1
|
Popek-Marciniec S, Styk W, Chocholska S, Szudy-Szczyrek A, Sidor K, Swiderska-Kolacz G, Hus M, Czerwik-Marcinkowska J, Zmorzynski S. Associations of ANGPT2 expression and its variants (rs1868554 and rs7825407) with multiple myeloma risk and outcome. Front Oncol 2025; 15:1468373. [PMID: 40115011 PMCID: PMC11922703 DOI: 10.3389/fonc.2025.1468373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
The growth of blood vessels from the existing vasculature has a significant impact on the course of multiple myeloma (MM). The ANGPT2 (angiopoietin-2) protein is encoded by the ANGPT2 gene and plays an important role in angiogenesis. The expression of proangiogenic proteins is influenced not only by microenvironmental factors but also by genetic changes. We analyzed two variants/polymorphisms of the ANGPT2 gene, rs1868554 (T>A) and rs7825407 (G>C). Both are located in the intron sequence and can affect the final mRNA sequence by modifying splicing. Purpose Therefore, we assessed the impact of selected variants on ANGPT2 gene expression at the mRNA and protein levels. Additionally, we evaluated the associations of the analyzed genetic changes with the clinical and laboratory parameters of the disease and the response to bortezomib/thalidomide-based therapies. We hypothesize that variants and expression of the ANGPT2 gene may be associated with a greater risk of MM development and may also affect the response to treatment in MM patients. Patients and methods Genomic DNA extracted from 103 newly diagnosed MM patients and 120 healthy blood donors was used to analyze ANGPT2 variants (via automated DNA sequencing). RNA was subjected to real-time PCR to determine ANGPT2 expression at the mRNA level. The concentration of angiopoietin-2 (in MM sera) was determined by ELISA. Results The results of our study showed that individuals with the AA genotype of rs1868554 and the CC genotype of rs7825407 had a greater risk of developing MM (OR=6.12, p=0.02 and OR=6.01, p=0.02, respectively). The ANGPT2 gene variants did not affect ANGPT2 expression at the mRNA level. However, ANGPT2 expression was positively correlated with CRP (Spearman's rho 0.26, p<0.05) and negatively correlated with LDH (Spearman's rho -0.25, p<0.05) in MM patients. Conclusion Our results showed that ANGPT2 expression at the mRNA level correlates with CRP, a negative prognostic factor in MM. The ANGPT2 protein is a proangiogenic factor, and its concentration is significantly greater in MM patients than in healthy individuals, which was also confirmed in our research. Therefore, this protein with VEGF and HB-EGF, should be considered in the future as a markers of angiogenesis in MM.
Collapse
Affiliation(s)
| | - Wojciech Styk
- Academic Laboratory of Psychological Tests, Medical University, Lublin, Poland
| | - Sylwia Chocholska
- Chair and Department of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, Lublin, Poland
| | - Aneta Szudy-Szczyrek
- Chair and Department of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, Lublin, Poland
| | - Katarzyna Sidor
- Academic Laboratory of Psychological Tests, Medical University, Lublin, Poland
| | | | - Marek Hus
- Chair and Department of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, Lublin, Poland
| | | | | |
Collapse
|
2
|
Zhao Y, Yu B, Wang Y, Tan S, Xu Q, Wang Z, Zhou K, Liu H, Ren Z, Jiang Z. Ang-1 and VEGF: central regulators of angiogenesis. Mol Cell Biochem 2025; 480:621-637. [PMID: 38652215 DOI: 10.1007/s11010-024-05010-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 04/09/2024] [Indexed: 04/25/2024]
Abstract
Angiopoietin-1 (Ang-1) and Vascular Endothelial Growth Factor (VEGF) are central regulators of angiogenesis and are often inactivated in various cardiovascular diseases. VEGF forms complexes with ETS transcription factor family and exerts its action by downregulating multiple genes. Among the target genes of the VEGF-ETS complex, there are a significant number encoding key angiogenic regulators. Phosphorylation of the VEGF-ETS complex releases transcriptional repression on these angiogenic regulators, thereby promoting their expression. Ang-1 interacts with TEK, and this phosphorylation release can be modulated by the Ang-1-TEK signaling pathway. The Ang-1-TEK pathway participates in the transcriptional activation of VEGF genes. In summary, these elements constitute the Ang-1-TEK-VEGF signaling pathway. Additionally, Ang-1 is activated under hypoxic and inflammatory conditions, leading to an upregulation in the expression of TEK. Elevated TEK levels result in the formation of the VEGF-ETS complex, which, in turn, downregulates the expression of numerous angiogenic genes. Hence, the Ang-1-dependent transcriptional repression is indirect. Reduced expression of many target genes can lead to aberrant angiogenesis. A significant overlap exists between the target genes regulated by Ang-1-TEK-VEGF and those under the control of the Ang-1-TEK-TSP-1 signaling pathway. Mechanistically, this can be explained by the replacement of the VEGF-ETS complex with the TSP-1 transcriptional repression complex at the ETS sites on target gene promoters. Furthermore, VEGF possesses non-classical functions unrelated to ETS and DNA binding. Its supportive role in TSP-1 formation may be exerted through the VEGF-CRL5-VHL-HIF-1α-VH032-TGF-β-TSP-1 axis. This review assesses the regulatory mechanisms of the Ang-1-TEK-VEGF signaling pathway and explores its significant overlap with the Ang-1-TEK-TSP-1 signaling pathway.
Collapse
Affiliation(s)
- Yuanqin Zhao
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, 421001, China
| | - Bo Yu
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, 421001, China
| | - Yanxia Wang
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, 421001, China
| | - Shiming Tan
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, 421001, China
| | - Qian Xu
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, 421001, China
| | - Zhaoyue Wang
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, 421001, China
| | - Kun Zhou
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, 421001, China
| | - Huiting Liu
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, 421001, China
| | - Zhong Ren
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, 421001, China
| | - Zhisheng Jiang
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, 421001, China.
| |
Collapse
|
3
|
Yu Y, Wang C, Wang Y, Shi H, Hu H, Du Y, Zhou Z. The conserved wobble uridine tRNA thiolase Ctu1 is required for angiogenesis and embryonic development. PLoS One 2024; 19:e0315854. [PMID: 39705244 DOI: 10.1371/journal.pone.0315854] [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: 09/08/2024] [Accepted: 12/01/2024] [Indexed: 12/22/2024] Open
Abstract
Cytosolic thiouridylase is a conserved cytoplasmic tRNA thiolase composed of two different subunits, CTU1 and CTU2. CTU2 serves as a scaffold protein, while CTU1 catalyzes the 2-thiolation at the 34th wobble uridine of the anticodon loop. tRNAGlnUUG, tRNAGluUUC, and tRNALysUUU are the tRNA substrates that are modified with a thiol group at the C2 positions (s2) by CTU1, and also with a methoxycarbonylmethyl group at the C5 positions (mcm5) by Elongator and ALKBH8. mcm5s2U34 modification of the three tRNAs, and their modifying enzymes are involved in human disease and development. Elongator mutant animals exhibit severe phenotypes, while the biological function of Ctu1 in vertebrate animal models remains poorly characterized. Here, we applied antisense morpholino oligonucleotides targeting cytosolic thiouridylase subunit1 (ctu1) transcripts in a zebrafish model and small interfereing RNA against CTU1 transcript in human endothelial cells to define the phenotypes. We found that deficiency of ctu1 causes impaired angiogenesis and development in zebrafish embryos, and CTU1 is involved in proliferation, migration, and tube formation of human endothelial cells. We employed single-cell RNA sequencing to acquire the transcriptomic atlas from ctu1 and control morphant zebrafish. Comprehensive bioinformatics analysis, including pseudo-time, RNA velocity, cell-cell communication, and gene regulatory network inference revealed that ctu1 deficiency leads to the arrest of cell cycle, and the defects of nerve development and erythrocyte differentiation and the attenuation of several pro-angiogenic signaling pathways, e.g., angpt-tek and dll4-notch. Our findings show for the first time that CTU1 is essential for angiogenesis and embryonic development in vertebrates.
Collapse
Affiliation(s)
- Yangziwei Yu
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
- Collaborative innovation Center for Biomedicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Chuqiao Wang
- Collaborative innovation Center for Biomedicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Yan Wang
- Collaborative innovation Center for Biomedicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Heng Shi
- Collaborative innovation Center for Biomedicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huiyuan Hu
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Yibin Du
- Shanghai World Foreign Language Academy, Shanghai, China
| | - Zhaoli Zhou
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
- Collaborative innovation Center for Biomedicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
- Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, China
| |
Collapse
|
4
|
Ostapowicz J, Ostrowska K, Golusiński W, Kulcenty K, Suchorska WM. Improving therapeutic strategies for Head and Neck Cancer: Insights from 3D hypoxic cell culture models in treatment response evaluation. Adv Med Sci 2024; 69:368-376. [PMID: 39047970 DOI: 10.1016/j.advms.2024.07.007] [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: 04/04/2024] [Revised: 06/04/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024]
Abstract
Hypoxia in the tumor core negatively affects the outcome of patients with head and neck squamous cell carcinoma (HNSCC). Nevertheless, its role in predicting treatment response requires further exploration. Typically, reduced oxygen levels in the tumor core correlate with diminished efficacy of radiotherapy, chemotherapy, and immunotherapy, which are commonly used for HNSCC patients' treatment. Understanding the mechanistic underpinnings of these varied treatment responses in HNSCC is crucial for enhancing therapeutic outcomes and extending patients' overall survival (OS) rates. Standard monolayer cell culture conditions have major limitations in mimicking tumor physiological features and the complexity of the tumor microenvironment. Three-dimensional (3D) cell cultures enable the recreation of the in vivo tumor attributes, encompassing oxygen and nutrient gradients, cellular morphology, and intracellular connections. It is vital to use the 3D model in treatment response studies to mimic the tumor microenvironment, as evidenced by the decreased sensitivity of 3D structures to anticancer therapy. Accordingly, the aim of the study was to delineate the utility of the 3D models of hypoxic head and neck tumors in drug screening and treatment response studies.
Collapse
Affiliation(s)
- Julia Ostapowicz
- Department of Electroradiology, Poznan University of Medical Sciences, Poznan, Poland; Radiobiology Laboratory, The Greater Poland Cancer Centre, Poznan, Poland; Doctoral School, Poznan University of Medical Sciences, Poznan, Poland.
| | - Kamila Ostrowska
- Radiobiology Laboratory, The Greater Poland Cancer Centre, Poznan, Poland; Department of Head and Neck Surgery, Poznan University of Medical Sciences, The Greater Poland Cancer Centre, Poznan, Poland
| | - Wojciech Golusiński
- Department of Head and Neck Surgery, Poznan University of Medical Sciences, The Greater Poland Cancer Centre, Poznan, Poland
| | - Katarzyna Kulcenty
- Radiobiology Laboratory, The Greater Poland Cancer Centre, Poznan, Poland
| | - Wiktoria M Suchorska
- Department of Electroradiology, Poznan University of Medical Sciences, Poznan, Poland; Radiobiology Laboratory, The Greater Poland Cancer Centre, Poznan, Poland
| |
Collapse
|
5
|
Benard C, Le Loarer F, Gomez-Mascard A, Azmani R, Garcia J, Perret R, de Pinieux G, Miquelestorena-Standley E, Weingertner N, Karanian M, Meurgey A, Michot A, Tirode F, Truffaux N, Macagno N, Bouvier C. Comprehensive Molecular Characterization of a Large Series of Calcified Chondroid Mesenchymal Neoplasms Widening Their Morphologic Spectrum. Am J Surg Pathol 2024; 48:991-1004. [PMID: 39016330 DOI: 10.1097/pas.0000000000002260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Recently, FN1 fusions to receptor tyrosine kinase genes have been identified in soft tissue tumors with calcified chondroid matrix named calcifying chondroid mesenchymal neoplasms (CCMNs). We collected 33 cases of CCMN from the French network for soft tissue and bone tumors. We performed whole-exome RNA sequencing, expression analysis, and genome-wide DNA methylation profiling in 33, 30, and 20 cases of CCMN compared with a control group of tumors, including noncalcified tenosynovial giant cell tumor (TGCT). Among them, 15 cases showed morphologic overlap with soft tissue chondroma, 8 cases with tophaceous pseudogout, and 10 cases with chondroid TGCT. RNA-sequencing revealed a fusion of FN1 in 76% of cases (25/33) with different 5' partners, including most frequently FGFR2 (14 cases), TEK or FGFR1. Among CCMN associated with FGFR1 fusions, 2 cases had overexpression of FGF23 without tumor-induced osteomalacia. Four CCMN had PDGFRA::USP8 fusions; 3 of which had histologic features of TGCT and were located in the hip, foot, and temporomandibular joint (TMJ). All cases with FN1::TEK fusion were located at TMJ and had histologic features of TGCT with or without chondroid matrix. They formed a distinct cluster on unsupervised clustering analyses based on whole transcriptome and genome-wide methylome data. Our study confirms the high prevalence of FN1 fusions in CCMN. In addition, through transcriptome and methylome analyses, we have identified a novel subgroup of tumors located at the TMJ, exhibiting TGCT-like features and FN1::TEK fusions.
Collapse
Affiliation(s)
- Clément Benard
- Department of Pathology, Timone University Hospital, Marseille
| | - François Le Loarer
- Department of Biopathology, Bergonié Institute, Regional Comprehensive Cancer Center
- UMR1312, University of Bordeaux, Inserm, BRIC, BoRdeaux Institute of Oncology
| | | | | | - Jeremy Garcia
- Department of Pathology, Timone University Hospital, Marseille
| | - Raul Perret
- Department of Biopathology, Bergonié Institute, Regional Comprehensive Cancer Center
- UMR1312, University of Bordeaux, Inserm, BRIC, BoRdeaux Institute of Oncology
| | | | | | | | | | | | - Audrey Michot
- UMR1312, University of Bordeaux, Inserm, BRIC, BoRdeaux Institute of Oncology
- Department of Surgery, Bergonié Institute, Bordeaux
| | | | - Nathalene Truffaux
- UMR1312, University of Bordeaux, Inserm, BRIC, BoRdeaux Institute of Oncology
| | - Nicolas Macagno
- UMR1251, Aix Marseille Univ, MMG, Marmara Institute, Timone University Hospital
| | - Corinne Bouvier
- Department of Pathology, Timone University Hospital, Marseille
- UMR7051, INP, Equipe 8 GlioME - Gliomagenèse et MicroEnvironnement, Aix Marseille Univ, Marseille, France
| |
Collapse
|
6
|
Nałęcz D, Świętek A, Hudy D, Wiczkowski K, Złotopolska Z, Strzelczyk JK. Assessment of Concentration KRT6 Proteins in Tumor and Matching Surgical Margin from Patients with Head and Neck Squamous Cell Carcinoma. Int J Mol Sci 2024; 25:7356. [PMID: 39000463 PMCID: PMC11242288 DOI: 10.3390/ijms25137356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/28/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) are one of the most frequently detected cancers in the world; not all mechanisms related to the expression of keratin in this type of cancer are known. The aim of this study was to evaluate type II cytokeratins (KRT): KRT6A, KRT6B, and KRT6C protein concentrations in 54 tumor and margin samples of head and neck squamous cell carcinoma (HNSCC). Moreover, we examined a possible association between protein concentration and the clinical and demographic variables. Protein concentrations were measured using enzyme-linked immunosorbent assay (ELISA). Significantly higher KRT6A protein concentration was found in HNSCC samples compared to surgical margins. An inverse relationship was observed for KRT6B and KRT6C proteins. We showed an association between the KRT6C protein level and clinical parameters T and N in tumor and margin samples. When analyzing the effect of smoking and drinking on KRT6A, KRT6B, and KRT6C levels, we demonstrated a statistically significant difference between regular or occasional tobacco and alcohol habits and patients who do not have any tobacco and alcohol habits in tumor and margin samples. Moreover, we found an association between KRT6B and KRT6C concentration and proliferative index Ki-67 and HPV status in tumor samples. Our results showed that concentrations of KRT6s were different in the tumor and the margin samples and varied in relation to clinical and demographic parameters. We add information to the current knowledge about the role of KRT6s isoforms in HNSCC. We speculate that variations in the studied isoforms of the KRT6 protein could be due to the presence and development of the tumor and its microenvironment. It is important to note that the analyses were performed in tumor and surgical margins and can provide more accurate information on the function in normal and cancer cells and regulation in response to various factors.
Collapse
Affiliation(s)
- Dariusz Nałęcz
- Department of Otolaryngology and Maxillofacial Surgery, St. Vincent De Paul Hospital, 1 Wójta Radtkego St., 81-348 Gdynia, Poland
| | - Agata Świętek
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 19 Jordana St., 41-808 Zabrze, Poland
- Silesia LabMed Research and Implementation Centre, Medical University of Silesia in Katowice, 19 Jordana St., 41-808 Zabrze, Poland
| | - Dorota Hudy
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 19 Jordana St., 41-808 Zabrze, Poland
| | - Karol Wiczkowski
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 19 Jordana St., 41-808 Zabrze, Poland
- Students' Scientific Association, Department of Medical and Molecular Biology, Medical University of Silesia, Katowice, 19 Jordana St., 41-808 Zabrze, Poland
| | - Zofia Złotopolska
- Department of Otolaryngology and Maxillofacial Surgery, St. Vincent De Paul Hospital, 1 Wójta Radtkego St., 81-348 Gdynia, Poland
| | - Joanna Katarzyna Strzelczyk
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 19 Jordana St., 41-808 Zabrze, Poland
| |
Collapse
|
7
|
de Lima MC, de Castro CC, Aguiar KEC, Monte N, da Costa Nunes GG, da Costa ACA, Rodrigues JCG, Guerreiro JF, Ribeiro-dos-Santos Â, de Assumpção PP, Burbano RMR, Fernandes MR, dos Santos SEB, dos Santos NPC. Molecular Profile of Important Genes for Radiogenomics in the Amazon Indigenous Population. J Pers Med 2024; 14:484. [PMID: 38793065 PMCID: PMC11122349 DOI: 10.3390/jpm14050484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/13/2024] [Accepted: 04/18/2024] [Indexed: 05/26/2024] Open
Abstract
Radiotherapy is focused on the tumor but also reaches healthy tissues, causing toxicities that are possibly related to genomic factors. In this context, radiogenomics can help reduce the toxicity, increase the effectiveness of radiotherapy, and personalize treatment. It is important to consider the genomic profiles of populations not yet studied in radiogenomics, such as the indigenous Amazonian population. Thus, our objective was to analyze important genes for radiogenomics, such as ATM, TGFB1, RAD51, AREG, XRCC4, CDK1, MEG3, PRKCE, TANC1, and KDR, in indigenous people and draw a radiogenomic profile of this population. The NextSeq 500® platform was used for sequencing reactions; for differences in the allelic frequency between populations, Fisher's Exact Test was used. We identified 39 variants, 2 of which were high impact: 1 in KDR (rs41452948) and another in XRCC4 (rs1805377). We found four modifying variants not yet described in the literature in PRKCE. We did not find any variants in TANC1-an important gene for personalized medicine in radiotherapy-that were associated with toxicities in previous cohorts, configuring a protective factor for indigenous people. We identified four SNVs (rs664143, rs1801516, rs1870377, rs1800470) that were associated with toxicity in previous studies. Knowing the radiogenomic profile of indigenous people can help personalize their radiotherapy.
Collapse
Affiliation(s)
- Milena Cardoso de Lima
- Oncology Research Center, Federal University of Pará, Belém 66073-005, PA, Brazil; (M.C.d.L.); (C.C.d.C.); (K.E.C.A.); (N.M.); (G.G.d.C.N.); (A.C.A.d.C.); (J.C.G.R.); (J.F.G.); (P.P.d.A.); (R.M.R.B.); (M.R.F.)
| | - Cinthia Costa de Castro
- Oncology Research Center, Federal University of Pará, Belém 66073-005, PA, Brazil; (M.C.d.L.); (C.C.d.C.); (K.E.C.A.); (N.M.); (G.G.d.C.N.); (A.C.A.d.C.); (J.C.G.R.); (J.F.G.); (P.P.d.A.); (R.M.R.B.); (M.R.F.)
| | - Kaio Evandro Cardoso Aguiar
- Oncology Research Center, Federal University of Pará, Belém 66073-005, PA, Brazil; (M.C.d.L.); (C.C.d.C.); (K.E.C.A.); (N.M.); (G.G.d.C.N.); (A.C.A.d.C.); (J.C.G.R.); (J.F.G.); (P.P.d.A.); (R.M.R.B.); (M.R.F.)
| | - Natasha Monte
- Oncology Research Center, Federal University of Pará, Belém 66073-005, PA, Brazil; (M.C.d.L.); (C.C.d.C.); (K.E.C.A.); (N.M.); (G.G.d.C.N.); (A.C.A.d.C.); (J.C.G.R.); (J.F.G.); (P.P.d.A.); (R.M.R.B.); (M.R.F.)
| | - Giovanna Gilioli da Costa Nunes
- Oncology Research Center, Federal University of Pará, Belém 66073-005, PA, Brazil; (M.C.d.L.); (C.C.d.C.); (K.E.C.A.); (N.M.); (G.G.d.C.N.); (A.C.A.d.C.); (J.C.G.R.); (J.F.G.); (P.P.d.A.); (R.M.R.B.); (M.R.F.)
| | - Ana Caroline Alves da Costa
- Oncology Research Center, Federal University of Pará, Belém 66073-005, PA, Brazil; (M.C.d.L.); (C.C.d.C.); (K.E.C.A.); (N.M.); (G.G.d.C.N.); (A.C.A.d.C.); (J.C.G.R.); (J.F.G.); (P.P.d.A.); (R.M.R.B.); (M.R.F.)
| | - Juliana Carla Gomes Rodrigues
- Oncology Research Center, Federal University of Pará, Belém 66073-005, PA, Brazil; (M.C.d.L.); (C.C.d.C.); (K.E.C.A.); (N.M.); (G.G.d.C.N.); (A.C.A.d.C.); (J.C.G.R.); (J.F.G.); (P.P.d.A.); (R.M.R.B.); (M.R.F.)
| | - João Farias Guerreiro
- Oncology Research Center, Federal University of Pará, Belém 66073-005, PA, Brazil; (M.C.d.L.); (C.C.d.C.); (K.E.C.A.); (N.M.); (G.G.d.C.N.); (A.C.A.d.C.); (J.C.G.R.); (J.F.G.); (P.P.d.A.); (R.M.R.B.); (M.R.F.)
- Laboratory of Human and Medical Genetics, Federal University of Pará, Belém 66075-110, PA, Brazil;
| | | | - Paulo Pimentel de Assumpção
- Oncology Research Center, Federal University of Pará, Belém 66073-005, PA, Brazil; (M.C.d.L.); (C.C.d.C.); (K.E.C.A.); (N.M.); (G.G.d.C.N.); (A.C.A.d.C.); (J.C.G.R.); (J.F.G.); (P.P.d.A.); (R.M.R.B.); (M.R.F.)
| | - Rommel Mario Rodríguez Burbano
- Oncology Research Center, Federal University of Pará, Belém 66073-005, PA, Brazil; (M.C.d.L.); (C.C.d.C.); (K.E.C.A.); (N.M.); (G.G.d.C.N.); (A.C.A.d.C.); (J.C.G.R.); (J.F.G.); (P.P.d.A.); (R.M.R.B.); (M.R.F.)
| | - Marianne Rodrigues Fernandes
- Oncology Research Center, Federal University of Pará, Belém 66073-005, PA, Brazil; (M.C.d.L.); (C.C.d.C.); (K.E.C.A.); (N.M.); (G.G.d.C.N.); (A.C.A.d.C.); (J.C.G.R.); (J.F.G.); (P.P.d.A.); (R.M.R.B.); (M.R.F.)
| | - Sidney Emanuel Batista dos Santos
- Oncology Research Center, Federal University of Pará, Belém 66073-005, PA, Brazil; (M.C.d.L.); (C.C.d.C.); (K.E.C.A.); (N.M.); (G.G.d.C.N.); (A.C.A.d.C.); (J.C.G.R.); (J.F.G.); (P.P.d.A.); (R.M.R.B.); (M.R.F.)
- Laboratory of Human and Medical Genetics, Federal University of Pará, Belém 66075-110, PA, Brazil;
| | - Ney Pereira Carneiro dos Santos
- Oncology Research Center, Federal University of Pará, Belém 66073-005, PA, Brazil; (M.C.d.L.); (C.C.d.C.); (K.E.C.A.); (N.M.); (G.G.d.C.N.); (A.C.A.d.C.); (J.C.G.R.); (J.F.G.); (P.P.d.A.); (R.M.R.B.); (M.R.F.)
| |
Collapse
|
8
|
Chen B, Han Y, Sheng S, Deng J, Vasquez E, Yau V, Meng M, Sun C, Wang T, Wang Y, Sheng M, Wu T, Wang X, Liu Y, Lin N, Zhang L, Shao W. An angiogenesis-associated gene-based signature predicting prognosis and immunotherapy efficacy of head and neck squamous cell carcinoma patients. J Cancer Res Clin Oncol 2024; 150:91. [PMID: 38347320 PMCID: PMC10861726 DOI: 10.1007/s00432-024-05606-8] [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: 09/17/2023] [Accepted: 01/02/2024] [Indexed: 02/15/2024]
Abstract
OBJECTIVES To develop a model that can assist in the diagnosis and prediction of prognosis for head and neck squamous cell carcinoma (HNSCC). MATERIALS AND METHODS Data from TCGA and GEO databases were used to generate normalized gene expression data. Consensus Cluster Plus was used for cluster analysis and the relationship between angiogenesis-associated gene (AAG) expression patterns, clinical characteristics and survival was examined. Support vector machine (SVM) and least absolute shrinkage and selection operator (LASSO) analyzes and multiple logistic regression analyzes were performed to determine the diagnostic model, and a prognostic nomogram was constructed using univariate and multivariate Cox regression analyses. ESTIMATE, XCELL, TIMER, QUANTISEQ, MCPCOUNTER, EPIC, CIBERSORT-ABS, CIBERSORT algorithms were used to assess the immune microenvironment of HNSCC patients. In addition, gene set enrichment analysis, treatment sensitivity analysis, and AAGs mutation studies were performed. Finally, we also performed immunohistochemistry (IHC) staining in the tissue samples. RESULTS We classified HNSCC patients into subtypes based on differences in AAG expression from TCGA and GEO databases. There are differences in clinical features, TME, and immune-related gene expression between two subgroups. We constructed a HNSCC diagnostic model based on nine AAGs, which has good sensitivity and specificity. After further screening, we constructed a prognostic risk signature for HNSCC based on six AAGs. The constructed risk score had a good independent prognostic significance, and it was further constructed into a prognostic nomogram together with age and stage. Different prognostic risk groups have differences in immune microenvironment, drug sensitivity, gene enrichment and gene mutation. CONCLUSION We have constructed a diagnostic and prognostic model for HNSCC based on AAG, which has good performance. The constructed prognostic risk score is closely related to tumor immune microenvironment and immunotherapy response.
Collapse
Affiliation(s)
- Bangjie Chen
- College & Hospital of Stomatology, Key Lab. of Oral Diseases Research of Anhui Province, Anhui Medical University, Hefei, China
- The First Affiliated Hospital (First Clinical Medical College), Anhui Medical University, Hefei, China
| | - Yanxun Han
- The First Affiliated Hospital (First Clinical Medical College), Anhui Medical University, Hefei, China
| | - Shuyan Sheng
- The First Affiliated Hospital (First Clinical Medical College), Anhui Medical University, Hefei, China
| | - Jianyi Deng
- The First Affiliated Hospital (First Clinical Medical College), Anhui Medical University, Hefei, China
| | | | - Vicky Yau
- Division of Oral and Maxillofacial Surgery, NewYork Presbyterian (Columbia Irving Medical Center), New York, USA
| | - Muzi Meng
- UK Program Site, American University of the Caribbean School of Medicine, Preston, UK
- Bronxcare Health System, New York, USA
| | - Chenyu Sun
- The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Tao Wang
- The Affiliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou, China
| | - Yu Wang
- The Affiliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou, China
| | - Mengfei Sheng
- College & Hospital of Stomatology, Key Lab. of Oral Diseases Research of Anhui Province, Anhui Medical University, Hefei, China
- Department of Microbiology and Parasitology (Anhui Provincial Laboratory of Pathogen Biology), School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Tiangang Wu
- College & Hospital of Stomatology, Key Lab. of Oral Diseases Research of Anhui Province, Anhui Medical University, Hefei, China
| | - Xinyi Wang
- The First Affiliated Hospital (First Clinical Medical College), Anhui Medical University, Hefei, China
| | - Yuchen Liu
- The First Affiliated Hospital (First Clinical Medical College), Anhui Medical University, Hefei, China
| | - Ning Lin
- The Affiliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou, China.
| | - Lei Zhang
- College & Hospital of Stomatology, Key Lab. of Oral Diseases Research of Anhui Province, Anhui Medical University, Hefei, China.
| | - Wei Shao
- College & Hospital of Stomatology, Key Lab. of Oral Diseases Research of Anhui Province, Anhui Medical University, Hefei, China.
- Department of Microbiology and Parasitology (Anhui Provincial Laboratory of Pathogen Biology), School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| |
Collapse
|
9
|
McWhorter R, Bonavida B. The Role of TAMs in the Regulation of Tumor Cell Resistance to Chemotherapy. Crit Rev Oncog 2024; 29:97-125. [PMID: 38989740 DOI: 10.1615/critrevoncog.2024053667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Tumor-associated macrophages (TAMs) are the predominant cell infiltrate in the immunosuppressive tumor microenvironment (TME). TAMs are central to fostering pro-inflammatory conditions, tumor growth, metastasis, and inhibiting therapy responses. Many cancer patients are innately refractory to chemotherapy and or develop resistance following initial treatments. There is a clinical correlation between the level of TAMs in the TME and chemoresistance. Hence, the pivotal role of TAMs in contributing to chemoresistance has garnered significant attention toward targeting TAMs to reverse this resistance. A prerequisite for such an approach requires a thorough understanding of the various underlying mechanisms by which TAMs inhibit response to chemotherapeutic drugs. Such mechanisms include enhancing drug efflux, regulating drug metabolism and detoxification, supporting cancer stem cell (CSCs) resistance, promoting epithelial-mesenchymal transition (EMT), inhibiting drug penetration and its metabolism, stimulating angiogenesis, impacting inhibitory STAT3/NF-κB survival pathways, and releasing specific inhibitory cytokines including TGF-β and IL-10. Accordingly, several strategies have been developed to overcome TAM-modulated chemoresistance. These include novel therapies that aim to deplete TAMs, repolarize them toward the anti-tumor M1-like phenotype, or block recruitment of monocytes into the TME. Current results from TAM-targeted treatments have been unimpressive; however, the use of TAM-targeted therapies in combination appears promising These include targeting TAMs with radiotherapy, chemotherapy, chemokine receptor inhibitors, immunotherapy, and loaded nanoparticles. The clinical limitations of these strategies are discussed.
Collapse
Affiliation(s)
| | - Benjamin Bonavida
- Department of Microbiology, Immunology, & Molecular Genetics, David Geffen School of Medicine at UCLA, Johnson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90025-1747, USA
| |
Collapse
|
10
|
Sayın Kocakap DB, Kaygusuz S, Aksoy E, Şahin Ö, Baççıoğlu A, Ekici A, Kalpaklıoğlu AF, Ekici MS, Gül S, Kaçmaz B, Ayaşlıoğlu Açıkgöz E, Alyılmaz Bekmez S, Rouse BT, Azkur AK. Adverse effect of VEGFR-2 (rs1870377) polymorphism on the clinical course of COVID-19 in females and males in an age-dependent manner. Microbes Infect 2023; 25:105188. [PMID: 37499788 DOI: 10.1016/j.micinf.2023.105188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/12/2023] [Accepted: 07/22/2023] [Indexed: 07/29/2023]
Abstract
The COVID-19 pandemic has affected people worldwide with varying clinical presentations ranging from mild to severe or fatal, and studies have found that age, gender, and some comorbidities can influence the severity of the disease. It would be valuable to have genetic markers that might help predict the likely outcome of infection. For this objective, genes encoding VEGFR-2 (rs1870377), CCR5Δ32 (rs333), and TLR3 (rs5743313) were analyzed for polymorphisms in the peripheral blood of 160 COVID-19 patients before COVID-19 vaccine was available in Türkiye. We observed that possession of the VEGFR-2 rs1870377 mutant allele increased the risk of severe/moderate disease in females and subjects ≥65 years of age, but was protective in males <65 years of age. Other significant results were that the CCR5Δ32 allele was protective against severe disease in subjects ≥65 years of age, while TLR3 rs5743313 polymorphism was found to be protective against severe/moderate illness in males <65 years of age. The VEGFR-2 rs1870377 mutant allele was a risk factor for severe/moderate disease, particularly in females over the age of 65. These findings suggest that genetic polymorphisms have an age- and sex-dependent influence on the severity of COVID-19, and the VEGFR-2 rs1870377 mutant allele could be a potential predictor of disease severity.
Collapse
Affiliation(s)
| | - Sedat Kaygusuz
- Department of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, Kırıkkale University, Kırıkkale, Türkiye
| | - Emel Aksoy
- Department of Virology, Faculty of Veterinary Medicine, Kırıkkale University, Kırıkkale, Türkiye
| | - Ömer Şahin
- Department of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, Kırıkkale University, Kırıkkale, Türkiye
| | - Ayşe Baççıoğlu
- Department of Allergy and Immunology, Faculty of Medicine, Kırıkkale University, Kırıkkale, Türkiye; Department of Pulmonary Diseases, Faculty of Medicine, Kırıkkale University, Kırıkkale, Türkiye
| | - Aydanur Ekici
- Department of Pulmonary Diseases, Faculty of Medicine, Kırıkkale University, Kırıkkale, Türkiye
| | - Ayşe Füsun Kalpaklıoğlu
- Department of Allergy and Immunology, Faculty of Medicine, Kırıkkale University, Kırıkkale, Türkiye; Department of Pulmonary Diseases, Faculty of Medicine, Kırıkkale University, Kırıkkale, Türkiye
| | - Mehmet Savaş Ekici
- Department of Pulmonary Diseases, Faculty of Medicine, Kırıkkale University, Kırıkkale, Türkiye
| | - Serdar Gül
- Department of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, Kırıkkale University, Kırıkkale, Türkiye
| | - Birgül Kaçmaz
- Department of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, Kırıkkale University, Kırıkkale, Türkiye
| | - Ergin Ayaşlıoğlu Açıkgöz
- Department of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, Kırıkkale University, Kırıkkale, Türkiye
| | - Sibel Alyılmaz Bekmez
- Department of Medical Genetics, Faculty of Medicine, Kırıkkale University, Kırıkkale, Türkiye
| | - Barry T Rouse
- College of Veterinary Medicine, University of Tennessee, Knoxville, TN, 37996, USA
| | - Ahmet Kürşat Azkur
- Department of Virology, Faculty of Veterinary Medicine, Kırıkkale University, Kırıkkale, Türkiye.
| |
Collapse
|
11
|
Xu H, Cao H, Zhang J, Jing C, Wang Z, Wu J, Du M, Xu X, Ma R. Serum VEGF levels as a predictor of recurrence in patients with advanced‑stage esophageal squamous cell carcinoma following curative esophagectomy followed by chemotherapy or concurrent radiotherapy. Mol Clin Oncol 2023; 19:86. [PMID: 37809347 PMCID: PMC10557093 DOI: 10.3892/mco.2023.2682] [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: 03/21/2023] [Accepted: 08/14/2023] [Indexed: 10/10/2023] Open
Abstract
The present study evaluated serum levels of vascular endothelial growth factor (VEGF) as a predictor of recurrence in patients with advanced-stage esophageal squamous cell carcinoma (ESCC) following curative esophagectomy followed by chemotherapy or concurrent radiotherapy. Patients with locally advanced resectable ESCC underwent R0 esophagectomy followed by chemotherapy or concurrent radiotherapy as an adjuvant. Serum VEGF levels in 173 patients, including 57 patients with recurrent disease, and 183 healthy controls were determined using a Luminex assay. The results demonstrated that the serum VEGF levels were significantly higher in 57 patients with locally advanced resectable ESCC at recurrence compared with the levels at pre-treatment (P<0.001). The patients with recurrence exhibited significantly higher serum VEGF levels during chemotherapy or concurrent radiotherapy than patients with no recurrence (P<0.05). Patients with low serum VEGF levels had a significantly longer survival time than those with high serum VEGF levels prior to treatment (P<0.01). The median survival times were 70 and 25 months in patients with locally advanced resectable ESCC with serum VEGF levels <161.75 and ≥161.75 pg/ml following treatment, respectively (P<0.01). Compared with patients with VEGF levels <147 pg/ml following treatment, patients with locally advanced resectable ESCC with VEGF levels ≥147 pg/ml had a significantly higher risk of recurrence (P<0.01). Patients with low serum VEGF levels (<147 pg/ml) had significantly higher recurrence-free survival rates than those with high serum VEGF levels (≥147 pg/ml) following treatment (P<0.01). The findings of the present study demonstrate that serum VEGF levels are a potential predictor of recurrence and of the treatment outcomes of chemotherapy or concurrent radiotherapy in patients with locally advanced resectable ESCC.
Collapse
Affiliation(s)
- Heng Xu
- Jiangsu Provincial Institute of Materia Medica, Nanjing Tech University, Nanjing, Jiangsu 210000, P.R. China
| | - Haixia Cao
- Research Center for Clinical Oncology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing, Jiangsu 210000, P.R. China
| | - Junying Zhang
- Research Center for Clinical Oncology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing, Jiangsu 210000, P.R. China
| | - Changwen Jing
- Research Center for Clinical Oncology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing, Jiangsu 210000, P.R. China
| | - Zhuo Wang
- Research Center for Clinical Oncology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing, Jiangsu 210000, P.R. China
| | - Jianzhong Wu
- Research Center for Clinical Oncology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing, Jiangsu 210000, P.R. China
| | - Mengjie Du
- Nanjing Runtai Clinical Laboratory, Nanjing, Jiangsu 210000, P.R. China
| | - Xuyun Xu
- Nanjing Runtai Clinical Laboratory, Nanjing, Jiangsu 210000, P.R. China
| | - Rong Ma
- Research Center for Clinical Oncology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing, Jiangsu 210000, P.R. China
| |
Collapse
|
12
|
Yang X, Chen X, Zhang S, Fan W, Zhong C, Liu T, Cheng G, Zhu L, Liu Q, Xi Y, Tan W, Lin D, Wu C. Collagen 1-mediated CXCL1 secretion in tumor cells activates fibroblasts to promote radioresistance of esophageal cancer. Cell Rep 2023; 42:113270. [PMID: 37851572 DOI: 10.1016/j.celrep.2023.113270] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 08/12/2023] [Accepted: 09/29/2023] [Indexed: 10/20/2023] Open
Abstract
Esophageal squamous-cell carcinoma (ESCC) is commonly treated with radiotherapy; however, radioresistance hinders its clinical effectiveness, and the underlying mechanism remains elusive. Here, we develop patient-derived xenografts (PDXs) from 19 patients with ESCC to investigate the mechanisms driving radioresistance. Using RNA sequencing, cytokine arrays, and single-cell RNA sequencing, we reveal an enrichment of cancer-associated fibroblast (CAF)-derived collagen type 1 (Col1) and tumor-cell-derived CXCL1 in non-responsive PDXs. Col1 not only promotes radioresistance by augmenting DNA repair capacity but also induces CXCL1 secretion in tumor cells. Additionally, CXCL1 further activates CAFs via the CXCR2-STAT3 pathway, establishing a positive feedback loop. Directly interfering with tumor-cell-derived CXCL1 or inhibiting the CXCL1-CXCR2 pathway effectively restores the radiosensitivity of radioresistant xenografts in vivo. Collectively, our study provides a comprehensive understanding of the molecular mechanisms underlying radioresistance and identifies potential targets to improve the efficacy of radiotherapy for ESCC.
Collapse
Affiliation(s)
- Xinyu Yang
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Xinjie Chen
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Shaosen Zhang
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Wenyi Fan
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; College of Chemistry and Molecular Engineering, Beijing National Laboratory for Molecular Sciences, Peking University, Beijing 100091, China; Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University (PKU), Beijing 100871, China
| | - Ce Zhong
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Tianyuan Liu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Guoyu Cheng
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Liang Zhu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Qingyi Liu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Yiyi Xi
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Wen Tan
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Dongxin Lin
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211166, China; Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou 510060, China.
| | - Chen Wu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211166, China; CAMS Oxford Institute, Chinese Academy of Medical Sciences, Beijing 100006, China.
| |
Collapse
|
13
|
Butkiewicz D, Krześniak M, Gdowicz-Kłosok A, Składowski K, Rutkowski T. DNA Double-Strand Break Response and Repair Gene Polymorphisms May Influence Therapy Results and Prognosis in Head and Neck Cancer Patients. Cancers (Basel) 2023; 15:4972. [PMID: 37894339 PMCID: PMC10605140 DOI: 10.3390/cancers15204972] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/05/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Radiotherapy and cisplatin-based chemotherapy belong to the main treatment modalities for head and neck squamous cell carcinoma (HNSCC) and induce cancer cell death by generating DNA damage, including the most severe double-strand breaks (DSBs). Alterations in DSB response and repair genes may affect individual DNA repair capacity and treatment sensitivity, contributing to the therapy resistance and poor prognosis often observed in HNSCC. In this study, we investigated the association of a panel of single-nucleotide polymorphisms (SNPs) in 20 DSB signaling and repair genes with therapy results and prognosis in 505 HNSCC patients treated non-surgically with DNA damage-inducing therapies. In the multivariate analysis, there were a total of 14 variants associated with overall, locoregional recurrence-free or metastasis-free survival. Moreover, we identified 10 of these SNPs as independent predictors of therapy failure and unfavorable prognosis in the whole group or in two treatment subgroups. These were MRE11 rs2155209, XRCC5 rs828907, RAD51 rs1801321, rs12593359, LIG4 rs1805388, CHEK1 rs558351, TP53 rs1042522, ATM rs1801516, XRCC6 rs2267437 and NBN rs2735383. Only CHEK1 rs558351 remained statistically significant after correcting for multiple testing. These results suggest that specific germline variants related to DSB response and repair may be potential genetic modifiers of therapy effects and disease progression in HNSCC treated with radiotherapy and cisplatin-based chemoradiation.
Collapse
Affiliation(s)
- Dorota Butkiewicz
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland
| | - Małgorzata Krześniak
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland
| | - Agnieszka Gdowicz-Kłosok
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland
| | - Krzysztof Składowski
- I Radiation and Clinical Oncology Department, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland
| | - Tomasz Rutkowski
- I Radiation and Clinical Oncology Department, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland
- Radiotherapy Department, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland
| |
Collapse
|
14
|
Gene polymorphisms and prognosis of head and neck squamous cell carcinoma: a systematic review. Rep Pract Oncol Radiother 2022; 27:1045-1057. [PMID: 36632296 PMCID: PMC9826662 DOI: 10.5603/rpor.a2022.0109] [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: 09/19/2022] [Accepted: 10/03/2022] [Indexed: 12/12/2022] Open
Abstract
Background Head and neck squamous cell carcinomas (HNSCCs) are associated with variable prognosis even with similar clinical characteristics and treatments. Gene polymorphisms have been suggested as prognostic factors for HNSCC which can justified this variable prognosis. So, the aim was to review literatures on gene polymorphisms and prognosis of HNSCCs. Materials and methods A systematic search was conducted using PubMed, Web of science, SCOPUS, Google Scholar and Cochrane library databases to find all related articles published up to December 2021 in the field of gene polymorphisms and HNSCC prognosis. Results Of 1029 initial searched articles, 71 articles were selected for inclusion in this systematic review. About 93 genes and 204 polymorphisms have been discussed in these articles. Among the most studied polymorphisms, the XRCC1 Arg399Gln and Arg194Trp polymorphisms were not associated with survival in most studies; the ERCC1 C19007T polymorphism had no significant association in any of the studies. Different gene polymorphisms of glutathione s-transferase family, including GSTM1 deletion, GSTT1 deletion and GSTP1 A313G, were not associated with survival in included studies. There are conflicting results regarding the association between polymorphisms such as ERCC2 A35931C, Asp312Asn, ERCC5 rs1047768 and rs17655 with HNSCC prognosis. Less studied polymorphisms, such as hOGG1 rs1052133 or the VEGF rs699947, were generally not associated with HNSCC prognosis. Conclusion Reviewed articles reported varied and contradictory results regarding the association of gene polymorphisms and HNSCC prognosis, which necessitates further studies along with meta-analysis on the results of such studies.
Collapse
|
15
|
Nasef SI, Ellawindy A, Askar AM, Hashem AA, Omar HH. Assessment of Angiopoietin-2 Single Nucleotide Polymorphism in Patients with Rheumatoid Arthritis. Inflammation 2022; 46:853-860. [PMID: 36562898 DOI: 10.1007/s10753-022-01773-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/04/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022]
Abstract
Rheumatoid arthritis (RA) is an inflammatory autoimmune disease that destroys joint cartilage and causes disability. Synovial inflammation, with angiogenesis, is an early event in the progression of the disease. Angiopoietin 2 (ANGPT2) is a cytokine with both inflammatory and angiogenic effects. Many genes can influence RA susceptibility and disease activity. The aim is to assess the relationship between ANGPT2 gene polymorphism (rs3020221) and RA. The study was a case-control study that included 212 RA patients and 238 age-and gender-matched healthy volunteers. RA disease activity was assessed using the Disease Activity Score 28 index. Erythrocyte sedimentation rate, C-reactive protein, rheumatoid factor, and antibody to cyclic citrullinated peptide were measured. ANGPT2 rs3020221 C > T SNP genotyping was done using real-time polymerase chain reaction (PCR). The TT genotype was more frequently represented in RA patients than in healthy controls (18.9% and 7.1%, respectively, p < 0.001) and increased the chance of developing RA four-fold, as compared to other genotypes (OR = 4.00, 95% CI = 2.09-7.63) (p < 0.001). The CT genotype was associated with elevated levels of the inflammatory markers ESR and CRP in RA patients (p = 0.012 and 0.037, respectively) as well as the DAS28 ESR Score (p < 0.001). The presence of the T allele either under the dominant model (for genotypes CT and TT) or the recessive model (for the genotype TT) predicts RA disease. Assessment of ANGPT2 gene polymorphism is useful to predict the patients with susceptibility to RA. The presence of T allele increased the risk of developing RA disease by two folds.
Collapse
Affiliation(s)
- Samah Ismail Nasef
- Department of Physical Medicine, Rheumatology, and Rehabilitation, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Alia Ellawindy
- Medical Genetic Unit- Department of Histology & Cell biology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Aya Mohamed Askar
- Department of Clinical Pathology, Faculty of Medicine, Suez Canal University, 4.5 Km Ring Road, P.O: 41111, Ismailia, Egypt
| | - Asmaa AbdelKreem Hashem
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Hanan Hassan Omar
- Department of Clinical Pathology, Faculty of Medicine, Suez Canal University, 4.5 Km Ring Road, P.O: 41111, Ismailia, Egypt.
| |
Collapse
|
16
|
Erkılınç G, Yasan H, Kumbul YÇ, Sivrice ME, Durgun M. Expression of prostate-specific membrane antigen in the neovasculature of primary tumors and lymph node metastasis of laryngeal squamous cell carcinomas. J Pathol Transl Med 2022; 56:134-143. [PMID: 35501674 PMCID: PMC9119807 DOI: 10.4132/jptm.2022.02.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 02/22/2022] [Indexed: 11/17/2022] Open
Abstract
Background Prostate-specific membrane antigen (PSMA) expression is encountered in tumor-associated neovascularization. Methods PSMA-antibody was applied to the paraffin blocks of 51 patients who were diagnosed with squamous cell carcinoma of the larynx and underwent laryngectomy and one who underwent lymph node dissection. The percentage of vascular expression in tumoral and extratumoral stroma and lymph nodes and intensity score in tumoral epithelium were evaluated and divided into groups according to the level of PSMA expression. Final PSMA expression was determined by multiplying intensity and percentage scores. Results The mean age was 61±10 years. Patients with perineural invasion, cartilage invasion, and local invasion exhibited higher PSMA expression scores. Age, tumor differentiation, tumor diameter, perineural invasion, tumor localization, capsular invasion, depth of invasion, surgical margin status, local invasion, nodal metastasis, TNM classification, and stage were similar in high and low PSMA expression groups. There was no PSMA expression in extratumoral vascular stroma. Significantly higher PSMA expression was observed in the vascular endothelium of metastatic lymph nodes compared with reactive lymph nodes. Patients with advanced-stage disease exhibited higher PSMA vascular expression scores compared to those with earlier stages (p<.001). PSMA expression was not correlated with overall survival, disease-specific survival, or disease-free survival (p>.05). Conclusions Our study suggests that higher PSMA expression is associated with cartilage invasion, local invasion, and advanced-stage of disease. PSMA expression can be utilized for detection of lymph node metastasis and has some predictive role in cases of neck metastasis.
Collapse
Affiliation(s)
- Gamze Erkılınç
- Department of Pathology, Süleyman Demirel Univesity, Çünür/Isparta, Turkey
- Corresponding Author: Gamze Erkılınç, MD, Department of Pathology, Süleyman Demirel Univesity, Süleyman Demirel Street, 32260, Çünür/Isparta, Turkey Tel: +90-2462113714, Fax: +90-2462112830, E-mail:
| | - Hasan Yasan
- Department of Otorhinolaryngology, Süleyman Demirel Univesity, Çünür/Isparta, Turkey
| | - Yusuf Çağdaş Kumbul
- Department of Otorhinolaryngology, Süleyman Demirel Univesity, Çünür/Isparta, Turkey
| | - Mehmet Emre Sivrice
- Department of Otorhinolaryngology, Süleyman Demirel Univesity, Çünür/Isparta, Turkey
| | - Meltem Durgun
- Department of Pathology, Süleyman Demirel Univesity, Çünür/Isparta, Turkey
| |
Collapse
|
17
|
Germline Variants in Angiogenesis-Related Genes Contribute to Clinical Outcome in Head and Neck Squamous Cell Carcinoma. Cancers (Basel) 2022; 14:cancers14071844. [PMID: 35406617 PMCID: PMC8997703 DOI: 10.3390/cancers14071844] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/28/2022] [Accepted: 04/01/2022] [Indexed: 12/20/2022] Open
Abstract
Simple Summary A high risk of relapse and treatment resistance are among the major challenges in locally advanced head and neck squamous cell carcinoma (HNSCC). Data show that common germline alterations in genes regulating angiogenesis may modulate treatment sensitivity, cancer progression, and prognosis, but relatively little is known about their role in HNSCC. Thus, our goal was to examine the effect of variation in these genes on survival outcomes in HNSCC patients receiving radiotherapy and cisplatin-based chemoradiotherapy. We identified genetic variants significantly affecting therapy results, constituting independent prognostic factors in these patients. Our results suggest that some polymorphisms in angiogenesis genes may be determinants of treatment efficacy and tumor aggressiveness in HNSCC, which may be of importance in standard therapy. These findings emphasize the potential value of the host genetic profile related to angiogenesis in assessing the risk of treatment failure. Abstract Fibroblast growth factor (FGF)/FGF receptor (FGFR), and platelet-derived growth factor (PDGF)/PDGF receptor (PDGFR) systems, as well as some matrix metalloproteinases (MMPs) and their tissue inhibitors (TIMPs), are involved in various steps of angiogenesis. Data indicate that common germline variations in angiogenesis-regulating genes may modulate therapy results and cancer progression. However, whether these variants affect clinical outcome in head and neck squamous cell carcinoma (HNSCC) is unclear. Hence, we assessed the relationship between FGF/FGFR, PDGF/PDGFR, MMP, and TIMP genetic variants and treatment outcomes in HNSCC patients receiving radiotherapy (RT) alone or combined with cisplatin-based chemotherapy. In multivariate analysis, FGF2 rs1048201 CC homozygotes showed a higher risk of death (p = 0.039), while PDGFRA rs2228230 T was strongly associated with an increased risk of locoregional relapse (HR 2.49, p = 0.001) in the combination treatment subgroup. In the RT alone subset, MMP2 rs243865 TT carriers had a higher risk of locoregional recurrence (HR 2.92, p = 0.019), whereas PDGFRB rs246395 CC homozygotes were at increased risk of metastasis (HR 3.06, p = 0.041). The MMP2 rs7201 C and TIMP2 rs7501477 T were associated with a risk of locoregional failure in the entire cohort (p = 0.032 and 0.045, respectively). Furthermore, rs1048201, rs2228230, rs246395, rs243865, rs7201, and rs7201/rs7501477 were independent indicators of an unfavorable outcome. This study demonstrates that the FGF2, PDGFRA, PDGFRB, MMP2, and TIMP2 variants may contribute to treatment failure and poor prognosis in HNSCC.
Collapse
|
18
|
Omar HH, Nasef SI, Anani MM. The Association of Angiopoietin-2 1064 C/T Rs3020221 Gene Polymorphism with Knee Osteoarthritis. Immunol Invest 2022; 51:1820-1832. [PMID: 35384768 DOI: 10.1080/08820139.2022.2058404] [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: 01/15/2023]
Abstract
Osteoarthritis (OA) is a common type of arthritis, affecting millions of people around the world. Angiopoietin-2 (Angpt-2) has a role in the development of chronic inflammatory diseases. We aimed to assess the serum Angpt-2 levels in knee OA patients and to investigate the association of Angpt-2 gene polymorphism(rs3020221 C/T) with knee OA susceptibility and severity. Angiopoietin-2(rs3020221C/T) gene polymorphism was identified in 254 knee OA patients and 227 healthy controls using real-time polymerase chain reaction. Serum Angpt-2 was measured using ELISA. The Arabic version of the Western Ontario and McMaster Universities Osteoarthritis (WOMAC) Index and Kellgren-Lawrence (KL) grading score were used to assess the clinical and radiological severity of OA and their relationship with Angpt-2(rs3020221C/T) gene polymorphism was investigated. Serum Angpt-2 levels were significantly higher in knee OA patients than in the controls (P = .001). OA patients with C/T genotype had a four times greater risk of developing OA than other genotypes (OR = 4.39, 95% CI = 2.85-6.76). Additionally, the T allele presented more in OA patients 224/508 (44%) with two times risk of developing OA (OR = 1.86, 95% CI = 1.43-2.43, p = .001). Angpt-2 SNP (rs3020221C/T) genotype C/T was significantly associated with elevated serum Angpt-2 levels (14.15 ± 5.62 ng/ml). The serum Angpt-2 levels are significantly elevated in OA patients and Angpt-2 gene polymorphism (rs3020221 C/T) may be a risk factor for OA development and both are associated with the severity of knee OA. Carriers of the C/T genotype have a significantly higher serum Angpt-2 levels and a greater risk of developing OA.
Collapse
Affiliation(s)
- Hanan H Omar
- Department of Clinical Pathology, Faculty of Medicine, Suez Canal University, Ismaila, Egypt
| | - Samah I Nasef
- Department of Physical Medicine, Rheumatology, and Rehabilitation, Faculty of Medicine, Suez Canal University, Ismaila, Egypt
| | - Maha M Anani
- Department of Clinical Pathology, Faculty of Medicine, Suez Canal University, Ismaila, Egypt
| |
Collapse
|
19
|
Larionova I, Kazakova E, Gerashchenko T, Kzhyshkowska J. New Angiogenic Regulators Produced by TAMs: Perspective for Targeting Tumor Angiogenesis. Cancers (Basel) 2021; 13:cancers13133253. [PMID: 34209679 PMCID: PMC8268686 DOI: 10.3390/cancers13133253] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/15/2021] [Accepted: 06/22/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Since the targeting of a single pro-angiogenic factor fails to improve oncological disease outcome, significant efforts have been made to identify new pro-angiogenic factors that could compensate for the deficiency of current therapy or act independently as single drugs. Our review aims to present the state-of-the art for well-known and recently described factors produced by macrophages that induce and regulate angiogenesis. A number of positive and negative regulators of angiogenesis in the tumor microenvironment are produced by tumor-associated macrophages (TAMs). Accumulating evidence has indicated that, apart from the well-known angiogenic factors, there are plenty of novel angiogenesis-regulating proteins that belong to different classes. We summarize the data regarding the direct or indirect mechanisms of the interaction of these factors with endothelial cells during angiogenesis. We highlight the recent findings that explain the limitations in the efficiency of current anti-angiogenic therapy approaches. Abstract Angiogenesis is crucial to the supply of a growing tumor with nutrition and oxygen. Inhibition of angiogenesis is one of the main treatment strategies for colorectal, lung, breast, renal, and other solid cancers. However, currently applied drugs that target VEGF or receptor tyrosine kinases have limited efficiency, which raises a question concerning the mechanism of patient resistance to the already developed drugs. Tumor-associated macrophages (TAMs) were identified in the animal tumor models as a key inducer of the angiogenic switch. TAMs represent a potent source not only for VEGF, but also for a number of other pro-angiogenic factors. Our review provides information about the activity of secreted regulators of angiogenesis produced by TAMs. They include members of SEMA and S100A families, chitinase-like proteins, osteopontin, and SPARC. The COX-2, Tie2, and other factors that control the pro-angiogenic activity of TAMs are also discussed. We highlight how these recent findings explain the limitations in the efficiency of current anti-angiogenic therapy. Additionally, we describe genetic and posttranscriptional mechanisms that control the expression of factors regulating angiogenesis. Finally, we present prospects for the complex targeting of the pro-angiogenic activity of TAMs.
Collapse
Affiliation(s)
- Irina Larionova
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050 Tomsk, Russia;
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia;
- Correspondence: (I.L.); (J.K.)
| | - Elena Kazakova
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050 Tomsk, Russia;
| | - Tatiana Gerashchenko
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia;
| | - Julia Kzhyshkowska
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050 Tomsk, Russia;
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
- German Red Cross Blood Service Baden-Württemberg—Hessen, 68167 Mannheim, Germany
- Correspondence: (I.L.); (J.K.)
| |
Collapse
|
20
|
Wang Z, Li X, Chen H, Han L, Ji X, Wang Q, Wei L, Miao Y, Wang J, Mao J, Zhang Z. Decreased HLF Expression Predicts Poor Survival in Lung Adenocarcinoma. Med Sci Monit 2021; 27:e929333. [PMID: 33979320 PMCID: PMC8127640 DOI: 10.12659/msm.929333] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background Lung adenocarcinoma (LUAD) is a type of non-small cell carcinoma. Its pathogenesis is being explored and there is no cure for the disease. Material/Methods The Gene Expression Omnibus (GEO) was searched to obtain data on expression of messenger RNA. GEO2R, an interactive web tool, was used to calculate the differentially expressed genes (DEGs) in LUAD. All the DEGs from different datasets were imported into VENNY 2.1 (https://bioinfogp.cnb.csic.es/tools/venny/index.html) to identify the intersection of the DEGs. An online analysis tool, the Database for Annotation, Visualization, and Integrated Discovery (DAVID), was used to help understand the biological meaning of DEG enrichment in LUAD. Cytoscape 3.7.2 was used to perform centrality analysis and visualize hub genes and related networks. Furthermore, the prognostic value of the hub genes was evaluated with the Kaplan-Meier plotter survival analysis tool. Results The GEO database was used to obtain RNA sequencing information for LUAD and normal tissue from the GSE118370, GSE136043, and GSE140797 datasets. A total of 376 DEGs were identified from GSE118370, 248 were identified from GSE136403, and 718 DEGs were identified from GSE140797. The 10 genes with the highest degrees of expression – the hub genes – were CAV1, TEK, SLIT2, RHOJ, DGSX, HLF, MEIS1, PTPRD, FOXF1, and ADRB2. In addition, Kaplan-Meier survival evaluation showed that CAV1, TEK, SLIT2, HLF, MEIS1, PTPRD, FOXF1, and ADRB2 were associated with favorable outcomes for LUAD. Conclusions CAV1, TEK, SLIT2, HLF, MEIS1, PTPRD, FOXF1, and ADRB2 are hub genes in the DEG interaction network for LUAD and are involved in the development of and prognosis for the disease. The mechanisms underlying these genes should be the subject of further studies.
Collapse
Affiliation(s)
- Zaiyan Wang
- Department of Respiratory Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China (mainland)
| | - Xiaoning Li
- Department of Respiratory Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China (mainland)
| | - Hao Chen
- Department of Respiratory Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China (mainland)
| | - Li Han
- Department of Respiratory Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China (mainland)
| | - Xiaobin Ji
- Department of Respiratory Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China (mainland)
| | - Qiubo Wang
- Department of Respiratory Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China (mainland)
| | - Li Wei
- Department of Respiratory Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China (mainland)
| | - Yafang Miao
- Department of Respiratory Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China (mainland)
| | - Jing Wang
- Department of Respiratory Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China (mainland)
| | - Jianfeng Mao
- Department of Respiratory Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China (mainland)
| | - Zeming Zhang
- Department of Respiratory Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China (mainland)
| |
Collapse
|