRIM28 was highly expressed in human atherosclerotic tissues, as well in cultured HASMCs stimulated by platelet-derived growth factor subunit B homodimer (PDGF-BB). Knockdown of TRIM28 by transfection with siRNA targeting TRIM28 (si-TRIM28) significantly suppressed the PDGF-BB-induced cell proliferation and migration of HASMCs. Besides, knockdown of TRIM28 inhibited the expressions of matrix metalloproteinase (MMP)-2 and MMP-9. The VSMC markers including α-smooth muscle actin (α-SMA), calponin and SM22α were upregulated in TRIM28 knocked down HASMCs. Furthermore, knockdown of TRIM28 blocked PDGF-BB-induced NF-κB activation in HASMCs. Collectively, knockdown of TRIM28 prevented PDGF-BB-induced phenotypic switching of HASMCs, which might be mediated by the regulation of NF-κB signaling pathway.
Collapse
Affiliation(s)
- Hongtao Liu
- Department of Cardiovascular Medicine, Shenzhen Longhua District Central Hospital, Longhua Central Hospital Affiliated Guangdong Medical University, Shenzhen, 518110, Guangdong Province, China.
| | - Hongwei Chen
- Department of Cardiovascular Medicine, Shenzhen Longhua District Central Hospital, Longhua Central Hospital Affiliated Guangdong Medical University, Shenzhen, 518110, Guangdong Province, China
| | - Xia Deng
- Pharmacy Department, Shenzhen Longhua District Central Hospital, Longhua Central Hospital Affiliated Guangdong Medical University, Shenzhen, 518110, Guangdong Province, China
| | - Yudong Peng
- Department of Institute of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei Province, China
| | - Qiutang Zeng
- Department of Institute of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei Province, China
| | - Zongren Song
- Department of Cardiovascular Medicine, Shenzhen Longhua District Central Hospital, Longhua Central Hospital Affiliated Guangdong Medical University, Shenzhen, 518110, Guangdong Province, China
| | - Wenping He
- Department of Cardiovascular Medicine, Shenzhen Longhua District Central Hospital, Longhua Central Hospital Affiliated Guangdong Medical University, Shenzhen, 518110, Guangdong Province, China
| | - Le Zhang
- Department of Cardiovascular Medicine, Shenzhen Longhua District Central Hospital, Longhua Central Hospital Affiliated Guangdong Medical University, Shenzhen, 518110, Guangdong Province, China
| | - Ting Xiao
- Department of Cardiovascular Medicine, Shenzhen Longhua District Central Hospital, Longhua Central Hospital Affiliated Guangdong Medical University, Shenzhen, 518110, Guangdong Province, China
| | - Gan Gao
- Department of Cardiovascular Medicine, Shenzhen Longhua District Central Hospital, Longhua Central Hospital Affiliated Guangdong Medical University, Shenzhen, 518110, Guangdong Province, China
| | - Bailin Li
- Department of Cardiovascular Medicine, Shenzhen Longhua District Central Hospital, Longhua Central Hospital Affiliated Guangdong Medical University, Shenzhen, 518110, Guangdong Province, China
| |
Collapse
|
47
|
Wang M, Wei J, Shang F, Zang K, Ji T. Platelet-derived growth factor B attenuates lethal sepsis through inhibition of inflammatory responses. Int Immunopharmacol 2019; 75:105792. [PMID: 31386981 DOI: 10.1016/j.intimp.2019.105792] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 07/25/2019] [Accepted: 07/26/2019] [Indexed: 11/29/2022]
Abstract
Sepsis is a systemic inflammatory response during infection and remains a major clinical problem with high morbidity and mortality. Platelet-derived growth factor B (PDGF-B) is a member belongs to PDGF family and has been recently reported higher expressed in survivors of severe sepsis patients. However, the exact role and underlying mechanisms of PDGF-BB in sepsis remains unclear. In this study, we found that PDGF-BB levels were significantly elevated in patients with sepsis, and higher PDGF-BB levels were negatively correlated with the levels of proinflammatory cytokines (TNF-α, IL-6, IL-1β, IL-8), and chemokines (CXCL-1 and CCL2). PDGF-BB was also found increased in experimental sepsis in mice. Blockade of PDGF-BB using Tyrphostin AG 1296 aggravated, whereas recombinant PDGF-BB treatment improved survival and tissues injury in both two murine models of CLP-induced sepsis and LPS- induced endotoxemia. PDGF-BB blockade increased, whereas PDGF-BB administration decreased the inflammatory responses, as reflected by proinflammatory cytokines (TNF-α, IL-6, IL-1β, IL-8), and chemokines (CXCL-1 and CCL2). PDGF-BB also showed inhibitory effect on immune cell activation and cytokines production in vivo and in vitro. Therefore, our findings suggest that PDGF-BB plays a protective role in sepsis by decreasing the production of pro-inflammatory cytokines and chemokines. PDGF-BB thus may be a potential therapeutic strategy for treating sepsis.
Collapse
Affiliation(s)
- Min Wang
- Department of Intensive Care Unit, The Affiliated Huai'an NO.1 People's Hospital of Nanjing Medical University, 6 Beijing West Road, Huai'an 223300, China.
| | - Jilou Wei
- Department of Intensive Care Unit, The Affiliated Huai'an NO.1 People's Hospital of Nanjing Medical University, 6 Beijing West Road, Huai'an 223300, China
| | - Futai Shang
- Department of Intensive Care Unit, The Affiliated Huai'an NO.1 People's Hospital of Nanjing Medical University, 6 Beijing West Road, Huai'an 223300, China
| | - Kui Zang
- Department of Intensive Care Unit, The Affiliated Huai'an NO.1 People's Hospital of Nanjing Medical University, 6 Beijing West Road, Huai'an 223300, China
| | - Ting Ji
- Department of Intensive Care Unit, The Affiliated Huai'an NO.1 People's Hospital of Nanjing Medical University, 6 Beijing West Road, Huai'an 223300, China
| |
Collapse
|
48
|
Minchenko OH, Viletska YM, Minchenko DO, Davydov VV. Insulin resistance in obese adolescents and adult men modifies the expression of proliferation related genes. UKRAINIAN BIOCHEMICAL JOURNAL 2019. [DOI: 10.15407/ubj91.03.065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|