1
|
Heabah NAEG, Darwish SA, Ibrahim FMK. Prognostic significance of annexin A2 and tumor associated macrophages (TAMs) in metastatic renal cell carcinoma and their relation to Sunitinib resistance. J Immunoassay Immunochem 2024; 45:1-19. [PMID: 38018145 DOI: 10.1080/15321819.2023.2285501] [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] [Indexed: 11/30/2023]
Abstract
Sunitinib, an antiangiogenic tyrosine kinase inhibitor, is the main treatment for metastatic renal cell carcinoma (mRCC). Development of resistance is a major obstacle against therapy success. The aim of this study was to assess annexin A2 and CD163+ tumor associated macrophages (TAMs) immunohistochemical expression in 50 mRCC cases as regard to patients' prognosis and Sunitinib response. Also, to assess the correlation between annexin A2 and TAMs expression. High annexin A2 expression and TAMs density were associated with serum calcium level (P = 0.024 and 0.037, respectively), larger tumor size (P < 0.001), high tumor grade (P = 0.014 and <0.001, respectively), and the presence of tumor necrosis (P < 0.001). High annexin A2 and TAMs expressions were related to shorter patients' overall survival (P = 0.009 and 0.001, respectively) and progression-free survival (P = 0.003 and 0.001, respectively). Annexin A2 was correlated with TAMs density (r = 0.890). Annexin A2 and TAMs are associated with poor prognostic parameters in mRCC patients, including high nuclear grade, increased tumor size, and the presence of tumor necrosis, together with shorter patients' survivals and poor response to Sunitinib. Annexin A2 expression is correlated with TAMs density suggesting immunomodulatory role of annexin A2.
Collapse
Affiliation(s)
| | - Sara A Darwish
- Clinical Oncology and Nuclear Medicine Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Fatma MKh Ibrahim
- Pathology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
2
|
Chen X, Xu Y, Wang M, Ren C. Development of Prognostic Indicator Based on AU-Rich Elements-Related Genes in Glioblastoma. World Neurosurg 2023; 175:e601-e613. [PMID: 37030479 DOI: 10.1016/j.wneu.2023.03.148] [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: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 04/10/2023]
Abstract
BACKGROUND AREs (AU-rich elements) are important cis-acting short sequences in the 3'UTR (3'-untranslated region) that affect messenger RNA stability and translation. However, there were no systematic researches about AREs-related genes to predict the survival of patients with GBM (glioblastoma). METHODS Differentially expressed genes were acquired from The Cancer Genome Atlas and Chinese Glioma Genome Atlas databases. Differentially expressed AREs-related genes were filtered by overlapping differentially expressed genes and AREs-related genes. The prognostic genes were selected to construct a risk model. Patients with GBM were categorized into 2 risk groups depending on the medium value of risk score. Gene Set Enrichment Analysis was performed to explore the potential biological pathways. We explored the correlation between the risk model and immune cells. The chemotherapy sensitivity was predicted in different risk groups. RESULTS A risk model was constructed by 10 differentially expressed AREs-related genes (GNS, ANKH, PTPRN2, NELL1, PLAUR, SLC9A2, SCARA3, MAPK1, HOXB2, and EN2), and it could accurately predict the prognosis of patients with GBM. Higher risk scores for patients with GBM had a lower survival probability. The predictive power of risk model was decent. The risk score and treatment type were regarded as independent prognostic indicators. The mainly Gene Set Enrichment Analysis enrichment pathways were primary immunodeficiency and chemokine signaling pathway. Six immune cells were significant different in the 2 risk groups. There were higher abundance of macrophages M2 and neutrophils and higher sensitivity of 11 chemotherapy drugs in the high-risk group. CONCLUSIONS The 10 biomarkers might be important prognostic markers and potential therapeutic targets for patients with GBM.
Collapse
Affiliation(s)
- Xiao Chen
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong, University, Xi'an, Shaanxi, China; Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ying Xu
- Health information Services, The First Affiliated Hospital of Xi'an Jiaotong, University, Xi'an, Shaanxi, China
| | - Maode Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong, University, Xi'an, Shaanxi, China; Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Chunying Ren
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong, University, Xi'an, Shaanxi, China; Gamma Knife Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
3
|
Večurkovská I, Mašlanková J, Tomečková V, Kaťuchová J, Kisková T, Fröhlichová L, Mareková M, Stupák M. Stage-Dependent Levels of Brain-Derived Neurotrophic Factor and Matrix Metalloproteinase 9 in the Prognosis of Colorectal Cancer. Biomedicines 2023; 11:1839. [PMID: 37509480 PMCID: PMC10377127 DOI: 10.3390/biomedicines11071839] [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: 05/19/2023] [Revised: 06/19/2023] [Accepted: 06/23/2023] [Indexed: 07/30/2023] Open
Abstract
PURPOSE The development of sensitive and non-invasive biomarkers for the early detection of CRC and determination of their role in the individual stages of CRC. METHODS MMP-9 expression in serum and tissue, and BDNF expression in plasma were detected using the ELISA method. MMP-9 and BDNF in the tissue were also determined by immunohistochemical staining. RESULTS To assess the balance between changes in survival and tumor progression, we compared BDNF/MMP-9 ratios in tissues of living and deceased individuals. The tissue BDNF/MMP-9 ratio (evaluated immunohistochemically) decreased significantly with the progression of the disease in living patients. The BDNF/MMP-9 ratio was statistically significantly reduced in stages II and III compared to the benign group. However, in deceased individuals, the ratio showed an opposite tendency. CONCLUSION The determination of the tissue BDNF/MMP9 ratio can be used as a prognostic biomarker of CRC.
Collapse
Affiliation(s)
- Ivana Večurkovská
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafarik University in Košice, Trieda SNP 1, 040 11 Košice, Slovakia
| | - Jana Mašlanková
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafarik University in Košice, Trieda SNP 1, 040 11 Košice, Slovakia
| | - Vladimíra Tomečková
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafarik University in Košice, Trieda SNP 1, 040 11 Košice, Slovakia
| | - Jana Kaťuchová
- 1st Department of Surgery, Faculty of Medicine, Pavol Jozef Šafarik University in Košice, Trieda SNP 1, 040 11 Košice, Slovakia
| | - Terézia Kisková
- Department of Animal Physiology, Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Šafarik University in Košice, Trieda SNP 1, 040 11 Košice, Slovakia
| | - Lucia Fröhlichová
- Department of Pathology, Louis Pasteur University Hospital, Rastislavova 43, 041 90 Košice, Slovakia
| | - Mária Mareková
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafarik University in Košice, Trieda SNP 1, 040 11 Košice, Slovakia
| | - Marek Stupák
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafarik University in Košice, Trieda SNP 1, 040 11 Košice, Slovakia
| |
Collapse
|
4
|
Christensen A, Grønhøj C, Jensen JS, Lelkaitis G, Kiss K, Juhl K, Charabi BW, Mortensen J, Kjær A, Von Buchwald C. Expression patterns of uPAR, TF and EGFR and their potential as targets for molecular imaging in oropharyngeal squamous cell carcinoma. Oncol Rep 2022; 48:147. [PMID: 35775375 PMCID: PMC9263836 DOI: 10.3892/or.2022.8359] [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: 02/28/2022] [Accepted: 05/05/2022] [Indexed: 11/24/2022] Open
Abstract
The clinical introduction of molecular imaging for the management of oropharyngeal squamous cell carcinoma (OPSCC) relies on the identification of relevant cancer-specific biomarkers. The application of three membrane-bound receptors, namely urokinase-type plasminogen activator receptor (uPAR), tissue factor (TF) and EGFR have been previously explored for targeted imaging and therapeutic strategies in a broad range of solid cancers. The present study aimed to investigate the expression patterns of uPAR, EGFR and TF by immunohistochemistry (IHC) to evaluate their potential for targeted imaging and prognostic value in OPSCC. In a retrospective cohort of 93 patients with primary OPSCC, who were balanced into the 45 human papillomavirus (HPV)-positive and 48 HPV-negative groups, the IHC-determined expression profiles of uPAR, TF and EGFR in large biopsy or tumor resection specimens were analyzed. Using the follow-up data, overall survival (OS) and recurrence-free survival were measured. Specifically, associations between survival outcome, biomarker expression and clinicopathological factors were examined using Cox proportional hazards model and log-rank test following Kaplan-Meier statistics. After comparing the expression pattern of biomarkers within the tumor compartment with that in the adjacent normal tissues, uPAR and TF exhibited a highly tumor-specific expression pattern, whereas EGFR showed a homogeneous expression within the tumor compartment as well as a consistent expression in the normal mucosal epithelium and salivary gland tissues. The positive expression rate of uPAR, TF and EGFR in the tumors was 98.9, 76.3 and 98.9%, respectively. No statistically significant association between biomarker expression and survival outcome could be detected. Higher uPAR expression levels had a trend towards reduced OS according to results from univariate analysis (P=0.07; hazard ratio=2.01; 95% CI=0.92-4.37). Taken together, these results suggest that uPAR, TF and EGFR may be suitable targets for molecular imaging and therapy in OPSCC. In particular, uPAR may be an attractive target owing to their high positive expression rates in tumors and a highly tumor-specific expression pattern.
Collapse
Affiliation(s)
- Anders Christensen
- Department of Otolaryngology, Head & Neck Surgery and Audiology, Rigshospitalet, University of Copenhagen, DK‑2100 Copenhagen, Denmark
| | - Christian Grønhøj
- Department of Otolaryngology, Head & Neck Surgery and Audiology, Rigshospitalet, University of Copenhagen, DK‑2100 Copenhagen, Denmark
| | - Jakob Schmidt Jensen
- Department of Otolaryngology, Head & Neck Surgery and Audiology, Rigshospitalet, University of Copenhagen, DK‑2100 Copenhagen, Denmark
| | - Giedrius Lelkaitis
- Department of Pathology, Rigshospitalet, University of Copenhagen, DK‑2100 Copenhagen, Denmark
| | - Katalin Kiss
- Department of Pathology, Rigshospitalet, University of Copenhagen, DK‑2100 Copenhagen, Denmark
| | - Karina Juhl
- Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet, University of Copenhagen, DK‑2100 Copenhagen, Denmark
| | - Birgitte Wittenborg Charabi
- Department of Otolaryngology, Head & Neck Surgery and Audiology, Rigshospitalet, University of Copenhagen, DK‑2100 Copenhagen, Denmark
| | - Jann Mortensen
- Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet, University of Copenhagen, DK‑2100 Copenhagen, Denmark
| | - Andreas Kjær
- Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet, University of Copenhagen, DK‑2100 Copenhagen, Denmark
| | - Christian Von Buchwald
- Department of Otolaryngology, Head & Neck Surgery and Audiology, Rigshospitalet, University of Copenhagen, DK‑2100 Copenhagen, Denmark
| |
Collapse
|
5
|
Ran Q, Xu D, Wang Q, Wang D. Hypermethylation of the Promoter Region of miR-23 Enhances the Metastasis and Proliferation of Multiple Myeloma Cells via the Aberrant Expression of uPA. Front Oncol 2022; 12:835299. [PMID: 35707350 PMCID: PMC9189361 DOI: 10.3389/fonc.2022.835299] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 05/02/2022] [Indexed: 11/24/2022] Open
Abstract
Multiple myeloma has a long course, with no obvious symptoms in the early stages. However, advanced stages are characterized by injury to the bone system and represent a severe threat to human health. The results of the present work indicate that the hypermethylation of miR-23 promoter mediates the aberrant expression of uPA/PLAU (urokinase plasminogen activator, uPA) in multiple myeloma cells. miR-23, a microRNA that potentially targets uPA’s 3’UTR, was predicted by the online tool miRDB. The endogenous expressions of uPA and miR-23 are related to disease severity in human patients, and the expression of miR-23 is negatively related to uPA expression. The hypermethylation of the promoter region of miR-23 is a promising mechanism to explain the low level of miR-23 or aberrant uPA expression associated with disease severity. Overexpression of miR-23 inhibited the expression of uPA by targeting the 3’UTR of uPA, not only in MM cell lines, but also in patient-derived cell lines. Overexpression of miR-23 also inhibited in vitro and in vivo invasion of MM cells in a nude mouse model. The results therefore extend our knowledge about uPA in MM and may assist in the development of more effective therapeutic strategies for MM treatment.
Collapse
Affiliation(s)
- Qijie Ran
- Department of Hematology, General Hospital of Central Theater Command, Wuhan, China
- *Correspondence: Qijie Ran, ; Dongsheng Wang,
| | - Dehong Xu
- Department of Hematology, General Hospital of Central Theater Command, Wuhan, China
| | - Qi Wang
- Department of Hematology, General Hospital of Central Theater Command, Wuhan, China
| | - Dongsheng Wang
- Department of Neurosurgery, The Fifth People’s Hospital of Dalian, Dalian, China
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian City, China
- *Correspondence: Qijie Ran, ; Dongsheng Wang,
| |
Collapse
|
6
|
Miles LA, Krajewski S, Baik N, Parmer RJ, Mueller BM. Plg-RKT Expression in Human Breast Cancer Tissues. Biomolecules 2022; 12:biom12040503. [PMID: 35454092 PMCID: PMC9028288 DOI: 10.3390/biom12040503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 12/12/2022] Open
Abstract
The plasminogen activation system regulates the activity of the serine protease, plasmin. The role of plasminogen receptors in cancer progression is being increasingly appreciated as key players in modulation of the tumor microenvironment. The interaction of plasminogen with cells to promote plasminogen activation requires the presence of proteins exposing C-terminal lysines on the cell surface. Plg-RKT is a structurally unique plasminogen receptor because it is an integral membrane protein that is synthesized with and binds plasminogen via a C-terminal lysine exposed on the cell surface. Here, we have investigated the expression of Plg-RKT in human breast tumors and human breast cancer cell lines. Breast cancer progression tissue microarrays were probed with anti-Plg-RKT mAB and we found that Plg-RKT is widely expressed in human breast tumors, that its expression is increased in tumors that have spread to draining lymph nodes and distant organs, and that Plg-RKT expression is most pronounced in hormone receptor (HR)-positive tumors. Plg-RKT was detected by Western blotting in human breast cancer cell lines. By flow cytometry, Plg-RKT cell surface expression was highest on the most aggressive tumor cell line. Future studies are warranted to address the functions of Plg-RKT in breast cancer.
Collapse
Affiliation(s)
- Lindsey A. Miles
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA 92037, USA; (L.A.M.); (N.B.)
| | | | - Nagyung Baik
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA 92037, USA; (L.A.M.); (N.B.)
| | - Robert J. Parmer
- Department of Medicine, Veterans Administration San Diego Healthcare System, University of California San Diego, San Diego, CA 92161, USA;
| | - Barbara M. Mueller
- San Diego Biomedical Research Institute, San Diego, CA 92121, USA
- Correspondence:
| |
Collapse
|
7
|
Ma K, Chen X, Liu W, Yang Y, Chen S, Sun J, Ma C, Wang T, Yang J. ANXA2 is correlated with the molecular features and clinical prognosis of glioma, and acts as a potential marker of immunosuppression. Sci Rep 2021; 11:20839. [PMID: 34675316 PMCID: PMC8531374 DOI: 10.1038/s41598-021-00366-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/11/2021] [Indexed: 12/15/2022] Open
Abstract
Recent studies have shown that ANXA2 is important in the development of many cancers, while its role in glioma-related immune response remains unclear. We aimed to comprehensively investigate its biological characteristics and clinical value in glioma. We analyzed 699 glioma samples from The Cancer Genome Atlas as training cohort and 325 samples from the Chinese Glioma Genome Atlas as validation cohort. All the statistical analyses and figures were generated with R. ANXA2 was overexpressed significantly in high-grade glioma, isocitrate dehydrogenase wild-type and mesenchymal-subtype glioma. ANXA2 was a special indicator of mesenchymal subtype. The survival analysis showed that highly-expressed ANXA2 was related to worse survival status as an independent factor of poor prognosis. Further gene ontology analysis showed that ANXA2 was mainly involved in immune response and inflammatory activities of glioma. Subsequent correlation analysis showed that ANXA2 was positively correlated with HCK, LCK, MHC II, STAT1 and interferon but negatively with IgG. Meanwhile, ANXA2 was positively related to the infiltration of tumor-related macrophages, regulatory T cells and myeloid-derived suppressor cells. Our study revealed that ANXA2 is a biomarker closely related to the malignant phenotype and poor prognosis of glioma, and plays an important role in immune response, inflammatory activity and immunosuppression.
Collapse
Affiliation(s)
- Kaiming Ma
- Department of Neurosurgery, Peking University Third Hospital, 49 North Garden Rd, Haidian District, Beijing, 100191, China
- Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Beijing, China
| | - Xin Chen
- Department of Neurosurgery, Peking University Third Hospital, 49 North Garden Rd, Haidian District, Beijing, 100191, China
- Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Beijing, China
| | - Weihai Liu
- Department of Neurosurgery, Peking University Third Hospital, 49 North Garden Rd, Haidian District, Beijing, 100191, China
- Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Beijing, China
| | - Yang Yang
- Department of Neurosurgery, Peking University Third Hospital, 49 North Garden Rd, Haidian District, Beijing, 100191, China
- Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Beijing, China
| | - Suhua Chen
- Department of Neurosurgery, Peking University Third Hospital, 49 North Garden Rd, Haidian District, Beijing, 100191, China
- Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Beijing, China
| | - Jianjun Sun
- Department of Neurosurgery, Peking University Third Hospital, 49 North Garden Rd, Haidian District, Beijing, 100191, China
- Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Beijing, China
| | - Changcheng Ma
- Department of Neurosurgery, Peking University Third Hospital, 49 North Garden Rd, Haidian District, Beijing, 100191, China
- Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Beijing, China
| | - Tao Wang
- Department of Neurosurgery, Peking University Third Hospital, 49 North Garden Rd, Haidian District, Beijing, 100191, China
- Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Beijing, China
| | - Jun Yang
- Department of Neurosurgery, Peking University Third Hospital, 49 North Garden Rd, Haidian District, Beijing, 100191, China.
- Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Beijing, China.
| |
Collapse
|
8
|
Abstract
Plasminogen is an abundant plasma protein that exists in various zymogenic forms. Plasmin, the proteolytically active form of plasminogen, is known for its essential role in fibrinolysis. To date, therapeutic targeting of the fibrinolytic system has been for 2 purposes: to promote plasmin generation for thromboembolic conditions or to stop plasmin to reduce bleeding. However, plasmin and plasminogen serve other important functions, some of which are unrelated to fibrin removal. Indeed, for >40 years, the antifibrinolytic agent tranexamic acid has been administered for its serendipitously discovered skin-whitening properties. Plasmin also plays an important role in the removal of misfolded/aggregated proteins and can trigger other enzymatic cascades, including complement. In addition, plasminogen, via binding to one of its dozen cell surface receptors, can modulate cell behavior and further influence immune and inflammatory processes. Plasminogen administration itself has been reported to improve thrombolysis and to accelerate wound repair. Although many of these more recent findings have been derived from in vitro or animal studies, the use of antifibrinolytic agents to reduce bleeding in humans has revealed additional clinically relevant consequences, particularly in relation to reducing infection risk that is independent of its hemostatic effects. The finding that many viruses harness the host plasminogen to aid infectivity has suggested that antifibrinolytic agents may have antiviral benefits. Here, we review the broadening role of the plasminogen-activating system in physiology and pathophysiology and how manipulation of this system may be harnessed for benefits unrelated to its conventional application in thrombosis and hemostasis.
Collapse
|