1
|
Gu W, Liu J, Liu Y, Wang Z. Identification of potential biomarkers for hypertension based on transcriptomic analysis in rats. Hypertens Res 2025:10.1038/s41440-025-02200-4. [PMID: 40240870 DOI: 10.1038/s41440-025-02200-4] [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: 10/09/2024] [Revised: 02/14/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025]
Abstract
Hypertension is a complex disorder influenced by genetic predisposition, neural and endocrine dysregulation, cardiovascular and renal dysfunction, and unhealthy lifestyles. It is a major risk factor for many diseases. However, the pathophysiological mechanisms underlying hypertension have not been systematically characterized to date. In this study, we compared physiological and molecular changes between spontaneously hypertensive rats (SHR) and Wistar Kyoto (WKY, control strain) models using RNA sequencing. Blood pressure increased significantly in SHR models over 3-15 weeks compared with WKY control rats. Furthermore, indicators of cardiac remodeling and fibrosis were elevated in SHR on echocardiography and immunohistochemical analyses. RNA sequencing findings revealed differentially expressed genes between SHRs and WKYs in each week, which were related to dysregulation of Epstein-Barr virus infection, fluid shear stress and atherosclerosis, RNA degradation, and endocrine resistance. Transcriptome analysis showed that differentially expressed genes related to hypertension were involved in the hypoxia inducible factor-1 (HIF-1) and interleukin-17 (IL-17) signaling pathways. Furthermore, Gene Ontology (GO) functional analysis showed that differentially expressed genes were mainly associated with catalytic activity and protein binding. The Venn diagram analysis identified KCNE1, Lad1, SLC9A3, and Frzb as potential targets of hypertension. In addition, the expression of these four genes exhibited excellent sensitivity and specificity, suggesting their potential diagnostic utility in hypertension. These findings support a theoretical basis for understanding hypertension and related heart remodeling.
Collapse
Affiliation(s)
- Wei Gu
- Coronary Heart Disease Center, Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jielin Liu
- Department of Hypertension Research, Beijing Anzhen Hospital, Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Ya Liu
- Department of Hypertension Research, Beijing Anzhen Hospital, Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Zuoguang Wang
- Department of Hypertension Research, Beijing Anzhen Hospital, Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China.
| |
Collapse
|
2
|
Dimayacyac-Esleta BRT, Mira FD, Zarate LM, Porras BJO, Juntilla DLA, Suñga LBL, Pondevida VB, Naval SS, Sayo TMS, Luna HGC, Prieto EI. Discovery of Key Candidate Protein Biomarkers in Early-Stage Nonsmall Cell Lung Carcinoma through Quantitative Proteomics. J Proteome Res 2025; 24:1701-1714. [PMID: 40014793 DOI: 10.1021/acs.jproteome.4c00764] [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: 03/01/2025]
Abstract
Difficulties in early-stage diagnosis are among the factors contributing to the high mortality of nonsmall cell lung carcinoma (NSCLC) patients. Unfortunately, diagnostic biomarkers are currently lacking, limiting options in the clinic. To discover proteins that have potential for biomarker applications, we performed an in-depth quantitative proteomic analysis on a cohort of Filipino early-stage NSCLC lung adenocarcinoma (LUAD) patients. Differentially expressed proteins (DEPs) were obtained by using tandem mass tag (TMT) labeling and mass spectrometry (MS)-based quantitative proteomics. A total of 6240 quantified proteins were identified with 3155 significantly upregulated and 1248 significantly downregulated. Integration of the proteomic result with curated transcriptome data allowed the identification of 33 proteins with biomarker potential. This study also provided insights into relevant pathways in NSCLC LUAD, such as protein translation and metabolic pathways. Interestingly, all of the enzymes in the hexosamine biosynthetic pathway (HBP) are found to be upregulated, suggesting its important role in NSCLC LUAD. It is worthwhile to look at the potential of targeting the metabolic vulnerability of NSCLC LUAD as a new strategy in drug development. All MS data were deposited into ProteomeXchange with the identifier PXD050598.
Collapse
Affiliation(s)
| | - Ferdinand D Mira
- Institute of Chemistry, University of the Philippines Diliman, Metro Manila 1101, Philippines
| | - Lorenzo M Zarate
- National Institute of Molecular Biology and Biotechnology, University of the Philippines Diliman, Metro Manila 1101, Philippines
| | - Ben Joshua O Porras
- National Institute of Molecular Biology and Biotechnology, University of the Philippines Diliman, Metro Manila 1101, Philippines
| | - Dave Laurence A Juntilla
- National Institute of Molecular Biology and Biotechnology, University of the Philippines Diliman, Metro Manila 1101, Philippines
| | - Lara Beatrice L Suñga
- Institute of Chemistry, University of the Philippines Diliman, Metro Manila 1101, Philippines
| | - Venus B Pondevida
- Institute of Chemistry, University of the Philippines Diliman, Metro Manila 1101, Philippines
| | - Sullian S Naval
- Lung Center of the Philippines, Metro Manila 1100, Philippines
| | | | | | - Eloise I Prieto
- National Institute of Molecular Biology and Biotechnology, University of the Philippines Diliman, Metro Manila 1101, Philippines
| |
Collapse
|
3
|
Yang H, Xiong R, Zhang R, Sun S, Pan Y, Zhao Q, Bie J, Luo Y, Song G, Liu K. LINC01305 and LAD1 Co-Regulate CTTN and N-WASP Phosphorylation, Mediating Cytoskeletal Reorganization to Promote ESCC Metastasis. Mol Carcinog 2025; 64:756-768. [PMID: 39835575 PMCID: PMC11890417 DOI: 10.1002/mc.23885] [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: 07/26/2024] [Revised: 12/29/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025]
Abstract
Esophageal squamous cell carcinoma (ESCC) is prone to metastasis and is a leading cause of mortality. The cytoskeleton is closely related to cell morphology and movement; however, little research has been conducted on ESCC metastasis. In this study, we found that the anchoring filament protein ladinin 1 (LAD1) specifically binds to LINC01305 for co-regulating the level of modulating cortactin proteins (CTTN) and neuronal Wiskott-Aldrich syndrome protein (N-WASP) phosphorylation, which mediates cytoskeletal reorganization and affects the metastasis of ESCC cells. Additionally, LINC01305 and LAD1 jointly promoted the epithelial-mesenchymal transition (EMT) process by activating the phosphoinositide-3-kinase-protein kinase B (PI3K/AKT) signaling pathway. Moreover, LINC01305 and LAD1 were related to the late clinical stage and lymph node metastasis of ESCC. Our study demonstrated that LINC01305 and LAD1 are major determinants of ESCC dissemination and revealed a novel molecular mechanism of cytoskeletal reorganization that controls ESCC metastasis. Trial Registration: N/A.
Collapse
Grants
- This work was supported by the National Institutes of the National Natural Science Foundation of China (82203851), the Sichuan Science and Technology Program (2024NSFSC1973, 2023YFS0473, MZGC20240072, MZGC20240071), the Nanchong Science and Technology Program (22SXQT0336, 20SXQT0328, 22SXQT0087, 22SXQT0340, 20SXQT0181), the Sichuan Medical Association Science and Technology Program (S2024009), and the North Sichuan Medical College Science and Technology Program (CBY24-KP03, CBY22-QDA01).
Collapse
Affiliation(s)
- Hang Yang
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital of Capital Medical University, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical CollegeNanchongChina
| | - Rong Xiong
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital of Capital Medical University, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
| | - Ruolan Zhang
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital of Capital Medical University, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
| | - Shan Sun
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital of Capital Medical University, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical CollegeNanchongChina
| | - Yingjie Pan
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital of Capital Medical University, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical CollegeNanchongChina
| | - Quanneng Zhao
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital of Capital Medical University, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
| | - Jun Bie
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital of Capital Medical University, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
| | - Yi Luo
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital of Capital Medical University, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
| | - Guiqin Song
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical CollegeNanchongChina
| | - Kang Liu
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital of Capital Medical University, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
| |
Collapse
|
4
|
Wan X, Deng Q, Chen A, Zhang X, Yang W. Bioinformatics analysis and experimental validation of the oncogenic role of COL11A1 in pan-cancer. 3 Biotech 2024; 14:290. [PMID: 39507058 PMCID: PMC11534945 DOI: 10.1007/s13205-024-04133-0] [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: 07/11/2024] [Accepted: 10/19/2024] [Indexed: 11/08/2024] Open
Abstract
The intricate expression patterns and oncogenic attributes of COL11A1 across different cancer types remain largely elusive. This study used several public databases (TCGA, GTEx, and CCLE) to investigate the pan-cancer landscape of COL11A1 expression, its prognostic implications, interplay with the immune microenvironment, and enriched signaling cascades. Concurrently, western blot analyses were performed to verify COL11A1 expression in lung adenocarcinoma (LUAD) cell lines and clinical samples. In addition, COL11A1 knockout cell lines were generated to scrutinize the functional consequences of COL11AI expression on cancer cell behavior by use MTT, colony formation, and scratch wound healing assays. A comprehensive database investigation revealed that COL11A1 was upregulated in a majority of tumor tissues and its expression was highly correlated with a patient's prognosis. Notably, genetic alterations in COL11A1 predominantly occurred as mutations, while its DNA methylation status inversely mirrored gene expression levels across multiple promoter regions. Our findings suggest that COL11A1 helps to modulate the tumor immune landscape and potentially acts through the epithelial-mesenchymal transition (EMT) pathway to exert its oncogenic function. Western blot analyses further substantiated the specific upregulation of COL11A1 in LUAD cell lines and tissues, suggesting a close association with the EMT process. Ablation of COL11A1 in cancer cells significantly reduced their proliferative, clonogenic, and migratory abilities, underscoring the functional significance of COL11A1 in tumor cell behavior. Collectively, this research revealed the prevalent overexpression of COL11A1 in pan-cancer tissues, its profound prognostic and microenvironmental correlations, and the mechanistic underpinnings of its tumor-promoting effects as mediated via EMT signaling. Our findings suggest that COL11A1 could serve as a prognostic and diagnostic biomarker and therapeutic target for cancer.
Collapse
Affiliation(s)
- Xiaofeng Wan
- Department of Laboratory, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui China
| | - Qingmei Deng
- Department of Laboratory, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui China
| | - Anling Chen
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031 Anhui China
- Science Island Branch, Graduate School of University of Science and Technology of China, Hefei, China
| | - Xinhui Zhang
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031 Anhui China
| | - Wulin Yang
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031 Anhui China
| |
Collapse
|
5
|
Ajiboye J, Uldry AC, Heller M, Naguleswaran A, Fan E, Van Voorhis WC, Hemphill A, Müller J. Molecular Targets of the 5-Amido-Carboxamide Bumped Kinase Inhibitor BKI-1748 in Cryptosporidium parvum and HCT-8 Host Cells. Int J Mol Sci 2024; 25:2707. [PMID: 38473953 PMCID: PMC10931551 DOI: 10.3390/ijms25052707] [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: 02/02/2024] [Revised: 02/22/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Cryptosporidium parvum is an apicomplexan parasite causing persistent diarrhea in humans and animals. Issuing from target-based drug development, calcium-dependent protein kinase 1 inhibitors, collectively named bumped kinase inhibitors (BKIs), with excellent efficacies in vitro and in vivo have been generated. Some BKIs including BKI-1748 share a core structure with similarities to the first-generation antiprotozoal drug quinine, which is known to exert notorious side effects. Unlike quinine, BKI-1748 rapidly interfered with C. parvum proliferation in the human colon tumor (HCT) cell line HCT-8 cells and caused dramatic effects on the parasite ultrastructure. To identify putative BKI targets in C. parvum and in host cells, we performed differential affinity chromatography with cell-free extracts from non-infected and infected HCT-8 cells using BKI-1748 and quinine epoxy-activated sepharose columns followed by mass spectrometry. C. parvum proteins of interest were identified in eluates from columns coupled to BKI-1748, or in eluates from both BKI-1748 and quinine columns. However, no C. parvum proteins could be identified binding exclusively to BKI-1748. In contrast, 25 BKI-1748-specific binding proteins originating from HCT-8 cells were detected. Moreover, 29 C. parvum and 224 host cell proteins were identified in both BKI-1748 as well as in quinine eluates. In both C. parvum and host cells, the largest subset of binding proteins was involved in RNA binding and modification, with a focus on ribosomal proteins and proteins involved in RNA splicing. These findings extend previous results, showing that BKI-1748 interacts with putative targets involved in common, essential pathways such as translation and RNA processing.
Collapse
Affiliation(s)
- Jubilee Ajiboye
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, 3012 Bern, Switzerland;
- Cellular, Molecular and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT 05405, USA
| | - Anne-Christine Uldry
- Proteomics and Mass Spectrometry Core Facility, Department for BioMedical Research (DBMR), University of Bern, Länggass-Strasse 122, 3012 Bern, Switzerland; (A.-C.U.); (M.H.)
| | - Manfred Heller
- Proteomics and Mass Spectrometry Core Facility, Department for BioMedical Research (DBMR), University of Bern, Länggass-Strasse 122, 3012 Bern, Switzerland; (A.-C.U.); (M.H.)
| | - Arunasalam Naguleswaran
- Institute of Molecular Pathology, Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, 3012 Bern, Switzerland;
| | - Erkang Fan
- Department of Biochemistry, University of Washington, Seattle, WA 98109, USA;
| | - Wesley C. Van Voorhis
- Center for Emerging and Re-Emerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA 98109, USA;
| | - Andrew Hemphill
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, 3012 Bern, Switzerland;
| | - Joachim Müller
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, 3012 Bern, Switzerland;
| |
Collapse
|