1
|
Boettger P, Schwertz H, Platsch H, Mueller-Werdan U, Werdan K, Buerke M. Intramural Administration of Translational Inhibitor Puromycin Upon Balloon Angioplasty Inhibits SMC Proliferation and Protein Synthesis-Vascular Proteome Profiling Analysis. Proteomics Clin Appl 2025; 19:e202400066. [PMID: 39993432 DOI: 10.1002/prca.202400066] [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: 07/04/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/26/2025]
Abstract
INTRODUCTION Percutaneous transluminal coronary angioplasty (PTCA) is an effective procedure to decrease the severity of stenotic coronary atherosclerotic lesions. However, its long-term success is limited by the formation of restenosis or neointima by increased proliferation of smooth muscle cells (SMCs) and synthesis of extracellular matrix. Polypeptide growth factors are potent SMC mitogens and are involved in SMC proliferation and extracellular matrix (ECM) synthesis. In this line, inhibition of de novo protein synthesis might be beneficial. METHODS We examined the effects of different concentrations of translational inhibitor puromycin on SMC proliferation and apoptosis, in vitro. Further, we examined the effects of local administration of puromycin in a rabbit balloon injury model of the iliac artery. RESULTS Injection of puromycin or its vehicle was performed with an infusion-balloon catheter directly into the vessel wall during angioplasty. PTA in the vehicle group resulted in neointima formation 3 weeks after the vascular intervention. In contrast, puromycin treatment resulted in a significant reduction of intima-media ratio. We observed decreased elastin and collagen III synthesis in puromycin-treated animals. With proteomics, we could demonstrate reduced protein expression of lamin, vimentin, alpha-1 antitrypsin, alpha-actin allowing puromycin treatment. In in vitro experiments, puromycin decreased SMCs proliferation (i.e., BrdU incorporation) following FCS stimulation. PERSPECTIVE Based on the data from our animal experiments, aministration of puromycin directly into the vessel wall during angioplasty may be effective in preventing or reducing restenosis in humans.
Collapse
Affiliation(s)
- Priyanka Boettger
- Department of Medicine II Cardiology, Intensive Care Medicine, Angiology, St. Marien Hospital, Siegen, Germany
- Department of Medicine III Martin-Luther-University Halle-Wittenberg, Halle/Saale, Germany
| | - Hansjörg Schwertz
- Molecular Medicine Program The University of Utah, Salt Lake City, Utah, USA
- Division of Occupational Medicine, The University of Utah, Salt Lake City, Utah, USA
- Department of Occupational Medicine, Billings Clinic Bozeman, Bozeman, Montana, USA
| | | | - Ursula Mueller-Werdan
- Department of Medicine III Martin-Luther-University Halle-Wittenberg, Halle/Saale, Germany
| | - Karl Werdan
- Department of Medicine III Martin-Luther-University Halle-Wittenberg, Halle/Saale, Germany
| | - Michael Buerke
- Department of Medicine II Cardiology, Intensive Care Medicine, Angiology, St. Marien Hospital, Siegen, Germany
- Department of Medicine III Martin-Luther-University Halle-Wittenberg, Halle/Saale, Germany
| |
Collapse
|
2
|
Zhu Z, Liu H, Feng L, Lu L, Zhu J, Liang Q, Lan Z, Ye Y, Wang S, Chen A, Yan J. Loss of ADAMTS5 promotes vascular calcification via versican/integrin β1/FAK signal. Atherosclerosis 2025; 404:119190. [PMID: 40215897 DOI: 10.1016/j.atherosclerosis.2025.119190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 03/29/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025]
Abstract
INTRODUCTION Extracellular matrix (ECM) proteases have been closely linked to the pathogenesis of vascular calcification. A disintegrin and metalloprotease with thrombospondin motifs-5 (ADAMTS5) is an ECM-degrading enzyme involved in ECM remodeling. Versican, a critical ECM component in the arteries, can be proteolytically cleaved by ADAMTS5 and activates integrin β1. However, whether ADAMTS5 is involved in the regulation of the pathogenesis of vascular calcification remains unclear. This study investigates the regulatory role of ADAMTS5 in vascular calcification and its mechanistic link to versican-integrin β1/FAK signaling. METHODS AND RESULTS Western blot, immunofluorescence, and immunohistochemistry analysis revealed that ADAMTS5 expression was significantly downregulated in rat and human vascular smooth muscle cells (VSMCs), as well as in rat and human arteries during vascular calcification. In addition, both pharmacological inhibition of ADAMTS5 and knockdown of ADAMTS5 by siRNA significantly aggravated mineral deposition in rat and human VSMCs under osteogenic conditions. Moreover, adenovirus-mediated ADAMTS5 overexpression markedly attenuated calcification of VSMCs and aortic calcification in rats with chronic kidney disease. Furthermore, inhibition of ADAMTS5 promoted aortic calcification in VitD3-overloaded mice. Mechanistically, overexpression of ADAMTS5 significantly reduced versican protein levels, and inhibited integrin β1 and FAK phosphorylation in rat VSMCs, but increased versikine protein levels. Moreover, either knockdown of versican or pharmacological inhibition of FAK phosphorylation repressed VSMC calcification mediated by loss of ADAMTS5. CONCLUSIONS We have demonstrated for the first time that ADAMTS5 deficiency promotes versican accumulation and activates integrin β1/FAK signaling. These findings suggest ADAMTS5 as a potential therapeutic target for vascular calcification.
Collapse
MESH Headings
- Animals
- Versicans/metabolism
- Humans
- ADAMTS5 Protein/genetics
- ADAMTS5 Protein/metabolism
- ADAMTS5 Protein/deficiency
- Signal Transduction
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Vascular Calcification/enzymology
- Vascular Calcification/pathology
- Vascular Calcification/genetics
- Integrin beta1/metabolism
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Male
- Cells, Cultured
- Disease Models, Animal
- Focal Adhesion Kinase 1/metabolism
- Mice, Inbred C57BL
- Rats, Sprague-Dawley
- Rats
- Phosphorylation
- Mice
- Aortic Diseases/enzymology
- Aortic Diseases/genetics
- Aortic Diseases/pathology
Collapse
Affiliation(s)
- Zhenyu Zhu
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, 510280, PR China; Department of Cardiology, Tongde Hospital of Zhejiang Province, PR China
| | - Hao Liu
- Division of Vascular and Interventional Radiology, Department of General Surgery, Nanfang Hospital, Southern Medical University, PR China
| | - Liyun Feng
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, 510280, PR China
| | - Lihe Lu
- Department of Pathophysiolgy, Zhongshan Medical School, Sun Yat-Sen University, Guangzhou, 510080, PR China
| | - Jiahui Zhu
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, 510280, PR China
| | - Qingchun Liang
- Department of Anesthesiology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510665, PR China
| | - Zirong Lan
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, 510280, PR China
| | - Yuanzhi Ye
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, 510280, PR China
| | - Siyi Wang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, 510280, PR China
| | - An Chen
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, 510280, PR China.
| | - Jianyun Yan
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, 510280, PR China.
| |
Collapse
|
3
|
Geng J, Zheng K, Wang P, Su B, Wei Q, Liu X. Focal Adhesion Regulation as a Strategy against Kidney Fibrosis. ACS Chem Biol 2025; 20:464-478. [PMID: 39818722 DOI: 10.1021/acschembio.4c00776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Chronic kidney fibrosis poses a significant global health challenge with effective therapeutic strategies remaining elusive. While cell-extracellular matrix (ECM) interactions are known to drive fibrosis progression, the specific role of focal adhesions (FAs) in kidney fibrosis is not fully understood. In this study, we investigated the role of FAs in kidney tubular epithelial cell fibrosis by employing precise nanogold patterning to modulate integrin distribution. We demonstrate that increasing ligand spacing disrupts integrin clustering, thereby inhibiting FA formation and attenuating fibrosis. Importantly, enhanced FA activity is associated with kidney fibrosis in both human disease specimens and murine models. Mechanistically, FAs regulate fibrosis through mechanotransduction pathways, and our in vivo experiments show that suppressing mechanotransduction significantly mitigates kidney fibrosis in mice. These findings highlight the potential of targeting FAs as a therapeutic strategy, offering new insights into clinical intervention in kidney fibrosis.
Collapse
Affiliation(s)
- Jiwen Geng
- Department of Nephrology, Kidney Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
- College of Polymer Science and Engineering, College of Biomedical Engineering, State Key Laboratory of Polymer Materials and Engineering Sichuan University, Chengdu 610065, China
| | - Kaikai Zheng
- College of Polymer Science and Engineering, College of Biomedical Engineering, State Key Laboratory of Polymer Materials and Engineering Sichuan University, Chengdu 610065, China
| | - Peng Wang
- College of Polymer Science and Engineering, College of Biomedical Engineering, State Key Laboratory of Polymer Materials and Engineering Sichuan University, Chengdu 610065, China
| | - Baihai Su
- Department of Nephrology, Kidney Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiang Wei
- College of Polymer Science and Engineering, College of Biomedical Engineering, State Key Laboratory of Polymer Materials and Engineering Sichuan University, Chengdu 610065, China
| | - Xiaojing Liu
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, and Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration, Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
| |
Collapse
|
4
|
Gong S, Li Y, Yan K, Shi Z, Leng J, Bao Y, Ning K. The Crosstalk Between Endothelial Cells, Smooth Muscle Cells, and Macrophages in Atherosclerosis. Int J Mol Sci 2025; 26:1457. [PMID: 40003923 PMCID: PMC11855868 DOI: 10.3390/ijms26041457] [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: 12/31/2024] [Revised: 02/02/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory vascular disease closely tied to cellular metabolism. Recent genome-wide association study data have suggested the significant roles of endothelial cells, smooth muscle cells, and macrophages in the regression and exacerbation of AS. However, the impact of cellular crosstalk and cellular metabolic derangements on disease progression in AS is vaguely understood. In this review, we analyze the roles of the three cell types in AS. We also summarize the crosstalk between the two of them, and the associated molecules and consequences involved. In addition, we emphasize potential therapeutic targets and highlight the importance of the three-cell co-culture model and extracellular vesicles in AS-related research, providing ideas for future studies.
Collapse
Affiliation(s)
- Sihe Gong
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Yanni Li
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Kaijie Yan
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Zhonghong Shi
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Jing Leng
- Preclinical Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China;
| | - Yimin Bao
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Ke Ning
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; (S.G.); (Y.L.); (K.Y.); (Z.S.)
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| |
Collapse
|
5
|
Wang X, Wang Y, Yuan T, Wang H, Zeng Z, Tian L, Cui L, Guo J, Chen Y. Network pharmacology provides new insights into the mechanism of traditional Chinese medicine and natural products used to treat pulmonary hypertension. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156062. [PMID: 39305743 DOI: 10.1016/j.phymed.2024.156062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 05/31/2024] [Accepted: 09/14/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a rare cardiovascular disease with high morbidity and mortality rates. It is characterized by increased pulmonary arterial pressure. Current research into relevant therapeutic drugs and targets for PH, however, is insufficient still. Traditional Chinese medicine (TCM) and natural products have a long history as therapeutics for PH. Network pharmacology is an approach that integrates drug-target interactions and signaling pathways based on biomarkers information obtained from drug and disease databases. The concept of network pharmacology shows many similarities with the TCM philosophy. Network pharmacology help elucidate the mechanisms of TCM in PH. This review presents representative applications of network pharmacology in the study of the mechanisms of TCM and natural products for the treatment of PH. METHODS In this review, we used ("pulmonary hypertension" OR "pulmonary arterial hypertension" OR "chronic thromboembolic pulmonary hypertension") AND ("network pharmacology" OR "systematic pharmacology") as keywords to search for reports from PubMed, Web of Science, and Google Scholar databases from ten years ago. The studies were screened and those chosen are summarized here. The TCM and natural products inPH and their corresponding targets and signaling pathways are described. Additionally, we discuss the application of network pharmacology in the study of TCM in PH to provide insights for future application strategies. RESULTS Network pharmacology have shown that AKT-related pathways, HIF-1 signaling pathway, MAPK signaling pathway, TGF-β-Smad pathway, cell cycle-related pathways and inflammation-related pathways are the main signaling pathways enriched in the PH targets of TCM. Reservatrol, curcumol, genistin, formononetin, wogonin, luteolin, baicalein, berberine, triptolide and tanshinone llA are active ingredients specific for PH treatment. A number of databases and tools specific for the treatment of PH are used in network pharmacology and natural product research. CONCLUSION Through the reasonable combination of molecular docking, omics technology and bioinformatics technology, the mechanism of multi-targets can be explained more comprehensively. Analyzing the complex mechanism of TCM from the clinical perspective may be a potential development trend of network pharmacology. Combination of predicted targets and traditional pharmacology improves efficiency of drug development.
Collapse
Affiliation(s)
- Xinyue Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yichen Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Tianyi Yuan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Hongjuan Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zuomei Zeng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Leiyu Tian
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Lidan Cui
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jian Guo
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Yucai Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
6
|
Yang H, Yang J, Zheng X, Chen T, Zhang R, Chen R, Cao T, Zeng F, Liu Q. The Hippo Pathway in Breast Cancer: The Extracellular Matrix and Hypoxia. Int J Mol Sci 2024; 25:12868. [PMID: 39684583 DOI: 10.3390/ijms252312868] [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: 11/07/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
As one of the most prevalent malignant neoplasms among women globally, the optimization of therapeutic strategies for breast cancer has perpetually been a research hotspot. The tumor microenvironment (TME) is of paramount importance in the progression of breast cancer, among which the extracellular matrix (ECM) and hypoxia are two crucial factors. The alterations of these two factors are predominantly regulated by the Hippo signaling pathway, which promotes tumor invasiveness, metastasis, therapeutic resistance, and susceptibility. Hence, this review focuses on the Hippo pathway in breast cancer, specifically, how the ECM and hypoxia impact the biological traits and therapeutic responses of breast cancer. Moreover, the role of miRNAs in modulating ECM constituents was investigated, and hsa-miR-33b-3p was identified as a potential therapeutic target for breast cancer. The review provides theoretical foundations and potential therapeutic direction for clinical treatment strategies in breast cancer, with the aspiration of attaining more precise and effective treatment alternatives in the future.
Collapse
Affiliation(s)
- Hanyu Yang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Jiaxin Yang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Xiang Zheng
- School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Tianshun Chen
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Ranqi Zhang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Rui Chen
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Tingting Cao
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Fancai Zeng
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Qiuyu Liu
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
- Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| |
Collapse
|
7
|
Katoh K. Signal Transduction Mechanisms of Focal Adhesions: Src and FAK-Mediated Cell Response. FRONT BIOSCI-LANDMRK 2024; 29:392. [PMID: 39614431 DOI: 10.31083/j.fbl2911392] [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: 06/17/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 12/01/2024]
Abstract
Cell-to-substrate adhesion sites, also known as focal adhesion sites (FAs), are complexes of different proteins on the cell surface. FAs play important roles in communication between cells and the extracellular matrix (ECM), leading to signal transduction involving different proteins that ultimately produce the cell response. This cell response involves cell adhesion, migration, motility, cell survival, and cell proliferation. The most important component of FAs are integrins. Integrins are transmembrane proteins that receive signals from the ECM and communicate them to the cytoplasm, thus activating several downstream proteins in a signaling cascade. Cellular Proto-oncogene tyrosine-protein kinase Src (c-Src) and focal adhesion kinase (FAK) are non-receptor tyrosine kinases that functionally interact to promote crucial roles in FAs. c-Src is a tyrosine kinase, activated by autophosphorylation and, in turn, activates another important protein, FAK. Activated FAK directly interacts with the cytoplasmic domain of integrin and activates other FA proteins by attaching to them. These proteins activated by FAK then activate other downstream pathways such as mitogen-activated protein kinase (MAPK) and Akt pathways involved in cell proliferation, migration, and cell survival. Src can induce detachment of FAK from the integrin to increase the focal adhesion turnover. As a result, the Src-FAK complex in FAs is critical for cell adhesion and survival mechanisms. Overexpression of FA proteins has been linked to a variety of pathological disorders, including cancers, growth retardation, and bone deformities. FAK and Src are overexpressed in various cancers. This review, which focuses on the roles of two important signaling proteins, c-Src and FAK, attempts to provide a thorough and up-to-date examination of the signal transduction mechanisms mediated by focal adhesions. The author also described that FAK and Src may serve as potential targets for future therapies against diseases associated with their overexpression, such as certain types of cancer.
Collapse
Affiliation(s)
- Kazuo Katoh
- Laboratory of Human Anatomy and Cell Biology, Faculty of Health Sciences, Tsukuba University of Technology, 305-8521 Tsukuba, Japan
| |
Collapse
|
8
|
Lv J, Shi S, Fu Z, Wang Y, Duan C, Hu S, Wu H, Zhang B, Li Y, Song Q. Exploring the inflammation-related mechanisms of Lingguizhugan decoction on right ventricular remodeling secondary to pulmonary arterial hypertension based on integrated strategy using UPLC-HRMS, systems biology approach, and experimental validation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155879. [PMID: 39032277 DOI: 10.1016/j.phymed.2024.155879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 05/27/2024] [Accepted: 07/13/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) and the consequent right heart dysfunction persist with high morbidity and mortality, and the mechanisms and pharmacologic interventions for chronic right-sided heart failure (RHF) have not been adequately investigated. Research has shown that prolonged inflammation is critical in precipitating the progression of PAH-associated right heart pathology. Some research demonstrated that Lingguizhugan decoction (LGZGD), as a classical Chinese medicine formula, had beneficial effects in alleviating PAH and RHF, while its underlying mechanisms involved are not fully elucidated. PURPOSE Based on that, this study aims to investigate the effects and underlying mechanisms of LGZGD on PAH-induced RHF. STUDY DESIGN In this study, we identified the serum constituents and deciphered the potential anti-inflammatory mechanism and crucial components of LGZGD using combined approaches of UPLC-HRMS, transcriptomic analysis, and molecular docking techniques. Finally, we used in vivo experiments to verify the expression of key targets in the monocrotaline (MCT)-induced RHF model and the intervene effect of LGZGD. RESULTS Integrated strategies based on UPLC-HRMS and systems biology approach combined with in vivo experimental validation showed that LGZGD could improve right heart fibrosis and dysfunction via regulating diverse inflammatory signaling pathways and the activity of immune cells, including chemokine family CCL2, CXCR4, leukocyte integrins family ITGAL, ITGB2, and M2 macrophage infiltration, as well as lipid peroxidation-associated HMOX1, NOX4, and 4-HNE. CONCLUSION The present research demonstrated for the first time that LGZGD might improve PAH-induced RHF through multiple anti-inflammatory signaling and inhibition of ferroptosis, which could provide certain directions for future research in related fields.
Collapse
Affiliation(s)
- Jiayu Lv
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shuqing Shi
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhenyue Fu
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China; College of Traditional Chinese Medicine, Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Yajiao Wang
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chenglin Duan
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shaowei Hu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Huaqin Wu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bingxuan Zhang
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yumeng Li
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Qingqiao Song
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
9
|
Reang J, Sharma V, Yadav V, Tonk RK, Majeed J, Sharma A, Sharma PC. Redefining the significance of quinoline containing compounds as potent VEGFR-2 inhibitors for cancer therapy. Med Chem Res 2024; 33:1079-1099. [DOI: 10.1007/s00044-024-03252-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/04/2024] [Indexed: 01/03/2025]
|
10
|
Kong D, Liu J, Lu J, Zeng C, Chen H, Duan Z, Yu K, Zheng X, Zou P, Zhou L, Lv Y, Zeng Q, Lu L, Li J, He Y. HMGB2 Release Promotes Pulmonary Hypertension and Predicts Severity and Mortality of Patients With Pulmonary Arterial Hypertension. Arterioscler Thromb Vasc Biol 2024; 44:e172-e195. [PMID: 38572649 DOI: 10.1161/atvbaha.123.319916] [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: 07/24/2023] [Accepted: 03/18/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a progressive and life-threatening disease characterized by pulmonary vascular remodeling, which involves aberrant proliferation and apoptosis resistance of the pulmonary arterial smooth muscle cells (PASMCs), resembling the hallmark characteristics of cancer. In cancer, the HMGB2 (high-mobility group box 2) protein promotes the pro-proliferative/antiapoptotic phenotype. However, the function of HMGB2 in PH remains uninvestigated. METHODS Smooth muscle cell (SMC)-specific HMGB2 knockout or HMGB2-OE (HMGB2 overexpression) mice and HMGB2 silenced rats were used to establish hypoxia+Su5416 (HySu)-induced PH mouse and monocrotaline-induced PH rat models, respectively. The effects of HMGB2 and its underlying mechanisms were subsequently elucidated using RNA-sequencing and cellular and molecular biology analyses. Serum HMGB2 levels were measured in the controls and patients with pulmonary arterial (PA) hypertension. RESULTS HMGB2 expression was markedly increased in the PAs of patients with PA hypertension and PH rodent models and was predominantly localized in PASMCs. SMC-specific HMGB2 deficiency or silencing attenuated PH development and pulmonary vascular remodeling in hypoxia+Su5416-induced mice and monocrotaline-treated rats. SMC-specific HMGB2 overexpression aggravated hypoxia+Su5416-induced PH. HMGB2 knockdown inhibited PASMC proliferation in vitro in response to PDGF-BB (platelet-derived growth factor-BB). In contrast, HMGB2 protein stimulation caused the hyperproliferation of PASMCs. In addition, HMGB2 promoted PASMC proliferation and the development of PH by RAGE (receptor for advanced glycation end products)/FAK (focal adhesion kinase)-mediated Hippo/YAP (yes-associated protein) signaling suppression. Serum HMGB2 levels were significantly increased in patients with PA hypertension, and they correlated with disease severity, predicting worse survival. CONCLUSIONS Our findings indicate that targeting HMGB2 might be a novel therapeutic strategy for treating PH. Serum HMGB2 levels could serve as a novel biomarker for diagnosing PA hypertension and determining its prognosis.
Collapse
MESH Headings
- Animals
- HMGB2 Protein/genetics
- HMGB2 Protein/metabolism
- Humans
- Vascular Remodeling
- Male
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Disease Models, Animal
- Pulmonary Artery/metabolism
- Pulmonary Artery/physiopathology
- Pulmonary Artery/pathology
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Rats
- Mice, Inbred C57BL
- Mice
- Cell Proliferation
- Severity of Illness Index
- Signal Transduction
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Arterial Hypertension/physiopathology
- Rats, Sprague-Dawley
- Female
- Cells, Cultured
- Middle Aged
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/physiopathology
Collapse
Affiliation(s)
- Deping Kong
- Departments of Cardiology (D.K., J. Liu, C.Z., H.C., X.Z., P.Z., L.Z., J. Li, Y.H.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital (D.K., Z.D., Y.L., Q.Z.), Shanghai Jiao Tong University School of Medicine, China
| | - Jing Liu
- Departments of Cardiology (D.K., J. Liu, C.Z., H.C., X.Z., P.Z., L.Z., J. Li, Y.H.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Junmi Lu
- Pathology (J. Lu), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Cheng Zeng
- Departments of Cardiology (D.K., J. Liu, C.Z., H.C., X.Z., P.Z., L.Z., J. Li, Y.H.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Hao Chen
- Departments of Cardiology (D.K., J. Liu, C.Z., H.C., X.Z., P.Z., L.Z., J. Li, Y.H.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zhenzhen Duan
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital (D.K., Z.D., Y.L., Q.Z.), Shanghai Jiao Tong University School of Medicine, China
| | - Ke Yu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Guangdong, China (K.Y.)
| | - Xialei Zheng
- Departments of Cardiology (D.K., J. Liu, C.Z., H.C., X.Z., P.Z., L.Z., J. Li, Y.H.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Pu Zou
- Departments of Cardiology (D.K., J. Liu, C.Z., H.C., X.Z., P.Z., L.Z., J. Li, Y.H.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Liufang Zhou
- Departments of Cardiology (D.K., J. Liu, C.Z., H.C., X.Z., P.Z., L.Z., J. Li, Y.H.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Cardiovascular Medicine, The Affiliated Hospital of Youjiang Medical College for Nationalities, Baise, Guangxi, China (L.Z.)
| | - Yicheng Lv
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital (D.K., Z.D., Y.L., Q.Z.), Shanghai Jiao Tong University School of Medicine, China
| | - Qingye Zeng
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital (D.K., Z.D., Y.L., Q.Z.), Shanghai Jiao Tong University School of Medicine, China
| | - Lin Lu
- Department of Cardiology, Rui Jin Hospital (L.L.), Shanghai Jiao Tong University School of Medicine, China
| | - Jiang Li
- Departments of Cardiology (D.K., J. Liu, C.Z., H.C., X.Z., P.Z., L.Z., J. Li, Y.H.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yuhu He
- Departments of Cardiology (D.K., J. Liu, C.Z., H.C., X.Z., P.Z., L.Z., J. Li, Y.H.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
11
|
Wu Q, Yuan K, Yao Y, Yao J, Shao J, Meng Y, Wu P, Shi H. LAMC1 attenuates neuronal apoptosis via FAK/PI3K/AKT signaling pathway after subarachnoid hemorrhage. Exp Neurol 2024; 376:114776. [PMID: 38609046 DOI: 10.1016/j.expneurol.2024.114776] [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: 01/17/2024] [Revised: 03/20/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024]
Abstract
BACKGROUND AND PURPOSE The poor prognosis in patients with subarachnoid hemorrhage (SAH) is often attributed to neuronal apoptosis. Recent evidence suggests that Laminin subunit gamma 1 (LAMC1) is essential for cell survival and proliferation. However, the effects of LAMC1 on early brain injury after SAH and the underlying mechanisms are unknown. The current study aimed to reveal the anti-neuronal apoptotic effect and the potential mechanism of LAMC1 in the rat and in the in vitro SAH models. METHODS The SAH model of Sprague-Dawley rats was established by endovascular perforation. Recombinant LAMC1 (rLAMC1) was administered intranasally 30 min after modeling. LAMC1 small interfering RNA (LAMC1 siRNA), focal adhesion kinase (FAK)-specific inhibitor Y15 and PI3K-specific inhibitor LY294002 were administered before SAH modeling to explore the neuroprotection mechanism of rLAMC1. HT22 cells were cultured and stimulated by oxyhemoglobin to establish an in vitro model of SAH. Subsequently, SAH grades, neurobehavioral tests, brain water content, blood-brain barrier permeability, western blotting, immunofluorescence, TUNEL, and Fluoro-Jade C staining were performed. RESULTS The expression of endogenous LAMC1 was markedly decreased after SAH, both in vitro and in vivo. rLAMC1 significantly reduced the brain water content and blood-brain barrier permeability, improved short- and long-term neurobehavior, and decreased neuronal apoptosis. Furthermore, rLAMC1 treatment significantly increased the expression of p-FAK, p-PI3K, p-AKT, Bcl-XL, and Bcl-2 and decreased the expression of Bax and cleaved caspase -3. Conversely, knockdown of endogenous LAMC1 aggravated the neurological impairment, suppressed the expression of Bcl-XL and Bcl-2, and upregulated the expression of Bax and cleaved caspase-3. Additionally, the administration of Y15 and LY294002 abolished the protective roles of rLAMC1. In vitro, rLAMC1 significantly reduced neuronal apoptosis, and the protective effects were also abolished by Y15 and LY294002. CONCLUSION Exogenous LAMC1 treatment improved neurological deficits after SAH in rats, and attenuated neuronal apoptosis in both in vitro and in vivo SAH models, at least partially through the FAK/PI3K/AKT pathway.
Collapse
Affiliation(s)
- Qiaowei Wu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Kaikun Yuan
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yanting Yao
- Department of Neurosurgery, Beidahuang Group General Hospital, Harbin, Heilongjiang, China
| | - Jinbiao Yao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jiang Shao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yuxiao Meng
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Pei Wu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
| | - Huaizhang Shi
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
| |
Collapse
|
12
|
Gao J, Cheng J, Xie W, Zhang P, Liu X, Wang Z, Zhang B. Prospects of focal adhesion kinase inhibitors as a cancer therapy in preclinical and early phase study. Expert Opin Investig Drugs 2024; 33:639-651. [PMID: 38676368 DOI: 10.1080/13543784.2024.2348068] [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: 12/01/2023] [Accepted: 04/23/2024] [Indexed: 04/28/2024]
Abstract
INTRODUCTION FAK, a nonreceptor cytoplasmic tyrosine kinase, plays a crucial role in tumor metastasis, drug resistance, tumor stem cell maintenance, and regulation of the tumor microenvironment. FAK has emerged as a promising target for tumor therapy based on both preclinical and clinical data. AREAS COVERED This paper aims to summarize the molecular mechanisms underlying FAK's involvement in tumorigenesis and progression. Encouraging results have emerged from ongoing clinical trials of FAK inhibitors. Additionally, we present an overview of clinical trials for FAK inhibitors, examining their potential as promising treatments. The pertinent studies gathered from databases including PubMed, ClinicalTrials.gov. EXPERT OPINION Since the first finding in 1990s, targeting FAK has became the focus of interests in many pharmaceutical companies. Through 30 years' discovery, the industry and academy gradually realized the features of FAK target which may not be a driver gene but a solid defense system for the cancer initiation and development. Currently, the ongoing clinical regimens involving FAK inhibition are all the combination strategies in which FAK inhibitors can further strengthen the cancer cell killing effects of other testing agents. The emerging positive signal in clinical trials foresee targeting FAK as class will be an effective mean to fight against cancers.
Collapse
Affiliation(s)
| | | | - Wanyu Xie
- InxMed (Shanghai) Co. Ltd, Shanghai, China
| | - Ping Zhang
- InxMed (Shanghai) Co. Ltd, Shanghai, China
| | - Xuebin Liu
- InxMed (Shanghai) Co. Ltd, Shanghai, China
| | - Zaiqi Wang
- InxMed (Shanghai) Co. Ltd, Shanghai, China
| | | |
Collapse
|
13
|
Zhang S, Zhang Q, Lu Y, Chen J, Liu J, Li Z, Xie Z. Roles of Integrin in Cardiovascular Diseases: From Basic Research to Clinical Implications. Int J Mol Sci 2024; 25:4096. [PMID: 38612904 PMCID: PMC11012347 DOI: 10.3390/ijms25074096] [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: 02/23/2024] [Revised: 03/28/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Cardiovascular diseases (CVDs) pose a significant global health threat due to their complex pathogenesis and high incidence, imposing a substantial burden on global healthcare systems. Integrins, a group of heterodimers consisting of α and β subunits that are located on the cell membrane, have emerged as key players in mediating the occurrence and progression of CVDs by regulating the physiological activities of endothelial cells, vascular smooth muscle cells, platelets, fibroblasts, cardiomyocytes, and various immune cells. The crucial role of integrins in the progression of CVDs has valuable implications for targeted therapies. In this context, the development and application of various integrin antibodies and antagonists have been explored for antiplatelet therapy and anti-inflammatory-mediated tissue damage. Additionally, the rise of nanomedicine has enhanced the specificity and bioavailability of precision therapy targeting integrins. Nevertheless, the complexity of the pathogenesis of CVDs presents tremendous challenges for monoclonal targeted treatment. This paper reviews the mechanisms of integrins in the development of atherosclerosis, cardiac fibrosis, hypertension, and arrhythmias, which may pave the way for future innovations in the diagnosis and treatment of CVDs.
Collapse
Affiliation(s)
- Shuo Zhang
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (S.Z.); (Q.Z.); (Y.L.); (J.C.); (J.L.); (Z.L.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Qingfang Zhang
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (S.Z.); (Q.Z.); (Y.L.); (J.C.); (J.L.); (Z.L.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Yutong Lu
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (S.Z.); (Q.Z.); (Y.L.); (J.C.); (J.L.); (Z.L.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Jianrui Chen
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (S.Z.); (Q.Z.); (Y.L.); (J.C.); (J.L.); (Z.L.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Jinkai Liu
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (S.Z.); (Q.Z.); (Y.L.); (J.C.); (J.L.); (Z.L.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Zhuohan Li
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (S.Z.); (Q.Z.); (Y.L.); (J.C.); (J.L.); (Z.L.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Zhenzhen Xie
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (S.Z.); (Q.Z.); (Y.L.); (J.C.); (J.L.); (Z.L.)
| |
Collapse
|
14
|
Tsare EPG, Klapa MI, Moschonas NK. Protein-protein interaction network-based integration of GWAS and functional data for blood pressure regulation analysis. Hum Genomics 2024; 18:15. [PMID: 38326862 PMCID: PMC11465932 DOI: 10.1186/s40246-023-00565-6] [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: 08/08/2023] [Accepted: 11/12/2023] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND It is valuable to analyze the genome-wide association studies (GWAS) data for a complex disease phenotype in the context of the protein-protein interaction (PPI) network, as the related pathophysiology results from the function of interacting polyprotein pathways. The analysis may include the design and curation of a phenotype-specific GWAS meta-database incorporating genotypic and eQTL data linking to PPI and other biological datasets, and the development of systematic workflows for PPI network-based data integration toward protein and pathway prioritization. Here, we pursued this analysis for blood pressure (BP) regulation. METHODS The relational scheme of the implemented in Microsoft SQL Server BP-GWAS meta-database enabled the combined storage of: GWAS data and attributes mined from GWAS Catalog and the literature, Ensembl-defined SNP-transcript associations, and GTEx eQTL data. The BP-protein interactome was reconstructed from the PICKLE PPI meta-database, extending the GWAS-deduced network with the shortest paths connecting all GWAS-proteins into one component. The shortest-path intermediates were considered as BP-related. For protein prioritization, we combined a new integrated GWAS-based scoring scheme with two network-based criteria: one considering the protein role in the reconstructed by shortest-path (RbSP) interactome and one novel promoting the common neighbors of GWAS-prioritized proteins. Prioritized proteins were ranked by the number of satisfied criteria. RESULTS The meta-database includes 6687 variants linked with 1167 BP-associated protein-coding genes. The GWAS-deduced PPI network includes 1065 proteins, with 672 forming a connected component. The RbSP interactome contains 1443 additional, network-deduced proteins and indicated that essentially all BP-GWAS proteins are at most second neighbors. The prioritized BP-protein set was derived from the union of the most BP-significant by any of the GWAS-based or the network-based criteria. It included 335 proteins, with ~ 2/3 deduced from the BP PPI network extension and 126 prioritized by at least two criteria. ESR1 was the only protein satisfying all three criteria, followed in the top-10 by INSR, PTN11, CDK6, CSK, NOS3, SH2B3, ATP2B1, FES and FINC, satisfying two. Pathway analysis of the RbSP interactome revealed numerous bioprocesses, which are indeed functionally supported as BP-associated, extending our understanding about BP regulation. CONCLUSIONS The implemented workflow could be used for other multifactorial diseases.
Collapse
Affiliation(s)
- Evridiki-Pandora G Tsare
- Department of General Biology, School of Medicine, University of Patras, Patras, Greece
- Metabolic Engineering and Systems Biology Laboratory, Institute of Chemical Engineering Sciences, Foundation for Research and Technology-Hellas (FORTH/ICE-HT), Patras, Greece
| | - Maria I Klapa
- Metabolic Engineering and Systems Biology Laboratory, Institute of Chemical Engineering Sciences, Foundation for Research and Technology-Hellas (FORTH/ICE-HT), Patras, Greece.
| | - Nicholas K Moschonas
- Department of General Biology, School of Medicine, University of Patras, Patras, Greece.
- Metabolic Engineering and Systems Biology Laboratory, Institute of Chemical Engineering Sciences, Foundation for Research and Technology-Hellas (FORTH/ICE-HT), Patras, Greece.
| |
Collapse
|
15
|
Comarița IK, Tanko G, Anghelache IL, Georgescu A. The siRNA-mediated knockdown of AP-1 restores the function of the pulmonary artery and the right ventricle by reducing perivascular and interstitial fibrosis and key molecular players in cardiopulmonary disease. J Transl Med 2024; 22:137. [PMID: 38317144 PMCID: PMC10845748 DOI: 10.1186/s12967-024-04933-1] [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: 11/10/2023] [Accepted: 01/26/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Pulmonary hypertension (PH) is a complex multifactorial vascular pathology characterized by an increased pulmonary arterial pressure, vasoconstriction, remodelling of the pulmonary vasculature, thrombosis in situ and inflammation associated with right-side heart failure. Herein, we explored the potential beneficial effects of treatment with siRNA AP-1 on pulmonary arterial hypertension (PAH), right ventricular dysfunction along with perivascular and interstitial fibrosis in pulmonary artery-PA, right ventricle-RV and lung in an experimental animal model of monocrotaline (MCT)-induced PAH. METHODS Golden Syrian hamsters were divided into: (1) C group-healthy animals taken as control; (2) MCT group obtained by a single subcutaneous injection of 60 mg/kg MCT at the beginning of the experiment; (3) MCT-siRNA AP-1 group received a one-time subcutaneous dose of MCT and subcutaneous injections containing 100 nM siRNA AP-1, every two weeks. All animal groups received water and standard chow ad libitum for 12 weeks. RESULTS In comparison with the MCT group, siRNA AP-1 treatment had significant beneficial effects on investigated tissues contributing to: (1) a reduction in TGF-β1/ET-1/IL-1β/TNF-α plasma concentrations; (2) a reduced level of cytosolic ROS production in PA, RV and lung and notable improvements regarding the ultrastructure of these tissues; a decrease of inflammatory and fibrotic marker expressions in PA (COL1A/Fibronectin/Vimentin/α-SMA/CTGF/Calponin/MMP-9), RV and lung (COL1A/CTGF/Fibronectin/α-SMA/F-actin/OB-cadherin) and an increase of endothelial marker expressions (CD31/VE-cadherin) in PA; (4) structural and functional recoveries of the PA [reduced Vel, restored vascular reactivity (NA contraction, ACh relaxation)] and RV (enlarged internal cavity diameter in diastole, increased TAPSE and PRVOFs) associated with a decrease in systolic and diastolic blood pressure, and heart rate; (5) a reduced protein expression profile of AP-1S3/ pFAK/FAK/pERK/ERK and a significant decrease in the expression levels of miRNA-145, miRNA-210, miRNA-21, and miRNA-214 along with an increase of miRNA-124 and miRNA-204. CONCLUSIONS The siRNA AP-1-based therapy led to an improvement of pulmonary arterial and right ventricular function accompanied by a regression of perivascular and interstitial fibrosis in PA, RV and lung and a down-regulation of key inflammatory and fibrotic markers in MCT-treated hamsters.
Collapse
Affiliation(s)
- Ioana Karla Comarița
- Institute of Cellular Biology and Pathology 'Nicolae Simionescu' of Romanian Academy, Bucharest, Romania
| | - Gabriela Tanko
- Institute of Cellular Biology and Pathology 'Nicolae Simionescu' of Romanian Academy, Bucharest, Romania
| | | | - Adriana Georgescu
- Institute of Cellular Biology and Pathology 'Nicolae Simionescu' of Romanian Academy, Bucharest, Romania.
| |
Collapse
|
16
|
Niida T, Yuki H, Suzuki K, Kinoshita D, Fujimoto D, Nakajima A, McNulty I, Lee H, Tanriverdi K, Nakamura S, Jang IK. Proteomics associated with coronary high-risk plaques by optical coherence tomography. J Thromb Thrombolysis 2024; 57:204-211. [PMID: 38296868 DOI: 10.1007/s11239-023-02938-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/16/2023] [Indexed: 02/02/2024]
Abstract
Biomarkers are widely used for the diagnosis and monitoring of cardiovascular disease. However, markers for coronary high-risk plaques have not been identified. The aim of this study was to identify proteins specific to coronary high-risk plaques. Fifty-one patients (71.2 ± 11.1 years, male: 66.7%) who underwent intracoronary optical coherence tomography imaging and provided blood specimens for proteomic analysis were prospectively enrolled. A total of 1470 plasma proteins were analyzed per patient using the Olink® Explore 1536 Reagent Kit. In patients with thin-cap fibroatheroma, the protein expression of Calretinin (CALB2), Corticoliberin (CRH) and Alkaline phosphatase, placental type (ALPP) were significantly increased, while the expression of Neuroplastin (NPTN), Folate receptor gamma (FOLR3) and Serpin A12 (SERPINA12) were significantly decreased. In patients with macrophage infiltration, the protein expressions of Fatty acid-binding protein, intestinal (FABP2), and Fibroblast growth factor 21 (FGF21) were significantly decreased. In patients with lipid-rich plaques, the protein expression of Interleukin-17 C (IL17C) was significantly increased, while the expression of Fc receptor-like protein 3 (FCRL3) was significantly decreased. These proteins might be useful markers in identifying patients with coronary high-risk plaques. Clinical Trial Registration: https://www.umin.ac.jp/ctr/ , UMIN000041692.
Collapse
Affiliation(s)
- Takayuki Niida
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Haruhito Yuki
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Keishi Suzuki
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daisuke Kinoshita
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daichi Fujimoto
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Akihiro Nakajima
- Interventional Cardiology Unit, New Tokyo Hospital, Chiba, Japan
| | - Iris McNulty
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hang Lee
- Biostatistics Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kahraman Tanriverdi
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sunao Nakamura
- Interventional Cardiology Unit, New Tokyo Hospital, Chiba, Japan
| | - Ik-Kyung Jang
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Allan and Gill Gray Professor of Medicine, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, GRB 800, Boston, MA, 02114, USA.
| |
Collapse
|
17
|
Rowe CJ, Nwaolu U, Salinas D, Hong J, Nunez J, Lansford JL, McCarthy CF, Potter BK, Levi BH, Davis TA. Inhibition of focal adhesion kinase 2 results in a macrophage polarization shift to M2 which attenuates local and systemic inflammation and reduces heterotopic ossification after polysystem extremity trauma. Front Immunol 2023; 14:1280884. [PMID: 38116014 PMCID: PMC10728492 DOI: 10.3389/fimmu.2023.1280884] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/16/2023] [Indexed: 12/21/2023] Open
Abstract
Introduction Heterotopic ossification (HO) is a complex pathology often observed in combat injured casualties who have sustained severe, high energy polytraumatic extremity injuries. Once HO has developed, prophylactic therapies are limited outside of surgical excision. Tourniquet-induced ischemia injury (IR) exacerbates trauma-mediated musculoskeletal tissue injury, inflammation, osteogenic progenitor cell development and HO formation. Others have shown that focal adhesion kinase-2 (FAK2) plays a key role in regulating early inflammatory signaling events. Therefore, we hypothesized that targeting FAK2 prophylactically would mitigate extremity trauma induced IR inflammation and HO formation. Methods We tested whether the continuous infusion of a FAK2 inhibitor (Defactinib, PF-573228; 6.94 µg/kg/min for 14 days) can mitigate ectopic bone formation (HO) using an established blast-related extremity injury model involving femoral fracture, quadriceps crush injury, three hours of tourniquet-induced limb ischemia, and hindlimb amputation through the fracture site. Tissue inflammation, infiltrating cells, osteogenic progenitor cell content were assessed at POD-7. Micro-computed tomography imaging was used to quantify mature HO at POD-56. Results In comparison to vehicle control-treated rats, FAK2 administration resulted in no marked wound healing complications or weight loss. FAK2 treatment decreased HO by 43%. At POD-7, marked reductions in tissue proinflammatory gene expression and assayable osteogenic progenitor cells were measured, albeit no significant changes in expression patterns of angiogenic, chondrogenic and osteogenic genes. At the same timepoint, injured tissue from FAK-treated rats had fewer infiltrating cells. Additionally, gene expression analyses of tissue infiltrating cells resulted in a more measurable shift from an M1 inflammatory to an M2 anti-inflammatory macrophage phenotype in the FAK2 inhibitor-treated group. Discussion Our findings suggest that FAK2 inhibition may be a novel strategy to dampen trauma-induced inflammation and attenuate HO in patients at high risk as a consequence of severe musculoskeletal polytrauma.
Collapse
Affiliation(s)
- Cassie J. Rowe
- Cell Biology and Regenerative Medicine Program, Department of Surgery, Uniformed Services University, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Uloma Nwaolu
- Cell Biology and Regenerative Medicine Program, Department of Surgery, Uniformed Services University, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Daniela Salinas
- Cell Biology and Regenerative Medicine Program, Department of Surgery, Uniformed Services University, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Jonathan Hong
- Center for Organogenesis Research and Trauma, University of Texas Southwestern, Dallas, TX, United States
| | - Johanna Nunez
- Center for Organogenesis Research and Trauma, University of Texas Southwestern, Dallas, TX, United States
| | - Jefferson L. Lansford
- Cell Biology and Regenerative Medicine Program, Department of Surgery, Uniformed Services University, Bethesda, MD, United States
| | - Conor F. McCarthy
- Cell Biology and Regenerative Medicine Program, Department of Surgery, Uniformed Services University, Bethesda, MD, United States
| | - Benjamin K. Potter
- Cell Biology and Regenerative Medicine Program, Department of Surgery, Uniformed Services University, Bethesda, MD, United States
| | - Benjamin H. Levi
- Center for Organogenesis Research and Trauma, University of Texas Southwestern, Dallas, TX, United States
| | - Thomas A. Davis
- Cell Biology and Regenerative Medicine Program, Department of Surgery, Uniformed Services University, Bethesda, MD, United States
| |
Collapse
|
18
|
Sun Z, Zhang L, Yin K, Zang G, Qian Y, Mao X, Li L, Jing Q, Wang Z. SIRT3-and FAK-mediated acetylation-phosphorylation crosstalk of NFATc1 regulates N ε-carboxymethyl-lysine-induced vascular calcification in diabetes mellitus. Atherosclerosis 2023; 377:43-59. [PMID: 37392543 DOI: 10.1016/j.atherosclerosis.2023.06.969] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 06/21/2023] [Accepted: 06/23/2023] [Indexed: 07/03/2023]
Abstract
BACKGROUND AND AIMS Arterial calcification is the predictor of cardiovascular risk in diabetic patients. Nε-carboxymethyl-lysine (CML), a toxic metabolite, is associated with accelerated vascular calcification in diabetes mellitus (DM). However, the mechanism remains elusive. This study aims to explore the key regulators involved in CML-induced vascular calcification in DM. METHODS We used Western blot and immuno-staining to test the expression and localization of nuclear factor of activated T cells, cytoplasmic 1 (NFATc1) in human samples, a diabetic apolipoprotein E-deficient (ApoE-/-) mouse model, and a vascular smooth muscle cells (VSMC) model. Further, we confirmed the regulator of NFATc1 phosphorylation and acetylation induced by CML. The role of NFATc1 in VSMCs calcification and osteogenic differentiation was explored in vivo and in vitro. RESULTS In diabetic patients, CML and NFATc1 levels increased in the severe calcified anterior tibial arteries. CML significantly promoted NFATc1 expression and nuclear translocation in VSMCs and mouse aorta. Knockdown of NFATc1 significantly inhibited CML-induced calcification. CML promoted NFATc1 acetylation at K549 by downregulating sirtuin 3 (SIRT3), which antagonized the focal adhesion kinase (FAK) induced NFATc1 phosphorylation at the Y270 site. FAK and SIRT3 affected the nuclear translocation of NFATc1 by regulating the acetylation-phosphorylation crosstalk. NFATc1 dephosphorylation mutant Y270F and deacetylation mutant K549R had opposite effects on VSMC calcification. SIRT3 overexpression and FAK inhibitor could reverse CML-promoted VSMC calcification. CONCLUSIONS CML enhances vascular calcification in DM through NFATc1. In this process, CML increases NFATc1 acetylation by downregulating SIRT3 to antagonize FAK-induced NFATc1 phosphorylation.
Collapse
Affiliation(s)
- Zhen Sun
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Lili Zhang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Kai Yin
- Department of General Practice, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Guangyao Zang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yongjiang Qian
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiang Mao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Lihua Li
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| | - Qing Jing
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Innovation Center for Intervention of Chronic Disease and Promotion of Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China.
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| |
Collapse
|
19
|
Wang Y, Gao J, Zhang L, Yang R, Zhang Y, Shan L, Li X, Ma K. Bioinformatics analysis of lncRNA-related ceRNA networks in the peripheral blood lymphocytes of Kazakh patients with essential hypertension in Xinjiang. Front Cardiovasc Med 2023; 10:1155767. [PMID: 37396592 PMCID: PMC10311024 DOI: 10.3389/fcvm.2023.1155767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/25/2023] [Indexed: 07/04/2023] Open
Abstract
Objective Here, we aimed to investigate long non-coding RNA (lncRNA) expression characteristics in the peripheral blood lymphocytes of Xinjiang Kazakh people with essential hypertension and the underlying regulatory mechanisms of competing endogenous RNAs (ceRNA). Methods From April 2016 to May 2019, six Kazakh patients with essential hypertension and six Kazakh healthy participants were randomly selected from the inpatient and outpatient cardiology departments of the First Affiliated Hospital of Shihezi University Medical College, Xinjiang. After detecting the expression levels of lncRNA and mRNA in the peripheral blood lymphocytes using gene chip technology, their levels in the hypertensive group were compared with those in the control group. Six differentially expressed lncRNAs were randomly selected for real-time PCR to verify the accuracy and reliability of the gene chip results. GO functional clustering and KEGG pathway analyses were performed for differentially expressed genes. The ceRNA regulatory network of lncRNA-miRNA-mRNA was constructed, followed by visualization of the results. The expressions of miR-139-5p and DCBLD2 after PVT1 overexpression in 293T cells were detected by qRT-PCR and Western blot. Results In the test group, 396 and 511 differentially expressed lncRNAs and mRNAs, respectively, were screened out. The trend of real-time PCR results was consistent with that of the microarray results. The differentially expressed mRNAs were found to be primarily involved in the adhesion spot, leukocyte migration via endothelial cells, gap junction, actin cytoskeleton regulation, and extracellular matrix-receptor interaction signaling pathways. By constructing the ceRNA regulatory network, we found that lncRNA PVT1-miR-139-5p-DCBLD2 has a potential ceRNA regulatory mechanism involved in the development of essential hypertension in Xinjiang Kazakh people. In 293T cells, lncRNA PVT1 overexpression inhibited miR-139-5p and DCBLD2 levels. Conclusions Our findings indicate that differentially expressed lncRNAs may be involved in the development of essential hypertension. lncRNA PVT1-miR-139-5p-DCBLD2 was indicated to comprise a potential ceRNA regulatory mechanism involved in the development of essential hypertension in the Xinjiang Kazakh population. Thus, it may act as a novel screening marker or therapeutic target for essential hypertension in this population.
Collapse
Affiliation(s)
- Yan Wang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China
| | - Jie Gao
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi, China
| | - Liang Zhang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi, China
| | - Rui Yang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi, China
| | - Yingying Zhang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi, China
| | - Liya Shan
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi, China
| | - Xinzhi Li
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China
- Department of Pathophysiology, Shihezi University School of Medicine, Shihezi, China
| | - Ketao Ma
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi, China
| |
Collapse
|
20
|
Scabertopin Derived from Elephantopus scaber L. Mediates Necroptosis by Inducing Reactive Oxygen Species Production in Bladder Cancer In Vitro. Cancers (Basel) 2022; 14:cancers14235976. [PMID: 36497458 PMCID: PMC9738305 DOI: 10.3390/cancers14235976] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/15/2022] [Accepted: 12/01/2022] [Indexed: 12/11/2022] Open
Abstract
Bladder cancer remains one of the most common malignant tumors that threatens human health worldwide. It imposes a heavy burden on patients and society due to the high medical costs associated with its easy metastasis and recurrence. Although several treatment options for bladder cancer are available, their clinical efficacy remains unsatisfactory. Therefore, actively exploring new drugs and their mechanisms of action for the clinical treatment of bladder cancer is very important. Scabertopin is one of the major sesquiterpene lactones found in Elephantopus scaber L. Sesquiterpene lactones are thought to have fairly strong anti-cancer efficacy. However, the anticancer effect of sesquiterpenoid scabertopin on bladder cancer and its mechanism are still unclear. The aim of this study is to evaluate the antitumor activity of scabertopin in bladder cancer and its potential molecular mechanism in vitro. Our results suggest that scabertopin can induce RIP1/RIP3-dependent necroptosis in bladder cancer cells by promoting the production of mitochondrial reactive oxygen species (ROS), inhibit the expression of MMP-9 by inhibiting the FAK/PI3K/Akt signaling pathway, and ultimately inhibit the migration and invasion ability of bladder cancer cells. At the same time, we also demonstrated that the half-inhibition concentration (IC50) of scabertopin on various bladder cancer cell lines (J82, T24, RT4 and 5637) is much lower than that on human ureteral epithelial immortalized cells (SV-HUC-1). The above observations indicate that scabertopin is a potential therapeutic agent for bladder cancer that acts by inducing necroptosis and inhibiting metastasis.
Collapse
|
21
|
Identification of Signal Pathways and Hub Genes of Pulmonary Arterial Hypertension by Bioinformatic Analysis. Can Respir J 2022; 2022:1394088. [PMID: 36072642 PMCID: PMC9444450 DOI: 10.1155/2022/1394088] [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: 05/26/2022] [Accepted: 07/19/2022] [Indexed: 11/30/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive and complex pulmonary vascular disease with poor prognosis. The aim of this study was to provide a new understanding of the pathogenesis of disease and potential treatment targets for patients with PAH based on multiple-microarray analysis.Two microarray datasets (GSE53408 and GSE113439) downloaded from the Gene Expression Omnibus (GEO) database were analysed. All the raw data were processed by R, and differentially expressed genes (DEGs) were screened out by the “limma” package. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed and visualized by R and Cytoscape software. Protein-protein interactions (PPI) of DEGs were analysed based on the NetworkAnalyst online tool. A total of 442 upregulated DEGs and 84 downregulated DEGs were identified. GO enrichment analysis showed that these DEGs were mainly enriched in mitotic nuclear division, organelle fission, chromosome segregation, nuclear division, and sister chromatid segregation. Significant KEGG pathway enrichment included ribosome biogenesis in eukaryotes, RNA transport, proteoglycans in cancer, dilated cardiomyopathy, rheumatoid arthritis, vascular smooth muscle contraction, focal adhesion, regulation of the actin cytoskeleton, and hypertrophic cardiomyopathy. The PPI network identified 10 hub genes including HSP90AA1, CDC5L, MDM2, LRRK2, CFTR, IQGAP1, CAND1, TOP2A, DDX21, and HIF1A. We elucidated potential biomarkers and therapeutic targets for PAH by bioinformatic analysis, which provides a theoretical basis for future study.
Collapse
|
22
|
Therapeutic targeting of mechanical stretch-induced FAK/ERK signaling by fisetin in hypertrophic scars. Eur J Pharmacol 2022; 932:175228. [PMID: 36007606 DOI: 10.1016/j.ejphar.2022.175228] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 08/15/2022] [Accepted: 08/17/2022] [Indexed: 11/23/2022]
Abstract
Hypertrophic scarring is a complex fibrotic disease with few treatment options. Mechanical stress has been proven to be crucial for hypertrophic scar (HS) formation. Here, we showed that the flavonoid small molecule fisetin, could dramatically ameliorate HS formation in a mechanical stretch-induced mouse model. In addition, in vitro and in vivo studies demonstrated that fisetin inhibited the stretch-induced profibrotic effects by suppressing the proliferation, activation, and collagen production of fibroblasts. Mechanistically, we revealed that fisetin obviously downregulated mechanical stretch-induced the phosphorylation of FAK and ERK, and reduced nuclear localization of ERK. This bioactivity of fisetin may result from its selective binding to the catalytic region of FAK, which was suggested by the molecular docking study and kinase binding assay. Taken together, these findings suggest that fisetin is a promising agent for the treatment of hypertrophic scars and other excessive fibrotic diseases.
Collapse
|
23
|
Wan Z, Wang K, Yin X, Guo X, Cheng G, Pan J. Renal Abscess Caused by Crizotinib: A Rare Case Report. Front Oncol 2022; 12:920990. [PMID: 35875128 PMCID: PMC9300938 DOI: 10.3389/fonc.2022.920990] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/08/2022] [Indexed: 11/13/2022] Open
Abstract
Crizotinib is a tyrosine kinase inhibitor that has been found to be effective in the treatment of c-ros oncogene 1-positive non-small cell lung cancer. Although this targeted agent for treating cancer has shown superiority to standard chemotherapy in some ways, this drug has adverse effects, such as the development of renal abscesses. Some associated renal damage may disappear with crizotinib withdrawal. Hence, we present the case of a 58-year-old man with non-small cell lung cancer on crizotinib therapy who developed bilateral renal abnormal space-occupying lesions, successively which were difficult to identify using various imaging methods; even PET-CT highly suspected the right renal masses as malignant. Finally, the right renal lesions were confirmed as renal abscesses by postoperative pathology. The left renal lesion was considered as renal cysts through the lesion disappearing after crizotinib withdrawal. There have been very few reports in this respect, especially proved by various methods and confirmed by postoperative pathology. It is important to recognize this drug-related complication in order to avoid incorrect diagnosis and inadequate therapy. It is necessary to monitor renal changes after taking crizotinib.
Collapse
Affiliation(s)
- Zhaojun Wan
- Department of Oncology, Rizhao Hospital of Traditional Chinese Medicine, Rizhao, China
- *Correspondence: Zhaojun Wan,
| | - Kai Wang
- Department of Oncology, The People’s Hospital of Rizhao City, Rizhao, China
| | - Xiangfu Yin
- Department of Pediatrics, Rizhao Hospital of Traditional Chinese Medicine, Rizhao, China
| | - Xiangting Guo
- Department of Rheumatology and Immunology, The People’s Hospital of Rizhao City, Rizhao, China
| | - Guoli Cheng
- Department of Oncology, Rizhao Hospital of Traditional Chinese Medicine, Rizhao, China
| | - Jihong Pan
- Department of Oncology, Rizhao Hospital of Traditional Chinese Medicine, Rizhao, China
| |
Collapse
|
24
|
Soleimani AA, Ghasmpour G, Mohammadi A, Gholizadeh M, Abkenar BR, Najafi M. Focal adhesion kinase-related pathways may be suppressed by metformin in vascular smooth muscle cells in high glucose conditions. Endocrinol Diabetes Metab 2022; 5:e351. [PMID: 35633523 PMCID: PMC9258994 DOI: 10.1002/edm2.351] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 05/09/2022] [Accepted: 05/13/2022] [Indexed: 12/16/2022] Open
Abstract
Introduction Cardiovascular diseases are known as one of the important causes of death in patients with diabetes mellitus. Metformin is used as an oral medication for reducing blood sugar. In this study, the effects of metformin were investigated on the FAK gene expression levels, pFAK protein values, cell viability and migration rate of VSMCs in high glucose conditions. Materials and methods The FAK gene expression levels and pFAK protein values were evaluated in VSMCs treated with different doses of metformin (1, 5 and 7 mM), based on cell viability using RT‐qPCR, western blotting and MTT techniques. The cellular migration was evaluated by scratch assay. Results The FAK gene expression levels reduced significantly in metformin‐treated VSMCs at 24 h and 48 h periods (p < .0008 and p < .0001, respectively). The pFAK protein values reduced significantly at 24 h (5 mM and 7 mM metformin doses) and 48 h periods (p < .001). In agreement with pFAK protein values, cellular migration reduced significantly at 24 h and 48 h periods (p < .001). Conclusion The results showed that metformin may suppress the proliferation and migration of VSMCs via FAK‐related pathways and may retard the progression of vessel stenosis in diabetes.
Collapse
Affiliation(s)
- Ali Akbar Soleimani
- Clinical Biochemistry Department, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ghasem Ghasmpour
- Clinical Biochemistry Department, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Asghar Mohammadi
- Clinical Biochemistry Department, Faculty of Medicine, Tarbiat Mdares University, Tehran, Iran
| | - Masoomeh Gholizadeh
- Clinical Biochemistry Department, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Golestan, Iran
| | - Borhan Rahimi Abkenar
- Clinical Biochemistry Department, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Najafi
- Clinical Biochemistry Department, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Wen D, Gao Y, Ho C, Yu L, Zhang Y, Lyu G, Hu D, Li Q, Zhang Y. Focusing on Mechanoregulation Axis in Fibrosis: Sensing, Transduction and Effecting. Front Mol Biosci 2022; 9:804680. [PMID: 35359592 PMCID: PMC8963247 DOI: 10.3389/fmolb.2022.804680] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 02/09/2022] [Indexed: 11/24/2022] Open
Abstract
Fibrosis, a pathologic process featured by the excessive deposition of connective tissue components, can affect virtually every organ and has no satisfactory therapy yet. Fibrotic diseases are often associated with organ dysfunction which leads to high morbidity and mortality. Biomechanical stmuli and the corresponding cellular response havebeen identified in fibrogenesis, as the fibrotic remodeling could be seen as the incapacity to reestablish mechanical homeostasis: along with extracellular matrix accumulating, the physical property became more “stiff” and could in turn induce fibrosis. In this review, we provide a comprehensive overview of mechanoregulation in fibrosis, from initialing cellular mechanosensing to intracellular mechanotransduction and processing, and ends up in mechanoeffecting. Our contents are not limited to the cellular mechanism, but further expand to the disorders involved and current clinical trials, providing an insight into the disease and hopefully inspiring new approaches for the treatment of tissue fibrosis.
Collapse
Affiliation(s)
- Dongsheng Wen
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ya Gao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chiakang Ho
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Yu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuguang Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guozhong Lyu
- Department of Burns and Plastic Surgery, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Dahai Hu
- Burns Centre of PLA, Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Qingfeng Li, ; Yifan Zhang,
| | - Yifan Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Qingfeng Li, ; Yifan Zhang,
| |
Collapse
|
26
|
Pomella S, Cassandri M, Braghini MR, Marampon F, Alisi A, Rota R. New Insights on the Nuclear Functions and Targeting of FAK in Cancer. Int J Mol Sci 2022; 23:ijms23041998. [PMID: 35216114 PMCID: PMC8874710 DOI: 10.3390/ijms23041998] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/03/2022] [Accepted: 02/09/2022] [Indexed: 02/01/2023] Open
Abstract
Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase over-expressed and activated in both adult and pediatric cancers, where it plays important roles in the regulation of pathogenesis and progression of the malignant phenotype. FAK exerts its functions in cancer by two different ways: a kinase activity in the cytoplasm, mainly dependent on the integrin signaling, and a scaffolding activity into the nucleus by networking with different gene expression regulators. For this reason, FAK has to be considered a target with high therapeutic values. Indeed, evidence suggests that FAK targeting could be effective, either alone or in combination, with other already available treatments. Here, we propose an overview of the novel insights about FAK’s structure and nuclear functions, with a special focus on the recent findings concerning the roles of this protein in cancer. Additionally, we provide a recent update on FAK inhibitors that are currently in clinical trials for patients with cancer, and discuss the challenge and future directions of drug-based anti-FAK targeted therapies.
Collapse
Affiliation(s)
- Silvia Pomella
- Department of Oncohematology, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (S.P.); (M.C.)
| | - Matteo Cassandri
- Department of Oncohematology, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (S.P.); (M.C.)
- Department of Radiotherapy, Policlinico Umberto I, Sapienza University of Rome, 00185 Rome, Italy;
| | - Maria Rita Braghini
- Unit of Molecular Genetics of Complex Phenotypes, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy;
| | - Francesco Marampon
- Department of Radiotherapy, Policlinico Umberto I, Sapienza University of Rome, 00185 Rome, Italy;
| | - Anna Alisi
- Unit of Molecular Genetics of Complex Phenotypes, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy;
- Correspondence: (A.A.); (R.R.); Tel.: +39-06-68592186 (A.A.); +39-06-68592648 (R.R.)
| | - Rossella Rota
- Department of Oncohematology, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (S.P.); (M.C.)
- Correspondence: (A.A.); (R.R.); Tel.: +39-06-68592186 (A.A.); +39-06-68592648 (R.R.)
| |
Collapse
|
27
|
Maudsley S, Leysen H, van Gastel J, Martin B. Systems Pharmacology: Enabling Multidimensional Therapeutics. COMPREHENSIVE PHARMACOLOGY 2022:725-769. [DOI: 10.1016/b978-0-12-820472-6.00017-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
28
|
OUP accepted manuscript. Brain 2022; 145:3179-3186. [DOI: 10.1093/brain/awac107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/25/2022] [Accepted: 03/13/2022] [Indexed: 11/15/2022] Open
|
29
|
Liu S, Lin Z. Vascular Smooth Muscle Cells Mechanosensitive Regulators and Vascular Remodeling. J Vasc Res 2021; 59:90-113. [PMID: 34937033 DOI: 10.1159/000519845] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 09/23/2021] [Indexed: 11/19/2022] Open
Abstract
Blood vessels are subjected to mechanical loads of pressure and flow, inducing smooth muscle circumferential and endothelial shear stresses. The perception and response of vascular tissue and living cells to these stresses and the microenvironment they are exposed to are critical to their function and survival. These mechanical stimuli not only cause morphological changes in cells and vessel walls but also can interfere with biochemical homeostasis, leading to vascular remodeling and dysfunction. However, the mechanisms underlying how these stimuli affect tissue and cellular function, including mechanical stimulation-induced biochemical signaling and mechanical transduction that relies on cytoskeletal integrity, are unclear. This review focuses on signaling pathways that regulate multiple biochemical processes in vascular mesangial smooth muscle cells in response to circumferential stress and are involved in mechanosensitive regulatory molecules in response to mechanotransduction, including ion channels, membrane receptors, integrins, cytoskeletal proteins, nuclear structures, and cascades. Mechanoactivation of these signaling pathways is closely associated with vascular remodeling in physiological or pathophysiological states.
Collapse
Affiliation(s)
- Shangmin Liu
- Ji Hua Institute of Biomedical Engineering Technology, Ji Hua Laboratory, Foshan, China, .,Medical Research Center, Guangdong Academy of Medical Sciences, Guangdong General Hospital, Guangzhou, China,
| | - Zhanyi Lin
- Ji Hua Institute of Biomedical Engineering Technology, Ji Hua Laboratory, Foshan, China.,Institute of Geriatric Medicine, Guangdong Academy of Medical Sciences, Guangdong General Hospital, Guangzhou, China
| |
Collapse
|
30
|
Hou S, Wang J, Li W, Hao X, Hang Q. Roles of Integrins in Gastrointestinal Cancer Metastasis. Front Mol Biosci 2021; 8:708779. [PMID: 34869579 PMCID: PMC8634653 DOI: 10.3389/fmolb.2021.708779] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
Integrins are a large family of heterodimeric transmembrane receptors which mediate cell adhesion and transmit signals to the cell interior. The mechanistic roles of integrins have long been an enigma in cancer, given its complexity in regulating different cellular behaviors. Recently, however, increasing research is providing new insights into its function and the underlying mechanisms, which collectively include the influences of altered integrin expression on the aberrant signaling pathways and cancer progression. Many studies have also demonstrated the potentiality of integrins as therapeutic targets in cancer treatment. In this review, we have summarized these recent reports and put a particular emphasis on the dysregulated expression of integrins and how they regulate related signaling pathways to facilitate the metastatic progression of gastrointestinal cancer, including gastric cancer (GC) and colorectal cancer (CRC), which will address the crucial roles of integrins in gastrointestinal cancer.
Collapse
Affiliation(s)
- Sicong Hou
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Jiaxin Wang
- Department of Clinical Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Wenqian Li
- Department of Clinical Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Xin Hao
- Department of Clinical Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Qinglei Hang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
31
|
Integrin αv and Vitronectin Prime Macrophage-Related Inflammation and Contribute the Development of Dry Eye Disease. Int J Mol Sci 2021; 22:ijms22168410. [PMID: 34445121 PMCID: PMC8395123 DOI: 10.3390/ijms22168410] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/31/2021] [Accepted: 08/03/2021] [Indexed: 12/27/2022] Open
Abstract
Cell signaling mediated by the αv integrin plays a pivotal role in macrophage activation in various inflammatory processes, but its involvement in the pathogenesis of dry eye disease (DED) remains unclear. In a murine model of DED, we found increased αv integrin expression in ocular surface macrophages. The αv integrins inhibitor c(RGDfK) ameliorated the corneal damage caused by DED, suggesting a pathogenic role for αv integrin. Because tear hyperosmolarity induces ocular inflammation in DED, a hyperosmolar culture of murine bone marrow-derived macrophages (BMDMs) is used to reproduce inflammation in vitro. However, the expression of proinflammatory cytokine mRNA was minimal, even though αv integrin was induced. In searching for components that are involved in αv integrin-mediated inflammation but that are missing from the culture model, we showed that the levels of vitronectin (VTN), a binding ligand of αv integrins, were increased in the tear fluid and conjunctival stroma of DED animals. The addition of VTN prominently enhanced hyperosmolarity-induced inflammation in BMDMs. Mechanistically, we showed that VTN/αv integrins mediated NF-κB activation to induce inflammatory gene expression in the BMDMs. Our findings indicate that interaction the of VTN with αv integrins is a crucial step in the inflammatory process in DED and suggests a novel therapeutic target.
Collapse
|
32
|
Murphy JM, Jeong K, Cioffi DL, Campbell PM, Jo H, Ahn EYE, Lim STS. Focal Adhesion Kinase Activity and Localization is Critical for TNF-α-Induced Nuclear Factor-κB Activation. Inflammation 2021; 44:1130-1144. [PMID: 33527321 PMCID: PMC8326009 DOI: 10.1007/s10753-020-01408-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/16/2020] [Accepted: 12/22/2020] [Indexed: 10/22/2022]
Abstract
While sustained nuclear factor-κB (NF-κB) activation is critical for proinflammatory molecule expression, regulators of NF-κB activity during chronic inflammation are not known. We investigated the role of focal adhesion kinase (FAK) on sustained NF-κB activation in tumor necrosis factor-α (TNF-α)-stimulated endothelial cells (ECs) both in vitro and in vivo. We found that FAK inhibition abolished TNF-α-mediated sustained NF-κB activity in ECs by disrupting formation of TNF-α receptor complex-I (TNFRC-I). Additionally, FAK inhibition diminished recruitment of receptor-interacting serine/threonine-protein kinase 1 (RIPK1) and the inhibitor of NF-κB (IκB) kinase (IKK) complex to TNFRC-I, resulting in elevated stability of IκBα protein. In mice given TNF-α, pharmacological and genetic FAK inhibition blocked TNF-α-induced IKK-NF-κB activation in aortic ECs. Mechanistically, TNF-α activated and redistributed FAK from the nucleus to the cytoplasm, causing elevated IKK-NF-κB activation. On the other hand, FAK inhibition trapped FAK in the nucleus of ECs even upon TNF-α stimulation, leading to reduced IKK-NF-κB activity. Together, these findings support a potential use for FAK inhibitors in treating chronic inflammatory diseases.
Collapse
Affiliation(s)
- James M Murphy
- Department of Biochemistry and Molecular Biology, University of South Alabama College of Medicine, 5851 N. USA Drive, Room 2366, Mobile, AL, 36688, USA
| | - Kyuho Jeong
- Department of Biochemistry and Molecular Biology, University of South Alabama College of Medicine, 5851 N. USA Drive, Room 2366, Mobile, AL, 36688, USA
| | - Donna L Cioffi
- Department of Biochemistry and Molecular Biology, University of South Alabama College of Medicine, 5851 N. USA Drive, Room 2366, Mobile, AL, 36688, USA
| | - Pamela Moore Campbell
- Department of Pathology, University of South Alabama College of Medicine, Mobile, AL, 36617, USA
| | - Hanjoong Jo
- Wallace H. Coulter Department of Bioengineering, Emory University and Georgia Institute of Technology, Atlanta, GA, 30322, USA
| | - Eun-Young Erin Ahn
- Department of Pathology, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Ssang-Taek Steve Lim
- Department of Biochemistry and Molecular Biology, University of South Alabama College of Medicine, 5851 N. USA Drive, Room 2366, Mobile, AL, 36688, USA.
| |
Collapse
|
33
|
Zalewski DP, Ruszel KP, Stępniewski A, Gałkowski D, Bogucki J, Kołodziej P, Szymańska J, Płachno BJ, Zubilewicz T, Feldo M, Kocki J, Bogucka-Kocka A. Identification of Transcriptomic Differences between Lower Extremities Arterial Disease, Abdominal Aortic Aneurysm and Chronic Venous Disease in Peripheral Blood Mononuclear Cells Specimens. Int J Mol Sci 2021; 22:3200. [PMID: 33801150 PMCID: PMC8004090 DOI: 10.3390/ijms22063200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/14/2021] [Accepted: 03/18/2021] [Indexed: 01/10/2023] Open
Abstract
Several human tissues are investigated in studies of molecular biomarkers associated with diseases development. Special attention is focused on the blood and its components due to combining abundant information about systemic responses to pathological processes as well as high accessibility. In the current study, transcriptome profiles of peripheral blood mononuclear cells (PBMCs) were used to compare differentially expressed genes between patients with lower extremities arterial disease (LEAD), abdominal aortic aneurysm (AAA) and chronic venous disease (CVD). Gene expression patterns were generated using the Ion S5XL next-generation sequencing platform and were analyzed using DESeq2 and UVE-PLS methods implemented in R programming software. In direct pairwise analysis, 21, 58 and 10 differentially expressed genes were selected from the comparison of LEAD vs. AAA, LEAD vs. CVD and AAA vs. CVD patient groups, respectively. Relationships between expression of dysregulated genes and age, body mass index, creatinine levels, hypertension and medication were identified using Spearman rank correlation test and two-sided Mann-Whitney U test. The functional analysis, performed using DAVID website tool, provides potential implications of selected genes in pathological processes underlying diseases studied. Presented research provides new insight into differences of pathogenesis in LEAD, AAA and CVD, and selected genes could be considered as potential candidates for biomarkers useful in diagnosis and differentiation of studied diseases.
Collapse
Affiliation(s)
- Daniel P. Zalewski
- Chair and Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland;
| | - Karol P. Ruszel
- Chair of Medical Genetics, Department of Clinical Genetics, Medical University of Lublin, 11 Radziwiłłowska St., 20-080 Lublin, Poland; (K.P.R.); (J.K.)
| | - Andrzej Stępniewski
- Ecotech Complex Analytical and Programme Centre for Advanced Environmentally Friendly Technologies, University of Marie Curie-Skłodowska, 39 Głęboka St., 20-612 Lublin, Poland;
| | - Dariusz Gałkowski
- Department of Pathology and Laboratory Medicine, Rutgers-Robert Wood Johnson Medical School, One Robert Wood Johnson Place, New Brunswick, NJ 08903-0019, USA;
| | - Jacek Bogucki
- Chair and Department of Organic Chemistry, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland;
| | - Przemysław Kołodziej
- Laboratory of Diagnostic Parasitology, Chair and Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland;
| | - Jolanta Szymańska
- Department of Integrated Paediatric Dentistry, Chair of Integrated Dentistry, Medical University of Lublin, 6 Chodźki St., 20-093 Lublin, Poland;
| | - Bartosz J. Płachno
- Department of Plant Cytology and Embryology, Institute of Botany, Faculty of Biology, Jagiellonian University in Kraków, 9 Gronostajowa St., 30-387 Cracow, Poland;
| | - Tomasz Zubilewicz
- Chair and Department of Vascular Surgery and Angiology, Medical University of Lublin, 11 Staszica St., 20-081 Lublin, Poland; (T.Z.); (M.F.)
| | - Marcin Feldo
- Chair and Department of Vascular Surgery and Angiology, Medical University of Lublin, 11 Staszica St., 20-081 Lublin, Poland; (T.Z.); (M.F.)
| | - Janusz Kocki
- Chair of Medical Genetics, Department of Clinical Genetics, Medical University of Lublin, 11 Radziwiłłowska St., 20-080 Lublin, Poland; (K.P.R.); (J.K.)
| | - Anna Bogucka-Kocka
- Chair and Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland;
| |
Collapse
|
34
|
Modulation of Insulin Sensitivity by Insulin-Degrading Enzyme. Biomedicines 2021; 9:biomedicines9010086. [PMID: 33477364 PMCID: PMC7830943 DOI: 10.3390/biomedicines9010086] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/14/2021] [Accepted: 01/15/2021] [Indexed: 12/15/2022] Open
Abstract
Insulin-degrading enzyme (IDE) is a highly conserved and ubiquitously expressed metalloprotease that degrades insulin and several other intermediate-size peptides. For many decades, IDE had been assumed to be involved primarily in hepatic insulin clearance, a key process that regulates availability of circulating insulin levels for peripheral tissues. Emerging evidence, however, suggests that IDE has several other important physiological functions relevant to glucose and insulin homeostasis, including the regulation of insulin secretion from pancreatic β-cells. Investigation of mice with tissue-specific genetic deletion of Ide in the liver and pancreatic β-cells (L-IDE-KO and B-IDE-KO mice, respectively) has revealed additional roles for IDE in the regulation of hepatic insulin action and sensitivity. In this review, we discuss current knowledge about IDE’s function as a regulator of insulin secretion and hepatic insulin sensitivity, both evaluating the classical view of IDE as an insulin protease and also exploring evidence for several non-proteolytic functions. Insulin proteostasis and insulin sensitivity have both been highlighted as targets controlling blood sugar levels in type 2 diabetes, so a clearer understanding the physiological functions of IDE in pancreas and liver could led to the development of novel therapeutics for the treatment of this disease.
Collapse
|
35
|
Dai L, Xie Y, Zhang W, Zhong X, Wang M, Jiang H, He Z, Liu X, Zeng H, Wang H. Weighted Gene Co-Expression Network Analysis Identifies ANGPTL4 as a Key Regulator in Diabetic Cardiomyopathy via FAK/SIRT3/ROS Pathway in Cardiomyocyte. Front Endocrinol (Lausanne) 2021; 12:705154. [PMID: 34616362 PMCID: PMC8488438 DOI: 10.3389/fendo.2021.705154] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/27/2021] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Diabetic cardiomyopathy (DbCM) is characterized by initial impairment of left ventricular relaxation followed by contractile dysfunction. Despite intensive research, the exact mechanism remains so far unsolved. METHODS We constructed weighted gene co-expression network analysis (WGCNA) to screen gene modules that were closely related with DbCM based on the GSE5606 dataset, which contained expression data of the cardiac left ventricle in a rodent model of streptozotocin (STZ)-induced DbCM. Then, the most related hub gene, angiopoietin-like 4 (ANGPTL4), was selected for functional ex vivo and in vitro assays. In our experiments, STZ-induced diabetic mice (C57BL/6J) and human cardiomyocytes (AC16) were used to study the functional roles and potential mechanisms of ANGPTL4 in DbCM. RESULTS WGCNA analysis revealed the yellow and green modules were most correlated with DbCM, and identified ANGPTL4 as one of the most significantly upregulated hub genes (ANGPTL4, ACOT1, DECR1, HMGCS2, and PDK4). Consistent with the bioinformatic analysis, the amount of ANGPTL4 was significantly upregulated in diabetic mouse heart. DbCM group, compared with the control group, had increased phosphorylation of focal adhesion kinase (FAK), reduced SIRT3 expression, increased SOD2 acetylation, upregulated NADPH oxidase activation, elevated reactive oxygen species (ROS) produciton, and enhanced apoptosis in the diabetic mouse heart. Moreover, ANGPTL4 induced apoptosis via FAK/SIRT3/ROS pathway in human cardiomyocytes (AC16) under high glucose condition in vitro.These effects were abrogated by treatment of two independent siRNA for ANGPTL4, whereas exogenous recombinant ANGPLT4 protein treatment exacerbated those effects in AC16. CONCLUSION We found ANGPTL4, ACOT1, DECR1, HMGCS2, and PDK4 were significantly increased in diabetic heart. ANGPTL4 could promote cardiac apoptosis via a FAK/SIRT3/ROS dependent signaling pathway in DbCM.
Collapse
Affiliation(s)
- Lei Dai
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Xie
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Wenjun Zhang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaodan Zhong
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Mengwen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Hongcheng Jiang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Zhen He
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaolei Liu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Hesong Zeng
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Hesong Zeng, ; Hongjie Wang,
| | - Hongjie Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Hesong Zeng, ; Hongjie Wang,
| |
Collapse
|
36
|
van Gastel J, Leysen H, Boddaert J, Vangenechten L, Luttrell LM, Martin B, Maudsley S. Aging-related modifications to G protein-coupled receptor signaling diversity. Pharmacol Ther 2020; 223:107793. [PMID: 33316288 DOI: 10.1016/j.pharmthera.2020.107793] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 11/26/2020] [Indexed: 02/06/2023]
Abstract
Aging is a highly complex molecular process, affecting nearly all tissue systems in humans and is the highest risk factor in developing neurodegenerative disorders such as Alzheimer's and Parkinson's disease, cardiovascular disease and Type 2 diabetes mellitus. The intense complexity of the aging process creates an incentive to develop more specific drugs that attenuate or even reverse some of the features of premature aging. As our current pharmacopeia is dominated by therapeutics that target members of the G protein-coupled receptor (GPCR) superfamily it may be prudent to search for effective anti-aging therapeutics in this fertile domain. Since the first demonstration of GPCR-based β-arrestin signaling, it has become clear that an enhanced appreciation of GPCR signaling diversity may facilitate the creation of therapeutics with selective signaling activities. Such 'biased' ligand signaling profiles can be effectively investigated using both standard molecular biological techniques as well as high-dimensionality data analyses. Through a more nuanced appreciation of the quantitative nature across the multiple dimensions of signaling bias that drugs possess, researchers may be able to further refine the efficacy of GPCR modulators to impact the complex aberrations that constitute the aging process. Identifying novel effector profiles could expand the effective pharmacopeia and assist in the design of precision medicines. This review discusses potential non-G protein effectors, and specifically their potential therapeutic suitability in aging and age-related disorders.
Collapse
Affiliation(s)
- Jaana van Gastel
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium; Faculty of Pharmacy, Biomedical and Veterinary Science, University of Antwerp, Antwerp, Belgium
| | - Hanne Leysen
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium; Faculty of Pharmacy, Biomedical and Veterinary Science, University of Antwerp, Antwerp, Belgium
| | - Jan Boddaert
- Molecular Pathology Group, Faculty of Medicine and Health Sciences, Laboratory of Cell Biology and Histology, Antwerp, Belgium
| | - Laura Vangenechten
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Louis M Luttrell
- Division of Endocrinology, Diabetes & Medical Genetics, Medical University of South Carolina, USA
| | - Bronwen Martin
- Faculty of Pharmacy, Biomedical and Veterinary Science, University of Antwerp, Antwerp, Belgium
| | - Stuart Maudsley
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium; Faculty of Pharmacy, Biomedical and Veterinary Science, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|