1
|
Wei S, Zhou J, Dong B. A novel risk model consisting of nine platelet-related gene signatures for predicting prognosis, immune features and drug sensitivity in glioma. Hereditas 2024; 161:52. [PMID: 39707577 DOI: 10.1186/s41065-024-00355-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Glioma is a malignancy with challenging clinical treatment and poor prognosis. Platelets are closely associated with tumor growth, propagation, invasion, and angiogenesis. However, the role of platelet-related genes in glioma treatment and prognosis remains unclear. RESULTS A prognostic risk model was established using nine platelet-related prognostic signature genes (CAPG, CLIC1, GLB1, GNG12, KIF20A, PDIA4, SULF2, TAGLN2, and WEE1), and the risk score of samples were calculated. Subsequently, the glioma samples were divided into high- and low-risk groups based on the median values of risk scores. scRNA-seq analysis revealed that the prognostic genes were primarily located in astrocytes and natural killer cells. The immune infiltration proportions of most immune cells differed significantly between high- and low-risk groups. Moreover, we found AZD7762 as a potential candidate for glioma treatment. CONCLUSION Nine platelet-related prognostic genes identified as prognostic signatures for glioma were closely associated with the TME and may aid in directing the clinical treatment and prognosis of gliomas.
Collapse
Affiliation(s)
- Sanlin Wei
- Dalian Medical University, Dalian, Liaoning Province, 116000, China
- Department of Neurosurgery, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116000, China
| | - Junke Zhou
- Department of Nephrology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116000, China
| | - Bin Dong
- Dalian Medical University, Dalian, Liaoning Province, 116000, China.
- Department of Neurosurgery, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116000, China.
| |
Collapse
|
2
|
Mei Y, Lv Q, Tan Z, Zhang Z, Ji Y, Chen S, Shen X. Decapping enzyme 2 is a novel immune-related biomarker that predicts poor prognosis in glioma. Biotechnol Genet Eng Rev 2024; 40:4262-4283. [PMID: 37191010 DOI: 10.1080/02648725.2023.2209409] [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: 02/21/2023] [Accepted: 04/27/2023] [Indexed: 05/17/2023]
Abstract
This study analyzed sequencing and clinical data from the Cancer Genome Atlas (TCGA) and gene expression synthesis, and used Chinese glioma Genome Atlas (CGGA) data for external validation. The expression of DCP2 in normal brain and tumor tissue was compared. We analyzed the clinical and molecular characteristics and prognostic value of DCP2 in glioma. In addition, DCP2 expression levels were evaluated in 30 glioma tissue samples and upregulated in glioma samples compared to normal brain tissue (p < 0.001). Multivariate data analysis from TCGA showed that increased DCP2 expression was an independent risk factor for overall survival and prognosis of glioma patients. As indicated by the analysis of the TCGA data set. The expression level of DCP2 is closely related to tumor immunity, including tumor immune cell infiltration, immune score, and co-expression of multiple immune-related genes. In addition, DCP2 was positively correlated with IL-6 and IL-7. Glioma cell proliferation and invasion were evaluated using cell viability, colony formation, wound healing, and transwell assays.Apoptosis and cell cycle were detected by flow cytometry. DCP2 promoted the proliferation, invasion and migration of glioma cells T98G and U251, inhibited apoptosis and blocked the S phase of the cell cycle. As a result of the altered expression of DCP2, a new prognostic biomarker may be identified that can improve patient survival.These findings suggest DCP2 as a potential biomarker for the prognosis of glioma and a candidate immunotherapy target.
Collapse
Affiliation(s)
- Yuran Mei
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Key Laboratory of Translational Cancer Research, Jiangxi Cancer Hospital, Nanchang, China
| | - Qiaoli Lv
- Jiangxi Key Laboratory of Translational Cancer Research, Jiangxi Cancer Hospital, Nanchang, China
| | - Zilong Tan
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhe Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yulong Ji
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shuhui Chen
- Department of Radiation Oncology, Jiangxi Cancer Hospital of Nanchang University, Nanchang, China
| | - Xiaoli Shen
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
3
|
Feng S, Zhu L, Qin Y, Kou K, Liu Y, Zhang G, Wang Z, Lu H, Sun R. Machine learning model reveals the role of angiogenesis and EMT genes in glioma patient prognosis and immunotherapy. Biol Direct 2024; 19:113. [PMID: 39533401 PMCID: PMC11555840 DOI: 10.1186/s13062-024-00565-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024] Open
Abstract
Gliomas represent a highly aggressive class of tumors located in the brain. Despite the availability of multiple treatment modalities, the prognosis for patients diagnosed with glioma remains unfavorable. Therefore, further exploration of new biomarkers is crucial to enhance the prognostic assessment of glioma and to investigate more effective treatment options. In this research, we utilized multiple machine learning techniques to assess the significance of genes related to angiogenesis and epithelial-mesenchymal transition (EMT) in the context of prognosis and treatment for glioma patients. The random forest algorithm highlighted the significance of CALU, and further analysis indicated that the effect of CALU on glioma progression may be regulated by MYC. Different machine learning approaches were employed in our investigation to uncover crucial genes associated with angiogenesis and EMT in glioma. Our findings verify the connection between these genes and the prognosis of patients with glioma, as well as the results of immunotherapeutic interventions. Notably, through experimental verification, we identified CALU as a new prognostic marker for glioma, and inhibiting the expression of CALU can impede the progression of glioma.
Collapse
Affiliation(s)
- Suyin Feng
- Department of Neurosurgery, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214062, China
- Department of Neurosurgery, Donghai County People's Hospital, Lianyungang, Jiangsu, 222000, China
- Neuroscience Center, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
- Donghai County People's Hospital - Jiangnan University Smart Healthcare Joint Laboratory, Donghai County People's Hospital, Lianyungang, Jiangsu, 222000, China
- Cardio-Cerebral Vascular Disease Prevention and Treatment Innovation Center, Donghai County People's Hospital, Lianyungang, Jiangsu, 222000, China
| | - Long Zhu
- Department of Neurosurgery, Donghai County People's Hospital, Lianyungang, Jiangsu, 222000, China
- Donghai County People's Hospital - Jiangnan University Smart Healthcare Joint Laboratory, Donghai County People's Hospital, Lianyungang, Jiangsu, 222000, China
- Cardio-Cerebral Vascular Disease Prevention and Treatment Innovation Center, Donghai County People's Hospital, Lianyungang, Jiangsu, 222000, China
| | - Yan Qin
- Department of Pathology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Kun Kou
- Department of Neurosurgery, Donghai County People's Hospital, Lianyungang, Jiangsu, 222000, China
| | - Yongtai Liu
- Department of Neurosurgery, Donghai County People's Hospital, Lianyungang, Jiangsu, 222000, China
| | - Guangmin Zhang
- Department of Neurosurgery, Donghai County People's Hospital, Lianyungang, Jiangsu, 222000, China
| | - Ziheng Wang
- The School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia.
- MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau SAR, 999078, China.
| | - Hua Lu
- Department of Neurosurgery, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214062, China.
| | - Runfeng Sun
- Donghai County People's Hospital - Jiangnan University Smart Healthcare Joint Laboratory, Donghai County People's Hospital, Lianyungang, Jiangsu, 222000, China.
- Cardio-Cerebral Vascular Disease Prevention and Treatment Innovation Center, Donghai County People's Hospital, Lianyungang, Jiangsu, 222000, China.
| |
Collapse
|
4
|
John Hamilton A, Lane S, Werry EL, Suri A, Bailey AW, Mercé C, Kadolsky U, Payne AD, Kassiou M, Treiger Sredni S, Saxena A, Gunosewoyo H. Synthesis and Antitumour Evaluation of Tricyclic Indole-2-Carboxamides against Paediatric Brain Cancer Cells. ChemMedChem 2024; 19:e202400098. [PMID: 38923350 DOI: 10.1002/cmdc.202400098] [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: 02/01/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 06/28/2024]
Abstract
Antitumour properties of some cannabinoids (CB) have been reported in the literature as early as 1970s, however there is no clear consensus to date on the exact mechanisms leading to cancer cell death. The indole-based WIN 55,212-2 and SDB-001 are both known as potent agonists at both CB1 and CB2 receptors, yet we demonstrate herein that only the former can exert in vitro antitumour effects when tested against a paediatric brain cancer cell line KNS42. In this report, we describe the synthesis of novel 3,4-fused tricyclic indoles and evaluate their functional potencies at both cannabinoid receptors, as well as their abilities to inhibit the growth or proliferation of KNS42 cells. Compared to our previously reported indole-2-carboxamides, these 3,4-fused tricyclic indoles had either completely lost activities, or, showed moderate-to-weak antagonism at both CB1 and CB2 receptors. Compound 23 displayed the most potent antitumour properties among the series. Our results further support the involvement of non-CB pathways for the observed antitumour activities of amidoalkylindole-based cannabinoids, in line with our previous findings. Transcriptomic analysis comparing cells treated or non-treated with compound 23 suggested the observed antitumour effects of 23 are likely to result mainly from disruption of the FOXM1-regulated cell cycle pathways.
Collapse
Affiliation(s)
| | - Samuel Lane
- School of Chemistry, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Eryn L Werry
- School of Chemistry, The University of Sydney, Sydney, NSW, 2006, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney NSW, 2006, Australia
| | - Amreena Suri
- Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, 60611, USA
| | - Anders W Bailey
- Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, 60611, USA
| | | | | | - Alan D Payne
- School of Molecular and Life Sciences, Curtin University, Bentley, WA, 6102, Australia
| | - Michael Kassiou
- School of Chemistry, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Simone Treiger Sredni
- Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, 60611, USA
- Department of Surgery, Northwestern University, Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Alka Saxena
- Genomics WA, QEII Campus, Nedlands, WA, 6009, Australia
| | - Hendra Gunosewoyo
- Curtin Medical School, Faculty of Health Sciences, Curtin University, Bentley, WA, 6102, Australia
| |
Collapse
|
5
|
Bu S, Singh A, Nguyen HC, Peddi B, Bhatt K, Ravendranathan N, Frisbee JC, Singh KK. Protein Disulfide Isomerase 4 Is an Essential Regulator of Endothelial Function and Survival. Int J Mol Sci 2024; 25:3913. [PMID: 38612722 PMCID: PMC11011381 DOI: 10.3390/ijms25073913] [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: 12/23/2023] [Revised: 03/29/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
Endothelial autophagy plays an important role in the regulation of endothelial function. The inhibition of endothelial autophagy is associated with the reduced expression of protein disulfide isomerase 4 (PDIA-4); however, its role in endothelial cells is not known. Here, we report that endothelial cell-specific loss of PDIA-4 leads to impaired autophagic flux accompanied by loss of endothelial function and apoptosis. Endothelial cell-specific loss of PDIA-4 also induced marked changes in endothelial cell architecture, accompanied by the loss of endothelial markers and the gain of mesenchymal markers consistent with endothelial-to-mesenchymal transition (EndMT). The loss of PDIA-4 activated TGFβ-signaling, and inhibition of TGFβ-signaling suppressed EndMT in PDIA-4-silenced endothelial cells in vitro. Our findings help elucidate the role of PDIA-4 in endothelial autophagy and endothelial function and provide a potential target to modulate endothelial function and/or limit autophagy and EndMT in (patho-)physiological conditions.
Collapse
Affiliation(s)
- Shuhan Bu
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, 1151 Richmond St. N., London, ON N6A 3K7, Canada; (S.B.); (A.S.); (H.C.N.); (B.P.); (K.B.); (N.R.); (J.C.F.)
| | - Aman Singh
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, 1151 Richmond St. N., London, ON N6A 3K7, Canada; (S.B.); (A.S.); (H.C.N.); (B.P.); (K.B.); (N.R.); (J.C.F.)
| | - Hien C. Nguyen
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, 1151 Richmond St. N., London, ON N6A 3K7, Canada; (S.B.); (A.S.); (H.C.N.); (B.P.); (K.B.); (N.R.); (J.C.F.)
- Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 3K7, Canada
| | - Bharatsinai Peddi
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, 1151 Richmond St. N., London, ON N6A 3K7, Canada; (S.B.); (A.S.); (H.C.N.); (B.P.); (K.B.); (N.R.); (J.C.F.)
| | - Kriti Bhatt
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, 1151 Richmond St. N., London, ON N6A 3K7, Canada; (S.B.); (A.S.); (H.C.N.); (B.P.); (K.B.); (N.R.); (J.C.F.)
| | - Naresh Ravendranathan
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, 1151 Richmond St. N., London, ON N6A 3K7, Canada; (S.B.); (A.S.); (H.C.N.); (B.P.); (K.B.); (N.R.); (J.C.F.)
| | - Jefferson C. Frisbee
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, 1151 Richmond St. N., London, ON N6A 3K7, Canada; (S.B.); (A.S.); (H.C.N.); (B.P.); (K.B.); (N.R.); (J.C.F.)
| | - Krishna K. Singh
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, 1151 Richmond St. N., London, ON N6A 3K7, Canada; (S.B.); (A.S.); (H.C.N.); (B.P.); (K.B.); (N.R.); (J.C.F.)
- Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 3K7, Canada
| |
Collapse
|
6
|
Hu X, Ma Y, Xia Y, Liu B. Integrated analysis of necroptosis related gene signature to predict clinical outcomes, immune status and drug sensitivity in lower grade Glioma. Heliyon 2024; 10:e23947. [PMID: 38192784 PMCID: PMC10772728 DOI: 10.1016/j.heliyon.2023.e23947] [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: 05/13/2023] [Revised: 11/05/2023] [Accepted: 12/16/2023] [Indexed: 01/10/2024] Open
Abstract
Background The treatment of lower grade gliomas (LGG) is currently the most challenging dilemma in the management of intracranial tumors. Necroptosis is a type of programmed cell death that is closely associated with tumor progression, However, the role of necroptosis related genes in LGG is not yet well elucidated. Methods Online databases were used to obtain gene expression and clinical information. After gene differential expression analysis, a risk score model based on prognostic differentially expressed necroptosis-related genes (DENGs) were constructed to predict prognosis for LGG patients. The validity of the risk score model was then assessed with Kaplan-Meier survival curve. The prognostic DENGs included in the risk score model were then subjected to gene expression analysis, functional enrichment analysis, consensus clustering analysis, and single cell sequencing analysis. Finally, we investigated the correlation of the risk score and immune infiltration in LGG tumor microenvironment and drug sensitivity for LGG patients in different risk groups. Results A survival risk score model was constructed based on seven prognostic DENGs, which demonstrated satisfactory performance in predicting the prognosis of LGG patients. According to functional enrichment analyses, these seven DENGs may play a regulatory role in LGG tumorigenesis through several immune and metabolic pathways. LGG patients could be categorized into two clusters with distinct prognosis and clinicopathologic characteristics based on the expression of seven DENGs. Single-cell sequencing analysis demonstrated that the DENG signature was differentially expressed in various types of cells in LGG and may play a vital role in oncogenesis. Additionally, drug sensitivity analysis suggested that the seven-gene signature could guide clinical medication for LGG patients. Conclusion Our study developed a reliable necroptosis-related signature to predict the prognosis of LGG patients. This gene signature may also help estimate immune status and anti-cancer drug sensitivity in LGG patients. Our findings may pave the way to enhance our understanding of necroptosis in LGG.
Collapse
Affiliation(s)
- Xiqi Hu
- Department of Neurosurgery, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, 570100, China
| | - Yanan Ma
- Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570100, China
| | - Ying Xia
- Department of Neurosurgery, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, 570100, China
| | - Bo Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410000, China
| |
Collapse
|
7
|
Kisling SG, Atri P, Shah A, Cox JL, Sharma S, Smith LM, Ghersi D, Batra SK. A Novel HOXA10-Associated 5-Gene-Based Prognostic Signature for Stratification of Short-term Survivors of Pancreatic Ductal Adenocarcinoma. Clin Cancer Res 2023; 29:3759-3770. [PMID: 37432996 PMCID: PMC10529249 DOI: 10.1158/1078-0432.ccr-23-0825] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 06/02/2023] [Accepted: 07/06/2023] [Indexed: 07/13/2023]
Abstract
PURPOSE Despite the significant association of molecular subtypes with poor prognosis in patients with pancreatic ductal adenocarcinoma (PDAC), few efforts have been made to identify the underlying pathway(s) responsible for this prognosis. Identifying a clinically relevant prognosis-based gene signature may be the key to improving patient outcomes. EXPERIMENTAL DESIGN We analyzed the transcriptomic profiles of treatment-naïve surgically resected short-term survivor (STS) and long-term survivor (LTS) tumors (GSE62452) for expression and survival, followed by validation in several datasets. These results were corroborated by IHC analysis of PDAC-resected STS and LTS tumors. The mechanism of this differential survival was investigated using CIBERSORT and pathway analyses. RESULTS We identified a short-surviving prognostic subtype of PDAC with a high degree of significance (P = 0.018). One hundred thirty genes in this novel subtype were found to be regulated by a master regulator, homeobox gene HOXA10, and a 5-gene signature derived from these genes, including BANF1, EIF4G1, MRPS10, PDIA4, and TYMS, exhibited differential expression in STSs and a strong association with poor survival. This signature was further associated with the proportion of T cells and macrophages found in STSs and LTSs, demonstrating a potential role in PDAC immunosuppression. Pathway analyses corroborated these findings, revealing that this HOXA10-driven prognostic signature is associated with immune suppression and enhanced tumorigenesis. CONCLUSIONS Overall, these findings reveal the presence of a HOXA10-associated prognostic subtype that can be used to differentiate between STS and LTS patients of PDAC and inform on the molecular interactions that play a role in this poor prognosis.
Collapse
Affiliation(s)
- Sophia G. Kisling
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, NE, 68198, USA
| | - Pranita Atri
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, NE, 68198, USA
| | - Ashu Shah
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, NE, 68198, USA
| | - Jesse L. Cox
- Department of Pathology and Microbiology, University of Nebraska Medical Center, NE, 68198, USA
| | - Sunandini Sharma
- Department of Pathology and Microbiology, University of Nebraska Medical Center, NE, 68198, USA
| | - Lynette M. Smith
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, NE, 68198, USA
| | - Dario Ghersi
- School of Interdisciplinary Informatics, College of Information Science & Technology, University of Nebraska Omaha, NE, 68182, USA
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, NE, 68198, USA
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, NE, 68198, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, NE, 68198, USA
| |
Collapse
|
8
|
Shikhevich S, Chadaeva I, Khandaev B, Kozhemyakina R, Zolotareva K, Kazachek A, Oshchepkov D, Bogomolov A, Klimova NV, Ivanisenko VA, Demenkov P, Mustafin Z, Markel A, Savinkova L, Kolchanov NA, Kozlov V, Ponomarenko M. Differentially Expressed Genes and Molecular Susceptibility to Human Age-Related Diseases. Int J Mol Sci 2023; 24:ijms24043996. [PMID: 36835409 PMCID: PMC9966505 DOI: 10.3390/ijms24043996] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/02/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
Mainstream transcriptome profiling of susceptibility versus resistance to age-related diseases (ARDs) is focused on differentially expressed genes (DEGs) specific to gender, age, and pathogeneses. This approach fits in well with predictive, preventive, personalized, participatory medicine and helps understand how, why, when, and what ARDs one can develop depending on their genetic background. Within this mainstream paradigm, we wanted to find out whether the known ARD-linked DEGs available in PubMed can reveal a molecular marker that will serve the purpose in anyone's any tissue at any time. We sequenced the periaqueductal gray (PAG) transcriptome of tame versus aggressive rats, identified rat-behavior-related DEGs, and compared them with their known homologous animal ARD-linked DEGs. This analysis yielded statistically significant correlations between behavior-related and ARD-susceptibility-related fold changes (log2 values) in the expression of these DEG homologs. We found principal components, PC1 and PC2, corresponding to the half-sum and the half-difference of these log2 values, respectively. With the DEGs linked to ARD susceptibility and ARD resistance in humans used as controls, we verified these principal components. This yielded only one statistically significant common molecular marker for ARDs: an excess of Fcγ receptor IIb suppressing immune cell hyperactivation.
Collapse
Affiliation(s)
- Svetlana Shikhevich
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
| | - Irina Chadaeva
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
| | - Bato Khandaev
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
- The Natural Sciences Department, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Rimma Kozhemyakina
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
| | - Karina Zolotareva
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
- The Natural Sciences Department, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Anna Kazachek
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
- The Natural Sciences Department, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Dmitry Oshchepkov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
- The Natural Sciences Department, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Anton Bogomolov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
- The Natural Sciences Department, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Natalya V. Klimova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
| | - Vladimir A. Ivanisenko
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
- The Natural Sciences Department, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Pavel Demenkov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
| | - Zakhar Mustafin
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
- The Natural Sciences Department, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Arcady Markel
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
- The Natural Sciences Department, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Ludmila Savinkova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
| | - Nikolay A. Kolchanov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
- The Natural Sciences Department, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Vladimir Kozlov
- Research Institute of Fundamental and Clinical Immunology (RIFCI) SB RAS, Novosibirsk 630099, Russia
| | - Mikhail Ponomarenko
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia
- Correspondence: ; Tel.: +7-(383)-363-4963 (ext. 1311)
| |
Collapse
|
9
|
Wang NC, Chen HW, Lin TY. Association of protein disulfide isomerase family A, member 4, and inflammation in people living with HIV. Int J Infect Dis 2023; 126:79-86. [PMID: 36375691 DOI: 10.1016/j.ijid.2022.11.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 10/25/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES Protein disulfide isomerase (PDI) family members are specific endoplasmic reticulum proteins associated with inflammation, obesity, and cancer. In HIV infection, the role of PDI family A, member 4 (PDIA4), is unclear. This study aimed to clarify the association between plasma PDIA4 levels and inflammation in people living with HIV (PLWH). METHODS In this study, 287 PLWH and 74 healthy participants were enrolled. The plasma PDIA4 values, demographic data, laboratory data, and other inflammatory markers were recorded. The association between PDIA4 level and inflammatory extent was analyzed using logistic regression and Spearman rank-order correlations. Other results were analyzed using Student's t-test or chi-square test. RESULTS In PLWH, the PDIA4 levels were positively associated with the inflammatory markers, interleukin 6 (r = 0.209, p = 0.001), and tumor necrosis factor-α (r = 0.162, p = 0.01) levels, but not with high-sensitivity C-reactive protein levels. Moreover, the plasma PDIA4 level of PLWH decreased after anti-viral treatment (p = 0.0001). CONCLUSION Plasma PDIA4 levels are closely associated with inflammation in PLWH and have a positive correlation with the viral load during anti-viral therapy.
Collapse
Affiliation(s)
- Ning-Chi Wang
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Hsuan-Wei Chen
- Division of Gastroenterology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Te-Yu Lin
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
10
|
Liu Y, He H, Song Z, Liu Z, Zhu K. Lin-28 Homolog B-Activated Protein Disulfide Isomerase A4 Regulates Cell Proliferation, Migration and Invasion of Glioma. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The goal of this study is to elucidate the role of protein disulfide isomerase A4 (PDIA4) in glioma, as well as its regulatory mechanism. Cell transfection was performed to adjust the expression level of PDIA4 and RNA-binding protein lin-28 homolog B (LIN28B). The expression of PDIA4
in human astrocytes and glioma cell lines was determined by quantitative real-time PCR and western blot. CCK-8, colony formation, Transwell and wound-healing assays were applied to determine the capabilities of cells to proliferate, invade and migrate. The connection between PDIA4 and LIN28B
was demonstrated by RNA immunoprecipitation (RIP) and RNA pull down assays. As a result, PDIA4 was elevated in glioma. PDIA4 depletion hugely suppressed cell proliferative ability, which was characterized by the reduced cell viability and colony formation, and declined contents of PCNA and
Ki67. Meanwhile, PDIA4 knockdown repressed the cell capabilities to migrate and invade, accompanied with downregulated MMP2 and MMP9. LIN28N was also found to be upregulated in glioma cells, and was verified to bind with PDIA4 and positively regulate PDIA4 expression. Additionally, LIN28B
overexpression partly hindered the suppressive impacts of PDIA4 knockdown on cell abilities to proliferate, migrate and invade. In conclusion, this study delineates that LIN28B-mediated PDIA4 plays a critical role in the progression of glioma.
Collapse
Affiliation(s)
- Yang Liu
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, P. R. China
| | - Hua He
- Department of Radiology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, P. R. China
| | - Zimu Song
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, P. R. China
| | - Zheng Liu
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, P. R. China
| | - Kai Zhu
- Department of Radiology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, 750004, P. R. China
| |
Collapse
|
11
|
Xing F, Song Z, Cheng Z. High expression of PDIA4 promotes malignant cell behavior and predicts reduced survival in cervical cancer. Oncol Rep 2022; 48:184. [PMID: 36082822 PMCID: PMC9478991 DOI: 10.3892/or.2022.8399] [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: 06/24/2022] [Accepted: 08/11/2022] [Indexed: 11/08/2022] Open
Abstract
The protein disulfide isomerase (PDI) gene family plays important roles in the maintenance of several cellular functions. Previous studies have showed that protein disulfide isomerase family A member 4 (PDIA4) is aberrantly expressed in several types of cancer, and correlates with prognosis of patients. However, the role of PDIA4 in cervical cancer remains unclear. In the present study, the expression pattern of PDIA4 from both public database and immunohistochemical analysis in cervical samples was analyzed. Cell Counting Kit-8 and Transwell assays were performed to determine the effect of PDIA4 on cervical cancer cell proliferation and migration. Gene set enrichment analysis (GSEA) was used to provide the associated enriched pathways of PDIA4 in regulating cervical tumorigenesis. It was observed that mRNA expression and protein level of PDIA4 were upregulated in cervical cancer tissues. High expression of PDIA4 was significantly associated with poor overall survival (P=0.0095) and relapse-free survival (P=0.0019) in The Cancer Genome Atlas cohort. Knockdown of PDIA4 inhibited cervical cancer cell proliferation and migration. Moreover, PDIA4 affected the expression of proliferation-related molecules (cyclin D1 and PCNA) and migration-related molecules (E-cadherin and Vimentin). Additionally, GSEA revealed that PDIA4 was significantly associated with gene signatures involving glycan biosynthesis, glycosaminoglycan degradation and protein export. In conclusion, the present findings highlighted the importance of PDIA4 in cervical oncogenesis, and suggested that targeting PDIA4 may be a potential therapeutic application for cervical cancer.
Collapse
Affiliation(s)
- Feng Xing
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, School of Clinical Medicine of Nanjing Medical University, Shanghai 200072, P.R. China
| | - Zhijiao Song
- Department of Obstetrics and Gynecology, Zhabei Central Hospital of Jing'an, Shanghai 200070, P.R. China
| | - Zhongping Cheng
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, School of Clinical Medicine of Nanjing Medical University, Shanghai 200072, P.R. China
| |
Collapse
|
12
|
Guo K, Duan X, Zhao J, Sun B, Liu X, Zhao Z. A novel necroptosis-related gene signature for predict prognosis of glioma based on single-cell and bulk RNA sequencing. Front Mol Biosci 2022; 9:984712. [PMID: 36111134 PMCID: PMC9469195 DOI: 10.3389/fmolb.2022.984712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Glioma is the most fatal neoplasm among the primary intracranial cancers. Necroptosis, a form of programmed cell death, is correlated with tumor progression and immune response. But, the role of necroptosis-related genes (NRGs) in glioma has not been well-uncovered.Methods: Single-cell and bulk RNA sequencing data, obtained from publicly accessed databases, were used to establish a necroptosis-related gene signature for predicting the prognosis of glioma patients. Multiple bioinformatics algorithms were conducted to evaluate the efficacy of the signature. The relative mRNA level of each signature gene was validated by quantitative real-time reverse transcription PCR (qRT-PCR) in glioma cell lines compared to human astrocytes.Results: In this predicted prognosis model, patients with a high risk score showed a shorter overall survival, which was verified in the testing cohorts. The signature risk score was positively related with immune cell infiltration and some immune check points, such as CD276 (B7-H3), CD152 (CTLA-4), CD223 (LAG-3), and CD274 (PD-L1). Single-cell RNA sequencing analysis confirmed that the glioma microenvironment consists of various immune cells with different markers. The eight NRGs of the signature were detected to be expressed in several immune cells. QRT-PCR results verified that all the eight signature genes were differentially expressed between human astrocytes and glioma cells.Conclusion: The eight NRGs correlate with the immune microenvironment of glioma according to our bioinformatics analysis. This necroptosis-related gene signature may evaluate the precise methodology of predicting prognosis of glioma and provide a novel thought in glioma investigation.
Collapse
Affiliation(s)
- Kai Guo
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Department of Neurosurgery, Affiliated Xing Tai People Hospital of Hebei Medical University, Xingtai, China
| | - Xinxin Duan
- Department of Oncology, Hebei General Hospital, Shijiazhuang, China
- Graduate School, North China University of Science and Technology, Tangshan, China
| | - Jiahui Zhao
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Boyu Sun
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaoming Liu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zongmao Zhao
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- *Correspondence: Zongmao Zhao,
| |
Collapse
|
13
|
Bioinformatics analysis of LMAN1 expression, clinical characteristics, and its effects on cell proliferation and invasion in glioma. Brain Res 2022; 1789:147952. [PMID: 35623391 DOI: 10.1016/j.brainres.2022.147952] [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: 03/09/2022] [Revised: 05/11/2022] [Accepted: 05/21/2022] [Indexed: 11/20/2022]
Abstract
Glioma is the most common primary central nervous system malignant tumor with high heterogeneity and poor prognosis. So far, the complex pathological process of glioma has not been fully elucidated, and there is a lack of effective biomarkers for the diagnosis and molecular targeted therapy of glioma. Using bioinformatics methods, 77 upregulated and 89 downregulated differentially expressed genes (DEGs) were detected by intersection analysis in different gene expression datasets of glioma cases from public databases. Then, GO and KEGG pathway analysis revealed that the biological functions of these upregulated DEGs were mainly focused on immune response, and the signaling pathways were mainly enriched in integrin family cell surface interactions. The overexpression of the LMAN1 gene of interest was then confirmed using the TCGA dataset and further verified by qRT-PCR in 29 clinical samples and 5 glioma cell lines. Furthermore, high expression of LMAN1 was found to be associated with higher WHO grade, IDH status, and 1p/19q co-deletion. Survival analysis showed that high expression of LMAN1 was associated with poor prognosis in glioma. Gene set enrichment analysis (GSEA) indicated that many cancer-related pathways were associated with LMAN1-high phenotype. Protein-protein interaction (PPI) analysis revealed significant interaction between LMAN1 and MCFD2, F8, and TMED10. Finally, cell experiments showed that LMAN1 knockdown significantly inhibited the proliferation, migration and invasion and promoted apoptosis in glioma cells. This study highlighted the malignant role of LMAN1 in gliomas and provided a potentially valuable biomarker for prognosis evaluation and molecular targeted therapy of glioma.
Collapse
|
14
|
Huang R, Li G, Wang K, Wang Z, Zeng F, Hu H, Jiang T. Comprehensive Analysis of the Clinical and Biological Significances of Endoplasmic Reticulum Stress in Diffuse Gliomas. Front Cell Dev Biol 2021; 9:619396. [PMID: 34307339 PMCID: PMC8301220 DOI: 10.3389/fcell.2021.619396] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 06/21/2021] [Indexed: 12/14/2022] Open
Abstract
Background As a critical organelle for protein and lipid synthesis, the dysfunction of endoplasmic reticulum has a significant impact on multiple biological processes of cells. Thus, in this study, we constructed an ER stress-related risk signature to investigate the functional roles of ER stress in gliomas. Methods A total of 626 samples from TCGA RNA-seq dataset (training cohort) and 310 samples from CGGA RNA-seq dataset (validation cohort) were enrolled in this study. Clinical information and genomic profiles were also obtained. The ER stress signature was developed by the LASSO regression model. The prognostic value of the risk signature was evaluated by Cox regression, Kaplan-Meier and ROC Curve analyses. Bioinformatics analysis and experiment in vitro were performed to explore the biological implication of this signature. Results We found that the ER stress-related signature was tightly associated with major clinicopathological features and genomic alterations of gliomas. Kaplan-Meier curve and Cox regression analysis indicated that ER stress activation was an independent prognostic factor for patients with glioma. Besides, we also constructed an individualized prognosis prediction model through Nomogram and ROC Curve analysis. Bioinformatics analysis suggested that ER stress activation also promoted the malignant progression of glioma and participated in the regulation of tumor immune microenvironment, especially the infiltration of macrophages in M2 phase. These results were further validated in IHC analysis and cell biology experiments. Conclusion The ER stress activation had a high prognostic value and could serve as a promising target for developing individualized treatment of glioma.
Collapse
Affiliation(s)
- Ruoyu Huang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Chinese Glioma Cooperative Group (CGCG), Beijing, China
| | - Guanzhang Li
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Chinese Glioma Cooperative Group (CGCG), Beijing, China
| | - Kuanyu Wang
- Chinese Glioma Cooperative Group (CGCG), Beijing, China.,Department of Gamma Knife Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Zhiliang Wang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Chinese Glioma Cooperative Group (CGCG), Beijing, China
| | - Fan Zeng
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Cooperative Group (CGCG), Beijing, China
| | - Huimin Hu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Cooperative Group (CGCG), Beijing, China
| | - Tao Jiang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Chinese Glioma Cooperative Group (CGCG), Beijing, China
| |
Collapse
|