1
|
Miyata T. Overview: PAI-1 inhibitors and clinical applications. Biomed J 2025:100874. [PMID: 40403845 DOI: 10.1016/j.bj.2025.100874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 05/09/2025] [Accepted: 05/19/2025] [Indexed: 05/24/2025] Open
Abstract
Plasminogen activator inhibitor-1 (PAI-1) is a protein involved in the fibrinolytic system and has been reported to be involved in various pathologies such as fibrosis and inflammation. We have developed small molecule inhibitors of human PAI-1 for clinical applications. A clinical candidate compound (TM5509) was finally identified among over 1,400 derivatives of a hit compound (TM5275) searched by the X-ray crystal structure information of human PAI-1 and the in silico approach on a big virtual chemical library. From pre-clinical studies using our PAI-1 inhibitors, new therapeutic concepts for clinical applications, such as anti-cancer and anti-aging, have been conceived, some oh which have been tested in various investigator-initiated clinical trials.
Collapse
Affiliation(s)
- Toshio Miyata
- Tohoku University Graduate School of Medicine, Japan.
| |
Collapse
|
2
|
Lin M, Wang L, Guan B, Tang S, Lin L, Wu K, Huang Q, He G, Zhang Z, Gao R, Liu X, Liu X, Chen Z, Liu L. Effect of PAI-1 inhibitor on pancreatic islet function and hepatic insulin resistance in db/db mice. Biochem Pharmacol 2025; 237:116906. [PMID: 40158817 DOI: 10.1016/j.bcp.2025.116906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/27/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
Type 2 diabetes mellitus (T2DM) is a global health challenge, necessitating innovative antidiabetic treatments. Levels of plasminogen activator inhibitor-1 (PAI-1) are elevated in patients with T2DM and may be an important but underappreciated risk factor for diabetes. However, its relationship with T2DM remains unclear. To this end, we developed a potent and highly specific PAI-1 inhibitor named PAItrap3. We aimed to elucidate the metabolic effects of PAItrap3 using a preclinical db/db mouse model. PAItrap3 was administered to mice intravenously, followed by an assessment of biochemical markers, histopathological examination of the liver and pancreas, and evaluation of the expression of hepatic proteins integral to insulin signaling. PAItrap3 demonstrated potent efficacy in alleviating hyperglycemia and enhancing glycemic control. This therapeutic action was supported by its ability to enhance β-cell function, consequently mitigating β-cell apoptosis and preserving their integrity. Furthermore, PAItrap3 alleviated hepatic insulin resistance through the regulation of lipid and glucose metabolism, thereby maintaining the delicate homeostasis of systemic lipid and glucose metabolism. These findings suggest that PAItrap3 is a promising therapeutic candidate for T2DM. The multifaceted benefits of PAItrap3 highlight its potential to vastly improve the effectiveness and specificity of T2DM treatment paradigms.
Collapse
Affiliation(s)
- Menghua Lin
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350000, China
| | - Lijing Wang
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350000, China
| | - Binbin Guan
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350000, China
| | - Shuzhi Tang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fujian Academy, University of Chinese Academy of Sciences, Fuzhou, Fujian 350108, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lu Lin
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350000, China
| | - Kejun Wu
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350000, China
| | - Qintao Huang
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350000, China
| | - Guanlian He
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350000, China
| | - Zhouyangyang Zhang
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350000, China
| | - Ruonan Gao
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350000, China
| | - Xiaoying Liu
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350000, China
| | - XiaoHong Liu
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350000, China
| | - Zhuo Chen
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fujian Academy, University of Chinese Academy of Sciences, Fuzhou, Fujian 350108, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Libin Liu
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou 350000, China.
| |
Collapse
|
3
|
Su H, Lin X, Paredong A, Yao C, Zhang Y, Geng M, Guan Y, Gong L, Jiang F, Lv Q, Shou S, Jin H. IL-6/GATA2/SERPINE1 pathway is implicated in regulating cellular senescence after acute kidney injury. Front Mol Biosci 2025; 12:1538526. [PMID: 40007559 PMCID: PMC11850272 DOI: 10.3389/fmolb.2025.1538526] [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: 12/03/2024] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
Purpose Acute kidney injury (AKI) secondary to Rhabdomyolysis syndrome represents a life-threatening complication, characterized by notably high incidence and mortality rates. The role of cellular senescence in the progression of AKI has increasingly garnered attention in recent years. Our previous research has demonstrated that remote ischemic postconditioning (RIPC) can attenuate renal cellular senescence and elevation of serum level of interleukin-6 (IL-6) induced by ischemia-reperfusion injury following crush injury. The objective of this study is to investigate the specific role of IL-6 in Rhabdomyolysis-induced AKI (RM-AKI). Methods We established a mouse model of RM-AKI by intramuscular injection of glycerol and simulated RM-AKI at the cellular level by treating Hk-2 cells with myoglobin. Tocilizumab (TCZ), a humanized monoclonal antibody against the interleukin-6 (IL-6) receptor, is a key substance. IL-6, a multifunctional cytokine, plays a crucial role in the occurrence and development of various kidney diseases. It can promote inflammatory responses, cell proliferation, fibrosis, and other processes. TCZ exerts a protective effect on the kidneys by specifically binding to the IL-6 receptor and blocking the signal transduction of IL-6. Additionally, the levels of IL-6 were detected by employing ELISA kits. RNA sequencing analysis was performed on cells treated with myoglobin and tocilizumab. Flow cytometry was utilized to assess cell cycle distribution and the percentage of senescent cells. The expression levels of SERPINE1, GATA2, p53, and p21 were determined by real-time quantitative PCR and Western blot. Additionally, a dual-luciferase reporter gene assay was conducted to validate the binding effect of SERPINE1 and GATA2. Results Transcriptome Analysis revealed that genes including GATA2 and SERPINE1 were downregulated in HK-2 cells following tocilizumab treatment. Inhibition of the IL-6 receptor by tocilizumab in these cells led to a reduction in cellular senescence, accompanied by decreased of the cell cycle regulatory proteins P53 and P21 in mRNA and protein levels, while alleviating cell cycle arrest. Additionally, a dual-luciferase reporter assay confirmed that GATA2 binds to the promoter of SERPINE1 (PAI-1), thereby initiating its transcription. Conclusion The IL-6/GATA2/SERPINE1 pathway mediates cellular senescence after acute kidney injury, and inhibiting IL-6 can alleviate AKI-induced cellular senescence, providing an important basis for exploring new therapeutic strategies.
Collapse
Affiliation(s)
- Hongshuang Su
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaoxi Lin
- Department of Emergency and Critical Care Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ayinuer Paredong
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
- Department of Critical Care Medicine, Hotan District People’s Hospital, Tianjin, China
| | - Congcong Yao
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan Zhang
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Mengke Geng
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuqian Guan
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Lichao Gong
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Feng Jiang
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Qi Lv
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Songtao Shou
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Heng Jin
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
4
|
Shaikh SB, Balaya RDA, Dagamajalu S, Bhandary YP, Unwalla H, Prasad TSK, Rahman I. A signaling pathway map of plasminogen activator inhibitor-1 (PAI-1/SERPINE-1): a review of an innovative frontier in molecular aging and cellular senescence. Cell Commun Signal 2024; 22:544. [PMID: 39543686 PMCID: PMC11566301 DOI: 10.1186/s12964-024-01910-5] [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: 05/15/2024] [Accepted: 10/26/2024] [Indexed: 11/17/2024] Open
Abstract
Plasminogen activator inhibitor-1 (PAI-1) is a vital regulator of the fibrinolytic mechanism and has been intricately involved in various physiological and clinical processes, including cancer, thrombosis, and wound healing. The PAI-1 signaling pathway is multifaceted, encompassing numerous signaling molecules and nodes. Recent studies have revealed a novel contribution of PAI-1 during cellular senescence. This review introduces a pathway resource detailing the signaling network events mediated by PAI-1. The literature curated on the PAI-1 system was manually compiled from various published studies, our analysis presents a signaling pathway network of PAI-1, which includes various events like enzyme catalysis, molecular association, gene regulation, protein expression, and protein translocation. This signaling network aims to provide a detailed analysis of the existing understanding of the PAI-1 signaling pathway in the context of cellular senescence across various research models. By developing this pathway, we aspire to deepen our understanding of aging and senescence research, ultimately contributing to the pursuit of effective therapeutic approaches for these complex chronic diseases.
Collapse
Affiliation(s)
- Sadiya Bi Shaikh
- Department of Environmental Medicine, University of Rochester Medical Centre, 601 Elmwood Avenue, Box 850, Rochester, NY, 14642, USA
| | | | - Shobha Dagamajalu
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, Karnataka, 575018, India
| | - Yashodhar Prabhakar Bhandary
- Division for Molecular Biology, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, Karnataka, 575018, India
| | - Hoshang Unwalla
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | | | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Centre, 601 Elmwood Avenue, Box 850, Rochester, NY, 14642, USA.
| |
Collapse
|
5
|
Wang W, Chen Y, Shen Y, Chen J, Yao X, Cheng Y, Xu J, Ma L, Chen Y, Zhang C. Secretory Phenotype in Peripheral Blood Mononuclear Cells of Elderly Patients with Rheumatoid Arthritis. Rejuvenation Res 2024; 27:122-130. [PMID: 38814828 DOI: 10.1089/rej.2024.0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2024] Open
Abstract
This study aims to investigate the expression differences of peripheral blood mononuclear cells (PBMCs) in patients with elderly rheumatoid arthritis (ERA). Differentially expressed genes (DEGs) of PBMCs between young patients with RA (RA_Y) and elderly patients with RA (RA_A) were identified by RNA sequencing using the DESeq2 package, followed by bioinformatics analysis. The overlapped targets of the current DEGs and proteomic differentially expressed proteins (another set of unpublished data) were identified and further validated. The bioinformatics analysis revealed significant transcriptomic heterogeneity between RA_A and RA_Y. A total of 348 upregulated and 363 downregulated DEGs were identified. Gene functional enrichment analysis indicated that the DEGs, which represented senescence phenotype for patients with ERA, were enriched in pathways such as Phosphatidylinositol3 kinase/AKT serine-threonine protein kinase (PI3K/Akt) signaling, Mitogen-activated protein kinases (MAPK) signaling, toll-like receptor family, neutrophil degranulation, and immune-related pathways. Gene set enrichment analysis further confirmed the activation of humoral immune response pathways in RA_A. Quantitative polymerase chain reaction validated the expression of five representative DEGs such as SPTA1, SPTB, VNN1, TNXB, and KRT1 in PBMCs of patients with ERA. Patients with ERA have significant senescence phenotype differences versus the young patients. The DEGs identified may facilitate exploring the biomarkers of senescence in RA.
Collapse
Affiliation(s)
- Wenlong Wang
- Department of Rheumatology and Immunology, First People's Hospital of Wenling, Wenling, P.R. China
| | - Yanjuan Chen
- Department of Geriatrics and Division of Rheumatology and Research, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, P.R. China
| | - Yidi Shen
- Department of Rheumatology and Immunology, Seventh People's Hospital of ShangHai University of Traditional Chinese Medicine, Shanghai, P.R. China
| | - Jian Chen
- Division of Traditional Medicine and Shanghai Integrated Traditional Chinese and Western Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China
| | - Xiaoyang Yao
- Department of Clinical Laboratory, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China
| | - Yongjun Cheng
- Department of Rheumatology and Immunology, First People's Hospital of Wenling, Wenling, P.R. China
| | - Jinzhong Xu
- Department of Clinical Pharmacy, First People's Hospital of Wenling, Wenling, P.R. China
| | - Lisha Ma
- Department of Clinical Laboratory, First People's Hospital of Wenling, Wenling, P.R. China
| | - Yong Chen
- Department of Rheumatology and Immunology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Chuanfu Zhang
- Department of Rheumatology and Immunology, Seventh People's Hospital of ShangHai University of Traditional Chinese Medicine, Shanghai, P.R. China
| |
Collapse
|
6
|
Fujita M, Sasada M, Iyoda T, Fukai F. Involvement of Matricellular Proteins in Cellular Senescence: Potential Therapeutic Targets for Age-Related Diseases. Int J Mol Sci 2024; 25:6591. [PMID: 38928297 PMCID: PMC11204155 DOI: 10.3390/ijms25126591] [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: 05/29/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Senescence is a physiological and pathological cellular program triggered by various types of cellular stress. Senescent cells exhibit multiple characteristic changes. Among them, the characteristic flattened and enlarged morphology exhibited in senescent cells is observed regardless of the stimuli causing the senescence. Several studies have provided important insights into pro-adhesive properties of cellular senescence, suggesting that cell adhesion to the extracellular matrix (ECM), which is involved in characteristic morphological changes, may play pivotal roles in cellular senescence. Matricellular proteins, a group of structurally unrelated ECM molecules that are secreted into the extracellular environment, have the unique ability to control cell adhesion to the ECM by binding to cell adhesion receptors, including integrins. Recent reports have certified that matricellular proteins are closely involved in cellular senescence. Through this biological function, matricellular proteins are thought to play important roles in the pathogenesis of age-related diseases, including fibrosis, osteoarthritis, intervertebral disc degeneration, atherosclerosis, and cancer. This review outlines recent studies on the role of matricellular proteins in inducing cellular senescence. We highlight the role of integrin-mediated signaling in inducing cellular senescence and provide new therapeutic options for age-related diseases targeting matricellular proteins and integrins.
Collapse
Affiliation(s)
- Motomichi Fujita
- Department of Molecular Patho-Physiology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda 278-8510, Chiba, Japan
| | - Manabu Sasada
- Clinical Research Center in Hiroshima, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-Ku, Hiroshima 734-8551, Japan
| | - Takuya Iyoda
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, 1-1-1 Daigaku-Doori, Sanyo-Onoda 756-0884, Yamaguchi, Japan
| | - Fumio Fukai
- Department of Molecular Patho-Physiology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda 278-8510, Chiba, Japan
| |
Collapse
|
7
|
Ibrahim AA, Fujimura T, Uno T, Terada T, Hirano KI, Hosokawa H, Ohta A, Miyata T, Ando K, Yahata T. Plasminogen activator inhibitor-1 promotes immune evasion in tumors by facilitating the expression of programmed cell death-ligand 1. Front Immunol 2024; 15:1365894. [PMID: 38779680 PMCID: PMC11109370 DOI: 10.3389/fimmu.2024.1365894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024] Open
Abstract
Background Increased levels of plasminogen activator inhibitor-1 (PAI-1) in tumors have been found to correlate with poor clinical outcomes in patients with cancer. Although abundant data support the involvement of PAI-1 in cancer progression, whether PAI-1 contributes to tumor immune surveillance remains unclear. The purposes of this study are to determine whether PAI-1 regulates the expression of immune checkpoint molecules to suppresses the immune response to cancer and demonstrate the potential of PAI-1 inhibition for cancer therapy. Methods The effects of PAI-1 on the expression of the immune checkpoint molecule programmed cell death ligand 1 (PD-L1) were investigated in several human and murine tumor cell lines. In addition, we generated tumor-bearing mice and evaluated the effects of a PAI-1 inhibitor on tumor progression or on the tumor infiltration of cells involved in tumor immunity either alone or in combination with immune checkpoint inhibitors. Results PAI-1 induces PD-L1 expression through the JAK/STAT signaling pathway in several types of tumor cells and surrounding cells. Blockade of PAI-1 impedes PD-L1 induction in tumor cells, significantly reducing the abundance of immunosuppressive cells at the tumor site and increasing cytotoxic T-cell infiltration, ultimately leading to tumor regression. The anti-tumor effect elicited by the PAI-1 inhibitor is abolished in immunodeficient mice, suggesting that PAI-1 blockade induces tumor regression by stimulating the immune system. Moreover, combining a PAI-1 inhibitor with an immune checkpoint inhibitor significantly increases tumor regression. Conclusions PAI-1 protects tumors from immune surveillance by increasing PD-L1 expression; hence, therapeutic PAI-1 blockade may prove valuable in treating malignant tumors.
Collapse
Affiliation(s)
- Abd Aziz Ibrahim
- Translational Molecular Therapeutics Laboratory, Division of Host Defense Mechanism, Tokai University School of Medicine, Kanagawa, Japan
| | - Taku Fujimura
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomoko Uno
- Department of Hematology and Oncology, Tokai University School of Medicine, Kanagawa, Japan
| | - Tomoya Terada
- Translational Molecular Therapeutics Laboratory, Division of Host Defense Mechanism, Tokai University School of Medicine, Kanagawa, Japan
| | - Ken-ichi Hirano
- Department of Immunology, Tokai University School of Medicine, Kanagawa, Japan
| | - Hiroyuki Hosokawa
- Department of Immunology, Tokai University School of Medicine, Kanagawa, Japan
| | - Akio Ohta
- Department of Immunology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Toshio Miyata
- Department of Molecular Medicine and Therapy, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Kiyoshi Ando
- Department of Hematology and Oncology, Tokai University School of Medicine, Kanagawa, Japan
| | - Takashi Yahata
- Translational Molecular Therapeutics Laboratory, Division of Host Defense Mechanism, Tokai University School of Medicine, Kanagawa, Japan
| |
Collapse
|
8
|
Qian C, Ito N, Tsuji K, Sato S, Kikuchi K, Yoshii T, Miyata T, Asou Y. A PAI-1 antagonist ameliorates hypophosphatemia in the Hyp vitamin D-resistant rickets model mouse. FEBS Open Bio 2024; 14:290-299. [PMID: 38050660 PMCID: PMC10839342 DOI: 10.1002/2211-5463.13745] [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: 08/01/2023] [Revised: 11/20/2023] [Accepted: 12/04/2023] [Indexed: 12/06/2023] Open
Abstract
Congenital fibroblast growth factor 23 (FGF23)-related hypophosphatemic rickets/osteomalacia is a rare bone metabolism disorder characterized by hypophosphatemia and caused by genetic abnormalities that result in excessive secretion of FGF23. Hyp mice are a model of X-linked hypophosphatemia (XLH) caused by deletion of the PHEX gene and excessive production of FGF23. The purpose of this study was to investigate the potential of TM5614 as a therapeutic agent for the treatment of congenital FGF23-related hypophosphatemic rickets and osteomalacia in humans by administering TM5614 to Hyp mice and examining its curative effect on hypophosphatemia. After a single oral administration of TM5614 10 mg·kg-1 to female Hyp mice starting at 17 weeks of age, the serum phosphate concentration increased with a peak at 6 h after administration. ELISA confirmed that TM5614 administration decreased the intact FGF23 concentration in the blood. Expression of 25-hydroxyvitamin D-1α-hydroxylase protein encoded by Cyp27b1 mRNA in the kidney was suppressed in Hyp mice, and treatment with 10 mg·kg-1 of TM5614 normalized the expression of 25-hydroxyvitamin D-1α-hydroxylase protein and Cyp27b1 mRNA in the kidneys of these mice. Our data indicate that oral administration of TM5614 ameliorates hypophosphatemia in Hyp mice, suggesting that TM5614 may be an effective treatment for congenital FGF23-related hypophosphatemic rickets and osteomalacia.
Collapse
Affiliation(s)
- Cheng Qian
- Department of Orthopedics SurgeryTokyo Medical and Dental UniversityJapan
| | - Nobuaki Ito
- Division of Nephrology and EndocrinologyThe University of Tokyo HospitalJapan
| | - Kunikazu Tsuji
- Department of Orthopedics SurgeryTokyo Medical and Dental UniversityJapan
| | - Shingo Sato
- Department of Orthopedics SurgeryTokyo Medical and Dental UniversityJapan
| | - Katsushi Kikuchi
- Department of Orthopedics SurgeryTokyo Medical and Dental UniversityJapan
| | - Toshitaka Yoshii
- Department of Orthopedics SurgeryTokyo Medical and Dental UniversityJapan
| | - Toshio Miyata
- United Centers for Advanced Research and Translational MedicineTohoku UniversitySendaiJapan
| | - Yoshinori Asou
- Department of Orthopedics SurgeryTokyo Medical and Dental UniversityJapan
- China‐Japan Friendship Institution of MedicineShanghai UniversityChina
| |
Collapse
|
9
|
Tian J, Jiang L, Li H, Dan J, Luo Y. The dual role of the DREAM/G2M pathway in non-tumorigenic immortalization of senescent cells. FEBS Open Bio 2024; 14:331-343. [PMID: 38073074 PMCID: PMC10839291 DOI: 10.1002/2211-5463.13748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/04/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023] Open
Abstract
Anti-aging and tumorigenesis share common genes and pathways, and thus targeting these genes as part of anti-aging interventions carries the risk of tumorigenesis. It is essential to understand the gene signatures that balance tumorigenesis and aging. To achieve this goal, we analyzed RNA-sequencing data from three non-tumorigenic immortalized cell lines that spontaneously escaped from senescence. By single sample gene set enrichment assay (ssGSEA) and GSEA analysis, we found that both cell growth signaling (E2F targets, MYC targets) and tumor surveillance mechanisms (DNA repair, G2M checkpoint, mitotic spindle) were up-regulated in all three cell lines, suggesting that these genes are potential signatures for non-tumorigenic immortalization. Further analysis revealed that the 182 commonly up-regulated genes in these three cell lines overlapped with the DREAM/G2M pathway, which is known to be the upstream regulator of E2F, Myc targets, DNA repair, G2M checkpoint and mitotic spindle pathways in its cell cycle activation or inhibitory form. By western blotting, quantitative PCR and co-immunoprecipitation, we verified that both forms of the DREAM pathway are up-regulated in all three cell lines; this pathway facilitates control of cell cycle progression, supporting a new mechanism for non-tumorigenic immortalization. Thus, we propose that the DREAM/G2M pathway plays important dual roles with respect to preventing tumorigenesis in the process of immortalization. Our data might serve as the basis for the identification of new signature pathways or gene biomarkers for non-tumorigenic immortalization, and may aid in the discovery of new targets for tumor-free anti-aging drug screening.
Collapse
Affiliation(s)
- Jie Tian
- Department of Pathophysiology, School of Basic MedicineGuizhou Medical UniversityGuiyangChina
| | - Liangxia Jiang
- Department of Pathophysiology, School of Basic MedicineGuizhou Medical UniversityGuiyangChina
| | - Haili Li
- School of Basic MedicineShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
| | - Juhua Dan
- Laboratory of Molecular Genetics of Aging & Tumor, Medical SchoolKunming University of Science and TechnologyChina
| | - Ying Luo
- Department of Pathophysiology, School of Basic MedicineGuizhou Medical UniversityGuiyangChina
| |
Collapse
|
10
|
Hirai T, Asano K, Ito I, Miyazaki Y, Sugiura H, Agirbasli M, Kobayashi S, Kobayashi M, Shimada D, Natsume I, Kawasaki T, Ohba T, Tajiri S, Sakamaki F, Mineshita M, Takihara T, Sekiya K, Tomii K, Tomioka H, Kita H, Nishizaka Y, Fukui M, Miyata T, Harigae H. A randomized double-blind placebo-controlled trial of an inhibitor of plasminogen activator inhibitor-1 (TM5614) in mild to moderate COVID-19. Sci Rep 2024; 14:165. [PMID: 38168544 PMCID: PMC10761996 DOI: 10.1038/s41598-023-50445-1] [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: 06/05/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024] Open
Abstract
An inhibitor of plasminogen activator inhibitor (PAI)-1, TM5614, inhibited thrombosis, inflammation, and fibrosis in several experimental mouse models. To evaluate the efficacy and safety of TM5614 in human COVID-19 pneumonia, phase IIa and IIb trials were conducted. In an open-label, single-arm trial, 26 Japanese COVID-19 patients with mild to moderate pneumonia were treated with 120-180 mg of TM5614 daily, and all were discharged without any notable side effects. Then, a randomized, double-blind, placebo-controlled trial was conducted in Japanese COVID-19 patients with mild to moderate pneumonia. The number of study participants was set to be 50 in each arm. Even after extension of the enrollment period, the number of study participants did not reach the initially intended sample size, and 75 patients were enrolled in the study. The total oxygenation scale from Day 1 to Day 14 as the primary endpoint was 1.5 in the TM5614 group vs 4.0 in the placebo group (p = 0.22), and the number of days of oxygen administration required as the secondary endpoint was 2.0 days in the TM5614 group vs 3.5 days in the placebo group (p = 0.34). Further studies will be necessary to verify the efficacy of PAI-1 inhibition for the treatment of COVID-19 pneumonia.Clinical trial registration: Two studies were conducted: a prospective, multicenter, open-label phase II study at https://jrct.niph.go.jp (jRCT2021200018) (First registration date 18/08/2020) and a prospective, multicenter, randomized, double-blind, placebo-controlled, phase II study at https://jrct.niph.go.jp (jRCT2021210006) (First registration date 28/05/2021).
Collapse
Affiliation(s)
- Toyohiro Hirai
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawaharacho, Sakyo, Kyoto, 606-8507, Japan.
| | - Koichiro Asano
- Division of Pulmonary Medicine, Department of Medicine, Tokai University School of Medicine, Kanagawa, Japan
| | - Isao Ito
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawaharacho, Sakyo, Kyoto, 606-8507, Japan
| | - Yasunari Miyazaki
- Department of Respiratory Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hisatoshi Sugiura
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mehmet Agirbasli
- Department of Cardiology, Istanbul Medeniyet University Hospital TR, Istanbul, Turkey
| | - Seiichi Kobayashi
- Department of Respiratory Medicine, Japanese Red Cross Ishinomaki Hospital, Ishinomaki, Japan
| | - Makoto Kobayashi
- Department of Respiratory Medicine, Osaki Citizen Hospital, Osaki, Japan
| | - Daishi Shimada
- Department of Infectious Disease Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Ichiro Natsume
- Department of Respiratory Internal Medicine, Yokosuka Kyosai Hospital, Yokosuka, Japan
| | - Tsutomu Kawasaki
- Department of Respiratory Medicine, Yokohama City Minato Red Cross Hospital, Yokohama, Japan
| | - Takehiko Ohba
- Department of Respiratory Medicine, Ome Municipal General Hospital, Ome, Japan
| | - Sakurako Tajiri
- Department of Respiratory Medicine, Tokai University Oiso Hospital, Oiso, Japan
| | - Fumio Sakamaki
- Department of Respiratory Medicine, Tokai University Hachioji Hospital, Hachioji, Japan
| | - Masamichi Mineshita
- Department of Respiratory Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Takahisa Takihara
- Department of Respiratory Medicine, Ebina General Hospital, Ebina, Japan
| | - Kiyoshi Sekiya
- Department of Allergy and Respiratory Medicine, National Organization Sagamihara National Hospital, Sagamihara, Japan
| | - Keisuke Tomii
- Department of Respiratory Medicine, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Hiromi Tomioka
- Department of Respiratory Medicine, Kobe City Medical Center West Hospital, Kobe, Japan
| | - Hideo Kita
- Department of Respiratory Medicine, Takatsuki Red Cross Hospital, Takatsuki, Japan
| | - Yasuo Nishizaka
- Department of Respiratory Medicine, Osaka Red Cross Hospital, Osaka, Japan
| | - Motonari Fukui
- Department of Respiratory Medicine, Medical Research Institute Kitano Hospital, Osaka, Japan
| | - Toshio Miyata
- Department of Molecular Medicine and Therapy, Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan.
| | - Hideo Harigae
- Department of Hematology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
11
|
Li S, Kong F, Xu X, Song S, Wu Y, Tong J. Identification and exploration of aging-related subtypes and distinctive role of SERPINE1 in heart failure based on single-cell and bulk RNA sequencing data. J Gene Med 2024; 26:e3631. [PMID: 38062883 DOI: 10.1002/jgm.3631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/19/2023] [Accepted: 10/27/2023] [Indexed: 01/30/2024] Open
Abstract
Aging is a major risk factor for heart failure (HF) and is the leading cause of death worldwide. Currently, the nature of the relationship between aging and HF is not entirely clear. Herein, this study aimed to explore new diagnostic biomarkers, molecular typing and therapeutic strategies for HF by investigating the biological significance of aging-related genes in HF. A total of 157 differentially expressed genes (DEGs) were screened totally between HF and normal samples, and functional enrichment analysis of DEGs revealed the strong association of HF progression with aging, immune processes and metabolism. Six HF-specific aging-related genes were further identified, and a diagnostic model was developed and validated for good diagnostic efficacy. In addition, we collected blood samples from 10 normal controls and 10 HF patients for RT-qPCR analysis to verify the bioinformation. We also identified two aging-associated subtypes with distinctly different immune infiltration and metabolic microenvironment. Further single-cell sequencing analysis conducted in the study identified SERPINE1 as a key gene in HF. The distinctive role of SERPINE1 fibroblasts was revealed, including three main findings: (I) fibroblasts had a higher proportion and expression of SERPINE1 levels in HF; (II) the ligand-receptor pair MDK-LRP1 made the most contributions in high interactions with other cell types in SERPINE1 fibroblasts; and (III) SERPINE1 fibroblasts were associated with the interaction of extracellular matrix and receptor and may be regulated by the transcription factor EGR1. In conclusion, this study highlights the importance of aging-related genes in diagnosing HF and regulating immune infiltration. We also identified different HF subtypes and a potentially crucial gene, which may provide a better understanding of the molecular-level mechanisms of aging-related HF and aid in developing effective therapeutic strategies.
Collapse
Affiliation(s)
- Shengnan Li
- Department of Cardiology, Zhongda Hospital of Southeast University, Nanjing, Jiangsu, China
| | - Fanliang Kong
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China
| | - Xuan Xu
- Department of Cardiology, Zhongda Hospital of Southeast University, Nanjing, Jiangsu, China
| | - Sifan Song
- Department of Cardiology, Zhongda Hospital of Southeast University, Nanjing, Jiangsu, China
| | - Yandan Wu
- Department of Cardiology, Zhongda Hospital of Southeast University, Nanjing, Jiangsu, China
| | - Jiayi Tong
- Department of Cardiology, Zhongda Hospital of Southeast University, Nanjing, Jiangsu, China
| |
Collapse
|
12
|
Alem F, Campos-Obando N, Narayanan A, Bailey CL, Macaya RF. Exogenous Klotho Extends Survival in COVID-19 Model Mice. Pathogens 2023; 12:1404. [PMID: 38133288 PMCID: PMC10746004 DOI: 10.3390/pathogens12121404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/18/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
A striking feature of COVID-19 disease is the broad spectrum of risk factors associated with case severity, as well as the diversity of clinical manifestations. While no central agent has been able to explain the pathogenesis of SARS-CoV-2 infection, the factors that most robustly correlate with severity are risk factors linked to aging. Low serum levels of Klotho, an anti-aging protein, strongly correlate with the pathogenesis of the same risk factors and manifestations of conditions similar to those expressed in severe COVID-19 cases. The current manuscript presents original research on the effects of the exogenous application of Klotho, an anti-aging protein, in COVID-19 model mice. Klotho supplementation resulted in a statistically significant survival benefit in parametric and non-parametric models. Further research is required to elucidate the mechanistic role Klotho plays in COVID-19 pathogenesis as well as the possible modulation SARS-CoV-2 may have on the biological aging process.
Collapse
Affiliation(s)
- Farhang Alem
- Biomedical Research Laboratory, George Mason University, 4400 University Dr., Fairfax, VA 22030, USA; (F.A.); (A.N.); (C.L.B.)
| | - Natalia Campos-Obando
- Formerly at Caja Costarricense de Seguro Social, San José P.O. Box 10105-1000, Costa Rica;
| | - Aarthi Narayanan
- Biomedical Research Laboratory, George Mason University, 4400 University Dr., Fairfax, VA 22030, USA; (F.A.); (A.N.); (C.L.B.)
| | - Charles L. Bailey
- Biomedical Research Laboratory, George Mason University, 4400 University Dr., Fairfax, VA 22030, USA; (F.A.); (A.N.); (C.L.B.)
| | - Roman F. Macaya
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, 665 Huntington Ave., Boston, MA 02115, USA
| |
Collapse
|
13
|
Van Wynsberghe J, Vanakker OM. Significance of Premature Vertebral Mineralization in Zebrafish Models in Mechanistic and Pharmaceutical Research on Hereditary Multisystem Diseases. Biomolecules 2023; 13:1621. [PMID: 38002303 PMCID: PMC10669475 DOI: 10.3390/biom13111621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
Zebrafish are increasingly becoming an important model organism for studying the pathophysiological mechanisms of human diseases and investigating how these mechanisms can be effectively targeted using compounds that may open avenues to novel treatments for patients. The zebrafish skeleton has been particularly instrumental in modeling bone diseases as-contrary to other model organisms-the lower load on the skeleton of an aquatic animal enables mutants to survive to early adulthood. In this respect, the axial skeletons of zebrafish have been a good read-out for congenital spinal deformities such as scoliosis and degenerative disorders such as osteoporosis and osteoarthritis, in which aberrant mineralization in humans is reflected in the respective zebrafish models. Interestingly, there have been several reports of hereditary multisystemic diseases that do not affect the vertebral column in human patients, while the corresponding zebrafish models systematically show anomalies in mineralization and morphology of the spine as their leading or, in some cases, only phenotype. In this review, we describe such examples, highlighting the underlying mechanisms, the already-used or potential power of these models to help us understand and amend the mineralization process, and the outstanding questions on how and why this specific axial type of aberrant mineralization occurs in these disease models.
Collapse
Affiliation(s)
- Judith Van Wynsberghe
- Center for Medical Genetics, Ghent University Hospital, 9000 Ghent, Belgium;
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- Ectopic Mineralization Research Group, 9000 Ghent, Belgium
| | - Olivier M. Vanakker
- Center for Medical Genetics, Ghent University Hospital, 9000 Ghent, Belgium;
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- Ectopic Mineralization Research Group, 9000 Ghent, Belgium
| |
Collapse
|
14
|
Suvakov S, Kattah AG, Gojkovic T, Enninga EAL, Pruett J, Jayachandran M, Sousa C, Santos J, Abou Hassan C, Gonzales-Suarez M, Garovic VD. Impact of Aging and Cellular Senescence in the Pathophysiology of Preeclampsia. Compr Physiol 2023; 13:5077-5114. [PMID: 37770190 DOI: 10.1002/cphy.c230003] [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: 10/03/2023]
Abstract
The incidence of hypertensive disorders of pregnancy is increasing, which may be due to several factors, including an increased age at pregnancy and more comorbid health conditions during reproductive years. Preeclampsia, the most severe hypertensive disorder of pregnancy, has been associated with an increased risk of future disease, including cardiovascular and kidney diseases. Cellular senescence, the process of cell cycle arrest in response to many physiologic and maladaptive stimuli, may play an important role in the pathogenesis of preeclampsia and provide a mechanistic link to future disease. In this article, we will discuss the pathophysiology of preeclampsia, the many mechanisms of cellular senescence, evidence for the involvement of senescence in the development of preeclampsia, as well as evidence that cellular senescence may link preeclampsia to the risk of future disease. Lastly, we will explore how a better understanding of the role of cellular senescence in preeclampsia may lead to therapeutic trials. © 2023 American Physiological Society. Compr Physiol 13:5077-5114, 2023.
Collapse
Affiliation(s)
- Sonja Suvakov
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrea G Kattah
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Tamara Gojkovic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Elizabeth A L Enninga
- Division of Research, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Jacob Pruett
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Ciria Sousa
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Janelle Santos
- Division of Research, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Coline Abou Hassan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Vesna D Garovic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
- Division of Research, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| |
Collapse
|
15
|
Qu JH, Tarasov KV, Tarasova YS, Chakir K, Lakatta EG. Transcriptome of Left Ventricle and Sinoatrial Node in Young and Old C57 Mice. FORTUNE JOURNAL OF HEALTH SCIENCES 2023; 6:332-356. [PMID: 37920273 PMCID: PMC10621664 DOI: 10.26502/fjhs.134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
Advancing age is the most important risk factor for cardiovascular diseases (CVDs). Two types of cells, within the heart pacemaker, sinoatrial node (SAN), and within the left ventricle (LV), control two crucial characteristics of heart function, heart beat rate and contraction strength. As age advances, the heart's structure becomes remodeled, and SAN and LV cell functions deteriorate, thus increasing the risk for CVDs. However, the different molecular features of age-associated changes in SAN and LV cells have never been compared in omics scale in the context of aging. We applied deep RNA sequencing to four groups of samples, young LV, old LV, young SAN and old SAN, followed by numerous bioinformatic analyses. In addition to profiling the differences in gene expression patterns between the two heart chambers (LV vs. SAN), we also identified the chamber-specific concordant or discordant age-associated changes in: (1) genes linked to energy production related to cardiomyocyte contraction, (2) genes related to post-transcriptional processing, (3) genes involved in KEGG longevity regulating pathway, (4) prolongevity and antilongevity genes recorded and curated in the GenAge database, and (5) CVD marker genes. Our bioinformatic analysis also predicted the regulation activities and mapped the expression of upstream regulators including transcription regulators and post-transcriptional regulator miRNAs. This comprehensive analysis promotes our understanding of regulation of heart functions and will enable discovery of gene-specific therapeutic targets of CVDs in advanced age.
Collapse
Affiliation(s)
- Jia-Hua Qu
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Kirill V Tarasov
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Yelena S Tarasova
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Khalid Chakir
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Edward G Lakatta
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| |
Collapse
|
16
|
Rana T, Jiang C, Banerjee S, Yi N, Zmijewski JW, Liu G, Liu RM. PAI-1 Regulation of p53 Expression and Senescence in Type II Alveolar Epithelial Cells. Cells 2023; 12:2008. [PMID: 37566086 PMCID: PMC10417428 DOI: 10.3390/cells12152008] [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: 05/25/2023] [Revised: 07/17/2023] [Accepted: 08/01/2023] [Indexed: 08/12/2023] Open
Abstract
Cellular senescence contributes importantly to aging and aging-related diseases, including idiopathic pulmonary fibrosis (IPF). Alveolar epithelial type II (ATII) cells are progenitors of alveolar epithelium, and ATII cell senescence is evident in IPF. Previous studies from this lab have shown that increased expression of plasminogen activator inhibitor 1 (PAI-1), a serine protease inhibitor, promotes ATII cell senescence through inducing p53, a master cell cycle repressor, and activating p53-p21-pRb cell cycle repression pathway. In this study, we further show that PAI-1 binds to proteasome components and inhibits proteasome activity and p53 degradation in human lung epithelial A549 cells and primary mouse ATII cells. This is associated with a senescence phenotype of these cells, manifested as increased p53 and p21 expression, decreased phosphorylated retinoblastoma protein (pRb), and increased senescence-associated beta-galactose (SA-β-gal) activity. Moreover, we find that, although overexpression of wild-type PAI-1 (wtPAI-1) or a secretion-deficient, mature form of PAI-1 (sdPAI-1) alone induces ATII cell senescence (increases SA-β-gal activity), only wtPAI-1 induces p53, suggesting that the premature form of PAI-1 is required for the interaction with the proteasome. In summary, our data indicate that PAI-1 can bind to proteasome components and thus inhibit proteasome activity and p53 degradation in ATII cells. As p53 is a master cell cycle repressor and PAI-1 expression is increased in many senescent cells, the results from this study will have a significant impact not only on ATII cell senescence/lung fibrosis but also on the senescence of other types of cells in different diseases.
Collapse
Affiliation(s)
- Tapasi Rana
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Chunsun Jiang
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Sami Banerjee
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Nengjun Yi
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jaroslaw W. Zmijewski
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Gang Liu
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Rui-Ming Liu
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
17
|
Patel TD, Nakka M, Grimm SL, Coarfa C, Gorelick DA. Functional genomic analysis of non-canonical DNA regulatory elements of the aryl hydrocarbon receptor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.01.538985. [PMID: 37205451 PMCID: PMC10187216 DOI: 10.1101/2023.05.01.538985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The aryl hydrocarbon receptor (AHR) is a ligand-dependent transcription factor that binds DNA and regulates genes in response to halogenated and polycyclic aromatic hydrocarbons. AHR also regulates the development and function of the liver and the immune system. In the canonical pathway, AHR binds a consensus DNA sequence, termed the xenobiotic response element (XRE), recruits protein coregulators, and regulates target gene expression. Emerging evidence suggests that AHR may regulate gene expression via an additional pathway, by binding to a non-consensus DNA sequence termed the non-consensus XRE (NC-XRE). The prevalence of NC-XRE motifs in the genome is not known. Studies using chromatin immunoprecipitation and reporter genes provide indirect evidence of AHR-NC-XRE interactions, but direct evidence for an AHR-NCXRE interaction that regulates transcription in a natural genomic context is lacking. Here, we analyzed AHR binding to NC-XRE DNA on a genome-wide scale in mouse liver. We integrated ChIP-seq and RNA-seq data and identified putative AHR target genes with NC-XRE motifs in regulatory regions. We also performed functional genomics at a single locus, the mouse Serpine1 gene. Deleting NC-XRE motifs from the Serpine1 promoter reduced the upregulation of Serpine1 by TCDD, an AHR ligand. We conclude that AHR upregulates Serpine1 via NC-XRE DNA. NC-XRE motifs are prevalent throughout regions of the genome where AHR binds. Taken together, our results suggest that AHR regulates genes via NC-XRE motifs. Our results will also improve our ability to identify AHR target genes and their physiologic relevance.
Collapse
Affiliation(s)
- Tajhal D Patel
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Manjula Nakka
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, USA
| | - Sandra L Grimm
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Cristian Coarfa
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Daniel A Gorelick
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
18
|
Callegari K, Dash S, Uchida H, Shingai Y, Liu C, Khodarkovskaya A, Lee Y, Ito A, Lopez A, Zhang T, Xiang J, Kluk MJ, Sanchez T. Molecular profiling of the stroke-induced alterations in the cerebral microvasculature reveals promising therapeutic candidates. Proc Natl Acad Sci U S A 2023; 120:e2205786120. [PMID: 37058487 PMCID: PMC10120001 DOI: 10.1073/pnas.2205786120] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 02/08/2023] [Indexed: 05/11/2023] Open
Abstract
Stroke-induced cerebral microvascular dysfunction contributes to aggravation of neuronal injury and compromises the efficacy of current reperfusion therapies. Understanding the molecular alterations in cerebral microvessels in stroke will provide original opportunities for scientific investigation of novel therapeutic strategies. Toward this goal, using a recently optimized method which minimizes cell activation and preserves endothelial cell interactions and RNA integrity, we conducted a genome-wide transcriptomic analysis of cerebral microvessels in a mouse model of stroke and compared these transcriptomic alterations with the ones observed in human, nonfatal, brain stroke lesions. Results from these unbiased comparative analyses have revealed the common alterations in mouse stroke microvessels and human stroke lesions and identified shared molecular features associated with vascular disease (e.g., Serpine1/Plasminogen Activator Inhibitor-1, Hemoxygenase-1), endothelial activation (e.g., Angiopoietin-2), and alterations in sphingolipid metabolism and signaling (e.g., Sphigosine-1-Phosphate Receptor 2). Sphingolipid profiling of mouse cerebral microvessels validated the transcript data and revealed the enrichment of sphingomyelin and sphingoid species in the cerebral microvasculature compared to brain and the stroke-induced increase in ceramide species. In summary, our study has identified novel molecular alterations in several microvessel-enriched, translationally relevant, and druggable targets, which are potent modulators of endothelial function. Our comparative analyses have revealed the presence of molecular features associated with cerebral microvascular dysfunction in human chronic stroke lesions. The results shared here provide a detailed resource for therapeutic discovery of candidates for neurovascular protection in stroke and potentially, other pathologies exhibiting cerebral microvascular dysfunction.
Collapse
Affiliation(s)
- Keri Callegari
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Sabyasachi Dash
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Hiroki Uchida
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Yuto Shingai
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Catherine Liu
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Anne Khodarkovskaya
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Yunkyoung Lee
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Akira Ito
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Amanda Lopez
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Tuo Zhang
- Genomics Resources Core Facility, Weill Cornell Medicine, New York, NY10065
| | - Jenny Xiang
- Genomics Resources Core Facility, Weill Cornell Medicine, New York, NY10065
| | - Michael J. Kluk
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Teresa Sanchez
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY10065
| |
Collapse
|
19
|
Jiang CS, Rana T, Jin LW, Farr SA, Morley JE, Qin H, Liu G, Liu RM. Aging, Plasminogen Activator Inhibitor 1, Brain Cell Senescence, and Alzheimer's Disease. Aging Dis 2023; 14:515-528. [PMID: 37008063 PMCID: PMC10017160 DOI: 10.14336/ad.2022.1220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/20/2022] [Indexed: 04/04/2023] Open
Abstract
The etiology for late-onset Alzheimer's disease (LOAD), which accounts for >95% of Alzheimer's disease (AD) cases, is unknown. Emerging evidence suggests that cellular senescence contributes importantly to AD pathophysiology, although the mechanisms underlying brain cell senescence and by which senescent cells promote neuro-pathophysiology remain unclear. In this study we show for the first time that the expression of plasminogen activator inhibitor 1 (PAI-1), a serine protease inhibitor, is increased, in correlation with the increased expression of cell cycle repressors p53 and p21, in the hippocampus/cortex of senescence accelerated mouse prone 8 (SAMP8) mice and LOAD patients. Double immunostaining results show that astrocytes in the brain of LOAD patients and SAMP8 mice express higher levels of senescent markers and PAI-1, compared to astrocytes in the corresponding controls. In vitro studies further show that overexpression of PAI-1 alone, intracellularly or extracellularly, induced senescence, whereas inhibition or silencing PAI-1 attenuated H2O2-induced senescence, in primary mouse and human astrocytes. Treatment with the conditional medium (CM) from senescent astrocytes induced neuron apoptosis. Importantly, the PAI-1 deficient CM from senescent astrocytes that overexpress a secretion deficient PAI-1 (sdPAI-1) has significantly reduced effect on neurons, compared to the PAI-1 containing CM from senescent astrocytes overexpressing wild type PAI-1 (wtPAI-1), although sdPAI-1 and wtPAI-1 induce similar degree of astrocyte senescence. Together, our results suggest that increased PAI-1, intracellularly or extracellularly, may contribute to brain cell senescence in LOAD and that senescent astrocytes can induce neuron apoptosis through secreting pathologically active molecules, including PAI-1.
Collapse
Affiliation(s)
- Chun-Sun Jiang
- Department of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA.
| | - Tapasi Rana
- Department of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA.
| | - Lee-Way Jin
- Department of Pathology and Laboratory Medicine, University of California, Davis, CA, USA.
| | - Susan A Farr
- Division of Geriatric Medicine, School of Medicine, Saint Louis University, St. Louis, MO, USA.
- Research and Development, Veterans Affairs Medical Center, St. Louis Missouri, MO, USA.
| | - John E Morley
- Division of Geriatric Medicine, School of Medicine, Saint Louis University, St. Louis, MO, USA.
| | - Hongwei Qin
- Department of Cell, Developmental and Integrative Biology, UAB, Birmingham, AL, USA.
| | - Gang Liu
- Department of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA.
| | - Rui-Ming Liu
- Department of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA.
| |
Collapse
|
20
|
Khalil R, Diab-Assaf M, Lemaitre JM. Emerging Therapeutic Approaches to Target the Dark Side of Senescent Cells: New Hopes to Treat Aging as a Disease and to Delay Age-Related Pathologies. Cells 2023; 12:915. [PMID: 36980256 PMCID: PMC10047596 DOI: 10.3390/cells12060915] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/05/2023] [Accepted: 03/07/2023] [Indexed: 03/19/2023] Open
Abstract
Life expectancy has drastically increased over the last few decades worldwide, with important social and medical burdens and costs. To stay healthy longer and to avoid chronic disease have become essential issues. Organismal aging is a complex process that involves progressive destruction of tissue functionality and loss of regenerative capacity. One of the most important aging hallmarks is cellular senescence, which is a stable state of cell cycle arrest that occurs in response to cumulated cell stresses and damages. Cellular senescence is a physiological mechanism that has both beneficial and detrimental consequences. Senescence limits tumorigenesis, lifelong tissue damage, and is involved in different biological processes, such as morphogenesis, regeneration, and wound healing. However, in the elderly, senescent cells increasingly accumulate in several organs and secrete a combination of senescence associated factors, contributing to the development of various age-related diseases, including cancer. Several studies have revealed major molecular pathways controlling the senescent phenotype, as well as the ones regulating its interactions with the immune system. Attenuating the senescence-associated secretory phenotype (SASP) or eliminating senescent cells have emerged as attractive strategies aiming to reverse or delay the onset of aging diseases. Here, we review current senotherapies designed to suppress the deleterious effect of SASP by senomorphics or to selectively kill senescent cells by "senolytics" or by immune system-based approaches. These recent investigations are promising as radical new controls of aging pathologies and associated multimorbidities.
Collapse
Affiliation(s)
- Roula Khalil
- IRMB, University Montpellier, INSERM, 34090 Montpellier, France;
| | - Mona Diab-Assaf
- Fanar Faculty of Sciences II, Lebanese University, Beirut P.O. Box 90656, Lebanon;
| | | |
Collapse
|
21
|
Amitani H, Chiba S, Amitani M, Michihara S, Takemoto R, Han L, Fujita N, Takahashi R, Inui A. Impact of Ninjin’yoeito on frailty and short life in klotho-hypomorphic (kl/kl) mice. Front Pharmacol 2022; 13:973897. [DOI: 10.3389/fphar.2022.973897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 10/03/2022] [Indexed: 11/13/2022] Open
Abstract
With the recent aging of society, the prevention of frailty has become an important issue because people desire both a long and healthy lifespan. Klotho-hypomorphic (kl/kl) mice are known to show phenotypes of premature aging. Ninjin’yoeito (NYT) is a traditional Japanese Kampo medicine used to treat patients with vulnerable constitution, fatigue or physical exhaustion caused by aging and illness. Recent studies have reported the potential efficacy of NYT against frailty. We therefore evaluated the effect of NYT on the gait function, activity, the histopathological status of organs and survival using kl/kl mice as a model of aging-related frailty. Two sets of 28-day-old male kl/kl mice were assigned to the vehicle (non-treated; NT), 3% or 5% NYT dietary groups. One set of groups (NT, n = 18; 3% NYT, n = 11; 5% NYT, n = 11) was subjected to the analysis of free walking, rotarod, and spontaneous activity tests at approximately 58 days old. Thereafter, we measured triceps surae muscles weight and myofiber cross-sectional area (CSA), and quantified its telomere content. In addition, we evaluated bone strength and performed histopathological examinations of organs. Survival was measured in the second set of groups (NT, 3% NYT and 5% NYT group, n = 8 each). In the walking test, several indicators such as gait velocity were improved in the NYT 3% group. Similar results were obtained for the latency to fall in the rotarod test and spontaneous motor activity. Triceps muscle mass, CSA and its telomere content were significantly improved in the NYT 3% group. Bone density, pulmonary alveolus destruction and testicular atrophy were also significantly improved in the NYT 3% group. Survival rate and body weight were both significantly improved in the NYT3% group compared with those in the NT group. Continuous administration of NYT from the early stage of aging improved not only gait performance, but also the survival in the aging-related frailty model. This effect may be associated with the improvements in aging-related organ changes such as muscle atrophy. Intervention with NYT against the progression of frailty may contribute to a longer, healthier life span among the elderly individuals.
Collapse
|
22
|
Sun Y, Li Q, Kirkland JL. Targeting senescent cells for a healthier longevity: the roadmap for an era of global aging. LIFE MEDICINE 2022; 1:103-119. [PMID: 36699942 PMCID: PMC9869767 DOI: 10.1093/lifemedi/lnac030] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/05/2022] [Indexed: 01/28/2023]
Abstract
Aging is a natural but relentless process of physiological decline, leading to physical frailty, reduced ability to respond to physical stresses (resilience) and, ultimately, organismal death. Cellular senescence, a self-defensive mechanism activated in response to intrinsic stimuli and/or exogenous stress, is one of the central hallmarks of aging. Senescent cells cease to proliferate, while remaining metabolically active and secreting numerous extracellular factors, a feature known as the senescence-associated secretory phenotype. Senescence is physiologically important for embryonic development, tissue repair, and wound healing, and prevents carcinogenesis. However, chronic accumulation of persisting senescent cells contributes to a host of pathologies including age-related morbidities. By paracrine and endocrine mechanisms, senescent cells can induce inflammation locally and systemically, thereby causing tissue dysfunction, and organ degeneration. Agents including those targeting damaging components of the senescence-associated secretory phenotype or inducing apoptosis of senescent cells exhibit remarkable benefits in both preclinical models and early clinical trials for geriatric conditions. Here we summarize features of senescent cells and outline strategies holding the potential to be developed as clinical interventions. In the long run, there is an increasing demand for safe, effective, and clinically translatable senotherapeutics to address healthcare needs in current settings of global aging.
Collapse
Affiliation(s)
- Yu Sun
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
- Department of Pharmacology, Institute of Aging Medicine, Binzhou Medical University, Yantai 264003, China
- Department of Medicine and VAPSHCS, University of Washington, Seattle, WA 98195, USA
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - James L Kirkland
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
23
|
Xiong H, Hua F, Dong Y, Lin Y, Ying J, Liu J, Wang X, Zhang L, Zhang J. DNA damage response and GATA4 signaling in cellular senescence and aging-related pathology. Front Aging Neurosci 2022; 14:933015. [PMID: 36177479 PMCID: PMC9513149 DOI: 10.3389/fnagi.2022.933015] [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: 04/30/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Aging is the continuous degradation of biological function and structure with time, and cellular senescence lies at its core. DNA damage response (DDR) can activate Ataxia telangiectasia-mutated serine/threonine kinase (ATM) and Rad3-related serine/threonine kinase (ATR), after which p53 activates p21, stopping the cell cycle and inducing cell senescence. GATA4 is a transcription factor that plays an important role in the development of many organs, such as the heart, testis, ovary, foregut, liver, and ventral pancreas. Studies have shown that GATA4 can also contribute to the DDR, leading to aging. Consistently, there is also evidence that the GATA4 signaling pathway is associated with aging-related diseases, including atherosclerosis and heart failure. This paper reviews the relationship between GATA4, DDR, and cellular senescence, as well as its effect on aging-related diseases.
Collapse
Affiliation(s)
- Hao Xiong
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Fuzhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Yao Dong
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yue Lin
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Jun Ying
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Jie Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Xifeng Wang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Xifeng Wang
| | - Lieliang Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
- *Correspondence: Lieliang Zhang
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
24
|
Catarinella G, Nicoletti C, Bracaglia A, Procopio P, Salvatori I, Taggi M, Valle C, Ferri A, Canipari R, Puri PL, Latella L. SerpinE1 drives a cell-autonomous pathogenic signaling in Hutchinson-Gilford progeria syndrome. Cell Death Dis 2022; 13:737. [PMID: 36028501 PMCID: PMC9418244 DOI: 10.1038/s41419-022-05168-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/02/2022] [Accepted: 08/05/2022] [Indexed: 01/21/2023]
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare, fatal disease caused by Lamin A mutation, leading to altered nuclear architecture, loss of peripheral heterochromatin and deregulated gene expression. HGPS patients eventually die by coronary artery disease and cardiovascular alterations. Yet, how deregulated transcriptional networks at the cellular level impact on the systemic disease phenotype is currently unclear. A genome-wide analysis of gene expression in cultures of primary HGPS fibroblasts identified SerpinE1, also known as Plasminogen Activator Inhibitor (PAI-1), as central gene that propels a cell-autonomous pathogenic signaling from the altered nuclear lamina. Indeed, siRNA-mediated downregulation and pharmacological inhibition of SerpinE1 by TM5441 could revert key pathological features of HGPS in patient-derived fibroblasts, including re-activation of cell cycle progression, reduced DNA damage signaling, decreased expression of pro-fibrotic genes and recovery of mitochondrial defects. These effects were accompanied by the correction of nuclear abnormalities. These data point to SerpinE1 as a novel potential effector and target for therapeutic interventions in HGPS pathogenesis.
Collapse
Affiliation(s)
| | - Chiara Nicoletti
- grid.479509.60000 0001 0163 8573Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037 USA
| | - Andrea Bracaglia
- grid.417778.a0000 0001 0692 3437IRCCS Fondazione Santa Lucia, Rome, Italy ,grid.6530.00000 0001 2300 0941PhD Program in Cellular and Molecular Biology, Department of Biology, University of Rome “Tor Vergata”, Rome, Italy
| | - Paola Procopio
- grid.417778.a0000 0001 0692 3437IRCCS Fondazione Santa Lucia, Rome, Italy ,grid.10253.350000 0004 1936 9756Present Address: BPC, Pharmakologisches Institut, Philipps-Universität Marburg, Marburg, Germany
| | - Illari Salvatori
- grid.417778.a0000 0001 0692 3437IRCCS Fondazione Santa Lucia, Rome, Italy ,grid.7841.aDepartment of Experimental Medicine, University of Rome “La Sapienza”, 00161 Rome, Italy
| | - Marilena Taggi
- grid.7841.aDAHFMO, Unit of Histology and Medical Embryology, Sapienza, University of Rome, Rome, Italy
| | - Cristiana Valle
- grid.417778.a0000 0001 0692 3437IRCCS Fondazione Santa Lucia, Rome, Italy ,grid.5326.20000 0001 1940 4177Institute of Translational Pharmacology, National Research Council of Italy, Rome, Italy
| | - Alberto Ferri
- grid.417778.a0000 0001 0692 3437IRCCS Fondazione Santa Lucia, Rome, Italy ,grid.5326.20000 0001 1940 4177Institute of Translational Pharmacology, National Research Council of Italy, Rome, Italy
| | - Rita Canipari
- grid.7841.aDAHFMO, Unit of Histology and Medical Embryology, Sapienza, University of Rome, Rome, Italy
| | - Pier Lorenzo Puri
- grid.479509.60000 0001 0163 8573Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037 USA
| | - Lucia Latella
- grid.417778.a0000 0001 0692 3437IRCCS Fondazione Santa Lucia, Rome, Italy ,grid.5326.20000 0001 1940 4177Institute of Translational Pharmacology, National Research Council of Italy, Rome, Italy
| |
Collapse
|
25
|
Jo A, Choi TG, Han JY, Tabor MH, Kolliputi N, Lockey RF, Cho SH. Age-Related Increase of Collagen/Fibrin Deposition and High PAI-1 Production in Human Nasal Polyps. Front Pharmacol 2022; 13:845324. [PMID: 35712705 PMCID: PMC9193225 DOI: 10.3389/fphar.2022.845324] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 05/12/2022] [Indexed: 11/13/2022] Open
Abstract
Objective: Our previous studies showed an age-related increased prevalence of nasal polyps (NP) and reduced production of S100A8/9 in elderly patients with chronic rhinosinusitis with NP (CRSwNP). In this study, we investigated an unbiased age-related gene expression profile in CRSwNP subjects and healthy controls, and further identified the differences in their tissue remodeling. Methods: Microarrays using NP and uncinate tissues from health controls (elderly, age ≥65 vs. non-elderly, age 18-49) were performed, and differentially regulated genes were analyzed. Quantitative real-time PCR (qPCR), Immunostaining, Periodic acid-Schiff (PAS), trichrome staining, Western blot, and ELISA were performed for further investigation. Results: Microarrays identified differentially expressed genes according to disease and age; 278 in NP vs. controls, 75 in non-elderly NP vs. non-elderly controls, and 32 in elderly NP vs. elderly controls. qPCR confirmed that the PLAT gene was downregulated and the SERPINB2 gene upregulated in NP vs. controls. The serous glandular cell-derived antimicrobial protein/peptide-related genes such as BPIFB3, BPIFB2, LPO, and MUC7 were remarkably reduced in NP, regardless of age. SERPINE1 gene (plasminogen activator inhibitor-1, PAI-1) expression was significantly increased in elderly NP versus elderly controls. IHC and western blot confirmed significantly decreased production of MUC7 and LPO in NP versus controls. There was a trend of age-related reduction of submucosal gland cells in normal controls. Trichrome and immunofluorescence staining demonstrated an age-related increase of collagen and fibrin deposition in NP, consistent with increased PAI-1 production. Conclusion: This study demonstrated age-related differential glandular remodeling patterns and fibrosis in NP and normal controls. PAI-1 expression was significantly increased in elderly NP versus elderly controls, suggesting PAI-1 as a potential treatment target in elderly NP.
Collapse
Affiliation(s)
- Ara Jo
- Division of Allergy-Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Tae Gyu Choi
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, South Korea
| | - Jung Yeon Han
- Division of Allergy-Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Mark H. Tabor
- Department of Otolaryngology-Head and Neck Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Narasaiah Kolliputi
- Division of Allergy-Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Richard F. Lockey
- Division of Allergy-Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Seong H. Cho
- Division of Allergy-Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
- Division of Allergy-Immunology, James A. Haley Veterans’ Hospital, Tampa, FL, United States
| |
Collapse
|
26
|
Badran M, Gozal D. PAI-1: A Major Player in the Vascular Dysfunction in Obstructive Sleep Apnea? Int J Mol Sci 2022; 23:5516. [PMID: 35628326 PMCID: PMC9141273 DOI: 10.3390/ijms23105516] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/05/2022] [Accepted: 05/12/2022] [Indexed: 02/04/2023] Open
Abstract
Obstructive sleep apnea is a chronic and prevalent condition that is associated with endothelial dysfunction, atherosclerosis, and imposes excess overall cardiovascular risk and mortality. Despite its high prevalence and the susceptibility of CVD patients to OSA-mediated stressors, OSA is still under-recognized and untreated in cardiovascular practice. Moreover, conventional OSA treatments have yielded either controversial or disappointing results in terms of protection against CVD, prompting the need for the identification of additional mechanisms and associated adjuvant therapies. Plasminogen activator inhibitor-1 (PAI-1), the primary inhibitor of tissue-type plasminogen activator (tPA) and urinary-type plasminogen activator (uPA), is a key regulator of fibrinolysis and cell migration. Indeed, elevated PAI-1 expression is associated with major cardiovascular adverse events that have been attributed to its antifibrinolytic activity. However, extensive evidence indicates that PAI-1 can induce endothelial dysfunction and atherosclerosis through complex interactions within the vasculature in an antifibrinolytic-independent matter. Elevated PAI-1 levels have been reported in OSA patients. However, the impact of PAI-1 on OSA-induced CVD has not been addressed to date. Here, we provide a comprehensive review on the mechanisms by which OSA and its most detrimental perturbation, intermittent hypoxia (IH), can enhance the transcription of PAI-1. We also propose causal pathways by which PAI-1 can promote atherosclerosis in OSA, thereby identifying PAI-1 as a potential therapeutic target in OSA-induced CVD.
Collapse
Affiliation(s)
- Mohammad Badran
- Department of Child Health and Child Health Research Institute, School of Medicine, University of Missouri, 400 N Keene St, Suite 010, Columbia, MO 65201, USA;
| | - David Gozal
- Department of Child Health and Child Health Research Institute, School of Medicine, University of Missouri, 400 N Keene St, Suite 010, Columbia, MO 65201, USA;
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65201, USA
| |
Collapse
|
27
|
Willis CM, Nicaise AM, Krzak G, Ionescu RB, Pappa V, D'Angelo A, Agarwal R, Repollés-de-Dalmau M, Peruzzotti-Jametti L, Pluchino S. Soluble factors influencing the neural stem cell niche in brain physiology, inflammation, and aging. Exp Neurol 2022; 355:114124. [DOI: 10.1016/j.expneurol.2022.114124] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 05/16/2022] [Accepted: 05/21/2022] [Indexed: 11/27/2022]
|
28
|
Almiñana C, Dubuisson F, Bauersachs S, Royer E, Mermillod P, Blesbois E, Guignot F. Unveiling how vitrification affects the porcine blastocyst: clues from a transcriptomic study. J Anim Sci Biotechnol 2022; 13:46. [PMID: 35303969 PMCID: PMC8932223 DOI: 10.1186/s40104-021-00672-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 12/24/2021] [Indexed: 12/13/2022] Open
Abstract
Background Currently, there is a high demand for efficient pig embryo cryopreservation procedures in the porcine industry as well as for genetic diversity preservation and research purposes. To date, vitrification (VIT) is the most efficient method for pig embryo cryopreservation. Despite a high number of embryos survives in vitro after vitrification/warming procedures, the in vivo embryo survival rates after embryo transfer are variable among laboratories. So far, most studies have focused on cryoprotective agents and devices, while the VIT effects on porcine embryonic gene expression remained unclear. The few studies performed were based on vitrified/warmed embryos that were cultured in vitro (IVC) to allow them to re–expand. Thus, the specific alterations of VIT, IVC, and the cumulative effect of both remained unknown. To unveil the VIT-specific embryonic alterations, gene expression in VIT versus (vs.) IVC embryos was analyzed. Additionally, changes derived from both VIT and IVC vs. control embryos (CO) were analyzed to confirm the VIT embryonic alterations. Three groups of in vivo embryos at the blastocyst stage were analyzed by RNA–sequencing: (1) VIT embryos (vitrified/warmed and cultured in vitro), (2) IVC embryos and (3) CO embryos. Results RNA–sequencing revealed three clearly different mRNA profiles for VIT, IVC and CO embryos. Comparative analysis of mRNA profiles between VIT and IVC identified 321, differentially expressed genes (DEG) (FDR < 0.006). In VIT vs. CO and IVC vs. CO, 1901 and 1519 DEG were found, respectively, with an overlap of 1045 genes. VIT-specific functional alterations were associated to response to osmotic stress, response to hormones, and developmental growth. While alterations in response to hypoxia and mitophagy were related to the sum of VIT and IVC effects. Conclusions Our findings revealed new insights into the VIT procedure-specific alterations of embryonic gene expression by first comparing differences in VIT vs. IVC embryos and second by an integrative transcriptome analysis including in vivo control embryos. The identified VIT alterations might reflect the transcriptional signature of the embryo cryodamage but also the embryo healing process overcoming the VIT impacts. Selected validated genes were pointed as potential biomarkers that may help to improve vitrification. Supplementary Information The online version contains supplementary material available at 10.1186/s40104-021-00672-1.
Collapse
Affiliation(s)
- C Almiñana
- UMR PRC, INRAE 0085, CNRS 7247, Université de Tours, IFCE, F, -37380, Nouzilly, France. .,Functional Genomics Group, Institute of Veterinary Anatomy, VetSuisse Faculty Zurich, University of Zurich, Zürich, Switzerland.
| | - F Dubuisson
- UMR PRC, INRAE 0085, CNRS 7247, Université de Tours, IFCE, F, -37380, Nouzilly, France
| | - S Bauersachs
- Functional Genomics Group, Institute of Veterinary Anatomy, VetSuisse Faculty Zurich, University of Zurich, Zürich, Switzerland
| | - E Royer
- UEPAO, INRAE, F, -37380, Nouzilly, France
| | - P Mermillod
- UMR PRC, INRAE 0085, CNRS 7247, Université de Tours, IFCE, F, -37380, Nouzilly, France
| | - E Blesbois
- UMR PRC, INRAE 0085, CNRS 7247, Université de Tours, IFCE, F, -37380, Nouzilly, France
| | - F Guignot
- UMR PRC, INRAE 0085, CNRS 7247, Université de Tours, IFCE, F, -37380, Nouzilly, France
| |
Collapse
|
29
|
Hamsanathan S, Gurkar AU. Lipids as Regulators of Cellular Senescence. Front Physiol 2022; 13:796850. [PMID: 35370799 PMCID: PMC8965560 DOI: 10.3389/fphys.2022.796850] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 01/03/2022] [Indexed: 12/11/2022] Open
Abstract
Lipids are key macromolecules that perform a multitude of biological functions ranging from maintaining structural integrity of membranes, energy storage, to signaling molecules. Unsurprisingly, variations in lipid composition and its levels can influence the functional and physiological state of the cell and its milieu. Cellular senescence is a permanent state of cell cycle arrest and is a hallmark of the aging process, as well as several age-related pathologies. Senescent cells are often characterized by alterations in morphology, metabolism, chromatin remodeling and exhibit a complex pro-inflammatory secretome (SASP). Recent studies have shown that the regulation of specific lipid species play a critical role in senescence. Indeed, some lipid species even contribute to the low-grade inflammation associated with SASP. Many protein regulators of senescence have been well characterized and are associated with lipid metabolism. However, the link between critical regulators of cellular senescence and senescence-associated lipid changes is yet to be elucidated. Here we systematically review the current knowledge on lipid metabolism and dynamics of cellular lipid content during senescence. We focus on the roles of major players of senescence in regulating lipid metabolism. Finally, we explore the future prospects of lipid research in senescence and its potential to be targeted as senotherapeutics.
Collapse
Affiliation(s)
- Shruthi Hamsanathan
- Aging Institute of UPMC, The University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Aditi U. Gurkar
- Aging Institute of UPMC, The University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Division of Geriatric Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, United States
- *Correspondence: Aditi U. Gurkar,
| |
Collapse
|
30
|
Hu C, Zhang X, Hu M, Teng T, Yuan Y, Song P, Kong C, Xu S, Ma Z, Tang Q. Fibronectin type III domain-containing 5 improves aging-related cardiac dysfunction in mice. Aging Cell 2022; 21:e13556. [PMID: 35166002 PMCID: PMC8920441 DOI: 10.1111/acel.13556] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 12/17/2021] [Accepted: 01/05/2022] [Indexed: 12/14/2022] Open
Abstract
Aging is an important risk factor for cardiovascular diseases, and aging‐related cardiac dysfunction serves as a major determinant of morbidity and mortality in elderly populations. Our previous study has identified fibronectin type III domain‐containing 5 (FNDC5) and its cleaved form, irisin, as the cardioprotectant against doxorubicin‐induced cardiomyopathy. Herein, aging or matched young mice were overexpressed with FNDC5 by adeno‐associated virus serotype 9 (AAV9) vectors, or subcutaneously infused with irisin to uncover the role of FNDC5 in aging‐related cardiac dysfunction. To verify the involvement of nucleotide‐binding oligomerization domain‐like receptor with a pyrin domain 3 (NLRP3) and AMP‐activated protein kinase α (AMPKα), Nlrp3 or Ampkα2 global knockout mice were used. Besides, young mice were injected with AAV9‐FNDC5 and maintained for 12 months to determine the preventive effect of FNDC5. Moreover, neonatal rat cardiomyocytes were stimulated with tumor necrosis factor‐α (TNF‐α) to examine the role of FNDC5 in vitro. We found that FNDC5 was downregulated in aging hearts. Cardiac‐specific overexpression of FNDC5 or irisin infusion significantly suppressed NLRP3 inflammasome and cardiac inflammation, thereby attenuating aging‐related cardiac remodeling and dysfunction. In addition, irisin treatment also inhibited cellular senescence in TNF‐α‐stimulated cardiomyocytes in vitro. Mechanistically, FNDC5 activated AMPKα through blocking the lysosomal degradation of glucagon‐like peptide‐1 receptor. More importantly, FNDC5 gene transfer in early life could delay the onset of cardiac dysfunction during aging process. We prove that FNDC5 improves aging‐related cardiac dysfunction by activating AMPKα, and it might be a promising therapeutic target to support cardiovascular health in elderly populations.
Collapse
Affiliation(s)
- Can Hu
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan China
| | - Xin Zhang
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan China
| | - Min Hu
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan China
| | - Teng Teng
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan China
| | - Yu‐Pei Yuan
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan China
| | - Peng Song
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan China
| | - Chun‐Yan Kong
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan China
| | - Si‐Chi Xu
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan China
| | - Zhen‐Guo Ma
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan China
| | - Qi‐Zhu Tang
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan China
| |
Collapse
|
31
|
Liu RM. Aging, Cellular Senescence, and Alzheimer's Disease. Int J Mol Sci 2022; 23:1989. [PMID: 35216123 PMCID: PMC8874507 DOI: 10.3390/ijms23041989] [Citation(s) in RCA: 131] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 01/10/2023] Open
Abstract
Aging is the greatest risk factor for late-onset Alzheimer's disease (LOAD), which accounts for >95% of Alzheimer's disease (AD) cases. The mechanism underlying the aging-related susceptibility to LOAD is unknown. Cellular senescence, a state of permanent cell growth arrest, is believed to contribute importantly to aging and aging-related diseases, including AD. Senescent astrocytes, microglia, endothelial cells, and neurons have been detected in the brain of AD patients and AD animal models. Removing senescent cells genetically or pharmacologically ameliorates β-amyloid (Aβ) peptide and tau-protein-induced neuropathologies, and improves memory in AD model mice, suggesting a pivotal role of cellular senescence in AD pathophysiology. Nonetheless, although accumulated evidence supports the role of cellular senescence in aging and AD, the mechanisms that promote cell senescence and how senescent cells contribute to AD neuropathophysiology remain largely unknown. This review summarizes recent advances in this field. We believe that the removal of senescent cells represents a promising approach toward the effective treatment of aging-related diseases, such as AD.
Collapse
Affiliation(s)
- Rui-Ming Liu
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294-0006, USA
| |
Collapse
|
32
|
Teissier T, Boulanger E, Cox LS. Interconnections between Inflammageing and Immunosenescence during Ageing. Cells 2022; 11:359. [PMID: 35159168 PMCID: PMC8834134 DOI: 10.3390/cells11030359] [Citation(s) in RCA: 131] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/13/2022] [Accepted: 01/15/2022] [Indexed: 02/04/2023] Open
Abstract
Acute inflammation is a physiological response to injury or infection, with a cascade of steps that ultimately lead to the recruitment of immune cells to clear invading pathogens and heal wounds. However, chronic inflammation arising from the continued presence of the initial trigger, or the dysfunction of signalling and/or effector pathways, is harmful to health. While successful ageing in older adults, including centenarians, is associated with low levels of inflammation, elevated inflammation increases the risk of poor health and death. Hence inflammation has been described as one of seven pillars of ageing. Age-associated sterile, chronic, and low-grade inflammation is commonly termed inflammageing-it is not simply a consequence of increasing chronological age, but is also a marker of biological ageing, multimorbidity, and mortality risk. While inflammageing was initially thought to be caused by "continuous antigenic load and stress", reports from the last two decades describe a much more complex phenomenon also involving cellular senescence and the ageing of the immune system. In this review, we explore some of the main sources and consequences of inflammageing in the context of immunosenescence and highlight potential interventions. In particular, we assess the contribution of cellular senescence to age-associated inflammation, identify patterns of pro- and anti-inflammatory markers characteristic of inflammageing, describe alterations in the ageing immune system that lead to elevated inflammation, and finally assess the ways that diet, exercise, and pharmacological interventions can reduce inflammageing and thus, improve later life health.
Collapse
Affiliation(s)
- Thibault Teissier
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK;
| | - Eric Boulanger
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167—RID-AGE—Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France;
| | - Lynne S. Cox
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK;
| |
Collapse
|
33
|
Hu MC, Moe OW. Phosphate and Cellular Senescence. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1362:55-72. [PMID: 35288873 PMCID: PMC10513121 DOI: 10.1007/978-3-030-91623-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Cellular senescence is one type of permeant arrest of cell growth and one of increasingly recognized contributor to aging and age-associated disease. High phosphate and low Klotho individually and synergistically lead to age-related degeneration in multiple organs. Substantial evidence supports the causality of high phosphate in cellular senescence, and potential contribution to human aging, cancer, cardiovascular, kidney, neurodegenerative, and musculoskeletal diseases. Phosphate can induce cellular senescence both by direct phosphotoxicity, and indirectly through downregulation of Klotho and upregulation of plasminogen activator inhibitor-1. Restriction of dietary phosphate intake and blockage of intestinal absorption of phosphate help suppress cellular senescence. Supplementation of Klotho protein, cellular senescence inhibitor, and removal of senescent cells with senolytic agents are potential novel strategies to attenuate phosphate-induced cellular senescence, retard aging, and ameliorate age-associated, and phosphate-induced disorders.
Collapse
Affiliation(s)
- Ming Chang Hu
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Departments of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Orson W Moe
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Departments of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Departments of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
34
|
Ismail AA, Shaker BT, Bajou K. The Plasminogen-Activator Plasmin System in Physiological and Pathophysiological Angiogenesis. Int J Mol Sci 2021; 23:ijms23010337. [PMID: 35008762 PMCID: PMC8745544 DOI: 10.3390/ijms23010337] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/20/2021] [Accepted: 12/23/2021] [Indexed: 12/20/2022] Open
Abstract
Angiogenesis is a process associated with the migration and proliferation of endothelial cells (EC) to form new blood vessels. It is involved in various physiological and pathophysiological conditions and is controlled by a wide range of proangiogenic and antiangiogenic molecules. The plasminogen activator–plasmin system plays a major role in the extracellular matrix remodeling process necessary for angiogenesis. Urokinase/tissue-type plasminogen activators (uPA/tPA) convert plasminogen into the active enzyme plasmin, which in turn activates matrix metalloproteinases and degrades the extracellular matrix releasing growth factors and proangiogenic molecules such as the vascular endothelial growth factor (VEGF-A). The plasminogen activator inhibitor-1 (PAI-1) is the main inhibitor of uPA and tPA, thereby an inhibitor of pericellular proteolysis and intravascular fibrinolysis, respectively. Paradoxically, PAI-1, which is expressed by EC during angiogenesis, is elevated in several cancers and is found to promote angiogenesis by regulating plasmin-mediated proteolysis and by promoting cellular migration through vitronectin. The urokinase-type plasminogen activator receptor (uPAR) also induces EC cellular migration during angiogenesis via interacting with signaling partners. Understanding the molecular functions of the plasminogen activator plasmin system and targeting angiogenesis via blocking serine proteases or their interactions with other molecules is one of the major therapeutic strategies scientists have been attracted to in controlling tumor growth and other pathological conditions characterized by neovascularization.
Collapse
Affiliation(s)
- Asmaa Anwar Ismail
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (A.A.I.); (B.T.S.)
- Human Genetics & Stem Cells Research Group, Research Institute of Sciences & Engineering, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Baraah Tariq Shaker
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (A.A.I.); (B.T.S.)
- Human Genetics & Stem Cells Research Group, Research Institute of Sciences & Engineering, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Khalid Bajou
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (A.A.I.); (B.T.S.)
- Human Genetics & Stem Cells Research Group, Research Institute of Sciences & Engineering, University of Sharjah, Sharjah 27272, United Arab Emirates
- Correspondence:
| |
Collapse
|
35
|
Cohen C, Le Goff O, Soysouvanh F, Vasseur F, Tanou M, Nguyen C, Amrouche L, Le Guen J, Saltel-Fulero O, Meunier T, Nguyen-Khoa T, Rabant M, Nochy D, Legendre C, Friedlander G, Childs BG, Baker DJ, Knebelmann B, Anglicheau D, Milliat F, Terzi F. Glomerular endothelial cell senescence drives age-related kidney disease through PAI-1. EMBO Mol Med 2021; 13:e14146. [PMID: 34725920 PMCID: PMC8573606 DOI: 10.15252/emmm.202114146] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 09/16/2021] [Accepted: 09/22/2021] [Indexed: 12/15/2022] Open
Abstract
The mechanisms underlying the development of glomerular lesions during aging are largely unknown. It has been suggested that senescence might play a role, but the pathophysiological link between senescence and lesion development remains unexplained. Here, we uncovered an unexpected role for glomerular endothelial cells during aging. In fact, we discovered a detrimental cross-talk between senescent endothelial cells and podocytes, through PAI-1. In vivo, selective inactivation of PAI-1 in endothelial cells protected glomeruli from lesion development and podocyte loss in aged mice. In vitro, blocking PAI-1 in supernatants from senescent endothelial cells prevented podocyte apoptosis. Consistently, depletion of senescent cells prevented podocyte loss in old p16 INK-ATTAC transgenic mice. Importantly, these experimental findings are relevant to humans. We showed that glomerular PAI-1 expression was predictive of poor outcomes in transplanted kidneys from elderly donors. In addition, we observed that in elderly patients, urinary PAI-1 was associated with age-related chronic kidney disease. Altogether, these results uncover a novel mechanism of kidney disease and identify PAI-1 as a promising biomarker of kidney dysfunction in allografts from elderly donors.
Collapse
Affiliation(s)
- Camille Cohen
- Université de Paris, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Département "Croissance et Signalisation", Paris, France
| | - Océane Le Goff
- Université de Paris, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Département "Croissance et Signalisation", Paris, France
| | - Frédéric Soysouvanh
- Institut de Radioprotection et de Sureté Nucléaire (IRSN), Laboratoire Radiobiologie des Expositions Médicale, Fontenay-aux-Roses, France
| | - Florence Vasseur
- Université de Paris, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Département "Croissance et Signalisation", Paris, France
| | - Marine Tanou
- Université de Paris, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Département "Croissance et Signalisation", Paris, France
| | - Clément Nguyen
- Université de Paris, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Département "Croissance et Signalisation", Paris, France
| | - Lucile Amrouche
- Université de Paris, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Département "Croissance et Signalisation", Paris, France
- Service de Néphrologie-Transplantation, Hôpital Necker Enfants Malades, AP-HP centre, Université de Paris, Paris, France
| | - Julien Le Guen
- Service de Gériatrie, Hôpital Européen Georges Pompidou, AP-HP Centre, Université de Paris, Paris, France
| | - Oriana Saltel-Fulero
- Service de Gériatrie, Hôpital Européen Georges Pompidou, AP-HP Centre, Université de Paris, Paris, France
| | - Tanguy Meunier
- Service de Gériatrie, Hôpital Européen Georges Pompidou, AP-HP Centre, Université de Paris, Paris, France
| | - Thao Nguyen-Khoa
- Université de Paris, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Département "Croissance et Signalisation", Paris, France
- Service de Biochimie, Hôpital Necker Enfants Malades, AP-HP Centre, Université de Paris, Paris, France
| | - Marion Rabant
- Université de Paris, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Département "Croissance et Signalisation", Paris, France
- Service d'Anatomo-Pathologie, AP-HP, Hôpital Necker Enfants Malades, AP-HP Centre, Université de Paris, Paris, France
| | - Dominique Nochy
- Service d'Anatomo-Pathologie, Hôpital Européen George Pompidou, AP-HP Centre, Université de Paris, Paris, France
| | - Christophe Legendre
- Université de Paris, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Département "Croissance et Signalisation", Paris, France
- Service de Néphrologie-Transplantation, Hôpital Necker Enfants Malades, AP-HP centre, Université de Paris, Paris, France
| | - Gérard Friedlander
- Université de Paris, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Département "Croissance et Signalisation", Paris, France
| | - Bennett G Childs
- Department of Pediatrics, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Daren J Baker
- Department of Pediatrics, Mayo Clinic College of Medicine, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Bertrand Knebelmann
- Université de Paris, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Département "Croissance et Signalisation", Paris, France
- Service de Néphrologie-Transplantation, Hôpital Necker Enfants Malades, AP-HP centre, Université de Paris, Paris, France
| | - Dany Anglicheau
- Université de Paris, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Département "Croissance et Signalisation", Paris, France
- Service de Néphrologie-Transplantation, Hôpital Necker Enfants Malades, AP-HP centre, Université de Paris, Paris, France
| | - Fabien Milliat
- Institut de Radioprotection et de Sureté Nucléaire (IRSN), Laboratoire Radiobiologie des Expositions Médicale, Fontenay-aux-Roses, France
| | - Fabiola Terzi
- Université de Paris, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Département "Croissance et Signalisation", Paris, France
| |
Collapse
|
36
|
Khan SS. The Central Role of Plasminogen Activator Inhibitor-1 in COVID-19: Thrombosis and Beyond. Am J Respir Cell Mol Biol 2021; 65:238-240. [PMID: 34086538 PMCID: PMC8485998 DOI: 10.1165/rcmb.2021-0208ed] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Sadiya S Khan
- Northwestern University Feinberg School of Medicine, 12244, Chicago, Illinois, United States;
| |
Collapse
|
37
|
Wang WJ, Chen XM, Cai GY. Cellular senescence and the senescence-associated secretory phenotype: Potential therapeutic targets for renal fibrosis. Exp Gerontol 2021; 151:111403. [PMID: 33984448 DOI: 10.1016/j.exger.2021.111403] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 12/25/2022]
Abstract
Renal fibrosis plays a crucial role in the progression of chronic kidney disease and end-stage renal disease. However, because the aetiology of this pathological process is complex and remains unclear, there is still no effective treatment. Cellular senescence and the senescence-associated secretory phenotype (SASP) have been reported to lead to renal fibrosis. This review first discusses the relationships among cellular senescence, the SASP and renal fibrosis. Then, the key role of the SASP in irreversible renal fibrosis, including fibroblast activation and abnormal extracellular matrix accumulation, is discussed, with the results of studies having indicated that inhibiting cellular senescence and the SASP might be a potential preventive and therapeutic strategy for renal fibrosis. Finally, we summarize promising therapeutic strategies revealed by existing research on senescent cells and the SASP, including emerging interventions targeting the SASP, caloric restriction and mimetics, and novel regeneration therapies with stem cells.
Collapse
Affiliation(s)
- Wen-Juan Wang
- School of Medicine, Nankai University, Tianjin 300071, China; Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Xiang-Mei Chen
- School of Medicine, Nankai University, Tianjin 300071, China; Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China.
| | - Guang-Yan Cai
- School of Medicine, Nankai University, Tianjin 300071, China; Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China.
| |
Collapse
|
38
|
Levine JA, Olivares S, Miyata T, Vaughan DE, Henkel AS. Inhibition of PAI-1 Promotes Lipolysis and Enhances Weight Loss in Obese Mice. Obesity (Silver Spring) 2021; 29:713-720. [PMID: 33594826 PMCID: PMC8842994 DOI: 10.1002/oby.23112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 11/30/2020] [Accepted: 12/14/2020] [Indexed: 12/20/2022]
Abstract
OBJECTIVE This study investigates the therapeutic potential of a small molecule inhibitor of plasminogen activator inhibitor-1 (PAI-1), TM5441, in reversing diet-induced obesity in mice. METHODS Wild-type C57BL/6J mice were fed a high-fat high-sugar (HFHS) diet for 8 weeks to induce obesity. After the first 8 weeks, TM5441 was added to the diet for an additional 8 weeks. In order to determine the efficacy of PAI-1 inhibition in conjunction with dietary modification, mice were fed an HFHS diet for 8 weeks to induce obesity and were then switched to a low-fat diet with or without TM5441 for an additional 2 to 8 weeks. RESULTS Obese mice showed weight reduction and significant improvement in hepatic steatosis when TM5441 was added to the HFHS diet. Obese mice that were treated with TM5441 in conjunction with dietary modification showed enhanced weight loss and a more rapid reversal of hepatic steatosis compared with obese mice treated with dietary modification alone. The enhanced weight loss among mice treated with TM5441 was associated with increased adipose tissue expression of adipose triglyceride lipase, phosphorylated hormone-sensitive lipase, and phosphorylated perilipin-1 as well as induction of adipose tissue lipolysis. CONCLUSIONS Pharmacologic PAI-1 inhibition stimulates adipose tissue lipolysis and enhances weight loss in obese mice.
Collapse
Affiliation(s)
- Joshua A. Levine
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Shantel Olivares
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
- Department of Medicine, Jesse Brown VA Medical Center, Chicago, IL
| | - Toshio Miyata
- Department of Molecular Medicine and Therapy, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Douglas E. Vaughan
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Anne S. Henkel
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
- Department of Medicine, Jesse Brown VA Medical Center, Chicago, IL
| |
Collapse
|
39
|
Di Micco R, Krizhanovsky V, Baker D, d'Adda di Fagagna F. Cellular senescence in ageing: from mechanisms to therapeutic opportunities. Nat Rev Mol Cell Biol 2021; 22:75-95. [PMID: 33328614 PMCID: PMC8344376 DOI: 10.1038/s41580-020-00314-w] [Citation(s) in RCA: 1204] [Impact Index Per Article: 301.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2020] [Indexed: 12/11/2022]
Abstract
Cellular senescence, first described in vitro in 1961, has become a focus for biotech companies that target it to ameliorate a variety of human conditions. Eminently characterized by a permanent proliferation arrest, cellular senescence occurs in response to endogenous and exogenous stresses, including telomere dysfunction, oncogene activation and persistent DNA damage. Cellular senescence can also be a controlled programme occurring in diverse biological processes, including embryonic development. Senescent cell extrinsic activities, broadly related to the activation of a senescence-associated secretory phenotype, amplify the impact of cell-intrinsic proliferative arrest and contribute to impaired tissue regeneration, chronic age-associated diseases and organismal ageing. This Review discusses the mechanisms and modulators of cellular senescence establishment and induction of a senescence-associated secretory phenotype, and provides an overview of cellular senescence as an emerging opportunity to intervene through senolytic and senomorphic therapies in ageing and ageing-associated diseases.
Collapse
Affiliation(s)
- Raffaella Di Micco
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Valery Krizhanovsky
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, Israel
| | - Darren Baker
- Department of Pediatrics, Mayo Clinic, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Fabrizio d'Adda di Fagagna
- IFOM - The FIRC Institute of Molecular Oncology, Milan, Italy.
- Istituto di Genetica Molecolare, Consiglio Nazionale delle Ricerche, Pavia, Italy.
| |
Collapse
|
40
|
Yahata T, Ibrahim AA, Hirano KI, Muguruma Y, Naka K, Hozumi K, Vaughan DE, Miyata T, Ando K. Targeting of plasminogen activator inhibitor-1 activity promotes elimination of chronic myeloid leukemia stem cells. Haematologica 2021; 106:483-494. [PMID: 32001531 PMCID: PMC7849585 DOI: 10.3324/haematol.2019.230227] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 01/24/2020] [Indexed: 12/12/2022] Open
Abstract
Therapeutic strategies that target leukemic stem cells (LSC) provide potential advantages in the treatment of chronic myeloid leukemia (CML). Here we showed that selective blockade of plasminogen activator inhibitor-1 (PAI-1) enhances the susceptibility of CML-LSC to tyrosine kinase inhibitor (TKI), which facilitates the eradication of CML-LSC and leads to sustained remission of the disease. We demonstrated for the first time that the TGF-−PAI-1 axis was selectively augmented in CMLLSC in the bone marrow (BM), thereby protecting CML-LSC from TKI treatment. Furthermore, the combined administration of the TKI imatib plus a PAI-1 inhibitor, in a mouse model of CML, significantly enhanced the eradication of CML cells in the BM and prolonged the survival of CML mice. The combined therapy of imatinib and a PAI-1 inhibitor prevented the recurrence of CML-like disease in serially transplanted recipients, indicating the elimination of CML-LSC. Interestingly, PAI-1 inhibitor treatment augmented membrane-type matrix metalloprotease-1 (MT1-MMP)-dependent motility of CML-LSC, and the anti-CML effect of PAI-1 inhibitor was extinguished by the neutralizing antibody for MT1-MMP, underlining the mechanistic importance of MT1-MMP. Our findings provide evidence of, and a rationale for, a novel therapeutic tactic, based on the blockade of PAI- 1 activity, for CML patients.
Collapse
|
41
|
Targeting senescent cell clearance: An approach to delay aging and age-associated disorders. TRANSLATIONAL MEDICINE OF AGING 2021. [DOI: 10.1016/j.tma.2020.12.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
42
|
Miller HA, Dean ES, Pletcher SD, Leiser SF. Cell non-autonomous regulation of health and longevity. eLife 2020; 9:62659. [PMID: 33300870 PMCID: PMC7728442 DOI: 10.7554/elife.62659] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/24/2020] [Indexed: 12/28/2022] Open
Abstract
As the demographics of the modern world skew older, understanding and mitigating the effects of aging is increasingly important within biomedical research. Recent studies in model organisms demonstrate that the aging process is frequently modified by an organism’s ability to perceive and respond to changes in its environment. Many well-studied pathways that influence aging involve sensory cells, frequently neurons, that signal to peripheral tissues and promote survival during the presence of stress. Importantly, this activation of stress response pathways is often sufficient to improve health and longevity even in the absence of stress. Here, we review the current landscape of research highlighting the importance of cell non-autonomous signaling in modulating aging from C. elegans to mammals. We also discuss emerging concepts including retrograde signaling, approaches to mapping these networks, and development of potential therapeutics.
Collapse
Affiliation(s)
- Hillary A Miller
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, United States
| | - Elizabeth S Dean
- Molecular & Integrative Physiology Department, University of Michigan, Ann Arbor, United States
| | - Scott D Pletcher
- Molecular & Integrative Physiology Department, University of Michigan, Ann Arbor, United States
| | - Scott F Leiser
- Molecular & Integrative Physiology Department, University of Michigan, Ann Arbor, United States.,Department of Internal Medicine, University of Michigan, Ann Arbor, United States
| |
Collapse
|
43
|
Aihemaiti A, Yamamoto N, Piao J, Oyaizu T, Ochi H, Sato S, Okawa A, Miyata T, Tsuji K, Ezura Y, Asou Y. A novel PAI-1 inhibitor prevents ageing-related muscle fiber atrophy. Biochem Biophys Res Commun 2020; 534:849-856. [PMID: 33213843 DOI: 10.1016/j.bbrc.2020.10.089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 10/29/2020] [Indexed: 11/30/2022]
Abstract
Sarcopenia is among the most common medical problems of the aging population worldwide and a major social concern. Here, we explored the therapeutic potential of TM5484, a novel orally available PAI-1 inhibitor, to prevent sarcopenia. The sarcopenic phenotypes of the calf muscle of 12- and 6-month-old middle-aged mice were compared. Although significant decline of isometric gastrocnemius muscle force was detected in the older untreated mice, those administered TM5484 had significantly greater calf muscle force, as determined using isometric measurements by electrical stimulation. Histological analysis indicated that cross-sectional gastrocnemius muscle fibers in untreated older mice were thinner than those in younger mice; however, TM5484-treated group showed thicker fibers than younger mice. Treatment with TM5484 for 6 months enhanced Igf1, Atrogin-1, Mt-Co1, and Chrna1 mRNA expression in the mice gastrocnemius muscle, with increased serum IGF-1 concentration. TM5484 induced dose-dependent Igf1, Atrogin-1, and Chrna1 expression in C2C12 myoblastic cells, confirming cell autonomous effect. Further, the presence of plasmin for 72 h caused significantly increased Igf1 expression in C2C12 cells. These findings suggest that oral PAI-1 inhibitors represent a promising therapeutic candidate for preventing sarcopenia progression in humans.
Collapse
Affiliation(s)
- Aidehamu Aihemaiti
- Department of Orthopaedics Surgery, Tokyo Medical and Dental University, Japan
| | - Naoki Yamamoto
- Department of Orthopaedics Surgery, Tokyo Medical and Dental University, Japan
| | - Jinying Piao
- Department of Orthopaedics Surgery, Tokyo Medical and Dental University, Japan
| | - Takuya Oyaizu
- Hyperbaric Medical Center, Tokyo Medical and Dental University, Japan
| | - Hiroki Ochi
- Department of Rehabilitation for Movement Functions, National Rehabilitation Center for Persons with Disabilities, Japan
| | - Shingo Sato
- Department of Orthopaedics Surgery, Tokyo Medical and Dental University, Japan
| | - Atsushi Okawa
- Department of Orthopaedics Surgery, Tokyo Medical and Dental University, Japan
| | - Toshio Miyata
- Department of Molecular Medicine and Therapy, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Japan
| | - Kunikazu Tsuji
- Department of Cartilage Regeneration, Tokyo Medical and Dental University, Japan
| | - Yoichi Ezura
- Department of Orthopaedics Surgery, Tokyo Medical and Dental University, Japan
| | - Yoshinori Asou
- Department of Nano-Bioscience, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Japan.
| |
Collapse
|
44
|
Urano T, Suzuki Y, Iwaki T, Sano H, Honkura N, Castellino FJ. Recognition of Plasminogen Activator Inhibitor Type 1 as the Primary Regulator of Fibrinolysis. Curr Drug Targets 2020; 20:1695-1701. [PMID: 31309890 DOI: 10.2174/1389450120666190715102510] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/17/2019] [Accepted: 06/24/2019] [Indexed: 01/12/2023]
Abstract
The fibrinolytic system consists of a balance between rates of plasminogen activation and fibrin degradation, both of which are finely regulated by spatio-temporal mechanisms. Three distinct inhibitors of the fibrinolytic system that differently regulate these two steps are plasminogen activator inhibitor type-1 (PAI-1), α2-antiplasmin, and thrombin activatable fibrinolysis inhibitor (TAFI). In this review, we focus on the mechanisms by which PAI-1 governs total fibrinolytic activity to provide its essential role in many hemostatic disorders, including fibrinolytic shutdown after trauma. PAI-1 is a member of the serine protease inhibitor (SERPIN) superfamily and inhibits the protease activities of plasminogen activators (PAs) by forming complexes with PAs, thereby regulating fibrinolysis. The major PA in the vasculature is tissue-type PA (tPA) which is secreted from vascular endothelial cells (VECs) as an active enzyme and is retained on the surface of VECs. PAI-1, existing in molar excess to tPA in plasma, regulates the amount of free active tPA in plasma and on the surface of VECs by forming a tPA-PAI-1 complex. Thus, high plasma levels of PAI-1 are directly related to attenuated fibrinolysis and increased risk for thrombosis. Since plasma PAI-1 levels are highly elevated under a variety of pathological conditions, including infection and inflammation, the fibrinolytic potential in plasma and on VECs is readily suppressed to induce fibrinolytic shutdown. A congenital deficiency of PAI-1 in humans, in turn, leads to life-threatening bleeding. These considerations support the contention that PAI-1 is the primary regulator of the initial step of fibrinolysis and governs total fibrinolytic activity.
Collapse
Affiliation(s)
- Tetsumei Urano
- Department of Medical Physiology, Hamamatsu University School of Medicine, 1-20-1, Handa-yama, Higashi-ku, Hamamatsu, 431-3192, Japan
| | - Yuko Suzuki
- Department of Medical Physiology, Hamamatsu University School of Medicine, 1-20-1, Handa-yama, Higashi-ku, Hamamatsu, 431-3192, Japan
| | - Takayuki Iwaki
- Department of Pharmacology, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, 431-3192, Japan
| | - Hideto Sano
- Department of Medical Physiology, Hamamatsu University School of Medicine, 1-20-1, Handa-yama, Higashi-ku, Hamamatsu, 431-3192, Japan
| | - Naoki Honkura
- Department of Medical Physiology, Hamamatsu University School of Medicine, 1-20-1, Handa-yama, Higashi-ku, Hamamatsu, 431-3192, Japan
| | - Francis J Castellino
- W.M. Keck Center for Transgene Research, Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, USA
| |
Collapse
|
45
|
Agoro R, Ni P, Noonan ML, White KE. Osteocytic FGF23 and Its Kidney Function. Front Endocrinol (Lausanne) 2020; 11:592. [PMID: 32982979 PMCID: PMC7485387 DOI: 10.3389/fendo.2020.00592] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 07/20/2020] [Indexed: 12/16/2022] Open
Abstract
Osteocytes, which represent up to 95% of adult skeletal cells, are deeply embedded in bone. These cells exhibit important interactive abilities with other bone cells such as osteoblasts and osteoclasts to control skeletal formation and resorption. Beyond this local role, osteocytes can also influence the function of distant organs due to the presence of their sophisticated lacunocanalicular system, which connects osteocyte dendrites directly to the vasculature. Through these networks, osteocytes sense changes in circulating metabolites and respond by producing endocrine factors to control homeostasis. One critical function of osteocytes is to respond to increased blood phosphate and 1,25(OH)2 vitamin D (1,25D) by producing fibroblast growth factor-23 (FGF23). FGF23 acts on the kidneys through partner fibroblast growth factor receptors (FGFRs) and the co-receptor Klotho to promote phosphaturia via a downregulation of phosphate transporters, as well as the control of vitamin D metabolizing enzymes to reduce blood 1,25D. In the first part of this review, we will explore the signals involved in the positive and negative regulation of FGF23 in osteocytes. In the second portion, we will bridge bone responses with the review of current knowledge on FGF23 endocrine functions in the kidneys.
Collapse
Affiliation(s)
- Rafiou Agoro
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Pu Ni
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Megan L. Noonan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Kenneth E. White
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
- Medicine/Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
46
|
Kashiwagi R, Sato R, Masumoto M, Yoshino M, Tanaka H. AS3288802, a highly selective antibody to active plasminogen activator inhibitor-1 (PAI-1), exhibits long efficacy duration in cynomolgus monkeys. Biologicals 2020; 67:21-28. [PMID: 32828642 DOI: 10.1016/j.biologicals.2020.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 07/26/2020] [Accepted: 08/03/2020] [Indexed: 10/23/2022] Open
Abstract
Antibodies have strong affinity to their target molecules, a characteristic that is utilized in antibody drugs. For antibody drugs, target molecule specificity and long duration pharmacokinetics, along with strong affinity to the target molecule are important characteristics. Plasminogen activator inhibitor-1 (PAI-1) is one of the key regulators of the fibrinolysis system, and the benefits of PAI-1 activity inhibition have been widely reported for multiple thrombosis and fibrosis-related diseases. Here, we generated a novel antibody, AS3288802, with high selectivity for active PAI-1. AS3288802 exhibited prolonged and strong inhibition of PAI-1 activity in cynomolgus monkey blood in vivo. Given that AS3288802 showed prolonged antigen inhibition activity due to its high target molecule selectivity, we propose that increasing target molecule selectivity may be a key strategy for lengthening the efficacy duration of antibody drugs. AS3288802 may be a promising anti-PAI-1 antibody drug with multiple clinical applications including thrombosis and fibrosis-related diseases.
Collapse
Affiliation(s)
- Risa Kashiwagi
- Astellas Pharma Inc., 2-5-1, Nihonbashi-Honcho, Chuo-ku, Tokyo, 103-8411
| | - Rui Sato
- Astellas Pharma Inc., 2-5-1, Nihonbashi-Honcho, Chuo-ku, Tokyo, 103-8411
| | - Mari Masumoto
- Astellas Pharma Inc., 2-5-1, Nihonbashi-Honcho, Chuo-ku, Tokyo, 103-8411
| | - Masayasu Yoshino
- Astellas Pharma Inc., 2-5-1, Nihonbashi-Honcho, Chuo-ku, Tokyo, 103-8411
| | - Hirotsugu Tanaka
- Astellas Innovation Management LLC, 1030 Massachusetts Avenue, Cambridge, MA, 02138, United States.
| |
Collapse
|
47
|
Maique J, Flores B, Shi M, Shepard S, Zhou Z, Yan S, Moe OW, Hu MC. High Phosphate Induces and Klotho Attenuates Kidney Epithelial Senescence and Fibrosis. Front Pharmacol 2020; 11:1273. [PMID: 32973510 PMCID: PMC7468469 DOI: 10.3389/fphar.2020.01273] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 07/31/2020] [Indexed: 12/18/2022] Open
Abstract
Cellular senescence is an irreversible cell growth arrest and is associated with aging and age-related diseases. High plasma phosphate (Pi) and deficiency of Klotho contribute to aging and kidney fibrosis, a pathological feature in the aging kidney and chronic kidney disease. This study examined the interactive role of Pi and Klotho in kidney senescence and fibrosis. Homozygous Klotho hypomorphic mice had high plasma Pi, undetectable Klotho in plasma and kidney, high senescence with massive collagen accumulation in kidney tubules, and fibrin deposits in peritubular capillaries. To examine the Pi effect on kidney senescence, a high (2%) Pi diet was given to wild-type mice. One week of high dietary Pi mildly increased plasma Pi, and upregulated kidney p16/p21 expression, but did not significantly decrease Klotho. Two weeks of high Pi intake led to increase in plasminogen activator inhibitor (PAI)-1, and decrease in kidney Klotho, but still without detectable increase in kidney fibrosis. More prolonged dietary Pi for 12 weeks exacerbated kidney senescence and fibrosis; more so in heterozygous Klotho hypomorphic mice compared to wild-type mice, and in mice with chronic kidney disease (CKD) on high Pi diet compared to CKD mice fed a normal Pi diet. In cultured kidney tubular cells, high Pi directly induced cellular senescence, injury and epithelial-mesenchymal transition, and enhanced H2O2-induced cellular senescence and injury, which were abrogated by Klotho. Fucoidan, a bioactive molecule with multiple biologic functions including senescence inhibition, blunted Pi-induced cellular senescence, oxidation, injury, epithelial-mesenchymal transition, and senescence-associated secretary phenotype. In conclusion, high Pi activates senescence through distinct but interconnected mechanisms: upregulating p16/p21 (early), and elevating plasminogen activator inhibitor-1 and downregulating Klotho (late). Klotho may be a promising agent to attenuate senescence and ameliorate age-associated, and Pi-induced kidney degeneration such as kidney fibrosis.
Collapse
Affiliation(s)
- Jenny Maique
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Brianna Flores
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Mingjun Shi
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Sierra Shepard
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Zhiyong Zhou
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Shirely Yan
- Departments of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Orson W Moe
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, United States.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States.,Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Ming Chang Hu
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, United States.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
48
|
Biomarkers of senescence in non-human primate adipose depots relate to aging. GeroScience 2020; 43:343-352. [PMID: 32705409 DOI: 10.1007/s11357-020-00230-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/07/2020] [Indexed: 10/23/2022] Open
Abstract
Forty-three female African green monkeys (Chlorocebus aethiops sabaeus) were selected to represent young adult to advanced geriatric ages (7-24 years) to exhibit a wide range of obesity status (8-53% body fat) and diverse metabolic syndrome criteria such as diabetes, dyslipidemia, and hypertension. Subcutaneous and visceral adipose tissues were collected and evaluated for the presence of senescence cells in both whole tissue and single-cell isolates from subcutaneous sources, utilizing senescence-associated β-galactosidase (SAβ-gal) staining. Plasma samples were analyzed for selected metabolic and inflammatory biomarkers related to the senescence-associated secretory profile. Our results indicated that tissue staining scores did not differ between subcutaneous and intra-abdominal visceral depots and were highly related within individuals. Tissue staining was significantly associated with chronological age; however, no associations with fatness or metabolic syndrome criteria were observed. Associations with age were unchanged when obesity status was included in regression models. Isolated cell staining did positively relate to age but not tissue staining, suggesting some of the SAβ-gal-positive cells were stromal vascular cells or small adipocytes, but that mature large adipocytes, filtered out in the cell isolation process, are also likely to exhibit positive SAβ-gal staining. Plasminogen activator inhibitor-1 (PAI-1) concentration in circulation was the sole inflammation-related biomarker that positively associated with age and is considered to be a marker of senescent cell burden. Our study is the largest, most comprehensive assessment of adipose SAβ-gal staining in a relevant animal model of human aging, and confirms that this senescence-associated biomarker specifically indicates an age-related process.
Collapse
|
49
|
Complement component C5a induces aberrant epigenetic modifications in renal tubular epithelial cells accelerating senescence by Wnt4/βcatenin signaling after ischemia/reperfusion injury. Aging (Albany NY) 2020; 11:4382-4406. [PMID: 31284268 PMCID: PMC6660044 DOI: 10.18632/aging.102059] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 06/24/2019] [Indexed: 12/30/2022]
Abstract
Epigenetic mechanisms, such as DNA methylation, affect tubular maladaptive response after Acute Kidney Injury (AKI) and accelerate renal aging. Upon ischemia/reperfusion (I/R) injury, Complement activation leads to C5a release that mediates damage; however, little is known about the effect of C5a-C5a Receptor (C5aR) interaction in Renal Tubular Epithelial Cells (RTEC). Through a whole-genome DNA methylation analysis in cultured RTEC, we found that C5a induced aberrant methylation, particularly in regions involved in cell cycle control, DNA damage and Wnt signaling. The most represented genes were BCL9, CYP1B1 and CDK6. C5a stimulation of RTEC led to up-regulation of SA-β Gal and cell cycle arrest markers such as p53 and p21. C5a increased also IL-6, MCP-1 and CTGF gene expression, consistent with SASP development. In accordance, in a swine model of renal I/R injury, we found the increased expression of Wnt4 and βcatenin correlating with SA-β Gal, p21, p16 and IL-6 positivity. Administration of Complement Inhibitor (C1-Inh), antagonized SASP by reducing SA-β Gal, p21, p16, IL-6 and abrogating Wnt4/βcatenin activation. Thus, C5a affects the DNA methylation of genes involved in tubular senescence. Targeting epigenetic programs and Complement may offer novels strategies to protect tubular cells from accelerated aging and to counteract progression to Chronic Kidney Disease
Collapse
|
50
|
Ariens R, Becattini C, Bender M, Bergmeier W, Castoldi E, Devreese K, Ellis M, Gailani D, Ignjatovic V, James PD, Kerrigan S, Lambert M, Lee LH, Levi M, Maugeri N, Meijers J, Melero-Martin J, Michelson AD, Mingozzi F, Neeves K, Ni H, Olsson AK, Prohászka Z, Ranson M, Riva N, Senis Y, van Ommen CH, Vaughan DE, Weisel J. Illustrated State-of-the-Art Capsules of the ISTH 2020 Congress. Res Pract Thromb Haemost 2020; 4:680-713. [PMID: 32685876 PMCID: PMC7354406 DOI: 10.1002/rth2.12368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/25/2020] [Accepted: 05/08/2020] [Indexed: 01/19/2023] Open
Abstract
The 2020 Congress of the International Society of Thrombosis and Haemostasis (ISTH) was held virtually July 12-15, 2019, due to the coronavirus disease 2019 pandemic. The congress convenes annually to discuss clinical and basic topics in hemostasis and thrombosis. Each year, the program includes State of Art (SOA) lectures given by prominent scientists. Presenters are asked to create Illustrated Capsules of their talks, which are concise illustrations with minimal explanatory text. Capsules cover major themes of the presentation, and these undergo formal peer review for inclusion in this article. Owing to the shift to a virtual congress this year, organizers reduced the program size. There were 39 SOA lectures virtually presented, and 29 capsules (9 from talks omitted from the virtual congress) were both submitted and successful in peer review, and are included in this article. Topics include the roles of the hemostatic system in inflammation, infection, immunity, and cancer, platelet function and signaling, platelet function disorders, megakaryocyte biology, hemophilia including gene therapy, phenotype tests in hemostasis, von Willebrand factor, anticoagulant factor V, computational driven discovery, endothelium, clinical and basic aspects of thrombotic microangiopathies, fibrinolysis and thrombolysis, antithrombotics in pediatrics, direct oral anticoagulant management, and thrombosis and hemostasis in pregnancy. Capsule authors invite virtual congress attendees to refer to these capsules during the live presentations and participate on Twitter in discussion. Research and Practice in Haemostasis and Thrombosis will release 2 tweets from @RPTHJournal during each presentation, using #IllustratedReview, #CoagCapsule and #ISTH2020. Readers are also welcome to utilize capsules for teaching and ongoing education.
Collapse
Affiliation(s)
- Robert Ariens
- Discovery and Translational Science Department Leeds Institute of Cardiovascular and Metabolic Medicine University of Leeds Leeds UK
| | - Cecilia Becattini
- Internal and Cardiovascular Medicine - Stroke Unit University of Perugia Perugia Italy
| | - Markus Bender
- Institute of Experimental Biomedicine - Chair I University Hospital and Rudolf Virchow Center Würzburg Germany
| | - Wolfgang Bergmeier
- Department of Biochemistry and Biophysics UNC Blood Research Center University of North Carolina at Chapel Hill Chapel Hill NC USA
| | - Elisabetta Castoldi
- Cardiovascular Research Institute Maastricht (CARIM) Maastricht University Maastricht The Netherlands
| | - Katrien Devreese
- Coagulation Laboratory Department of Laboratory Medicine Ghent University Hospital Ghent University Ghent Belgium
- Coagulation Laboratory Department of Diagnostic Sciences Ghent University Hospital Ghent University Ghent Belgium
| | - Martin Ellis
- Hematology Institute and Blood Bank Meir Medical Center and Sackler School of Medicine Tel Aviv University Tel Aviv Israel
| | - David Gailani
- Department of Pathology, Microbiology and Immunology Vanderbilt University Medical Center Nashville TN USA
| | - Vera Ignjatovic
- Haematology Research Team Murdoch Children's Research Institute Department of Paediatrics The University of Melbourne Parkville Vic. Australia
| | | | - Steven Kerrigan
- Royal College of Surgeons in Ireland School of Pharmacy and Biomolecular Sciences Irish Centre for Vascular Biology Dublin Ireland
| | - Michele Lambert
- Department of Pediatrics Perelman School of Medicine at the University of Pennsylvania Philadelphia PA USA
| | - Lai Heng Lee
- Department of Haematology Singapore General Hospital SingHealth Singapore City Singapore
| | - Marcel Levi
- University College London Hospitals London UK
| | - Norma Maugeri
- San Raffaele Scientific Institute and Vita-Salute San Raffaele University Milano Italy
| | - Joost Meijers
- Department of Experimental Vascular Medicine Amsterdam University Medical Centers University of Amsterdam Amsterdam The Netherlands
- Department of Molecular and Cellular Hemostasis Sanquin Research Amsterdam The Netherlands
| | | | - Alan D Michelson
- Boston Children's Hospital and Harvard Medical School Boston MA USA
| | | | - Keith Neeves
- Department of Chemical and Biological Engineering Colorado School of Mines Golden CO USA
| | - Heyu Ni
- Department of Laboratory Medicine and Keenan Research Centre for Biomedical Science of St. Michael's Hospital University of Toronto Toronto ON Canada
| | - Anna-Karin Olsson
- Department of Medical Biochemistry and Microbiology Uppsala University Uppsala Sweden
| | - Zoltán Prohászka
- Research Laboratory 3rd Department of Internal Medicine MTA-SE Research Group of Immunology and Hematology Hungarian Academy of Sciences and Semmelweis University Budapest Hungary
| | - Marie Ranson
- School of Chemistry and Molecular Bioscience University of Wollongong Wollongong NSW Australia
| | - Nicoletta Riva
- Department of Pathology Faculty of Medicine and Surgery University of Malta Msida Malta
| | - Yotis Senis
- Directeur de Recherche Etablissement Français du Sang Grand Est Inserm UMR-S1255 Université de Strasbourg Strasbourg France
| | - Cornelia H van Ommen
- Department of Pediatric Hematology Oncology Erasmus MC Sophia Children's Hospital Rotterdam The Netherlands
| | | | - John Weisel
- Department of Cell and Developmental Biology Perelman School of Medicine University of Pennsylvania Philadelphia PA USA
| |
Collapse
|