1
|
Ma S, Zhao N, Dong X, Wang Y, Song L, Zheng R, Zhi X, Ma C, Cheng S, Li J, Liu Y, Xiao T. Liquid biopsy-derived extracellular vesicle protein biomarkers for diagnosis and prognostic assessment of lung squamous cell carcinoma. Cancer Cell Int 2025; 25:161. [PMID: 40275246 PMCID: PMC12023671 DOI: 10.1186/s12935-025-03792-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 04/17/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND For patients with nodules detected in imaging that are indeterminate for malignancy, achieving accurate, early, and non-invasive diagnosis of Lung Squamous Cell Carcinoma (LUSC) remains a significant challenge. Therefore, we aimed to establish diagnostic and prognostic models by identifying plasma extracellular vesicles (EVs) associated protein biomarkers specific to LUSC. METHODS This study employed a novel nanomaterial, NaY, for the enrichment of EVs from plasma. Validation was conducted through transmission electron microscopy, nanoparticle tracking analyses, and Western blotting. Machine learning algorithms were utilized to compute protein biomarkers associated with LUSC and establish a diagnostic model. Additionally, a prognostic prediction model for LUSC was developed using a combination of 101 machine learning algorithms. Risk scoring of patients was performed to explore the underlying reasons for prognostic differences between high and low-risk groups. RESULTS The results of three experiments demonstrate that the new nanomaterial NaY effectively enriches EVs from plasma. Analysis of the enriched profile reveals pathways related to glycolysis/gluconeogenesis and carbon metabolism enriched in plasma EVs of LUSC patients. Thirty-eight LSCC-related EV biomarkers were identified, from which five proteins (TUBB3, RPS7, RPLP1, KRT2, and VTN) were selected to establish a diagnostic model distinguishing between benign and LUSC nodules. The diagnostic efficacy of RPS7 and VTN was further validated in independent samples using ELISA experiments. Furthermore, DPYD, GALK1, CDC23, UBE2L3, RHEB, and PSME1 were determined as potential prognostic biomarkers. Subsequently, risk scores were computed for each sample, classifying all patients into high and low-risk groups. Enrichment analysis revealed that EVs from the high-risk group contained proteins promoting cell proliferation and invasion, while those from the low-risk group were enriched in immune-related protein biomarkers. CONCLUSIONS The novel nanomaterial NaY effectively enriches EVs from plasma. Utilizing plasma EV biomarkers, the diagnostic model demonstrates strong discriminative ability between benign and malignant pulmonary nodules in patients.
Collapse
Affiliation(s)
- Sheng Ma
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Na Zhao
- Department of Proteomics, Tianjin Key Laboratory of Clinical Multi-Omics, Tianjin, China
| | - Xin Dong
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Beijing, China
| | - Yaru Wang
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei Song
- Department of Proteomics, Tianjin Key Laboratory of Clinical Multi-Omics, Tianjin, China
| | - Ruiqi Zheng
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaochen Zhi
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Congcong Ma
- Department of Proteomics, Tianjin Key Laboratory of Clinical Multi-Omics, Tianjin, China
| | - Shujun Cheng
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Li
- Department of Proteomics, Tianjin Key Laboratory of Clinical Multi-Omics, Tianjin, China.
| | - Yutao Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Ting Xiao
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
2
|
Zeng C, Xu C, Liu S, Wang Y, Wei Y, Qi Y, Wang Y, Wang J, Ma F. Integrated bulk and single-cell transcriptomic analysis unveiled a novel cuproptosis-related lipid metabolism gene molecular pattern and a risk index for predicting prognosis and antitumor drug sensitivity in breast cancer. Discov Oncol 2025; 16:318. [PMID: 40085377 PMCID: PMC11909392 DOI: 10.1007/s12672-025-02044-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 03/04/2025] [Indexed: 03/16/2025] Open
Abstract
Breast cancer is the second most prevalent malignant tumor worldwide and is highly heterogeneous. Cuproptosis, a newly identified form of cell death, is intimately connected to lipid metabolism. This study investigated breast cancer heterogeneity through the lens of cuproptosis-related lipid metabolism genes (CLMGs), with the goal of predicting patient prognosis, immunotherapy efficacy, and sensitivity to anticancer drugs. By utilizing transcriptomic data from The Cancer Genome Atlas (TCGA) for breast cancer, we identified 682 CLMGs and applied the nonnegative matrix factorization (NMF) method to categorize breast cancer patients into four distinct clusters: cluster 1, ''immune-cold and stroma-poor''; cluster 2, ''immune-infiltrated''; cluster 3, ''stroma-rich''; and cluster 4, ''moderate infiltration''. We subsequently developed a risk model based on CLMGs that incorporates ACSL1, ATP2B4, ATP7B, ENPP6, HSPH1, PIP4K2C, SRD5A3, and ULBP1. This model demonstrated excellent prognostic predictive performance in both the internal (testing and entire sets) and external (GSE20685 and Kaplan-Meier Plotter sets) validation sets. High-risk patients presented lower expression levels of immune checkpoint-related genes and lower immunophenoscores (IPSs), whereas low-risk patients presented higher CD8+ T-cell infiltration levels and IPSs. Furthermore, the risk index was positively correlated with tumor cell stemness and could predict sensitivity to anticancer drugs. We also confirmed that SRD5A3 was highly expressed in breast cancer and participated in promoting the proliferation and migration of breast cancer cells. In conclusion, the results of this study provide new insights and strategies for assessing prognosis and implementing precision treatment for breast cancer through the lens of CLMGs.
Collapse
Affiliation(s)
- Cheng Zeng
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chang Xu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shuning Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yuanyi Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yuhan Wei
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yalong Qi
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yue Wang
- Department of Oncology, Wujin Hospital Affiliated With Jiangsu University, Changzhou, 213000, Jiangsu Province, China
- Department of Oncology, Wujin Clinical College of Xuzhou Medical University, Changzhou, 213000, Jiangsu Province, China
| | - Jiani Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Fei Ma
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
3
|
Ma B, Zhang F, Raza SHA, Wu Z, Su Q, Zhang Y, Wang Z, ALMatrafi TA, Aloufi BH, Ghamry HI, Shukry M, Hou S, Gui L. Palm kernel meal regulates the expression of genes involved in the amino acid metabolism in the liver of Tibetan sheep. BMC Vet Res 2024; 20:333. [PMID: 39044234 PMCID: PMC11264444 DOI: 10.1186/s12917-024-04193-7] [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: 02/12/2024] [Accepted: 07/15/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND Palm kernel meal (PKM) is a by-product of oil palm kernel after oil extraction, which is widely used in animal feeds due to its high energy content. This study aimed to investigate the impact of supplementing Tibetan sheep with PKM on their hepatic phenotype, oxidative stress and immune response. A total of 120 Tibetan lambs (Initial weight = 12.37 ± 0.92 kg) were randomly assigned into four groups: control group (C group, 0% PKM diet), low group (L group, 15% PKM diet), middle group (M group, 18% PKM diet), and high group (H group, 21% PKM diet) on a dry matter basis. The feeding experiment was performed for 130 d, including a 10 d adaption period. RESULTS Results showed that the level of GSH-Px were higher in the H and M groups than in the C and L groups (P < 0.05). The levels of IgM and TNF-α were higher in the M group when compared to those on the C group (P < 0.05). The level of IgA was significantly higher in the M group than in the H group (P < 0.05). Additionally, compared with the others groups, the hepatocytes in the M group displayed a radial arrangement, forming hepatic plates that were centered around the central vein. The transcriptome results revealed that proteasome 26 S subunit, ATPase 3 (PSMC3), proteasome 26 S subunit, ATPase 5 (PSMC5), proteasome 26 S subunit ubiquitin receptor, non-ATPase 4 (PSMD4), proteasome activator subunit 1 (PSME1), acyl-CoA dehydrogenase short/branched chain (ACADSB), enoyl-CoA hydratase, short chain 1 (ECHS1), serine dehydratase (SDS), ornithine transcarbamylase (OTC), and phenylalanine hydroxylase (PAH) were the hub genes regulating the amino acid metabolism in the liver. CONCLUSIONS In summary, dietary 18% PMK supplementation contributed to improve the hepatic phenotype, oxidative stress and immune response through regulating the expression of related genes.
Collapse
Affiliation(s)
- Boyan Ma
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, 810016, Qinghai Province, People's Republic of China
| | - Fengshuo Zhang
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, 810016, Qinghai Province, People's Republic of China
| | - Sayed Haidar Abbas Raza
- Guangdong Provincial Key Laboratory of Food Quality and Safety, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region, Shaoguan University, Shaoguan, 512005, China
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Zhenling Wu
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, 810016, Qinghai Province, People's Republic of China
| | - Quyangangmao Su
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, 810016, Qinghai Province, People's Republic of China
| | - Yu Zhang
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, 810016, Qinghai Province, People's Republic of China
| | - Zhiyou Wang
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, 810016, Qinghai Province, People's Republic of China
| | | | - Bandar Hamad Aloufi
- Biology Department, Faculty of Science, University of Ha'il, Ha'il, Saudi Arabia
| | - Heba I Ghamry
- Nutrition and Food Science, Department of Biology, College of Science, King Khalid University, P.O. Box 960, Abha, 61421, Saudi Arabia
| | - Mustafa Shukry
- Department of Physiology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| | - Shengzhen Hou
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, 810016, Qinghai Province, People's Republic of China
| | - Linsheng Gui
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, 810016, Qinghai Province, People's Republic of China.
| |
Collapse
|
4
|
Yedla P, Bhamidipati P, Syed R, Amanchy R. Working title: Molecular involvement of p53-MDM2 interactome in gastrointestinal cancers. Cell Biochem Funct 2024; 42:e4075. [PMID: 38924101 DOI: 10.1002/cbf.4075] [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: 02/16/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024]
Abstract
The interaction between murine double minute 2 (MDM2) and p53, marked by transcriptional induction and feedback inhibition, orchestrates a functional loop dictating cellular fate. The functional loop comprising p53-MDM2 axis is made up of an interactome consisting of approximately 81 proteins, which are spatio-temporally regulated and involved in DNA repair mechanisms. Biochemical and genetic alterations of the interactome result in dysregulation of the p53-mdm2 axis that leads to gastrointestinal (GI) cancers. A large subset of interactome is well known and it consists of proteins that either stabilize p53 or MDM2 and proteins that target the p53-MDM2 complex for ubiquitin-mediated destruction. Upstream signaling events brought about by growth factors and chemical messengers invoke a wide variety of posttranslational modifications in p53-MDM2 axis. Biochemical changes in the transactivation domain of p53 impact the energy landscape, induce conformational switching, alter interaction potential and could change solubility of p53 to redefine its co-localization, translocation and activity. A diverse set of chemical compounds mimic physiological effectors and simulate biochemical modifications of the p53-MDM2 interactome. p53-MDM2 interactome plays a crucial role in DNA damage and repair process. Genetic aberrations in the interactome, have resulted in cancers of GI tract (pancreas, liver, colorectal, gastric, biliary, and esophageal). We present in this article a review of the overall changes in the p53-MDM2 interactors and the effectors that form an epicenter for the development of next-generation molecules for understanding and targeting GI cancers.
Collapse
Affiliation(s)
- Poornachandra Yedla
- Division of Applied Biology, CSIR-IICT (Indian Institute of Chemical Technology), Ministry of Science and Technology (GOI), Hyderabad, Telangana, India
- Department of Pharmacogenomics, Institute of Translational Research, Asian Healthcare Foundation, Hyderabad, Telangana, India
| | - Pranav Bhamidipati
- Division of Applied Biology, CSIR-IICT (Indian Institute of Chemical Technology), Ministry of Science and Technology (GOI), Hyderabad, Telangana, India
- Department of Life Sciences, Imperial College London, London, UK
| | - Riyaz Syed
- Division of Applied Biology, CSIR-IICT (Indian Institute of Chemical Technology), Ministry of Science and Technology (GOI), Hyderabad, Telangana, India
| | - Ramars Amanchy
- Division of Applied Biology, CSIR-IICT (Indian Institute of Chemical Technology), Ministry of Science and Technology (GOI), Hyderabad, Telangana, India
| |
Collapse
|
5
|
Wen P, Sun Y, Jiang TX, Qiu XB. PA200-Mediated Proteasomal Protein Degradation and Regulation of Cellular Senescence. Int J Mol Sci 2024; 25:5637. [PMID: 38891826 PMCID: PMC11171664 DOI: 10.3390/ijms25115637] [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: 03/10/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 06/21/2024] Open
Abstract
Cellular senescence is closely related to DNA damage, proteasome inactivity, histone loss, epigenetic alterations, and tumorigenesis. The mammalian proteasome activator PA200 (also referred to as PSME4) or its yeast ortholog Blm10 promotes the acetylation-dependent degradation of the core histones during transcription, DNA repair, and spermatogenesis. According to recent studies, PA200 plays an important role in senescence, probably because of its role in promoting the degradation of the core histones. Loss of PA200 or Blm10 is a major cause of the decrease in proteasome activity during senescence. In this paper, recent research progress on the association of PA200 with cellular senescence is summarized, and the potential of PA200 to serve as a therapeutic target in age-related diseases is discussed.
Collapse
Affiliation(s)
- Pei Wen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; (P.W.); (Y.S.)
| | - Yan Sun
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; (P.W.); (Y.S.)
| | - Tian-Xia Jiang
- Ministry of Education Key Laboratory of Cell Proliferation & Regulation Biology, College of Life Sciences, Beijing Normal University, 19 Xinjiekouwai Avenue, Beijing 100875, China
| | - Xiao-Bo Qiu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; (P.W.); (Y.S.)
- Ministry of Education Key Laboratory of Cell Proliferation & Regulation Biology, College of Life Sciences, Beijing Normal University, 19 Xinjiekouwai Avenue, Beijing 100875, China
| |
Collapse
|
6
|
Nguyen TX, Agazzi A, McGill S, Weidt S, Han QH, Gelemanović A, McLaughlin M, Savoini G, Eckersall PD, Burchmore R. Proteomic changes associated with maternal dietary low ω6:ω3 ratio in piglets supplemented with seaweed Part II: Ileum proteomes. J Proteomics 2023; 270:104739. [PMID: 36174954 DOI: 10.1016/j.jprot.2022.104739] [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: 03/25/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 02/01/2023]
Abstract
This study evaluates how long-term dietary low ω6:ω3 ratio in sows and offspring's seaweed (SW) intake affects piglet intestinal function and growth through modifying ileum proteome. Sows were assigned to either control diet (CR, ω6:ω3 ratio = 13:1) or treatment diet (LR, ω6:ω3 = 4:1) during gestation and lactation (n = 8 each). The male weaned offspring were received a basal diet with or without SW powder supplementation (4 g/kg) for 21 days, denoted as SW and CT groups, respectively. In total, four groups of weaned piglets were formed following maternal and offspring's diets combination, represented by CRCT, CRSW, LRCT, and LRSW (n = 10 each). Piglet ileum tissue was collected on day 22 post-weaning and analysed using TMT-based quantitative proteomics. The differentially abundant proteins (n = 300) showed the influence of maternal LR diet on protein synthesis, cell proliferation, and cell cycle regulation. In contrast, the SW diet lowered the inflammation severity and promoted ileal tissue development in CRSW piglets but reduced the fat absorption capacity in LRSW piglets. These results uncovered the mechanism behind the anti-inflammation and intestinal-boosting effects of maternal LR diet in piglets supplemented with SW.
Collapse
Affiliation(s)
- Thi Xuan Nguyen
- Università degli Studi di Milano, Via dell'Università, 6, 26900 Lodi, Italy; University of Glasgow, Bearsden Rd, G61 1QH, United Kingdom; Vietnam National University of Agriculture, Hanoi, Viet Nam.
| | - Alessandro Agazzi
- Università degli Studi di Milano, Via dell'Università, 6, 26900 Lodi, Italy
| | - Suzanne McGill
- University of Glasgow, Bearsden Rd, G61 1QH, United Kingdom
| | - Stefan Weidt
- University of Glasgow, Bearsden Rd, G61 1QH, United Kingdom
| | - Quang Hanh Han
- University of Glasgow, Bearsden Rd, G61 1QH, United Kingdom; Vietnam National University of Agriculture, Hanoi, Viet Nam
| | - Andrea Gelemanović
- Mediterranean Institute for Life Sciences (MedILS), Meštrovićevo šetalište 45, 21000 Split, Croatia
| | | | - Giovanni Savoini
- Università degli Studi di Milano, Via dell'Università, 6, 26900 Lodi, Italy
| | | | | |
Collapse
|
7
|
Wu C, Zhong R, Sun X, Shi J. PSME2 identifies immune-hot tumors in breast cancer and associates with well therapeutic response to immunotherapy. Front Genet 2022; 13:1071270. [PMID: 36583022 PMCID: PMC9793949 DOI: 10.3389/fgene.2022.1071270] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
Breast cancer (BrCa) is a heterogeneous disease, which leads to unsatisfactory prognosis in females worldwide. Previous studies have proved that tumor immune microenvironment (TIME) plays crucial roles in oncogenesis, progression, and therapeutic resistance in Breast cancer. However, biomarkers related to TIME features have not been fully discovered. Proteasome activator complex subunit 2 (PSME2) is a member of proteasome activator subunit gene family, which is critical to protein degradation mediated by the proteasome. In the current research, we comprehensively analyzed the expression and immuno-correlations of Proteasome activator complex subunit 2 in Breast cancer. Proteasome activator complex subunit 2 was significantly upregulated in tumor tissues but associated with well prognosis. In addition, Proteasome activator complex subunit 2 was overexpressed in HER2-positive Breast cancer but not related to other clinicopathological features. Interestingly, Proteasome activator complex subunit 2 was positively related to immune-related processes and identified immuno-hot TIME in Breast cancer. Specifically, Proteasome activator complex subunit 2 was positively correlated with immunomodulators, tumor-infiltrating immune cells (TIICs), immune checkpoints, and tumor mutation burden (TMB) levels. Moreover, the positive correlation between Proteasome activator complex subunit 2 and PD-L1 expression was confirmed in a tissue microarray (TMA) cohort. Furthermore, in an immunotherapy cohort of Breast cancer, patients with pathological complete response (pCR) expressed higher Proteasome activator complex subunit 2 compared with those with non-pathological complete response. In conclusion, Proteasome activator complex subunit 2 is upregulated in tumor tissues and correlated with the immuno-hot tumor immune microenvironment, which can be a novel biomarker for the recognition of tumor immune microenvironment features and immunotherapeutic response in Breast cancer.
Collapse
Affiliation(s)
- Cen Wu
- Department of General Surgery, Rudong County People’s Hospital, Nantong, China
| | - Ren Zhong
- Department of General Surgery, Rudong County People’s Hospital, Nantong, China
| | - Xiaofei Sun
- Department of General Surgery, Rudong County People’s Hospital, Nantong, China
| | - Jiajie Shi
- Departments of Breast Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
8
|
Hu L, Yang K, Chen Y, Sun C, Wang X, Zhu S, Yang S, Cao G, Xiong M, Chen B. Survival nomogram for different grades of gastric cancer patients based on SEER database and external validation cohort. Front Oncol 2022; 12:951444. [PMID: 36185304 PMCID: PMC9523147 DOI: 10.3389/fonc.2022.951444] [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: 05/24/2022] [Accepted: 08/25/2022] [Indexed: 12/02/2022] Open
Abstract
Background Influencing factors varied among gastric cancer (GC) for different differentiation grades which affect the prognosis accordingly. This study aimed to develop a nomogram to effectively identify the overall survival (OS). Methods Totally, 9,568 patients with GC were obtained from the SEER database as the training cohort and internal validation cohort. We then retrospectively enrolled patients diagnosed with GC to construct the external validation cohort from the First Affiliated Hospital of Anhui Medical University. The prognostic factors were integrated into the multivariate Cox regression to construct a nomogram. To test the accuracy of the model, we used the calibration curves, receiver operating characteristics (ROC) curves, C-index, and decision curve analysis (DCA). Results Race chemotherapy, tumor size, and other four factors were significantly associated with the prognosis of Grade III GC Patients. On this basis, we developed a nomogram. The discrimination of the nomogram revealed good prognostic accuracy The results of the area under the curve (AUC) calculated by ROC for five-year survival were 0.828 and 0.758 in the training set and external validation cohort, higher than that of the TNM staging system. The calibration plot revealed that the estimated risk was close to the actual risk. DCA also suggested an excellent predictive value of the nomogram. Similar results were obtained in Grade-I and Grade-II GC patients. Conclusions The nomogram developed in this study and other findings could help individualize the treatment of GC patients and assist clinicians in their shared decision-making with patients.
Collapse
Affiliation(s)
- Lei Hu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Clinical Medicine, School of the First Clinical Medicine, Anhui Medical University, Hefei, China
| | - Kang Yang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Public Health Clinical Center, Hefei, China
| | - Yue Chen
- Department of Clinical Medicine, School of the First Clinical Medicine, Anhui Medical University, Hefei, China
| | - Chenyu Sun
- AMITA Health Saint Joseph Hospital Chicago, Chicago, IL, United States
| | - Xu Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shaopu Zhu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shiyi Yang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Guodong Cao
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- *Correspondence: Guodong Cao, ; Maoming Xiong, ; Bo Chen,
| | - Maoming Xiong
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- *Correspondence: Guodong Cao, ; Maoming Xiong, ; Bo Chen,
| | - Bo Chen
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of surgery, the People’s Hospital of Hanshan County, Ma’anshan City, China
- *Correspondence: Guodong Cao, ; Maoming Xiong, ; Bo Chen,
| |
Collapse
|
9
|
Zahedi S, Carvalho AS, Ejtehadifar M, Beck HC, Rei N, Luis A, Borralho P, Bugalho A, Matthiesen R. Assessment of a Large-Scale Unbiased Malignant Pleural Effusion Proteomics Study of a Real-Life Cohort. Cancers (Basel) 2022; 14:cancers14184366. [PMID: 36139528 PMCID: PMC9496668 DOI: 10.3390/cancers14184366] [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: 08/09/2022] [Revised: 08/29/2022] [Accepted: 09/01/2022] [Indexed: 11/29/2022] Open
Abstract
Simple Summary Pleural effusion (PE) occurs as a consequence of various pathologies. Malignant effusion due to lung cancer is one of the most frequent causes. A method for accurate differentiation of malignant from benign PE is an unmet clinical need. Proteomics profiling of PE has shown promising results. However, mass spectrometry (MS) analysis typically involves the tedious elimination of abundant proteins before analysis, and clinical annotation of proteomics profiled cohorts is limited. This study compares the proteomes of malignant PE and nonmalignant PE, identifies lung cancer malignant markers in agreement with other studies, and identifies markers strongly associated with patient survival. Abstract Background: Pleural effusion (PE) is common in advanced-stage lung cancer patients and is related to poor prognosis. Identification of cancer cells is the standard method for the diagnosis of a malignant PE (MPE). However, it only has moderate sensitivity. Thus, more sensitive diagnostic tools are urgently needed. Methods: The present study aimed to discover potential protein targets to distinguish malignant pleural effusion (MPE) from other non-malignant pathologies. We have collected PE from 97 patients to explore PE proteomes by applying state-of-the-art liquid chromatography-mass spectrometry (LC-MS) to identify potential biomarkers that correlate with immunohistochemistry assessment of tumor biopsy or with survival data. Functional analyses were performed to elucidate functional differences in PE proteins in malignant and benign samples. Results were integrated into a clinical risk prediction model to identify likely malignant cases. Sensitivity, specificity, and negative predictive value were calculated. Results: In total, 1689 individual proteins were identified by MS-based proteomics analysis of the 97 PE samples, of which 35 were diagnosed as malignant. A comparison between MPE and benign PE (BPE) identified 58 differential regulated proteins after correction of the p-values for multiple testing. Furthermore, functional analysis revealed an up-regulation of matrix intermediate filaments and cellular movement-related proteins. Additionally, gene ontology analysis identified the involvement of metabolic pathways such as glycolysis/gluconeogenesis, pyruvate metabolism and cysteine and methionine metabolism. Conclusion: This study demonstrated a partial least squares regression model with an area under the curve of 98 and an accuracy of 0.92 when evaluated on the holdout test data set. Furthermore, highly significant survival markers were identified (e.g., PSME1 with a log-rank of 1.68 × 10−6).
Collapse
Affiliation(s)
- Sara Zahedi
- iNOVA4Health, NOVA Medical School (NMS), Faculdade de Ciências Médicas (FCM), Universidade Nova de Lisboa, 1150-082 Lisbon, Portugal
| | - Ana Sofia Carvalho
- iNOVA4Health, NOVA Medical School (NMS), Faculdade de Ciências Médicas (FCM), Universidade Nova de Lisboa, 1150-082 Lisbon, Portugal
| | - Mostafa Ejtehadifar
- iNOVA4Health, NOVA Medical School (NMS), Faculdade de Ciências Médicas (FCM), Universidade Nova de Lisboa, 1150-082 Lisbon, Portugal
| | - Hans C. Beck
- Department of Clinical Biochemistry, Odense University Hospital, 5000 Odense, Denmark
| | - Nádia Rei
- iNOVA4Health, NOVA Medical School (NMS), Faculdade de Ciências Médicas (FCM), Universidade Nova de Lisboa, 1150-082 Lisbon, Portugal
| | - Ana Luis
- Hospital CUF Descobertas, CUF Oncologia, 1998-018 Lisbon, Portugal
| | - Paula Borralho
- Hospital CUF Descobertas, CUF Oncologia, 1998-018 Lisbon, Portugal
| | - António Bugalho
- iNOVA4Health, NOVA Medical School (NMS), Faculdade de Ciências Médicas (FCM), Universidade Nova de Lisboa, 1150-082 Lisbon, Portugal
- Hospital CUF Descobertas, CUF Oncologia, 1998-018 Lisbon, Portugal
- Correspondence: (A.B.); (R.M.)
| | - Rune Matthiesen
- iNOVA4Health, NOVA Medical School (NMS), Faculdade de Ciências Médicas (FCM), Universidade Nova de Lisboa, 1150-082 Lisbon, Portugal
- Correspondence: (A.B.); (R.M.)
| |
Collapse
|
10
|
Yazgili AS, Ebstein F, Meiners S. The Proteasome Activator PA200/PSME4: An Emerging New Player in Health and Disease. Biomolecules 2022; 12:1150. [PMID: 36009043 PMCID: PMC9406137 DOI: 10.3390/biom12081150] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 11/24/2022] Open
Abstract
Proteasomes comprise a family of proteasomal complexes essential for maintaining protein homeostasis. Accordingly, proteasomes represent promising therapeutic targets in multiple human diseases. Several proteasome inhibitors are approved for treating hematological cancers. However, their side effects impede their efficacy and broader therapeutic applications. Therefore, understanding the biology of the different proteasome complexes present in the cell is crucial for developing tailor-made inhibitors against specific proteasome complexes. Here, we will discuss the structure, biology, and function of the alternative Proteasome Activator 200 (PA200), also known as PSME4, and summarize the current evidence for its dysregulation in different human diseases. We hereby aim to stimulate research on this enigmatic proteasome regulator that has the potential to serve as a therapeutic target in cancer.
Collapse
Affiliation(s)
- Ayse Seda Yazgili
- Comprehensive Pneumology Center (CPC), Helmholtz Center Munich, Max-Lebsche Platz 31, 81377 Munich, Germany
| | - Frédéric Ebstein
- Institut für Medizinische Biochemie und Molekularbiologie (IMBM), Universitätsmedizin Greifswald, Ferdinand-Sauerbruch-Straße, Klinikum DZ/7, 17475 Greifswald, Germany
| | - Silke Meiners
- Research Center Borstel/Leibniz Lung Center, Parkallee 1-40, 23845 Borstel, Germany
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), 23845 Sülfeld, Germany
- Institute of Experimental Medicine, Christian-Albrechts University Kiel, 24118 Kiel, Germany
| |
Collapse
|
11
|
Ge S, Huang H, Huang W, Ji R, Chen J, Wu S, Wang L, Huang T, Sheng Y, Yan H, Lu C, Ma L. PSME4 Activates mTOR Signaling and Promotes the Malignant Progression of Hepatocellular Carcinoma. Int J Gen Med 2022; 15:885-895. [PMID: 35115815 PMCID: PMC8801729 DOI: 10.2147/ijgm.s344360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/10/2022] [Indexed: 12/27/2022] Open
Abstract
Background As hepatocellular carcinoma (HCC) having the second-highest mortality rate globally, the early diagnosis and prognosis of HCC have always been the focus of various studies. Although PSME4 has been reported to be closely related to several malignancies, its role in HCC remains unclear. Materials and Methods The TCGA-LIHC database and HCC tissues were used to explore the expression of PSME4 in HCC. Gene set enrichment analysis (GSEA) was used to forecast the biological behavior of HCC cells that PSME4 might be involved in regulation. In addition, CCK-8, colony formation and flow cytometry assays were used to explore the effect of PSME4 on HCC cells. Furthermore, the underlying PSME4-related signaling pathways in HCC were further confirmed using GSEA. Results We found that the expression of PSME4 in HCC tissues was significantly higher than that in adjacent normal tissues, and patients with high PSME4 expression have a poor prognosis. CCK-8, colony formation and flow cytometry assays shown that knockdown of PSME4 inhibits HCC cell proliferation of HCC cells, promotes cell apoptosis and moves the cell cycle away from the S phase. Mechanistically, PSME4 may promote the development of HCC through mTOR signaling pathway. Conclusion The high expression of PSME4 in HCC promotes the proliferation of HCC cells via the mTOR signalling pathway. Therefore, PSME4 is an emerging tumour marker for the early diagnosis and prognosis of HCC.
Collapse
Affiliation(s)
- Sijia Ge
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Jiangsu, People’s Republic of China
| | - Hua Huang
- Department of Pathology, Affiliated Hospital of Nantong University, Jiangsu, People’s Republic of China
| | - Wei Huang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Jiangsu, People’s Republic of China
| | - Ran Ji
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Jiangsu, People’s Republic of China
| | - Jing Chen
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Jiangsu, People’s Republic of China
| | - Shuzhen Wu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Jiangsu, People’s Republic of China
| | - Liyang Wang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Jiangsu, People’s Republic of China
| | - Tianxin Huang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Jiangsu, People’s Republic of China
| | - Yu Sheng
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Jiangsu, People’s Republic of China
| | - Haiou Yan
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Jiangsu, People’s Republic of China
| | - Cuihua Lu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Jiangsu, People’s Republic of China
- Cuihua Lu, Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, People’s Republic of China, Email
| | - Lin Ma
- Department of Gastroenterology, Affiliated Haian Hospital of Nantong University, Jiangsu, People’s Republic of China
- Correspondence: Lin Ma, Department of Gastroenterology, Affiliated Haian Hospital of Nantong University, Nantong, 226600, Jiangsu, People’s Republic of China, Email
| |
Collapse
|